1
|
Xu S, Luo C, Chen D, Tang L, Cheng Q, Chen L, Liu Z. circMMD reduction following tumor treating fields inhibits glioblastoma progression through FUBP1/FIR/DVL1 and miR-15b-5p/FZD6 signaling. J Exp Clin Cancer Res 2023; 42:64. [PMID: 36932454 PMCID: PMC10021944 DOI: 10.1186/s13046-023-02642-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Tumor treating fields (TTF) is the latest treatment for GBM. Circular RNA (circRNA) has been demonstrated to play critical roles in tumorigenesis. However, the molecular mechanism of TTF remained largely unknown and the role of circRNA in TTF was not reported. The aim of this study was to elucidate the role and mechanism of circMMD in TTF treatment of GBM. METHODS Divergent primer was designed to verify the existence of circMMD in GBM cells. The prognostic role of circMMD was explored in glioma specimens. The knockdown and overexpressed plasmids were used to evaluate the effect of circMMD on GBM cell proliferation and TTF efficacy. RNA pull-down and RNA immunoprecipitation were performed to identify binding proteins of circMMD. Subcutaneous and intracranial tumor models were established to validate findings in vivo. RESULTS The expression of circMMD was elevated in GBM and its high expression indicated poor prognoses. TTF intervention could reduce circMMD synthesis, which suppressed GBM proliferation and increased TTF-mediated apoptosis. The reduction of circMMD promoted the interaction between FUBP1 and FIR, which decreased DVL1 transcription. Meanwhile, decreased circMMD would promote the activity of miR-15b-5p to degrade FZD6. Finally, the diminished expression of DVL1 and FZD6 expression suppressed the activation of Wnt/β-catenin pathway. CONCLUSIONS Our study revealed a novel mechanism of TTF that TTF-mediated reduction of circMMD could inhibit Wnt/β-catenin pathway to suppress GBM proliferation.
Collapse
Affiliation(s)
- Shengchao Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chengke Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, 410008, China
| | - Lu Tang
- Department of Anesthesiology, Xiangya Hospital, Central South University, ChangshaHunan, 410008, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ling Chen
- Department of Neurosurgery, Chinese People's Liberation Army of China (PLA) General Hospital, Medical School of Chinese PLA, Institute of Neurosurgery of Chinese PLA, Beijing, 100853, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, No.87, Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
2
|
Seervai RNH, Sinha A, Kulkarni RP. Mechanisms of dermatologic toxicities to immune checkpoint inhibitor cancer therapies. Clin Exp Dermatol 2022; 47:1928-1942. [PMID: 35844072 DOI: 10.1111/ced.15332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
Abstract
The discovery of immune checkpoint inhibition (ICI) sparked a revolution in the era of targeted anticancer therapy. While monoclonal antibodies targeting the CTLA-4 and PD-1 axes have improved survival in patients with advanced cancers, these immunotherapies are associated with a wide spectrum of dermatologic immune-related adverse events (irAEs). Several publications have addressed the clinical and histopathologic classification of these skin-directed irAEs, their impact on antitumor immunity and survival, and the critical role of supportive oncologic dermatology in their management. Here, we review the current understanding of the mechanistic drivers of immune-related skin toxicities with a focus on inflammatory, immunobullous, melanocyte/pigment-related reactions. We detail the specific immune-based mechanisms that may underlie different cutaneous reactions. We also discuss potential mechanisms as they relate to non-cutaneous irAEs and potential overlap with cutaneous irAEs, techniques to study differences in immune-related versus de novo skin reactions, and how treatment of these adverse events impacts cancer treatment, patient quality of life, and overall survival. An improved understanding of the mechanistic basis of cutaneous irAEs will allow us to develop and utilize blood-based biomarkers that could help ultimately predict onset and/or severity of these irAEs and to implement rational mechanistic-based treatment strategies that are targeted to the irAEs while potentially avoiding abrogating anti-tumor effect of ICIs.
Collapse
Affiliation(s)
- Riyad N H Seervai
- Department of Internal Medicine, Providence Portland Medical Center, Portland, Oregon, 97213.,Medical Scientist Training Program, Baylor College of Medicine, 77030, Houston, Texas, USA.,Department of Dermatology, Baylor College of Medicine, 77030, Houston, Texas, USA
| | - Avilasha Sinha
- Department of Dermatology, Baylor College of Medicine, 77030, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, 77030, Houston, Texas, USA
| | - Rajan P Kulkarni
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon 97239, USA.,Department of Biomedical Engineering, Oregon Health and Science University, 97239, Portland, OR.,Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, 97239, Portland, OR.,Operative Care Division, VA Portland Health Care System, 92739, Portland, OR
| |
Collapse
|
3
|
Liang F, Zhu L, Wang C, Yang Y, He Z. BSA-MnO 2-SAL multifunctional nanoparticle-mediated M 1 macrophages polarization for glioblastoma therapy. RSC Adv 2021; 11:35331-35341. [PMID: 35493189 PMCID: PMC9043005 DOI: 10.1039/d1ra06705b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/24/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is a type of brain tumour with a very high fatality rate. Owing to the presence of the blood-brain barrier (BBB), it is difficult for drugs to reach the tumour site; thus, there has been little progress in GBM chemotherapeutics. Furthermore, the malignant growth of tumours largely depends on the tumour microenvironment. GBM is especially prevalent in slightly acidic, hydrogen peroxide (H2O2)-rich, hypoxic, and immunosuppressive microenvironments. Tumour-supporting macrophages (M2 macrophages) are a type of immune cell that promote tumour growth. Therefore, targeting M2 macrophages and repolarizing them into tumour-suppressor macrophages (M1 macrophages) are important strategies for GBM treatment. Salinomycin (SAL) is an anti-tumour drug that can improve the tumour immune microenvironment. Interestingly, we found that SAL promoted the expression of M1 macrophages in vitro, but its ability was limited in vivo because of the presence of the BBB. In this study, we combined SAL and MnO2 to design bovine serum albumin-MnO2-SAL (BMS), a nanoparticle that responds to acidic and H2O2-rich microenvironments. Our experimental results showed that BMS reduced GBM growth efficiency and had the ability to penetrate the BBB. It also enhanced the repolarization ability of SAL owing to the production of Mn2+ after decomposition, which could be applied in Magnetic Resonance Imaging (MRI). This study demonstrated that the multifunctional nanoparticle BMS is of great significance in inhibiting orthotopic GBM growth and improving immunosuppressive microenvironments.
Collapse
Affiliation(s)
- Fuming Liang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University 1 Friendship Road 400016 Chongqing China .,CAS Key Laborytory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology 100190 Beijing China
| | - Ling Zhu
- CAS Key Laborytory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology 100190 Beijing China
| | - Chen Wang
- CAS Key Laborytory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology 100190 Beijing China
| | - Yanlian Yang
- CAS Key Laborytory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology 100190 Beijing China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University 1 Friendship Road 400016 Chongqing China
| |
Collapse
|