1
|
Kulkarni GC, Saha R, Peters CJ. Ion channel expression and function in glioblastoma multiforme (GBM): pathophysiological mechanisms and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119982. [PMID: 40328081 DOI: 10.1016/j.bbamcr.2025.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
Glioblastoma Multiforme (GBM) is a highly malignant and diffusely invasive WHO Grade IV brain tumor arising from glial and neural stem cells. GBM is characterized by rapid proliferation and migration, aggressive invasion of local brain parenchyma, a hypoxic microenvironment, resistance to apoptosis and high vascular remodeling and angiogenesis. These hallmarks contribute to a near universal tumor recurrence after treatment or resection and poor patient prognosis. Ion channels, a superfamily of proteins responsible for permitting ion flux across otherwise impermeant membranes, show extensive remodeling in GBM with aberrant function mechanistically linked to manipulation of each of these hallmarks. In this review, we will discuss the known links between ion channel expression and activity and cellular processes that are enhanced or perturbed during GBM formation or progression. We will also discuss the extent to which basic or translational findings on ion channels in GBM samples or cell lines have shown preclinical promise towards the development of improved therapeutics against GBMs.
Collapse
Affiliation(s)
- Gauri C Kulkarni
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Rayna Saha
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Christian J Peters
- Department of Anatomy and Cell Biology, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Ma K, Wang S, Ma Y, Zeng L, Xu K, Mu N, Lai Y, Shi Y, Yang C, Chen B, Quan Y, Li L, Lu Y, Yang Y, Liu Y, Hu R, Wang X, Chen Y, Bian X, Feng H, Li F, Chen T. Increased oxygen stimulation promotes chemoresistance and phenotype shifting through PLCB1 in gliomas. Drug Resist Updat 2024; 76:101113. [PMID: 39053384 DOI: 10.1016/j.drup.2024.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/24/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
Gliomas, the most common CNS (central nerve system) tumors, face poor survival due to severe chemoresistance exacerbated by hypoxia. However, studies on whether altered hypoxic conditions benefit for chemo-sensitivity and how gliomas react to increased oxygen stimulation are limited. In this study, we demonstrated that increased oxygen stimulation promotes glioma growth and chemoresistance. Mechanically, increased oxygen stimulation upregulates miR-1290 levels. miR-1290, in turn, downregulates PLCB1, while PLCB1 facilitates the proteasomal degradation of β-catenin and active-β-catenin by increasing the proportion of ubiquitinated β-catenin in a destruction complex-independent mechanism. This process inhibits PLCB1 expression, leads to the accumulation of active-β-catenin, boosting Wnt signaling through an independent mechanism and ultimately promoting chemoresistance in glioma cells. Pharmacological inhibition of Wnt by WNT974 could partially inhibit glioma volume growth and prolong the shortened survival caused by increased oxygen stimulation in a glioma-bearing mouse model. Moreover, PLCB1, a key molecule regulated by increased oxygen stimulation, shows promising predictive power in survival analysis and has great potential to be a biomarker for grading and prognosis in glioma patients. These results provide preliminary insights into clinical scenarios associated with altered hypoxic conditions in gliomas, and introduce a novel perspective on the role of the hypoxic microenvironment in glioma progression. Furthermore, the outcomes reveal the potential risks of utilizing hyperbaric oxygen treatment (HBOT) in glioma patients, particularly when considering HBOT as a standalone option to ameliorate neuro-dysfunctions or when combining HBOT with a single chemotherapy agent without radiotherapy.
Collapse
Affiliation(s)
- Kang Ma
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi Wang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yingjie Ma
- Medical Data Science Academy, Chongqing Medical University, Chongqing, China
| | - Lan Zeng
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Kai Xu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ning Mu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Lai
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yaning Shi
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuanyan Yang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Beike Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yulian Quan
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Li
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongling Lu
- Medical Research Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yan Liu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Rong Hu
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoming Wang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yujie Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Hua Feng
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Fei Li
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Tunan Chen
- Glioma Medical Research Center and Department of Neurosurgery, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
3
|
Qi L, Du Y, Huang Y, Kogiso M, Zhang H, Xiao S, Abdallah A, Suarez M, Niu L, Liu ZG, Lindsay H, Braun FK, Stephen C, Davies PJ, Teo WY, Adenkunle A, Baxter P, Su JM, Li XN. CD57 defines a novel cancer stem cell that drive invasion of diffuse pediatric-type high grade gliomas. Br J Cancer 2024; 131:258-270. [PMID: 38834745 PMCID: PMC11263392 DOI: 10.1038/s41416-024-02724-5] [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: 06/14/2023] [Revised: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Diffuse invasion remains a primary cause of treatment failure in pediatric high-grade glioma (pHGG). Identifying cellular driver(s) of pHGG invasion is needed for anti-invasion therapies. METHODS Ten highly invasive patient-derived orthotopic xenograft (PDOX) models of pHGG were subjected to isolation of matching pairs of invasive (HGGINV) and tumor core (HGGTC) cells. RESULTS pHGGINV cells were intrinsically more invasive than their matching pHGGTC cells. CSC profiling revealed co-positivity of CD133 and CD57 and identified CD57+CD133- cells as the most abundant CSCs in the invasive front. In addition to discovering a new order of self-renewal capacities, i.e., CD57+CD133- > CD57+CD133+ > CD57-CD133+ > CD57-CD133- cells, we showed that CSC hierarchy was impacted by their spatial locations, and the highest self-renewal capacities were found in CD57+CD133- cells in the HGGINV front (HGGINV/CD57+CD133- cells) mediated by NANOG and SHH over-expression. Direct implantation of CD57+ (CD57+/CD133- and CD57+/CD133+) cells into mouse brains reconstituted diffusely invasion, while depleting CD57+ cells (i.e., CD57-CD133+) abrogated pHGG invasion. CONCLUSION We revealed significantly increased invasive capacities in HGGINV cells, confirmed CD57 as a novel glioma stem cell marker, identified CD57+CD133- and CD57+CD133+ cells as a new cellular driver of pHGG invasion and suggested a new dual-mode hierarchy of HGG stem cells.
Collapse
Affiliation(s)
- Lin Qi
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 510080, China
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuchen Du
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yulun Huang
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Neurosurgery and Brain and Nerve Research Laboratory, the First Affiliated Hospital, and Dushu Lake Hospital, Soochow University Medical School, Suzhou, 215007, China
| | - Mari Kogiso
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Huiyuan Zhang
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sophie Xiao
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Aalaa Abdallah
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Milagros Suarez
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Long Niu
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Zhi-Gang Liu
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
- Cancer Center, Affiliated Dongguan Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Holly Lindsay
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Frank K Braun
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Clifford Stephen
- Center for Epigenetics & Disease Prevention, Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Peter J Davies
- Center for Epigenetics & Disease Prevention, Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Wan Yee Teo
- The Laboratory of Pediatric Brain Tumor Research Office, SingHealth Duke-NUS Academic Medical Center, Singapore, 169856, Singapore
| | - Adesina Adenkunle
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Patricia Baxter
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jack Mf Su
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiao-Nan Li
- Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Ann & Robert H. Lurie Children's Hospital of Chicago, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA.
- Robert H. Laurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
4
|
Ignatova TN, Chaitin HJ, Kukekov NV, Suslov ON, Dulatova GI, Hanafy KA, Vrionis FD. Gliomagenesis is orchestrated by the Oct3/4 regulatory network. J Neurosurg Sci 2024; 68:148-156. [PMID: 34342203 DOI: 10.23736/s0390-5616.21.05437-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a lethal brain tumor characterized by developmental hierarchical phenotypic heterogeneity, therapy resistance and recurrent growth. Neural stem cells (NSCs) from human central nervous system (CNS), and glioblastoma stem cells from patient-derived GBM (pdGSC) samples were cultured in both 2D well-plate and 3D monoclonal neurosphere culture system (pdMNCS). The pdMNCS model shows promise to establish a relevant 3D-tumor environment that maintains GBM cells in the stem cell phase within suspended neurospheres. METHODS Utilizing the pdMNCS, we examined GBM cell-lines for a wide spectrum of developmental cancer stem cell markers, including the early blastocyst inner-cell mass (ICM)-specific Nanog, Oct3/4,B, and CD133. RESULTS We observed that MNCS epigenotype is recapitulated using gliomasphere-derived cells. CD133, the marker of GSC is robustly expressed in 3D-gliomaspheres and localized within the plasma membrane compartment. Conversely, gliomasphere cultures grown in conventional 2D culture quickly lost CD133 expression, indicating its variable expression is dependent on cell-culture conditions. Incomplete differentiation of cytoskeleton microtubules and intermediate filaments (IFs) of patient derived cells, similar to commercially available GBM cell lines, was seen. Subsequently, in order to determine whether Oct3/4 it was necessary for CD133 expression and cancer stemness, we transfected 2D and 3D culture with siRNA against Oct3/4 and found a significant reduction in gliomasphere formation. CONCLUSIONS These results suggest that expression of Oct3/4,A- and CD133 suppress differentiation of GSCs.
Collapse
Affiliation(s)
- Tatyana N Ignatova
- Department of Neurosurgery, University of Tennessee, Health Science Center, Memphis, TN, USA
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA
| | - Hersh J Chaitin
- College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Nickolay V Kukekov
- Department of Pathology and Center for Neurobiology and Behavior, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Oleg N Suslov
- McKnight Brain Institute, Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Galina I Dulatova
- Department of Neurosurgery, University of Tennessee, Health Science Center, Memphis, TN, USA
| | - Khalid A Hanafy
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA
| | - Frank D Vrionis
- Marcus Neuroscience Institute, Boca Raton Regional Hospital and Florida Atlantic University, Boca Raton, FL, USA -
| |
Collapse
|
5
|
Kim HJ, Batara DC, Jeon YJ, Lee S, Beck S, Kim SH. The impact of MEIS1 TALE homeodomain transcription factor knockdown on glioma stem cell growth. Anim Cells Syst (Seoul) 2024; 28:93-109. [PMID: 38487309 PMCID: PMC10939110 DOI: 10.1080/19768354.2024.2327340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
Myeloid ecotropic virus insertion site 1 (MEIS1) is a HOX co-factor necessary for organ development and normal hematopoiesis. Recently, MEIS1 has been linked to the development and progression of various cancers. However, its role in gliomagenesis particularly on glioma stem cells (GSCs) remains unclear. Here, we demonstrate that MEIS1 is highly upregulated in GSCs compared to normal, and glioma cells and to its differentiated counterparts. Inhibition of MEIS1 expression by shRNA significantly reduced GSC growth in both in vitro and in vivo experiments. On the other hand, integrated transcriptomics analyses of glioma datasets revealed that MEIS1 expression is correlated to cell cycle-related genes. Clinical data analysis revealed that MEIS1 expression is elevated in high-grade gliomas, and patients with high MEIS1 levels have poorer overall survival outcomes. The findings suggest that MEIS1 is a prognostic biomarker for glioma patients and a possible target for developing novel therapeutic strategies against GBM.
Collapse
Affiliation(s)
- Hyun-Jin Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Don Carlo Batara
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Samuel Beck
- Department of Dermatology, Center for Aging Research, Chobanian & Avedisian School of Medicine, Boston University, Boston, USA
| | - Sung-Hak Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
6
|
Mahmoudian E, Jahani-Asl A. Establishing Brain Tumor Stem Cell Culture from Patient Brain Tumors and Imaging Analysis of Patient-Derived Xenografts. Methods Mol Biol 2024; 2736:177-192. [PMID: 37243860 DOI: 10.1007/7651_2023_482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Brain tumor stem cells (BTSCs) have been isolated from different types of brain tumors including glioblastoma. Although BTSCs share common characteristics with neural stem cells (NSCs), such as capacity to self-renew and undergo long-term proliferation, they have tumor-propagating capabilities. A small population of BTSC can give rise to secondary tumor when transplanted into severe immunodeficient (SCID) mice. The histological and cytological features, as well as genetic heterogeneity of the xenografted tumors in mice, closely resemble those of primary tumors in patients. Patient-derived xenografts (PDX), therefore, provide a clinically relevant model to study brain tumors. Here, we describe our protocol for establishing BTSC cultures following surgical excision of human brain tumors and the procedures to conduct PDX studies in SCID mice. We also provide our detailed step-by-step protocol on in vivo imaging system (IVIS) of the PDX tumors as a noninvasive method to trace the cells and tumor volume.
Collapse
Affiliation(s)
- Elham Mahmoudian
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.
- Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
7
|
Pöhlking C, Beier S, Formanski JP, Friese M, Schreiber M, Schwalbe B. Isolation of Cells from Glioblastoma Multiforme Grade 4 Tumors for Infection with Zika Virus prME and ME Pseudotyped HIV-1. Int J Mol Sci 2023; 24:ijms24054467. [PMID: 36901897 PMCID: PMC10002608 DOI: 10.3390/ijms24054467] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/01/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
This study aimed to isolate cells from grade 4 glioblastoma multiforme tumors for infection experiments with Zika virus (ZIKV) prME or ME enveloped HIV-1 pseudotypes. The cells obtained from tumor tissue were successfully cultured in human cerebrospinal fluid (hCSF) or a mixture of hCSF/DMEM in cell culture flasks with polar and hydrophilic surfaces. The isolated tumor cells as well as the U87, U138, and U343 cells tested positive for ZIKV receptors Axl and Integrin αvβ5. Pseudotype entry was detected by the expression of firefly luciferase or green fluorescent protein (gfp). In prME and ME pseudotype infections, luciferase expression in U-cell lines was 2.5 to 3.5 logarithms above the background, but still two logarithms lower than in the VSV-G pseudotype control. Infection of single cells was successfully detected in U-cell lines and isolated tumor cells by gfp detection. Even though prME and ME pseudotypes had low infection rates, pseudotypes with ZIKV envelopes are promising candidates for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Celine Pöhlking
- Department of Virology, LG-Schreiber, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Sebastian Beier
- Department of Virology, LG-Schreiber, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Jan Patrick Formanski
- Department of Virology, LG-Schreiber, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Michael Friese
- Department of Pathology and Neuropathology, Asklepios Kliniken Hamburg GmbH, Asklepios Klinik Nord, Standort Heidberg, 22417 Hamburg, Germany
| | - Michael Schreiber
- Department of Virology, LG-Schreiber, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
- Correspondence:
| | - Birco Schwalbe
- Department of Neurosurgery, Asklepios Kliniken Hamburg GmbH, Asklepios Klinik Nord, Standort Heidberg, 22417 Hamburg, Germany
| |
Collapse
|
8
|
Swati K, Agrawal K, Raj S, Kumar R, Prakash A, Kumar D. Molecular mechanism(s) of regulations of cancer stem cell in brain cancer propagation. Med Res Rev 2022; 43:441-463. [PMID: 36205299 DOI: 10.1002/med.21930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 06/01/2022] [Accepted: 09/11/2022] [Indexed: 11/12/2022]
Abstract
Brain tumors are most often diagnosed with solid neoplasms and are the primary reason for cancer-related deaths in both children and adults worldwide. With recent developments in the progression of novel targeted chemotherapies, the prognosis of malignant glioma remains dismal. However, the high recurrence rate and high mortality rate remain unresolved and are closely linked to the biological features of cancer stem cells (CSCs). Research on tumor biology has reached a new age with more understanding of CSC features. CSCs, a subpopulation of whole tumor cells, are now regarded as candidate therapeutic targets. Therefore, in the diagnosis and treatment of tumors, recognizing the biological properties of CSCs is of considerable significance. Here, we have discussed the concept of CSCs and their significant role in brain cancer growth and propagation. We have also discussed personalized therapeutic development and immunotherapies for brain cancer by specifically targeting CSCs.
Collapse
Affiliation(s)
- Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Kirti Agrawal
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, India.,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, India
| | - Sibi Raj
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, India.,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, India
| | - Rajeev Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, India.,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
9
|
A Quality Control Mechanism of Splice Site Selection Abrogated under Stress and in Cancer. Cancers (Basel) 2022; 14:cancers14071750. [PMID: 35406522 PMCID: PMC8996931 DOI: 10.3390/cancers14071750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Splicing and alternative splicing play a major role in regulating gene expression, and mis-regulation of splicing can lead to several diseases, including cancer. The aim of this review is to summarize the current knowledge of a quality control mechanism of splice site selection termed Suppression of Splicing (SOS), proposed to protect cells from splicing at the numerous intronic unused 5′ splice sites, and emphasize its relevance to cancer. This relevance stems from the finding that SOS is abrogated under stress and in cancer resulting in the expression of thousands of aberrant nonsense mRNAs that may be toxic to cells. These findings highlight the unexplored potential of such aberrant isoforms as novel targets for cancer diagnosis and therapies. Abstract Latent 5’ splice sites, highly abundant in human introns, are not normally used. This led to the proposal of a quality control mechanism, Suppression of Splicing (SOS), which protects cells from splicing at the numerous intronic latent sites, and whose activation can generate nonsense mRNAs. SOS was shown to be independent of Nonsense-Mediated mRNA Decay (NMD). Efforts to decipher the SOS mechanism revealed a pivotal role for initiator-tRNA, independent of protein translation. Recently, nucleolin (a multifunctional protein) was found to directly and specifically bind the initiator-tRNA in the nucleus and was shown to be a protein component of SOS, enabling an updated model of the SOS mechanism. Importantly, SOS is abrogated under stress and in cancer (e.g., in breast cancer cells and gliomas), generating thousands of nonsense mRNAs due to activation of latent splicing. The resulting affected human genes cover a variety of functional groups, including genes involved in cell proliferation and differentiation. Furthermore, in oligodendroglioma, the extent of activation of latent splicing increases with the severity of the cancer. Interesting examples are genes expressing aberrant nonsense mRNAs in both breast cancer and glioma, due to latent splicing activation. These findings highlight the unexplored potential of such aberrant isoforms as novel targets for cancer diagnosis and therapies.
Collapse
|
10
|
Seifert C, Balz E, Herzog S, Korolev A, Gaßmann S, Paland H, Fink MA, Grube M, Marx S, Jedlitschky G, Tzvetkov MV, Rauch BH, Schroeder HWS, Bien-Möller S. PIM1 Inhibition Affects Glioblastoma Stem Cell Behavior and Kills Glioblastoma Stem-like Cells. Int J Mol Sci 2021; 22:ijms222011126. [PMID: 34681783 PMCID: PMC8541331 DOI: 10.3390/ijms222011126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022] Open
Abstract
Despite comprehensive therapy and extensive research, glioblastoma (GBM) still represents the most aggressive brain tumor in adults. Glioma stem cells (GSCs) are thought to play a major role in tumor progression and resistance of GBM cells to radiochemotherapy. The PIM1 kinase has become a focus in cancer research. We have previously demonstrated that PIM1 is involved in survival of GBM cells and in GBM growth in a mouse model. However, little is known about the importance of PIM1 in cancer stem cells. Here, we report on the role of PIM1 in GBM stem cell behavior and killing. PIM1 inhibition negatively regulates the protein expression of the stem cell markers CD133 and Nestin in GBM cells (LN-18, U-87 MG). In contrast, CD44 and the astrocytic differentiation marker GFAP were up-regulated. Furthermore, PIM1 expression was increased in neurospheres as a model of GBM stem-like cells. Treatment of neurospheres with PIM1 inhibitors (TCS PIM1-1, Quercetagetin, and LY294002) diminished the cell viability associated with reduced DNA synthesis rate, increased caspase 3 activity, decreased PCNA protein expression, and reduced neurosphere formation. Our results indicate that PIM1 affects the glioblastoma stem cell behavior, and its inhibition kills glioblastoma stem-like cells, pointing to PIM1 targeting as a potential anti-glioblastoma therapy.
Collapse
Affiliation(s)
- Carolin Seifert
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Ellen Balz
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Susann Herzog
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Anna Korolev
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Sebastian Gaßmann
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Heiko Paland
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Matthias A. Fink
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Markus Grube
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Sascha Marx
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Gabriele Jedlitschky
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Mladen V. Tzvetkov
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Bernhard H. Rauch
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Pharmacology and Toxicology, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Henry W. S. Schroeder
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Sandra Bien-Möller
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
- Correspondence: ; Tel.: +49-03834-865646
| |
Collapse
|
11
|
Radiotherapy versus combination radiotherapy-bevacizumab for the treatment of recurrent high-grade glioma: a systematic review. Acta Neurochir (Wien) 2021; 163:1921-1934. [PMID: 33796887 PMCID: PMC8195900 DOI: 10.1007/s00701-021-04794-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/25/2021] [Indexed: 02/07/2023]
Abstract
Background High-grade gliomas (HGG) comprise the most common primary adult brain cancers and universally recur. Combination of re-irradiation therapy (reRT) and bevacizumab (BVZ) therapy for recurrent HGG is common, but its reported efficacy is mixed. Objective To assess clinical outcomes after reRT ± BVZ in recurrent HGG patients receiving stereotactic radiosurgery (SRS), hypofractionated radiosurgery (HFSRT), or fully fractionated radiotherapy (FFRT). Methods We performed a systematic review of PubMed, Web of Science, Scopus, Embase, and Cochrane databases, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. We identified studies reporting outcomes for patients with recurrent HGG treated via reRT ± BVZ. Cohorts were stratified by BVZ treatment status and re-irradiation modality (SRS, HFSRT, and FFRT). Outcome variables were overall survival (OS), progression-free survival (PFS), and radiation necrosis (RN). Results Data on 1399 patients was analyzed, with 954 patients receiving reRT alone and 445 patients receiving reRT + BVZ. All patients initially underwent standard-of-care therapy for their primary HGG. In a multivariate analysis that adjusted for median patient age, WHO grade, RT dosing, reRT fractionation regimen, time between primary and re-irradiation, and re-irradiation target volume, BVZ therapy was associated with significantly improved OS (2.51, 95% CI [0.11, 4.92] months, P = .041) but no significant improvement in PFS (1.40, 95% CI [− 0.36, 3.18] months, P = .099). Patients receiving BVZ also had significantly lower rates of RN (2.2% vs 6.5%, P < .001). Conclusions Combination of reRT + BVZ may improve OS and reduce RN rates in recurrent HGG, but further controlled studies are needed to confirm these effects. Supplementary Information The online version contains supplementary material available at 10.1007/s00701-021-04794-3.
Collapse
|
12
|
Karlsson J, Tzeng SY, Hemmati S, Luly KM, Choi O, Rui Y, Wilson DR, Kozielski KL, Quiñones-Hinojosa A, Green JJ. Photocrosslinked Bioreducible Polymeric Nanoparticles for Enhanced Systemic siRNA Delivery as Cancer Therapy. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2009768. [PMID: 34650390 PMCID: PMC8513781 DOI: 10.1002/adfm.202009768] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Indexed: 05/05/2023]
Abstract
Clinical translation of polymer-based nanocarriers for systemic delivery of RNA has been limited due to poor colloidal stability in the blood stream and intracellular delivery of the RNA to the cytosol. To address these limitations, this study reports a new strategy incorporating photocrosslinking of bioreducible nanoparticles for improved stability extracellularly and rapid release of RNA intracellularly. In this design, the polymeric nanocarriers contain ester bonds for hydrolytic degradation and disulfide bonds for environmentally triggered small interfering RNA (siRNA) release in the cytosol. These photocrosslinked bioreducible nanoparticles (XbNPs) have a shielded surface charge, reduced adsorption of serum proteins, and enable superior siRNA-mediated knockdown in both glioma and melanoma cells in high-serum conditions compared to non-crosslinked formulations. Mechanistically, XbNPs promote cellular uptake and the presence of secondary and tertiary amines enables efficient endosomal escape. Following systemic administration, XbNPs facilitate targeting of cancer cells and tissue-mediated siRNA delivery beyond the liver, unlike conventional nanoparticle-based delivery. These attributes of XbNPs facilitate robust siRNA-mediated knockdown in vivo in melanoma tumors colonized in the lungs following systemic administration. Thus, biodegradable polymeric nanoparticles, via photocrosslinking, demonstrate extended colloidal stability and efficient delivery of RNA therapeutics under physiological conditions, and thereby potentially advance systemic delivery technologies for nucleic acid-based therapeutics.
Collapse
Affiliation(s)
- Johan Karlsson
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemistry-Ångström Laboratory, Uppsala University, Uppsala SE-75121, Sweden
| | - Stephany Y Tzeng
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Shayan Hemmati
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kathryn M Luly
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Olivia Choi
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuan Rui
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - David R Wilson
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kristen L Kozielski
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Functional Interfaces, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen 76344, Germany
| | | | - Jordan J Green
- Department of Biomedical Engineering and Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Departments of Materials Science and Engineering, Neurosurgery, Oncology, Ophthalmology, and Chemical and Biomolecular Engineering, Sidney Kimmel Comprehensive Cancer Center, The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
13
|
Tai PA, Liu YL, Wen YT, Lin CM, Huynh TT, Hsiao M, Wu ATH, Wei L. The Development and Applications of a Dual Optical Imaging System for Studying Glioma Stem Cells. Mol Imaging 2020; 18:1536012119870899. [PMID: 31478435 PMCID: PMC6724491 DOI: 10.1177/1536012119870899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma multiforme represents one of the deadliest brain tumor types, manifested by a high rate of recurrence and poor prognosis. The presence of glioma stem cells (GSCs) can repopulate the tumor posttreatment and resist therapeutics. A better understanding of GSC biology is essential for developing more effective interventions. We established a CD133 promoter-driven dual reporter, expressing green fluorescent protein (GFP) and firefly luciferase (CD133-LG), capable for in vitro and in vivo imaging of CD133+ GSCs. We first demonstrated the reporter enabled in vitro analyses of GSCs. DBTRG-05MG (Denver Brain Tumor Research Group 05) carrying CD133-LG (DBTRG-05MG-CD133-LG) system reported increased GFP/luciferase activities in neurospheres. Additionally, we identified and isolated CD133+/GFP+ cells with increased tumorigenic properties, stemness markers, Notch1, β-catenin, and Bruton’s tyrosine kinase (Btk). Furthermore, prolonged temozolomide (TMZ) treatment enriched GSCs (reflected by increased percentage of CD133+ cells). Subsequently, Btk inhibitor, ibrutinib, suppressed GSC generation and stemness markers. Finally, we demonstrated real-time evaluation of anti-GSC function of ibrutinib in vivo with TMZ-enriched GSCs. Tumorigenesis was noninvasively monitored by bioluminescence imaging and mice that received ibrutinib showed a significantly lower tumor burden, indicating ibrutinib as a potential GSC inhibitor. In conclusion, we established a dual optical imaging system which enables the identification of CD133+ GSCs and screening for anti-GSC drugs.
Collapse
Affiliation(s)
- Po-An Tai
- 1 Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City.,2 Department of Surgery, School of Medicine, Buddhist Tzu Chi University, Hualien County
| | - Yen-Lin Liu
- 3 Department of Pediatrics, Taipei Medical University Hospital, Taipei.,4 Pediatric Brain Tumor Program, Taipei Cancer Center, Taipei Medical University, Taipei.,5 Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei
| | - Ya-Ting Wen
- 6 The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei.,7 Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei
| | - Chien-Min Lin
- 8 Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei.,9 Division of Neurosurgery, Department of Surgery, Shuang-Ho Hospital, Taipei Medical University, Taipei.,10 Taipei Neuroscience Institute, Taipei Medical University, Taipei
| | - Thanh-Tuan Huynh
- 11 Center for Molecular Biomedicine, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Michael Hsiao
- 12 Genomics Research Center, Academia Sinica, Taipei
| | - Alexander T H Wu
- 6 The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei.,13 Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei
| | - Li Wei
- 1 Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City.,14 Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei
| |
Collapse
|
14
|
Richard SA, Jia-Hao Z. Elucidating the pathogenic and biomarker potentials of FOXG1 in glioblastoma. Oncol Rev 2020; 14:444. [PMID: 32395201 PMCID: PMC7204822 DOI: 10.4081/oncol.2020.444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GB) is an extremely pugnacious brain cancer originating from neural stem (NS) cell-like cells. Forkhead box G1 (FOXG1; previously recognized as BF-1, qin, Chicken Brain Factor 1, or XBF-1 and renamed FOXG1 for mouse and human, and FoxG1 for other chordates) is an evolutionary preserved transcription factor driven from the forkhead box group of proteins FOXG1 modulates the speed of neurogenesis by maintaining progenitor cells in a proliferative mode as well as obstructing their differentiation into neurons during the initial periods of cortical formation. FOXG1 has been implicated in the formation of central nervous system (CNS) tumors and precisely GBs. Pathophysiologically, joint actions of FOXG1 and phosphatidylinositol- 3-kinases (PI3K) intermediate in intrinsic resistance of human GB cells to transforming growth factor-beta (TGF-β) stimulation of cyclin-dependent kinase inhibitor 1(p21Cip1) as well as growth inhibition. FOXG1 and NOTCH signaling pathways may functionally interrelate at different stages to facilitate gliomagenesis. Furthermore, FoxG1 actively contributed to the formation of transcription suppression complexes with corepressors of the Groucho/transducin-like Enhancer of split (Gro/TLEs). Also, FOXG1 was stimulated by Gro/TLE1 and abridged by Grg6. FOXG1 silencing in brain tumor-initiating cells (BTICs) also resulted in diminished secretion of markers characteristic undifferentiated natural neural stem/progenitor cells (NSPC) states, such as Oligodendrocyte transcription factor (OLIG2), (sex determining region Y)-box 2. (SOX2) and B lymphoma Mo-MLV insertion region 1 homolog (BMI1). This review therefore focuses on the pathogenic and biomarker potentials of FOXG1 in GB.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, P.R. China.,Department of Medicine, Princefield University, Ho-Volta Region, Ghana, West Africa
| | - Zhou Jia-Hao
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, P.R. China
| |
Collapse
|
15
|
Rodriguez V, Bailey R, Larion M, Gilbert MR. Retinoid receptor turnover mediated by sumoylation, ubiquitination and the valosin-containing protein is disrupted in glioblastoma. Sci Rep 2019; 9:16250. [PMID: 31700049 PMCID: PMC6838077 DOI: 10.1038/s41598-019-52696-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 08/30/2019] [Indexed: 02/06/2023] Open
Abstract
Resistance to therapeutic use of retinoids in glioma has been observed for over 20 years; however, the exact mechanism of resistance remains unknown. To understand retinoic acid resistance in glioma, we studied the turnover mechanism of retinoid receptor proteins in neural stem cells and glioma stem-like cells. Here, we show that in normal neural stem cells, proteasomal degradation of retinoid receptors involves sumoylation, ubiquitination and recognition by the valosin-containing protein (VCP/p97/Cdc48). We find that Sumo1 modification has a dual role to stabilize the retinoid receptor from unwanted degradation and signal additional modification via ubiquitination. Subsequently, the modified receptor binds to the VCP chaperone and both proteins are degraded by the proteasome. Additionally, we reveal that all trans retinoic acid (ATRA) induces VCP expression, creating a positive feedback loop that enhances degradation. In contrast, the pathway is impaired in the glioma stem-like cells resulting in the accumulation of sumoylated and high molecular weight forms of retinoid receptors that lack transcriptional activity and fail to be recognized by the proteasome. Moreover, modified receptor accumulation occurs before ATRA treatment; therefore, the transcritptional defect in glioma is due to a block in the proteasomal degradation pathway that occurs after the sumo modification step.
Collapse
Affiliation(s)
- Virginia Rodriguez
- Neuro-oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | - Rolanda Bailey
- Neuro-oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mioara Larion
- Neuro-oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark R Gilbert
- Neuro-oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Disulfiram modulates ROS accumulation and overcomes synergistically cisplatin resistance in breast cancer cell lines. Biomed Pharmacother 2019; 113:108727. [DOI: 10.1016/j.biopha.2019.108727] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/21/2019] [Accepted: 02/21/2019] [Indexed: 01/16/2023] Open
|
17
|
Cenciarini M, Valentino M, Belia S, Sforna L, Rosa P, Ronchetti S, D'Adamo MC, Pessia M. Dexamethasone in Glioblastoma Multiforme Therapy: Mechanisms and Controversies. Front Mol Neurosci 2019; 12:65. [PMID: 30983966 PMCID: PMC6449729 DOI: 10.3389/fnmol.2019.00065] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/26/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant of the glial tumors. The world-wide estimates of new cases and deaths annually are remarkable, making GBM a crucial public health issue. Despite the combination of radical surgery, radio and chemotherapy prognosis is extremely poor (median survival is approximately 1 year). Thus, current therapeutic interventions are highly unsatisfactory. For many years, GBM-induced brain oedema and inflammation have been widely treated with dexamethasone (DEX), a synthetic glucocorticoid (GC). A number of studies have reported that DEX also inhibits GBM cell proliferation and migration. Nevertheless, recent controversial results provided by different laboratories have challenged the widely accepted dogma concerning DEX therapy for GBM. Here, we have reviewed the main clinical features and genetic and epigenetic abnormalities underlying GBM. Finally, we analyzed current notions and concerns related to DEX effects on cerebral oedema, cancer cell proliferation and migration and clinical outcome.
Collapse
Affiliation(s)
- Marta Cenciarini
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Simona Ronchetti
- Section of Pharmacology, Department of Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Mauro Pessia
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy.,Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| |
Collapse
|
18
|
Zhang H, Ma Y, Wang H, Xu L, Yu Y. MMP-2 expression and correlation with pathology and MRI of glioma. Oncol Lett 2018; 17:1826-1832. [PMID: 30675244 PMCID: PMC6341586 DOI: 10.3892/ol.2018.9806] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023] Open
Abstract
The expression of matrix metalloproteinase-2 (MMP-2) in brain glioma and its correlation with patients' clinicopathological characteristics and magnetic resonance imaging (MRI) features were investigated. A total of 104 patients with brain glioma admitted and treated in the First Affiliated Hospital of Anhui Medical University from June 2010 to September 2014 were randomly enrolled. MRI examination was performed before operation. Immunohistochemistry (IHC) was used to detect the expression levels of MMP-2 in brain glioma tissues and paired normal brain tissues after operation and to analyze the associations of MMP-2 expression with the clinicopathological characteristics of brain glioma and survival time of patients. The relationship between MMP-2 expression and preoperative MRI features of glioma was analyzed. The positive rate of MMP-2 expression in brain glioma was 73.08% (76/104), while that in paired normal brain tissues was only 12.5% (13/104), obviously lower than that in brain glioma tissues (P<0.05). The MMP-2 expression in the body of glioma was not related to the patients' sex, age, tumor location and pathological type (P>0.05), but there was a significant correlation with the tumor diameter and pathological grade of the patients (P<0.05). Analysis by Cox model suggested that tumor diameter, pathological grade and MMP-2 were independent prognostic factors for glioma (P<0.05). The overall survival (OS) of patients in the positive MMP-2 expression group was 16.4 months, while the OS in the negative MMP-2 expression group was 20.16 months, and the difference between the two groups was statistically significant (P<0.05). The positive expression of MMP-2 in glioma was closely related to the uniformity of MRI signal for tumor, tumor diameter, severity of peritumoral edema, degree of enhancement and pathological grade of tumor (P<0.05). MMP-2 is highly expressed in brain glioma, and it is a negative factor for prognosis. Therefore, the MRI manifestations of glioma can reflect to some extent the intensity of MMP-2 expression.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yunxia Ma
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Haibao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Liyan Xu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
19
|
Rouigari M, Dehbashi M, Ghaedi K, Pourhossein M. Targetome Analysis Revealed Involvement of MiR-126 in Neurotrophin Signaling Pathway: A Possible Role in Prevention of Glioma Development. CELL JOURNAL 2018; 20:150-156. [PMID: 29633591 PMCID: PMC5893285 DOI: 10.22074/cellj.2018.4901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/14/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVES For the first time, we used molecular signaling pathway enrichment analysis to determine possible involvement of miR-126 and IRS-1 in neurotrophin pathway. MATERIALS AND METHODS In this prospective study, Validated and predicted targets (targetome) of miR-126 were collected following searching miRtarbase (http://mirtarbase.mbc.nctu.edu.tw/) and miRWalk 2.0 databases, respectively. Then, approximate expression of miR-126 targeting in Glioma tissue was examined using UniGene database (http://www.ncbi. nlm.nih.gov/unigene). In silico molecular pathway enrichment analysis was carried out by DAVID 6.7 database (http://david. abcc.ncifcrf.gov/) to explore which signaling pathway is related to miR-126 targeting and how miR-126 attributes to glioma development. RESULTS MiR-126 exerts a variety of functions in cancer pathogenesis via suppression of expression of target gene including PI3K, KRAS, EGFL7, IRS-1 and VEGF. Our bioinformatic studies implementing DAVID database, showed the involvement of miR-126 target genes in several signaling pathways including cancer pathogenesis, neurotrophin functions, Glioma formation, insulin function, focal adhesion production, chemokine synthesis and secretion and regulation of the actin cytoskeleton. CONCLUSIONS Taken together, we concluded that miR-126 enhances the formation of glioma cancer stem cell probably via down regulation of IRS-1 in neurotrophin signaling pathway.
Collapse
Affiliation(s)
- Maedeh Rouigari
- Isfahan Neuroscience Research Center (INRC), Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moein Dehbashi
- Isfahan Neuroscience Research Center (INRC), Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
- Genetics Division, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Kamran Ghaedi
- Cell and Molecular Biology Division, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Meraj Pourhossein
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences Isfahan, Iran
- Department of Food Science and Technology, Food Security Research Center, School of Nutrition and Food Science, Isfahan, Iran.
| |
Collapse
|
20
|
Soeda A, Lathia J, Williams BJ, Wu Q, Gallagher J, Androutsellis-Theotokis A, Giles AJ, Yang C, Zhuang Z, Gilbert MR, Rich JN, Park DM. The p38 signaling pathway mediates quiescence of glioma stem cells by regulating epidermal growth factor receptor trafficking. Oncotarget 2018; 8:33316-33328. [PMID: 28410196 PMCID: PMC5464870 DOI: 10.18632/oncotarget.16741] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 03/19/2017] [Indexed: 12/21/2022] Open
Abstract
EGFR pathway is upregulated in malignant gliomas, and its downstream signaling is important for self-renewal of glioma cancer stem-like cells (GSC). p38 mitogen-activated protein kinase (MAPK) signaling, a stress-activated signaling cascade with suppressive and permissive effects on tumorigenesis, can promote internalization and ubiquitin ligase mediated degradation of EGFR. In this study, we investigated the role of p38 MAPK signaling on the self-renewal of GSCs with the hypothesis that inhibition may lead to enhanced self-renewal capacity by retention of EGFR. Inhibition of p38 MAPK pathway led to increase in EGFR expression but surprisingly, reduced proliferation. Additional functional evaluation revealed that p38 inhibition was associated with decrease in cell death and maintenance of undifferentiated state. Further probing the effect of p38 inhibition demonstrated attenuation of EGFR downstream signaling activity in spite of prolonged surface expression of the receptor. In vitro observations were confirmed in xenograft in vivo experiments. These data suggest that p38 MAPK control of EGFR signaling activity may alter GSC cell cycle state by regulating quiescence and passage into transit amplifying state.
Collapse
Affiliation(s)
- Akio Soeda
- Department of Neurosurgery, Gifu University, Gifu, Japan
| | - Justin Lathia
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Brian J Williams
- Department of Neurosurgery, University of Louisville, Louisville, KY, USA
| | - Qiulian Wu
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Joseph Gallagher
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Amber J Giles
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Chunzhang Yang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Jeremy N Rich
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Deric M Park
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
21
|
Liu Y, Shi L, Liu Y, Li P, Jiang G, Gao X, Zhang Y, Jiang C, Zhu W, Han H, Ju F. Activation of PPARγ mediates icaritin-induced cell cycle arrest and apoptosis in glioblastoma multiforme. Biomed Pharmacother 2018; 100:358-366. [PMID: 29453045 DOI: 10.1016/j.biopha.2018.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most prevalent primary malignancy of the brain. This study was designed to investigate whether icaritin exerts anti-neoplastic activity against GBM in vitro. MATERIALS AND METHODS Cell Counting Kit-8 (CCK-8) assay was utilized to examine the viability of GBM cells. The apoptotic cell population was measured by flow cytometry analysis. Cell cycle distribution was detected by flow cytometry as well. Western blot analysis was performed to examine the level of biomarker proteins in GBM cells. Levels of PPARγ mRNA and protein were detected by qPCR and western blot analysis, respectively. To examine the role of PPARγ in the anti-neoplastic activity of icaritin, PPARγ antagonist GW9662 or PPARγ siRNA was used. The activity of PPARγ was determined by DNA binding and luciferase assays. RESULTS Our findings revealed that icaritin markedly suppresses cell growth in a dose-dependent and time-dependent fashion. The cell population at the G0/G1 phase of the cell cycle was significantly increased following icaritin treatment. Meanwhile, icaritin promoted apoptotic cell death in T98G and U87MG cells. Further investigation showed upregulation of PPARγ played a key role in the anti-neoplastic activities of icaritin. Moreover, our result demonstrated activation of AMPK signaling by icaritin mediated the modulatory effect of icaritin on PPARγ. CONCLUSION Our results suggest the PPARγ may mediate anti-neoplastic activities against GBM.
Collapse
Affiliation(s)
- Yongji Liu
- Department of Neurosurgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China.
| | - Ling Shi
- Department of Neurosurgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Yuan Liu
- Department of Neurosurgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Peng Li
- Department of Emergency, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Guoping Jiang
- Department of Neurosurgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Xiaoning Gao
- Department of Radiology, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Yongbin Zhang
- Department of Radiology, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Chuanwu Jiang
- Department of Radiology, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Weiping Zhu
- Department of Rheumatology and Immunology, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, Shandong, China
| | - Hongxing Han
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong, China
| | - Fang Ju
- Department of Oncology, Qingdao Central Hospital, Qingdao, Shandong, China.
| |
Collapse
|
22
|
Jahan N, Lee JM, Shah K, Wakimoto H. Therapeutic targeting of chemoresistant and recurrent glioblastoma stem cells with a proapoptotic variant of oncolytic herpes simplex virus. Int J Cancer 2017; 141:1671-1681. [PMID: 28567859 PMCID: PMC5796532 DOI: 10.1002/ijc.30811] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 02/21/2017] [Accepted: 05/15/2017] [Indexed: 01/14/2023]
Abstract
Temozolomide (TMZ) chemotherapy, in combination with maximal safe resection and radiotherapy, is the current standard of care for patients with glioblastoma (GBM). Despite this multimodal approach, GBM inevitably relapses primarily due to resistance to chemo-radiotherapy, and effective treatment is not available for recurrent disease. In this study we identified TMZ resistant patient-derived primary and previously treated recurrent GBM stem cells (GSC), and investigated the therapeutic activity of a pro-apoptotic variant of oHSV (oHSV-TRAIL) in vitro and in vivo. We show that oHSV-TRAIL modulates cell survival and MAP Kinase proliferation signaling pathways as well as DNA damage response pathways in both primary and recurrent TMZ-resistant GSC. Utilizing real time in vivo imaging and correlative immunohistochemistry, we show that oHSV-TRAIL potently inhibits tumor growth and extends survival of mice bearing TMZ-insensitive recurrent intracerebral GSC tumors via robust and selective induction of apoptosis-mediated death in tumor cells, resulting in cures in 40% of the treated mice. In comparison, the anti-tumor effects in a primary chemoresistant GSC GBM model exhibiting a highly invasive phenotype were significant but less prominent. This work thus demonstrates the ability of oHSV-TRAIL to overcome the therapeutic resistance and recurrence of GBM, and provides a basis for its testing in a GBM clinical trial.
Collapse
Affiliation(s)
- Nusrat Jahan
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jae M. Lee
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Hiroaki Wakimoto
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
23
|
Colwell N, Larion M, Giles AJ, Seldomridge AN, Sizdahkhani S, Gilbert MR, Park DM. Hypoxia in the glioblastoma microenvironment: shaping the phenotype of cancer stem-like cells. Neuro Oncol 2017; 19:887-896. [PMID: 28339582 PMCID: PMC5570138 DOI: 10.1093/neuonc/now258] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common and aggressive malignant primary brain tumor. Cellular heterogeneity is a characteristic feature of the disease and contributes to the difficulty in formulating effective therapies. Glioma stem-like cells (GSCs) have been identified as a subpopulation of tumor cells that are thought to be largely responsible for resistance to treatment. Intratumoral hypoxia contributes to maintenance of the GSCs by supporting the critical stem cell traits of multipotency, self-renewal, and tumorigenicity. This review highlights the interaction of GSCs with the hypoxic tumor microenvironment, exploring the mechanisms underlying the contribution of GSCs to tumor vessel dynamics, immune modulation, and metabolic alteration.
Collapse
Affiliation(s)
- Nicole Colwell
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Mioara Larion
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Amber J Giles
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Ashlee N Seldomridge
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Saman Sizdahkhani
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Deric M Park
- Neuro-Oncology Branch, National Cancer Institute and National Institute of Neurological Disorders and Stroke, Bethesda, Maryland ; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| |
Collapse
|
24
|
Notch3 signaling-mediated melanoma-endothelial crosstalk regulates melanoma stem-like cell homeostasis and niche morphogenesis. J Transl Med 2017; 97:725-736. [PMID: 28165469 PMCID: PMC5446297 DOI: 10.1038/labinvest.2017.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/25/2016] [Accepted: 12/22/2016] [Indexed: 12/27/2022] Open
Abstract
Melanoma is among the most virulent cancers, owing to its propensity to metastasize and its resistance to current therapies. The treatment failure is largely attributed to tumor heterogeneity, particularly subpopulations possessing stem cell-like properties, ie, melanoma stem-like cells (MSLCs). Evidence indicates that the MSLC phenotype is malleable and may be acquired by non-MSLCs through phenotypic switching upon appropriate stimuli, the so-called 'dynamic stemness'. Since the phenotypic characteristics and functional integrity of MSLCs depend on their vascular niche, using a two-dimensional (2D) melanoma-endothelium co-culture model, where the MSLC niche is recapitulated in vitro, we identified Notch3 signaling pathway as a micro-environmental cue governing MSLC phenotypic plasticity via pathway-specific gene expression arrays. Accordingly, lentiviral shRNA-mediated Notch3 knockdown (KD) in melanoma cell lines exhibiting high levels of endogenous Notch3 led to retarded/abolished tumorigenicity in vivo through both depleting MSLC fractions, evinced by MSLC marker downregulation (eg, CD133 and CD271); and impeding the MSLC niche, corroborated by the attenuated tumor angiogenesis as well as vasculogenic mimicry. In contrast, Notch3 KD affected neither tumor growth nor MSLC subsets in a melanoma cell line with relatively low endogenous Notch3 expression. Thus, Notch3 signaling may facilitate MSLC plasticity and niche morphogenesis in a cell context-dependent manner. Our findings illustrate Notch3 as a molecular switch driving melanoma heterogeneity, and provide the biological rationale for Notch inhibition as a promising therapeutic option.
Collapse
|
25
|
Zhang C, Mukherjee S, Tucker-Burden C, Ross JL, Chau MJ, Kong J, Brat DJ. TRIM8 regulates stemness in glioblastoma through PIAS3-STAT3. Mol Oncol 2017; 11:280-294. [PMID: 28100038 PMCID: PMC5332279 DOI: 10.1002/1878-0261.12034] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant form of primary brain tumor, and GBM stem-like cells (GSCs) contribute to the rapid growth, therapeutic resistance, and clinical recurrence of these fatal tumors. STAT3 signaling supports the maintenance and proliferation of GSCs, yet regulatory mechanisms are not completely understood. Here, we report that tri-partite motif-containing protein 8 (TRIM8) activates STAT3 signaling to maintain stemness and self-renewing capabilities of GSCs. TRIM8 (also known as 'glioblastoma-expressed ring finger protein') is expressed equally in GBM and normal brain tissues, despite its hemizygous deletion in the large majority of GBMs, and its expression is highly correlated with stem cell markers. Experimental knockdown of TRIM8 reduced GSC self-renewal and expression of SOX2, NESTIN, and p-STAT3, and promoted glial differentiation. Overexpression of TRIM8 led to higher expression of p-STAT3, c-MYC, SOX2, NESTIN, and CD133, and enhanced GSC self-renewal. We found that TRIM8 activates STAT3 by suppressing the expression of PIAS3, an inhibitor of STAT3, most likely through E3-mediated ubiquitination and proteasomal degradation. Interestingly, we also found that STAT3 activation upregulates TRIM8, providing a mechanism for normalized TRIM8 expression in the setting of hemizygous gene deletion. These data demonstrate that bidirectional TRIM8-STAT3 signaling regulates stemness in GSC.
Collapse
Affiliation(s)
- Changming Zhang
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA.,Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan, China
| | - Subhas Mukherjee
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Carol Tucker-Burden
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - James L Ross
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA.,Graduate Program in Cancer Biology, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Monica J Chau
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Jun Kong
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel J Brat
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
26
|
Han H, Xu B, Hou P, Jiang C, Liu L, Tang M, Yang X, Zhang Y, Liu Y. Icaritin Sensitizes Human Glioblastoma Cells to TRAIL-Induced Apoptosis. Cell Biochem Biophys 2017; 72:533-42. [PMID: 25577511 DOI: 10.1007/s12013-014-0499-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been considered to be one of the most promising candidates in research on treatments for cancer, because it induces apoptosis in a wide variety of cancer cells but not in most normal human cell types. However, many cells including glioblastoma (GBM) cells are resistant to TRAIL-induced apoptosis, which limits the potential application of TRAIL in cancer therapy. Icaritin, a hydrolytic product of icariin from Epimedium Genus, has been identified as a potential therapeutic and preventive agent in renal cell carcinoma and breast cancer. In this study, we investigated whether Icaritin treatment could modulate TRAIL-induced apoptosis in GBM. The effect of icaritin on TRAIL sensitivity was assessed in human GBM U87 and U373 cells. The underlying regulatory cascades were approached by biochemical and pharmacological strategies. We found that nontoxic concentration of icaritin alone had no significant effect on the level of apoptosis, but a combination treatment of TRAIL and icaritin caused a significantly more profound apoptosis. The sensitization was accompanied by c-FLIP down-regulation and inhibition of NF-κB activity. Studies have further demonstrated that silencing NF-κB alone was sufficient to down-regulate c-FLIP expression and sensitized both tested cells to TRAIL-induced apoptosis. These data suggest that icaritin sensitizes TRAIL-induced tumor cell apoptosis via suppression of NF-κB-dependent c-FLIP expression, providing in vitro evidence supporting the notion that icaritin is a potential sensitizer of TRAIL in anticancer therapy against human GBM.
Collapse
Affiliation(s)
- Hongxing Han
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Neurology, Linyi People's Hospital, Linyi, Shandong, China
| | - Bo Xu
- Department of Neurology, Provincial Hospital of Shandong University, Jinan, Shandong, China.,Department of Neurology, The Second Affiliated Hospital, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Pengzhi Hou
- Department of Neurosurgery, Huangdao Traditional Chinese Medicine Hospital, Qingdao, Shandong, China
| | - Chuanwu Jiang
- Department of Radiology, Qingdao Hiser Medical Center, Qingdao, Shandong, China.,Department of Radiology, Provincial Hospital of Shandong University, Jinan, Shandong, China
| | - Longxi Liu
- Department of Neurosurgery, Qingdao Hiser Medical Center, Qingdao, Shandong, China
| | - Ming Tang
- Department of Neurology, Qingdao Hiser Medical Center, Qingdao, Shandong, China
| | - Xiuli Yang
- Department of Neurology, Qingdao Hiser Medical Center, Qingdao, Shandong, China
| | - Yunxu Zhang
- Department of Neurosurgery, Qingdao Hiser Medical Center, Qingdao, Shandong, China
| | - Yongji Liu
- Department of Neurosurgery, Qingdao Hiser Medical Center, Qingdao, Shandong, China.
| |
Collapse
|
27
|
Ferreira WAS, Pinheiro DDR, Costa Junior CAD, Rodrigues-Antunes S, Araújo MD, Leão Barros MB, Teixeira ACDS, Faro TAS, Burbano RR, Oliveira EHCD, Harada ML, Borges BDN. An update on the epigenetics of glioblastomas. Epigenomics 2016; 8:1289-305. [PMID: 27585647 DOI: 10.2217/epi-2016-0040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glioblastomas, also known as glioblastoma multiforme (GBM), are the most aggressive and malignant type of primary brain tumor in adults, exhibiting notable variability at the histopathological, genetic and epigenetic levels. Recently, epigenetic alterations have emerged as a common hallmark of many tumors, including GBM. Considering that a deeper understanding of the epigenetic modifications that occur in GBM may increase the knowledge regarding the tumorigenesis, progression and recurrence of this disease, in this review we discuss the recent major advances in GBM epigenetics research involving histone modification, glioblastoma stem cells, DNA methylation, noncoding RNAs expression, including their main alterations and the use of epigenetic therapy as a valid option for GBM treatment.
Collapse
Affiliation(s)
- Wallax Augusto Silva Ferreira
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Danilo do Rosário Pinheiro
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Carlos Antonio da Costa Junior
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Symara Rodrigues-Antunes
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Mariana Diniz Araújo
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Mariceli Baia Leão Barros
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Adriana Corrêa de Souza Teixeira
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Thamirys Aline Silva Faro
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | | | | | - Maria Lúcia Harada
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| | - Bárbara do Nascimento Borges
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA)-Belém, Pará, Brazil
| |
Collapse
|
28
|
Verkhratsky A, Steardo L, Parpura V, Montana V. Translational potential of astrocytes in brain disorders. Prog Neurobiol 2016; 144:188-205. [PMID: 26386136 PMCID: PMC4794425 DOI: 10.1016/j.pneurobio.2015.09.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/03/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Abstract
Fundamentally, all brain disorders can be broadly defined as the homeostatic failure of this organ. As the brain is composed of many different cells types, including but not limited to neurons and glia, it is only logical that all the cell types/constituents could play a role in health and disease. Yet, for a long time the sole conceptualization of brain pathology was focused on the well-being of neurons. Here, we challenge this neuron-centric view and present neuroglia as a key element in neuropathology, a process that has a toll on astrocytes, which undergo complex morpho-functional changes that can in turn affect the course of the disorder. Such changes can be grossly identified as reactivity, atrophy with loss of function and pathological remodeling. We outline the pathogenic potential of astrocytes in variety of disorders, ranging from neurotrauma, infection, toxic damage, stroke, epilepsy, neurodevelopmental, neurodegenerative and psychiatric disorders, Alexander disease to neoplastic changes seen in gliomas. We hope that in near future we would witness glial-based translational medicine with generation of deliverables for the containment and cure of disorders. We point out that such as a task will require a holistic and multi-disciplinary approach that will take in consideration the concerted operation of all the cell types in the brain.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Luca Steardo
- Department of Psychiatry, University of Naples, SUN, Largo Madonna delle Grazie, Naples, Italy
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine and Atomic Force Microscopy & Nanotechnology Laboratories, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vedrana Montana
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
29
|
Pavon LF, Sibov TT, de Oliveira DM, Marti LC, Cabral FR, de Souza JG, Boufleur P, Malheiros SM, de Paiva Neto MA, da Cruz EF, Chudzinski-Tavassi AM, Cavalheiro S. Mesenchymal stem cell-like properties of CD133+ glioblastoma initiating cells. Oncotarget 2016; 7:40546-40557. [PMID: 27244897 PMCID: PMC5130027 DOI: 10.18632/oncotarget.9658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/16/2016] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma is composed of dividing tumor cells, stromal cells and tumor initiating CD133+ cells. Recent reports have discussed the origin of the glioblastoma CD133+ cells and their function in the tumor microenvironment. The present work sought to investigate the multipotent and mesenchymal properties of primary highly purified human CD133+ glioblastoma-initiating cells. To accomplish this aim, we used the following approaches: i) generation of tumor subspheres of CD133+ selected cells from primary cell cultures of glioblastoma; ii) analysis of the expression of pluripotency stem cell markers and mesenchymal stem cell (MSC) markers in the CD133+ glioblastoma-initiating cells; iii) side-by-side ultrastructural characterization of the CD133+ glioblastoma cells, MSC and CD133+ hematopoietic stem cells isolated from human umbilical cord blood (UCB); iv) assessment of adipogenic differentiation of CD133+ glioblastoma cells to test their MSC-like in vitro differentiation ability; and v) use of an orthotopic glioblastoma xenograft model in the absence of immune suppression. We found that the CD133+ glioblastoma cells expressed both the pluripotency stem cell markers (Nanog, Mush-1 and SSEA-3) and MSC markers. In addition, the CD133+ cells were able to differentiate into adipocyte-like cells. Transmission electron microscopy (TEM) demonstrated that the CD133+ glioblastoma-initiating cells had ultrastructural features similar to those of undifferentiated MSCs. In addition, when administered in vivo to non-immunocompromised animals, the CD133+ cells were also able to mimic the phenotype of the original patient's tumor. In summary, we showed that the CD133+ glioblastoma cells express molecular signatures of MSCs, neural stem cells and pluripotent stem cells, thus possibly enabling differentiation into both neural and mesodermal cell types.
Collapse
Affiliation(s)
- Lorena Favaro Pavon
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
- Hospital Israelita Albert Einstein (HIAE), Experimental Research, São Paulo, Brazil
| | - Tatiana Tais Sibov
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | | | - Luciana C. Marti
- Hospital Israelita Albert Einstein (HIAE), Experimental Research, São Paulo, Brazil
- Allergy and Immunopathology Graduate Program, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Francisco Romero Cabral
- Hospital Israelita Albert Einstein (HIAE), Experimental Research, São Paulo, Brazil
- Faculdade de Ciências Médicas da São Casa de São Paulo, São Paulo, Brazil
| | - Jean Gabriel de Souza
- Biochemistry and Biophysics Laboratory, Butantan Institute, Neuro-Oncology Program, São Paulo, Brazil
| | - Pamela Boufleur
- Biochemistry and Biophysics Laboratory, Butantan Institute, Neuro-Oncology Program, São Paulo, Brazil
| | - Suzana M.F. Malheiros
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
- Hospital Israelita Albert Einstein (HIAE), Neuro-Oncology Program, São Paulo, Brazil
| | - Manuel A. de Paiva Neto
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Edgard Ferreira da Cruz
- Discipline of Nephrology, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | | | - Sérgio Cavalheiro
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| |
Collapse
|
30
|
Triscott J, Rose Pambid M, Dunn SE. Concise review: bullseye: targeting cancer stem cells to improve the treatment of gliomas by repurposing disulfiram. Stem Cells 2016; 33:1042-6. [PMID: 25588723 DOI: 10.1002/stem.1956] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/19/2014] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs) are thought to be at the root of cancer recurrence because they resist conventional therapies and subsequently reinitiate tumor cell growth. Thus, targeting CSCs could be the bullseye to successful cancer therapeutics in the future. Brain tumors are some of the most challenging types of cancer to treat and the median survival following the initial diagnosis is 12-18 months. Among the different types of brain tumors, glioblastoma (GBM) is considered the most aggressive and remains extremely difficult to treat. Despite surgery, radiation, and chemotherapy, most patients develop refractory disease. Temozolomide (TMZ) is a chemotherapy used to treat GBM however resistance develops in most patients. The underlying mechanisms for TMZ resistance (TMZ-resistant) involve the expression of DNA repair gene O(6)-methylguanine-DNA methyltransferase. CSC genes such as Sox-2, BMI-1, and more recently Y-box binding protein-1 also play a role in resistance. In order to develop novel therapies for GBM, libraries of small interfering RNAs and off-patent drugs have been screened. Over the past few years, several independent laboratories identified disulfiram (DSF) as an off-patent drug that kills GBM CSCs. Reportedly DSF has several modes of action including its ability to inhibit aldehyde dehydrogenases, E3 ligase, polo-like kinase 1, and NFkB. Due to the fact that GBM is a disease of heterogeneity, chemotherapy with multitargeting properties may be the way of the future. In broader terms, DSF kills CSCs from a range of different cancer types further supporting the idea of repurposing it for "target practice."
Collapse
Affiliation(s)
- Joanna Triscott
- Experimental Medicine Program, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
31
|
Zhang J, Hao G, Yao C, Hu S, Hu C, Zhang B. Paramagnetic albumin decorated CuInS2/ZnS QDs for CD133+ glioma bimodal MR/fluorescence targeted imaging. J Mater Chem B 2016; 4:4110-4118. [PMID: 32264613 DOI: 10.1039/c6tb00834h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A sensitive, specific, accurate and biocompatible molecular nanoprobe is constructed, by rational design of the structure and an advanced surface engineering strategy, with MR/fluorescence imaging modalities for CD133+ glioma bimodal targeted imaging.
Collapse
Affiliation(s)
- Jing Zhang
- Imaging Center
- The First Affiliated Hospital of Soochow University
- Suzhou 215006
- China
| | - Guangyu Hao
- Imaging Center
- The First Affiliated Hospital of Soochow University
- Suzhou 215006
- China
| | - Chenfei Yao
- Imaging Center
- The First Affiliated Hospital of Soochow University
- Suzhou 215006
- China
| | - Su Hu
- Imaging Center
- The First Affiliated Hospital of Soochow University
- Suzhou 215006
- China
| | - Chunhong Hu
- Imaging Center
- The First Affiliated Hospital of Soochow University
- Suzhou 215006
- China
| | - Bingbo Zhang
- Institute of Photomedicine
- Shanghai Skin Disease Hospital
- The Institute for Biomedical Engineering & Nano Science
- Tongji University School of Medicine
- Shanghai 200443
| |
Collapse
|
32
|
Afzali A, Emadi-Baygi M, Nikpour P, Nazemroaya F, Kheirollahi M. Expression of ZFX gene correlated with the central features of the neoplastic phenotype in human brain tumors with distinct phenotypes. Adv Biomed Res 2015; 4:179. [PMID: 26605218 PMCID: PMC4617010 DOI: 10.4103/2277-9175.164000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/14/2015] [Indexed: 11/27/2022] Open
Abstract
Background: The zinc finger transcription factor zinc finger protein, X-linked (ZFX) acts as an important director of self-renewal in several stem cell types. Moreover, ZFX expression abnormally increases in various cancers and relates to tumor grade. We performed this study, to examine its role in the pathogenesis of astrocytoma and meningioma. Materials and Methods: We used real-time reverse transcription polymerase chain reaction method for evaluation of ZFX expression in 25 astrocytoma tumoral tissue and 25 meningioma tumoral tissues with different WHO grades. Furthermore, the association of gene expression with various clinic-pathological characteristics was examined. Results: We found that there is a significant association between gene expression and different tumor grades, the presence or absence of invasion, forming and nonforming of glomeruloid vessels, the age over or under 50 and the presence or absence of calcification in astrocytomas. This is the first report that shows that ZFX was directly correlated with the central features of the neoplastic phenotype, including the growth of cancer cells, angiogenesis, and invasion. Conclusion: Regarding all the above-mentioned studies, it is highly plausible that silencing the expression of ZFX gene in gliomas has a major role in the therapeutic interventions of the disease in future.
Collapse
Affiliation(s)
- Azita Afzali
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Modjtaba Emadi-Baygi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, School of Medicine, Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemehe Nazemroaya
- Department of Genetics and Molecular Biology, School of Medicine, Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Majid Kheirollahi
- Department of Genetics and Molecular Biology, School of Medicine, Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
33
|
Auffinger B, Spencer D, Pytel P, Ahmed AU, Lesniak MS. The role of glioma stem cells in chemotherapy resistance and glioblastoma multiforme recurrence. Expert Rev Neurother 2015; 15:741-52. [PMID: 26027432 DOI: 10.1586/14737175.2015.1051968] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioma stem cells (GSCs) constitute a slow-dividing, small population within a heterogeneous glioblastoma. They are able to self-renew, recapitulate a whole tumor, and differentiate into other specific glioblastoma multiforme (GBM) subpopulations. Therefore, they have been held responsible for malignant relapse after primary standard therapy and the poor prognosis of recurrent GBM. The failure of current therapies to eliminate specific GSC subpopulations has been considered a major factor contributing to the inevitable recurrence in GBM patients after treatment. Here, we discuss the molecular mechanisms of chemoresistance of GSCs and the reasons why complete eradication of GSCs is so difficult to achieve. We will also describe the targeted therapies currently available for GSCs and possible mechanisms to overcome such chemoresistance and avoid therapeutic relapse.
Collapse
Affiliation(s)
- Brenda Auffinger
- The Brain Tumor Center, The University of Chicago, 5841 South Maryland Ave, M/C 3026, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
34
|
Zhou X, Zheng C, Shi Q, Li X, Shen Z, Yu R. Isolation, cultivation and identification of brain glioma stem cells by magnetic bead sorting. Neural Regen Res 2015; 7:985-92. [PMID: 25722686 PMCID: PMC4341279 DOI: 10.3969/j.issn.1673-5374.2012.13.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 03/24/2012] [Indexed: 12/28/2022] Open
Abstract
This study describes a detailed process for obtaining brain glioma stem cells from freshly dissected human brain glioma samples using an immunomagnetic bead technique combined with serum-free media pressure screening. Furthermore, the proliferation, differentiation and self-renewal biological features of brain glioma stem cells were identified. Results showed that a small number of CD133 positive tumor cells isolated from brain glioma samples survived as a cell suspension in serum-free media and proliferated. Subcultured CD133 positive cells maintained a potent self-renewal and proliferative ability, and expressed the stem cell-specific markers CD133 and nestin. After incubation with fetal bovine serum, the number of glial fibrillary acidic protein and microtubule associated protein 2 positive cells increased significantly, indicating that the cultured brain glioma stem cells can differentiate into astrocytes and neurons. Western blot analysis showed that tumor suppressor phosphatase and tensin homolog was highly expressed in tumor spheres compared with the differentiated tumor cells. These experimental findings indicate that the immunomagnetic beads technique is a useful method to obtain brain glioma stem cells from human brain tumors.
Collapse
Affiliation(s)
- Xiuping Zhou
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou 221002, Jiangsu Province, China ; Jiangsu Key Laboratory of Brain Disease Biology, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Chao Zheng
- Graduate School, Xuzhou Medical College, Xuzhou 221002, Jiangsu Province, China
| | - Qiong Shi
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou 221002, Jiangsu Province, China ; Jiangsu Key Laboratory of Brain Disease Biology, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Zhigang Shen
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Rutong Yu
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China ; Laboratory of Neurosurgery, Xuzhou Medical College, Xuzhou 221002, Jiangsu Province, China ; Jiangsu Key Laboratory of Brain Disease Biology, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| |
Collapse
|
35
|
Kim SH, Kim EJ, Hitomi M, Oh SY, Jin X, Jeon HM, Beck S, Jin X, Kim JK, Park CG, Chang SY, Yin J, Kim T, Jeon YJ, Song J, Lim YC, Lathia JD, Nakano I, Kim H. The LIM-only transcription factor LMO2 determines tumorigenic and angiogenic traits in glioma stem cells. Cell Death Differ 2015; 22:1517-25. [PMID: 25721045 DOI: 10.1038/cdd.2015.7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 12/30/2014] [Accepted: 01/14/2015] [Indexed: 01/23/2023] Open
Abstract
Glioblastomas (GBMs) maintain their cellular heterogeneity with glioma stem cells (GSCs) producing a variety of tumor cell types. Here we interrogated the oncogenic roles of Lim domain only 2 (LMO2) in GBM and GSCs in mice and human. High expression of LMO2 was found in human patient-derived GSCs compared with the differentiated progeny cells. LMO2 is required for GSC proliferation both in vitro and in vivo, as shRNA-mediated LMO2 silencing attenuated tumor growth derived from human GSCs. Further, LMO2 is sufficient to induce stem cell characteristics (stemness) in mouse premalignant astrocytes, as forced LMO2 expression facilitated in vitro and in vivo growth of astrocytes derived from Ink4a/Arf null mice and acquisition of GSC phenotypes. A subset of mouse and human GSCs converted into vascular endothelial-like tumor cells both in vitro and in vivo, which phenotype was attenuated by LMO2 silencing and promoted by LMO2 overexpression. Mechanistically, the action of LMO2 for induction of glioma stemness is mediated by transcriptional regulation of Jagged1 resulting in activation of the Notch pathway, whereas LMO2 directly occupies the promoter regions of the VE-cadherin gene for a gain of endothelial cellular phenotype. Subsequently, selective ablation of human GSC-derived VE-cadherin-expressing cells attenuated vascular formation in mouse intracranial tumors, thereby significantly prolonging mouse survival. Clinically, LMO2 expression was elevated in GBM tissues and inversely correlated with prognosis of GBM patients. Taken together, our findings describe novel dual roles of LMO2 to induce tumorigenesis and angiogenesis, and provide potential therapeutic targets in GBMs.
Collapse
Affiliation(s)
- S-H Kim
- 1] School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea [2] Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - E-J Kim
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - M Hitomi
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - S-Y Oh
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - X Jin
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - H-M Jeon
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - S Beck
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - X Jin
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - J-K Kim
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - C G Park
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - S-Y Chang
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| | - J Yin
- Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang 410-769, Republic of Korea
| | - T Kim
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y-J Jeon
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - J Song
- Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Y C Lim
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 143-752, Republic of Korea
| | - J D Lathia
- 1] Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA [2] Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA [3] Case Comprehensive Cancer Center, Cleveland, OH 44195, USA
| | - I Nakano
- 1] Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA [2] James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - H Kim
- School of Life Sciences and Biotechnology and Institute of Life Science and Natural Resources, Korea University, Seoul 136-713, Republic of Korea
| |
Collapse
|
36
|
Qu M, Qiu BO, Xiong W, Chen D, Wu A. Expression of a-disintegrin and metalloproteinase 10 correlates with grade of malignancy in human glioma. Oncol Lett 2015; 9:2157-2162. [PMID: 26137031 DOI: 10.3892/ol.2015.2993] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/05/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to determine the expression of a-disintegrin and metalloproteinase 10 (ADAM10) in human glioma tissues from surgical specimens and discuss its possible significance in glioma biology. A total of 43 glioma specimens obtained from patients between 2007 and 2010 were collected and a series of assays were performed. Of these, 22 cases were low-grade gliomas, while 21 cases were high-grade gliomas. In addition, 20 cases of meningioma were used as the control group. Reverse transcription-polymerase chain reaction (RT-PCR), western blot analysis and immunohistochemistry were used to determine the mRNA and protein expression levels of ADAM10. Besides the quantitative analysis, histological observations were also performed to localize ADAM10 expression in glioma cells. The RT-PCR and western blot analysis results demonstrated increased ADAM10 expression in the low-grade glioma samples compared with the control (P<0.05), while ADAM10 expression was further increased in the high-grade glioma samples (P<0.01 vs. control; P<0.05 vs. low-grade glioma), indicating that the mRNA and protein expression levels of ADAM10 were malignancy-dependent. The immunohistochemical analysis revealed that the ADAM10 protein was located on both the tumor cell membrane and blood vessel walls within tumor tissues. In conclusion, these results indicated that ADAM10 expression correlates with the grade of malignancy in human glioma from surgical specimens. In addition, the fact that ADAM10 protein was expressed on cell membranes and blood vessel walls within tumor tissues, indicates that its expression may be associated with invasive tumor growth and peritumoral edema formation.
Collapse
Affiliation(s)
- Min Qu
- Department of Neurosurgery, The Central Hospital of Dalian City, Dalian, Liaoning 116033, P.R. China
| | - B O Qiu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Wende Xiong
- Department of Neurosurgery, The Central Hospital of Dalian City, Dalian, Liaoning 116033, P.R. China
| | - Dong Chen
- Department of Neurosurgery, The Central Hospital of Dalian City, Dalian, Liaoning 116033, P.R. China
| | - Anhua Wu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
37
|
Zheng D, Liao S, Zhu G, Luo G, Xiao S, He J, Pei Z, Li G, Zhou Y. CD38 is a putative functional marker for side population cells in human nasopharyngeal carcinoma cell lines. Mol Carcinog 2015; 55:300-11. [PMID: 25630761 DOI: 10.1002/mc.22279] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 11/21/2014] [Accepted: 12/05/2014] [Indexed: 11/09/2022]
Abstract
Cancer stem cells (CSCs) are thought to be responsible for cancer progression and therapeutic resistance but identification of this subpopulation requires selective markers. Fortunately, side population (SP) cells analysis brings a novel method to CSCs study. In this study, we identified SP cells, which are demonstrated rich in CSCs, in four nasopharyngeal carcinoma (NPC) cell lines. We investigated SP cells from HK-1 NPC cell line and showed CSCs characteristics in this subpopulation. SP cells displayed greater proliferation and invasion and expressed high levels of CSCs markers than NSP cells. Furthermore, our microRNA microarray analysis of SP versus NSP cells revealed that CD38-related miRNAs were down-regulated in SP cell, but the mRNA and protein level of CD38 were highly expressed in SP cells. We further searched for molecules interacting with CD38 and identified ZAP70, which was also well expressed in SP cells at both mRNA and protein levels. Our results uncover a CD38 pathway that may regulate the proliferation and migration of SP cells from HK-1 NPC cell line.
Collapse
Affiliation(s)
- Danwei Zheng
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Shan Liao
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Guangchao Zhu
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Gengqiu Luo
- Department of Pathology, Basic School of Medicine, Central South University, Changsha, Hunan Province, P. R. China
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Junyu He
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Zhen Pei
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Guiyuan Li
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Yanhong Zhou
- Human Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P. R. China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P. R. China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| |
Collapse
|
38
|
Abstract
Cancers are composed of heterogeneous combinations of cells that exhibit distinct phenotypic characteristics and proliferative potentials. Because most cancers have a clonal origin, cancer stem cells (CSCs) must generate phenotypically diverse progenies including mature CSCs that can self-renew indefinitely and differentiated cancer cells that possess limited proliferative potential. However, no convincing evidence exists to suggest that only single CSCs are representative of patients' tumors. To investigate the CSCs' diversity, we established 4 subclones from a glioblastoma patient. These subclones were subsequently propagated and analyzed. The morphology, the self-renewal and proliferative capacities of the subclones differed. Fluorescence-activated cell sorting and cDNA-microarray analyses revealed that each subclone was composed of distinct populations of cells. Moreover, the sensitivities of the subclones to an inhibitor of epidermal growth factor receptor were dissimilar. In a mouse model featuring xenografts of the subclones, the progression and invasion of tumors and animal survival were also different. Here, we present clear evidence that a brain tumor contains heterogeneous subclones that exhibit dissimilar morphologies and self-renewal and proliferative capacities. Our results suggest that single cell-derived subclones from a patient can produce phenotypically heterogeneous self-renewing progenies in both in vitro and in vivo settings.
Collapse
|
39
|
Wee S, Niklasson M, Marinescu VD, Segerman A, Schmidt L, Hermansson A, Dirks P, Forsberg-Nilsson K, Westermark B, Uhrbom L, Linnarsson S, Nelander S, Andäng M. Selective calcium sensitivity in immature glioma cancer stem cells. PLoS One 2014; 9:e115698. [PMID: 25531110 PMCID: PMC4274094 DOI: 10.1371/journal.pone.0115698] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/26/2014] [Indexed: 12/22/2022] Open
Abstract
Tumor-initiating cells are a subpopulation in aggressive cancers that exhibit traits shared with stem cells, including the ability to self-renew and differentiate, commonly referred to as stemness. In addition, such cells are resistant to chemo- and radiation therapy posing a therapeutic challenge. To uncover stemness-associated functions in glioma-initiating cells (GICs), transcriptome profiles were compared to neural stem cells (NSCs) and gene ontology analysis identified an enrichment of Ca2+ signaling genes in NSCs and the more stem-like (NSC-proximal) GICs. Functional analysis in a set of different GIC lines regarding sensitivity to disturbed homeostasis using A23187 and Thapsigargin, revealed that NSC-proximal GICs were more sensitive, corroborating the transcriptome data. Furthermore, Ca2+ drug sensitivity was reduced in GICs after differentiation, with most potent effect in the NSC-proximal GIC, supporting a stemness-associated Ca2+ sensitivity. NSCs and the NSC-proximal GIC line expressed a larger number of ion channels permeable to potassium, sodium and Ca2+. Conversely, a higher number of and higher expression levels of Ca2+ binding genes that may buffer Ca2+, were expressed in NSC-distal GICs. In particular, expression of the AMPA glutamate receptor subunit GRIA1, was found to associate with Ca2+ sensitive NSC-proximal GICs, and decreased as GICs differentiated along with reduced Ca2+ drug sensitivity. The correlation between high expression of Ca2+ channels (such as GRIA1) and sensitivity to Ca2+ drugs was confirmed in an additional nine novel GIC lines. Calcium drug sensitivity also correlated with expression of the NSC markers nestin (NES) and FABP7 (BLBP, brain lipid-binding protein) in this extended analysis. In summary, NSC-associated NES+/FABP7+/GRIA1+ GICs were selectively sensitive to disturbances in Ca2+ homeostasis, providing a potential target mechanism for eradication of an immature population of malignant cells.
Collapse
Affiliation(s)
- Shimei Wee
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Maria Niklasson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Voichita Dana Marinescu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Segerman
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Linnéa Schmidt
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Annika Hermansson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Peter Dirks
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Bengt Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Lene Uhrbom
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sven Nelander
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Michael Andäng
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- * E-mail:
| |
Collapse
|
40
|
Baxter PA, Lin Q, Mao H, Kogiso M, Zhao X, Liu Z, Huang Y, Voicu H, Gurusiddappa S, Su JM, Adesina AM, Perlaky L, Dauser RC, Leung HCE, Muraszko KM, Heth JA, Fan X, Lau CC, Man TK, Chintagumpala M, Li XN. Silencing BMI1 eliminates tumor formation of pediatric glioma CD133+ cells not by affecting known targets but by down-regulating a novel set of core genes. Acta Neuropathol Commun 2014; 2:160. [PMID: 25526772 PMCID: PMC4289398 DOI: 10.1186/s40478-014-0160-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 10/30/2014] [Indexed: 01/08/2023] Open
Abstract
Clinical outcome of children with malignant glioma remains dismal. Here, we examined the role of over-expressed BMI1, a regulator of stem cell self-renewal, in sustaining tumor formation in pediatric glioma stem cells. Our investigation revealed BMI1 over-expression in 29 of 54 (53.7%) pediatric gliomas, 8 of 8 (100%) patient derived orthotopic xenograft (PDOX) mouse models, and in both CD133+ and CD133− glioma cells. We demonstrated that lentiviral-shRNA mediated silencing of suppressed cell proliferation in vitro in cells derived from 3 independent PDOX models and eliminated tumor-forming capacity of CD133+ and CD133− cells derived from 2 PDOX models in mouse brains. Gene expression profiling showed that most of the molecular targets of BMI1 ablation in CD133+ cells were different from that in CD133- cells. Importantly, we found that silencing BMI1 in CD133+ cells derived from 3 PDOX models did not affect most of the known genes previously associated with the activated BMI1, but modulated a novel set of core genes, including RPS6KA2, ALDH3A2, FMFB, DTL, API5, EIF4G2, KIF5c, LOC650152, C20ORF121, LOC203547, LOC653308, and LOC642489, to mediate the elimination of tumor formation. In summary, we identified the over-expressed BMI1 as a promising therapeutic target for glioma stem cells, and suggest that the signaling pathways associated with activated BMI1 in promoting tumor growth may be different from those induced by silencing BMI1 in blocking tumor formation. These findings highlighted the importance of careful re-analysis of the affected genes following the inhibition of abnormally activated oncogenic pathways to identify determinants that can potentially predict therapeutic efficacy.
Collapse
|
41
|
Shi QY, Zhang SJ, Liu L, Chen QS, Yu LN, Zhang FJ, Yan M. Sevoflurane promotes the expansion of glioma stem cells through activation of hypoxia-inducible factors in vitro. Br J Anaesth 2014; 114:825-30. [PMID: 25492570 DOI: 10.1093/bja/aeu402] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Growing evidences indicate that inhalational anaesthetics can enhance the growth and malignant potential of tumour cells and may affect tumour recurrence after surgery. Tumour stem cells play a vital role in tumour recurrence. This study investigates the effect of sevoflurane on glioma stem cells (GSCs) in vitro and the underlying molecular mechanisms in this process. METHODS Cultured GSCs were exposed to clinically relevant concentrations and durations of sevoflurane exposure. Cell proliferation and self-renewal capacity were determined. Expression of the stem cell marker CD133, vascular endothelial growth factor (VEGF), hypoxia-inducible factors (HIFs), and phosphorylated Akt, which is a protein kinase invoved in multiple cellular processes, were measured using western blotting. Small interfering RNAs and an Akt inhibitor were used to investigate specific pathways. RESULTS Compared with controls, cells exposed to 2% sevoflurane for 6 h induced a larger number of proliferated cells (31.2±7.6% vs 19.0±5.8%; P<0.01). Levels of CD133, VEGF, HIF-1α, HIF-2α, and p-Akt were up-regulated by sevoflurane in a time- and concentration-dependent manner. Small interfering RNA against HIFs decreased the percentage of proliferating GSCs after sevoflurane exposure and pre-treatment of cells with an Akt inhibitor abrogated the expression of HIFs induced by sevoflurane. CONCLUSIONS Sevoflurane can promote the expansion of human GSCs through HIFs in vitro. Inhaled anaesthetics may enhance tumour growth through tumour stem cells.
Collapse
Affiliation(s)
- Q Y Shi
- Department of Anaesthesiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - S J Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - L Liu
- Department of Anaesthesiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Q S Chen
- Department of Anaesthesiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - L N Yu
- Department of Anaesthesiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - F J Zhang
- Department of Anaesthesiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - M Yan
- Department of Anaesthesiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| |
Collapse
|
42
|
Angiopep-2-conjugated liposomes encapsulating γ-secretase inhibitor for targeting glioblastoma stem cells. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2014. [DOI: 10.1007/s40005-014-0151-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Zhu J, Wang H, Fan Y, Hu Y, Ji X, Sun Q, Liu H. Knockdown of nuclear factor erythroid 2-related factor 2 by lentivirus induces differentiation of glioma stem-like cells. Oncol Rep 2014; 32:1170-8. [PMID: 25017049 DOI: 10.3892/or.2014.3320] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/06/2014] [Indexed: 11/06/2022] Open
Abstract
Glioma stem cells (GSCs) are key in the progression and recurrence of glioblastoma. Inducing the differentiation of GSCs is an important therapeutic target for glioblastoma. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been reported to be important in maintaining the stem cell status of GSCs; however, its association with differentiation has not been studied. Herein, we knocked down Nrf2 from GSCs to investigate the role of Nrf2 in the differentiation of GSCs. First, Nrf2 expression was observed at different stages of differentiation; then, Nrf2 was knocked down and the association of Nrf2 with differentiation degree was observed in vitro. Finally, GSCs were planted in nude mice to study the association of Nrf2 with differentiation in vivo. The expression of Nrf2 decreased with the differentiation process. Following Nrf2 knockdown, the proportion of sphere-like colonies decreased and the dendritic cells in spheres increased; the expression of Nrf2 significantly decreased while the expression of differentiation marker glial fibrillary acidic protein (GFAP) and βIII-tubulin increased both at the protein and the gene level. In the xenografts of nude mice, the differentiation of tumor cells was improved. These results suggest that Nrf2 is a key factor inhibiting the differentiation of GSCs, and knockdown of Nrf2 may promote the differentiation process, providing a therapy target for GSCs.
Collapse
Affiliation(s)
- Jianhong Zhu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Youwu Fan
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yangchun Hu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiangjun Ji
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qing Sun
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Huandong Liu
- Department of Neurosurgery, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
44
|
Transcription factors FOXG1 and Groucho/TLE promote glioblastoma growth. Nat Commun 2014; 4:2956. [PMID: 24356439 PMCID: PMC3984242 DOI: 10.1038/ncomms3956] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/18/2013] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain cancer, with a median survival of less than two years. GBM displays a cellular complexity that includes brain tumour-initiating cells (BTICs), which are considered as potential key targets for GBM therapies. Here we show that the transcription factors FOXG1 and Groucho/TLE are expressed in poorly differentiated astroglial cells in human GBM specimens and in primary cultures of GBM-derived BTICs, where they form a complex. FOXG1 knockdown in BTICs causes downregulation of neural stem/progenitor and proliferation markers, increased replicative senescence, upregulation of astroglial differentiation genes, and decreased BTIC-initiated tumour growth upon intracranial transplantation into host mice. These effects are phenocopied by Groucho/TLE knockdown or dominant-inhibition of the FOXG1:Groucho/TLE complex. These results provide evidence that transcriptional programs regulated by FOXG1 and Groucho/TLE are important for BTIC-initiated brain tumour growth, implicating FOXG1 and Groucho/TLE in GBM tumorigenesis.
Collapse
|
45
|
Bjørnbak C, Brøchner CB, Larsen LA, Johansen JS, Møllgård K. Brain barriers and a subpopulation of astroglial progenitors of developing human forebrain are immunostained for the glycoprotein YKL-40. J Histochem Cytochem 2014; 62:369-88. [PMID: 24595665 DOI: 10.1369/0022155414528514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
YKL-40, a glycoprotein involved in cell differentiation, has been associated with neurodevelopmental disorders, angiogenesis, neuroinflammation and glioblastomas. We evaluated YKL-40 protein distribution in the early human forebrain using double-labeling immunofluorescence and immunohistochemistry. Immunoreactivity was detected in neuroepithelial cells, radial glial end feet, leptomeningeal cells and choroid plexus epithelial cells. The subpial marginal zone was YKL-40-positive, particularly in the hippocampus, from an early beginning stage in its development. Blood vessels in the intermediate and subventricular zones showed specific YKL-40 reactivity confined to pericytes. Furthermore, a population of YKL-40-positive, small, rounded cells was identified in the ventricular and subventricular zones. Real-time quantitative RT-PCR analysis showed strong YKL-40 mRNA expression in the leptomeninges and the choroid plexuses, and weaker expression in the telencephalic wall. Immunohistochemistry revealed a differential distribution of YKL-40 across the zones of the developing telencephalic wall. We show that YKL-40 is associated with sites of the brain barrier systems and propose that it is involved in controlling local angiogenesis and access of peripheral cells to the forebrain via secretion from leptomeningeal cells, choroid plexus epithelium and pericytes. Furthermore, we suggest that the small, rounded, YKL-40-positive cells represent a subpopulation of astroglial progenitors, and that YKL-40 could be involved in the differentiation of a particular astrocytic lineage.
Collapse
Affiliation(s)
- Camilla Bjørnbak
- Department of Cellular and Molecular Medicine (CB,CBB,LAL,KM), Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
46
|
Mak AB, Schnegg C, Lai CY, Ghosh S, Yang MH, Moffat J, Hsu MY. CD133-targeted niche-dependent therapy in cancer: a multipronged approach. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1256-62. [PMID: 24589338 DOI: 10.1016/j.ajpath.2014.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/05/2014] [Accepted: 01/16/2014] [Indexed: 02/07/2023]
Abstract
Cancer treatment continues to be challenged by the development of therapeutic resistances and relapses in the clinical setting, which are largely attributed to tumor heterogeneity, particularly the existence of cancer stem cells (CSCs). Thus, targeting the CSC subpopulation may represent an effective therapeutic strategy. However, despite advances in identifying and characterizing CD133(+) CSCs in various human cancers, efforts to translate these experimental findings to clinical modalities have been slow in the making, especially in light of the growing awareness of CSC plasticity and the foreseeable pitfall of therapeutically targeting CSC base sorely on a surface marker. We, and others, have demonstrated that the CD133(+) CSCs reside in complex vascular niches, where reciprocal signaling between the CD133(+) CSCs and their microenvironment may govern niche morphogenesis and homeostasis. Herein, we discuss the multifaceted functional role of the CD133(+) cells in the context of their niche, and the potential of targeting CD133 as a niche-dependent approach in effective therapy.
Collapse
Affiliation(s)
- Anthony B Mak
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts; Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Caroline Schnegg
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts
| | - Chiou-Yan Lai
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Subrata Ghosh
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts
| | - Moon Hee Yang
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts
| | - Jason Moffat
- Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Mei-Yu Hsu
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts; Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| |
Collapse
|
47
|
Happold C, Roth P, Silginer M, Florea AM, Lamszus K, Frei K, Deenen R, Reifenberger G, Weller M. Interferon-β Induces Loss of Spherogenicity and Overcomes Therapy Resistance of Glioblastoma Stem Cells. Mol Cancer Ther 2014; 13:948-61. [DOI: 10.1158/1535-7163.mct-13-0772] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Lasorella A, Benezra R, Iavarone A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat Rev Cancer 2014; 14:77-91. [PMID: 24442143 DOI: 10.1038/nrc3638] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inhibitor of DNA binding (ID) proteins are transcriptional regulators that control the timing of cell fate determination and differentiation in stem and progenitor cells during normal development and adult life. ID genes are frequently deregulated in many types of human neoplasms, and they endow cancer cells with biological features that are hijacked from normal stem cells. The ability of ID proteins to function as central 'hubs' for the coordination of multiple cancer hallmarks has established these transcriptional regulators as therapeutic targets and biomarkers in specific types of human tumours.
Collapse
Affiliation(s)
- Anna Lasorella
- Institute for Cancer Genetics, Department of Pathology and Pediatrics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 241, New York, 10065 New York, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Pathology and Neurology, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| |
Collapse
|
49
|
Shin DH, Xuan S, Kim WY, Bae GU, Kim JS. CD133 antibody-conjugated immunoliposomes encapsulating gemcitabine for targeting glioblastoma stem cells. J Mater Chem B 2014; 2:3771-3781. [DOI: 10.1039/c4tb00185k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
50
|
Wolpert F, Tritschler I, Steinle A, Weller M, Eisele G. A disintegrin and metalloproteinases 10 and 17 modulate the immunogenicity of glioblastoma-initiating cells. Neuro Oncol 2013; 16:382-91. [PMID: 24327582 DOI: 10.1093/neuonc/not232] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There are emerging reports that the family of a disintegrin and metalloproteinases (ADAM) are involved in the maintenance of the malignant phenotype of glioblastomas. Notably, ADAM proteases 10 and 17 might impair the immune recognition of glioma cells via the activating immunoreceptor NKG2D by cleavage of its ligands from the cell surface. Glioblastoma-initiating cells (GIC) with stem cell properties have been identified as an attractive target for immunotherapy. However, GIC immunogenicity seems to be low. METHODS AND RESULTS Here,we show that ADAM10 and ADAM17 are expressed on the cell surface of GIC and contribute to an immunosuppressive phenotype by cleavage of ULBP2. The cell surface expression of ULBP2 is enhanced upon blocking ADAM10 and ADAM17, and treatment with ADAM10 and ADAM17specific inhibitors leads to enhanced immunerecognition of GIC by natural killer cells. CONCLUSIONS Therefore, ADAM10 and ADAM17 constitute suitable targets to boost an immune response against GIC.
Collapse
Affiliation(s)
- Fabian Wolpert
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland (F.W., I.T., M.W., G.E.); Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany (A.S.)
| | | | | | | | | |
Collapse
|