1
|
Li H, Ouyang J, Wang X, Qian C. Platycodin D Enhances Glioma Sensitivity to Temozolomide by Inhibition of the Wnt/β-Catenin Pathway. Drug Des Devel Ther 2025; 19:1811-1824. [PMID: 40093646 PMCID: PMC11910054 DOI: 10.2147/dddt.s503167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background Temozolomide (TMZ) is a first-line chemotherapeutic agent for gliomas. However, its efficacy is limited by drug resistance. Platycodin D (PD) exhibits notable anti-glioma activity The objective of this study was to investigate the potential of PD to augment glioma sensitivity to TMZ and the underlying mechanisms. Methods Cell viability and proliferation were assessed using CCK-8 and clonogenic assays, respectively, while flow cytometry was used to detect apoptosis. Cell migration and invasion were assessed using Transwell assays. Western blotting and immunohistochemistry analyses were performed to determine protein expression levels. A xenograft glioma model was established to investigate the in vivo effects of PD. Results PD augmented glioma cell sensitivity to TMZ, as evidenced by heightened inhibition of cell growth, colony formation, migration, and invasion, accompanied by elevated apoptosis. Treatment with PD or a combination of PD and TMZ robustly suppressed the expression of active β-catenin and c-Myc, which was reversed by the β-catenin activator, SKL2001. In vivo experiments demonstrated that PD amplified the anti-glioma efficacy of TMZ, resulting in diminished Ki67 expression and substantially reduced expression of active β-catenin and c-Myc in the tumor tissue. Conclusion PD augmented glioma cell sensitivity to TMZ by modulating Wnt/β-catenin pathway. Our findings demonstrate the potential of PD as an innovative therapeutic agent to enhance glioma treatment, especially in TMZ-resistant gliomas.
Collapse
MESH Headings
- Temozolomide/pharmacology
- Glioma/drug therapy
- Glioma/pathology
- Glioma/metabolism
- Humans
- Wnt Signaling Pathway/drug effects
- Cell Proliferation/drug effects
- Saponins/pharmacology
- Animals
- Mice
- Triterpenes/pharmacology
- Apoptosis/drug effects
- Cell Survival/drug effects
- Drug Screening Assays, Antitumor
- beta Catenin/metabolism
- beta Catenin/antagonists & inhibitors
- Dose-Response Relationship, Drug
- Mice, Nude
- Antineoplastic Agents, Alkylating/pharmacology
- Tumor Cells, Cultured
- Structure-Activity Relationship
- Mice, Inbred BALB C
- Brain Neoplasms/drug therapy
- Brain Neoplasms/pathology
- Brain Neoplasms/metabolism
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Cell Movement/drug effects
- Cell Line, Tumor
Collapse
Affiliation(s)
- Haima Li
- Department of Neurosurgery, The Nuclear Industry 215 hospital of Shaanxi Province, Xianyang, Shaanxi, 712000, People’s Republic of China
| | - Jia Ouyang
- Department of Neurosurgery, Peking University People’s Hospital, Beijing, 100044, People’s Republic of China
| | - Xuelian Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, 710038, People’s Republic of China
| | - Chao Qian
- Department of Neurosurgery, The Nuclear Industry 215 hospital of Shaanxi Province, Xianyang, Shaanxi, 712000, People’s Republic of China
| |
Collapse
|
2
|
Tang ZQ, Ye YR, Shen Y. Molecular Mechanisms and Strategies for Inducing Neuronal Differentiation in Glioblastoma Cells. Cell Reprogram 2025; 27:24-32. [PMID: 39880036 DOI: 10.1089/cell.2024.0087] [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] [Indexed: 01/31/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a highly invasive brain tumor, and traditional treatments combining surgery with radiochemotherapy have limited effects, with tumor recurrence being almost inevitable. Given the lack of proliferative capacity in neurons, inducing terminal differentiation of GBM cells or glioma stem cells (GSCs) into neuron-like cells has emerged as a promising strategy. This approach aims to suppress their proliferation and self-renewal capabilities through differentiation. This review summarizes the methods involved in recent research on the neuronal differentiation of GBM cells or GSCs, including the regulation of transcription factors, signaling pathways, miRNA, and the use of small molecule drugs, among various strategies. It also outlines the interconnections between the mechanisms studied, hoping to provide ideas for exploring new therapeutic avenues for GBM and the development of differentiation-inducing drugs for GBM.
Collapse
Affiliation(s)
- Zhao-Qi Tang
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, China
| | - Yan-Rong Ye
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shen
- Department of Pharmacy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Deng X, Zhang Q, Jin F, Lu F, Duan G, Han L, Zhu M, Yang Z, Zhang G. Ailanthone disturbs cross-talk between cancer cells and tumor-associated macrophages via HIF1-α/LINC01956/FUS/β-catenin signaling pathway in glioblastoma. Cancer Cell Int 2024; 24:397. [PMID: 39639311 PMCID: PMC11619249 DOI: 10.1186/s12935-024-03594-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND An increasing number of studies have focused on ailanthone (aila) due to its antitumor activity. However, the role of ailanthone in glioblastoma(GBM) has not been investigated before. This study aims to explore the biological function and the underlying mechanism of ailanthone in GBM. METHODS The microarray analysis was used to screen out down-stream long non-coding RNAs (lncRNAs) targeted by ailanthone. Real-time PCR(RT-PCR) assay was used to examine LINC01956 expression levels. Colony-formation, Methylthiazolyldiphenyl-tetrazolium bromide(MTT), cell-cycle, organoids culture and in-vivo tumorigenesis assays were used to examine cell growth in vitro and in vivo. Boyden assay was used to examine cell invasion ability in vitro. RNA immunoprecipitation and RNA-protein pull-down assays were used to examine the interaction between LINC01956 and FUS protein. Chromatin Immunoprecipitation(ChIP) assay was used to examine HIF1-α-binding sites in the LINC01956 promoter. RESULTS Ailanthone decreased GBM cell growth in vitro and in vivo via inducing ferroptosis. Ailanthone treatment exhibited blood‒brain barrier(BBB) permeability and specifically targeted the tumor area. LINC01956 was identified as a down-stream target of Ailanthone. LINC01956 exerted as an onco-lncRNA in GBM. M2 polarization of macrophages induced by exosomes derived from glioma cells overexpressing LINC01956 accelerated GBM progression. Mechanistically, we found that LINC01956 bound to FUS and reduced its ubiquitination. LINC01956 evoked nuclear translocation of phosphorylated (p)-β-catenin by recruiting FUS. Furthermore, under hypoxic conditions, LINC01956 was regulated by HIF-1α. Ailanthone decreased the expression of LINC01956 via suppressing HIF-1α. CONCLUSION Taken together, our data revealed for the first time that ailanthone regulated HIF-1α/LINC01956/FUS/β-catenin signaling pathway and thereby inhibited GBM progression.
Collapse
Affiliation(s)
- Xubin Deng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fa Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fengfei Lu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guosheng Duan
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Luwei Han
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Meiling Zhu
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Zhengyan Yang
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Gong Zhang
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China.
| |
Collapse
|
4
|
Li M, Lu L, Xiao Q, Maalim AA, Nie B, Liu Y, Kahlert UD, Shu K, Lei T, Zhu M. Bioengineer mesenchymal stem cell for treatment of glioma by IL-12 mediated microenvironment reprogramming and nCD47-SLAMF7 mediated phagocytosis regulation of macrophages. EXPLORATION (BEIJING, CHINA) 2024; 4:20240027. [PMID: 39713206 PMCID: PMC11657999 DOI: 10.1002/exp.20240027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/07/2024] [Indexed: 12/24/2024]
Abstract
High expression of cellular self-activated immunosuppressive molecules and extensive infiltration of suppressive immune cells in the tumor microenvironment are the main factors contributing to glioma's resistance to immunotherapy. Nonetheless, technology to modify the expression of glioma cellular self-molecules through gene editing requires further development. This project advances cell therapy strategies to reverse the immunosuppressive microenvironment of glioma (TIME). Bone marrow-derived mesenchymal stem cells (MSCs) are engineered to express bioactive proteins and demonstrate tumor-homing characteristics upon activation by TGF-β. These MSCs are designed to secrete the anti-tumor immune cytokine IL-12 and the nCD47-SLAMF7 fusion protein, which regulates T-cell activity and macrophage phagocytosis. The engineered MSCs are then injected in situ into the glioma site, circumventing the blood-brain barrier to deliver high local concentrations of bioactive proteins. This approach aims to enhance the M1 polarization of infiltrating macrophages, stimulate macrophage-mediated tumor cell phagocytosis, activate antigen-presenting cells, and promote effector CD8+ T cell infiltration, effectively controlling glioma. Additionally, the engineered MSCs may serve as a universal treatment for other tumors that express TGF-β at high levels. This study proposes a novel treatment strategy for the clinical management of glioma patients.
Collapse
Affiliation(s)
- Man Li
- Department of Anesthesiology and Pain MedicineHubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Healthand Wuhan Clinical Research Center for Geriatric AnesthesiaTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Lisen Lu
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Qungen Xiao
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Ali Abdi Maalim
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Bin Nie
- Department of Anesthesiology and Pain MedicineHubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Healthand Wuhan Clinical Research Center for Geriatric AnesthesiaTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Yanchao Liu
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Ulf D. Kahlert
- Molecular and Experimental SurgeryClinic for General‐, Visceral‐, Vascular and Transplant SurgeryFaculty of Medicine and University Hospital MagdeburgOtto‐von‐Guericke UniversityMagdeburgGermany
| | - Kai Shu
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Ting Lei
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Mingxin Zhu
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| |
Collapse
|
5
|
Tang Q, Ren T, Bai P, Wang X, Zhao L, Zhong R, Sun G. Novel strategies to overcome chemoresistance in human glioblastoma. Biochem Pharmacol 2024; 230:116588. [PMID: 39461382 DOI: 10.1016/j.bcp.2024.116588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Temozolomide (TMZ) is currently the first-line chemotherapeutic agent for the treatment of glioblastoma multiforme (GBM). However, the inherent heterogeneity of GBM often results in suboptimal outcomes, particularly due to varying degrees of resistance to TMZ. Over the past several decades, O6-methylguanine-DNA methyltransferase (MGMT)-mediated DNA repair pathway has been extensively investigated as a target to overcome TMZ resistance. Nonetheless, the combination of small molecule covalent MGMT inhibitors with TMZ and other chemotherapeutic agents has frequently led to adverse clinical effects. Recently, additional mechanisms contributing to TMZ resistance have been identified, including epidermal growth factor receptor (EGFR) mutations, overactivation of intracellular signalling pathways, energy metabolism reprogramming or survival autophagy, and changes in tumor microenvironment (TME). These findings suggest that novel therapeutic strategies targeting these mechanisms hold promise for overcoming TMZ resistance in GBM patients. In this review, we summarize the latest advancements in understanding the mechanisms underlying intrinsic and acquired TMZ resistance. Additionally, we compile various small-molecule compounds with potential to mitigate chemoresistance in GBM. These mechanism-based compounds may enhance the sensitivity of GBM to TMZ and related chemotherapeutic agents, thereby improving overall survival rates in clinical practice.
Collapse
Affiliation(s)
- Qing Tang
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
6
|
Mastrantuono E, Ghibaudi M, Matias D, Battaglia G. The multifaceted therapeutical role of low-density lipoprotein receptor family in high-grade glioma. Mol Oncol 2024; 18:2966-2976. [PMID: 39276062 PMCID: PMC11619799 DOI: 10.1002/1878-0261.13730] [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: 02/01/2024] [Revised: 07/25/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
The diverse roles of the low-density lipoprotein receptor family (LDLR) have been associated with many processes critical to maintaining central nervous system (CNS) health and contributing to neurological diseases or cancer. In this review, we provide a comprehensive understanding of the LDLR's involvement in common brain tumors, specifically high-grade gliomas, emphasizing the receptors' critical role in the pathophysiology and progression of these tumors due to LDLR's high expression. We delve into LDLR's role in regulating cellular uptake and transport through the brain barrier. Additionally, we highlight LDLR's role in activating several signaling pathways related to tumor proliferation, migration, and invasion, engaging readers with an in-depth understanding of the molecular mechanisms at play. By synthesizing current research findings, this review underscores the significance of LDLR during tumorigenesis and explores its potential as a therapeutic target for high-grade gliomas. The collective insights presented here contribute to a deeper appreciation of LDLR's multifaceted roles and implications for physiological and pathological states, opening new avenues for tumor treatment.
Collapse
Affiliation(s)
- Elisa Mastrantuono
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de MedicinaUniversidade de LisboaPortugal
- Institute for Bioengineering of CataloniaBarcelona Institute of Science and TechnologySpain
| | - Matilde Ghibaudi
- Institute for Bioengineering of CataloniaBarcelona Institute of Science and TechnologySpain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER‐BBN)MadridSpain
| | - Diana Matias
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de MedicinaUniversidade de LisboaPortugal
| | - Giuseppe Battaglia
- Institute for Bioengineering of CataloniaBarcelona Institute of Science and TechnologySpain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER‐BBN)MadridSpain
- Catalan Institution for Research and Advanced StudiesPasseig de Lluís CompanysBarcelonaSpain
| |
Collapse
|
7
|
Hedayati N, Mafi A, Farahani A, Hashemi M, Nabavi N, Alimohammadi M, Rahimzadeh P, Taheriazam A, Farahani N. The importance of the circRNA/Wnt axis in gliomas: Biological functions and clinical opportunities. Pathol Res Pract 2024; 261:155510. [PMID: 39116573 DOI: 10.1016/j.prp.2024.155510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Gliomas are among the most common cancers in the central nervous system, arising through various signaling pathways. One significant pathway is Wnt signaling, a tightly regulated process that plays a crucial role in gliomagenesis and development. The current study aims to explore the relationship between circular RNAs (circRNAs) and the Wnt/β-catenin signaling pathway in gliomas, considering the growing recognition of circRNAs in disease pathogenesis. A comprehensive review of recent research was conducted to investigate the roles of circRNAs in gliomas, focusing on their expression patterns and interactions with the Wnt signaling pathway. The analysis included studies examining circRNAs' function as microRNA sponges and their impact on glioma biology. The findings reveal that circRNAs are differentially expressed in gliomas and significantly influence the occurrence, growth, and metastasis of these tumors. Specifically, circRNAs interact with the Wnt signaling pathway, affecting glioma development and progression. This interaction highlights the importance of circRNAs in glioma pathophysiology. Understanding the regulatory network involving circRNAs and Wnt signaling offers valuable insights into glioma pathophysiology. CircRNAs hold promise as diagnostic and prognostic biomarkers and may serve as targets for novel therapeutic strategies in glioma treatment.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aryan Farahani
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
8
|
Vercouillie N, Ren Z, Terras E, Lammens T. Long Non-Coding RNAs in Neuroblastoma: Pathogenesis, Biomarkers and Therapeutic Targets. Int J Mol Sci 2024; 25:5690. [PMID: 38891878 PMCID: PMC11171840 DOI: 10.3390/ijms25115690] [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: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Neuroblastoma is the most common malignant extracranial solid tumor of childhood. Recent studies involving the application of advanced high-throughput "omics" techniques have revealed numerous genomic alterations, including aberrant coding-gene transcript levels and dysfunctional pathways, that drive the onset, growth, progression, and treatment resistance of neuroblastoma. Research conducted in the past decade has shown that long non-coding RNAs, once thought to be transcriptomic noise, play key roles in cancer development. With the recent and continuing increase in the amount of evidence for the underlying roles of long non-coding RNAs in neuroblastoma, the potential clinical implications of these RNAs cannot be ignored. In this review, we discuss their biological mechanisms of action in the context of the central driving mechanisms of neuroblastoma, focusing on potential contributions to the diagnosis, prognosis, and treatment of this disease. We also aim to provide a clear, integrated picture of future research opportunities.
Collapse
Affiliation(s)
- Niels Vercouillie
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium; (N.V.); (Z.R.); (E.T.)
| | - Zhiyao Ren
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium; (N.V.); (Z.R.); (E.T.)
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Eva Terras
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium; (N.V.); (Z.R.); (E.T.)
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Tim Lammens
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium; (N.V.); (Z.R.); (E.T.)
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
9
|
Tahmasebi Dehkordi H, Khaledi F, Ghasemi S. Immunological processes of enhancers and suppressors of long non-coding RNAs associated with brain tumors and inflammation. Int Rev Immunol 2024; 43:178-196. [PMID: 37974420 DOI: 10.1080/08830185.2023.2280581] [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: 07/16/2022] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Immunological processes, such as inflammation, can both cause tumor suppression and cancer progression. Moreover, deregulated levels of long non-coding RNA (lncRNA) expression in the brain may cause inflammation and lead to the growth of tumors. Like other biological processes, the immune system's role in cancer is complicated, varies, and can help or hurt the cancer's maintenance. According to research, inflammation and brain cancer are correlated via several signaling pathways. A variety of lncRNAs have recently been revealed to influence cancer by modulating inflammatory pathways. As a result, lncRNAs have the potential to influence carcinogenesis, tumor formation, or tumor suppression via an increase or decrease in inflammation functions. Although the study and targeting of lncRNAs have made great progress in the treatment of cancer, there are definitely limitations and challenges. Using new technologies like nanocarriers and cell-penetrating peptides (CPPs) to target treatments without hurting healthy body tissues has shown to be very effective. In this review article, we have collected significantly related lncRNAs and their inhibitory or stimulating roles in inflammation and brain cancer for the first time. However, there are limitations, such as side effects and damage to normal tissues. With the advancement of new targeting technologies, these lncRNAs may be candidates for the specific targeting therapy of brain cancers by limiting inflammation or stimulating the immune system against them in the future.
Collapse
Affiliation(s)
- Hossein Tahmasebi Dehkordi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Khaledi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
10
|
Pećina-Šlaus N, Hrašćan R. Glioma Stem Cells-Features for New Therapy Design. Cancers (Basel) 2024; 16:1557. [PMID: 38672638 PMCID: PMC11049195 DOI: 10.3390/cancers16081557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
On a molecular level, glioma is very diverse and presents a whole spectrum of specific genetic and epigenetic alterations. The tumors are unfortunately resistant to available therapies and the survival rate is low. The explanation of significant intra- and inter-tumor heterogeneity and the infiltrative capability of gliomas, as well as its resistance to therapy, recurrence and aggressive behavior, lies in a small subset of tumor-initiating cells that behave like stem cells and are known as glioma cancer stem cells (GCSCs). They are responsible for tumor plasticity and are influenced by genetic drivers. Additionally, GCSCs also display greater migratory abilities. A great effort is under way in order to find ways to eliminate or neutralize GCSCs. Many different treatment strategies are currently being explored, including modulation of the tumor microenvironment, posttranscriptional regulation, epigenetic modulation and immunotherapy.
Collapse
Affiliation(s)
- Nives Pećina-Šlaus
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| | - Reno Hrašćan
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia;
| |
Collapse
|
11
|
Lai Y, Wu Y, Chen X, Gu W, Zhou G, Weng M. MRI-based Machine Learning Radiomics Can Predict CSF1R Expression Level and Prognosis in High-grade Gliomas. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:209-229. [PMID: 38343263 PMCID: PMC10976932 DOI: 10.1007/s10278-023-00905-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 03/02/2024]
Abstract
The purpose of this study is to predict the mRNA expression of CSF1R in HGG non-invasively using MRI (magnetic resonance imaging) omics technology and to evaluate the correlation between the established radiomics model and prognosis. We investigated the predictive value of CSF1R in the Cancer Genome Atlas (TCGA) and The Cancer Imaging Archive (TCIA) database. The Support vector machine (SVM) and the Logistic regression (LR) algorithms were used to create a radiomics_score (Rad_score), respectively. The effectiveness and performance of the radiomics model was assessed in the training (n = 89) and tenfold cross-validation sets. We further analyzed the correlation between Rad_score and macrophage-related genes using Spearman correlation analysis. A radiomics nomogram combining the clinical factors and Rad_score was constructed to validate the radiomic signatures for individualized survival estimation and risk stratification. The results showed that CSF1R expression was markedly elevated in HGG tissues, which was related to worse prognosis. CSF1R expression was closely related to the abundance of infiltrating immune cells, such as macrophages. We identified nine features for establishing a radiomics model. The radiomics model predicting CSF1R achieved high AUC in training (0.768 in SVM and 0.792 in LR) and tenfold cross-validation sets (0.706 in SVM and 0.717 in LR). Rad_score was highly associated with tumor-related macrophage genes. A radiomics nomogram combining the Rad_score and clinical factors was constructed and revealed satisfactory performance. MRI-based Rad_score is a novel way to predict CSF1R expression and prognosis in high-grade glioma patients. The radiomics nomogram could optimize individualized survival estimation for HGG patients.
Collapse
Affiliation(s)
- Yuling Lai
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yiyang Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenchao Gu
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Ibaraki, Japan.
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | - Guoxia Zhou
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| | - Meilin Weng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Isa AI. Exploring signaling pathway crosstalk in glioma by mapping miRNA and WNT pathways: A review. Int J Biol Macromol 2024; 257:128722. [PMID: 38092099 DOI: 10.1016/j.ijbiomac.2023.128722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/26/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Glioma is a significant healthcare burden; nevertheless, the particular genetic regulatory mechanism underpinning its onset and progression is still unknown. Recent research has focused in large part on trying to determine the underlying molecular pathways that contribute to the malignancy of this disease because of the difficulties in treating it. Many tumors have been linked to changes in the expression of microRNAs (miRNAs). miRNAs play a critical role in cancer development by controlling a wide variety of targets and signaling cascades. A rising body of evidence emphasizes WNT pathway dysregulation in glioma, despite the fact that it is dysregulated in many malignancies. Here, we give a detailed analysis of the roles played by miRNAs in the WNT pathway by glioma. We also demonstrate how the WNT pathway cooperates with miRNAs to control a variety of functions, including cell proliferation, invasion, migration, and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Adamu Imam Isa
- Department of Physiology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| |
Collapse
|
13
|
Einafshar E, Mobasheri L, Hasanpour M, Rashidi R, Ghorbani A. Pro-apoptotic effect of chloroform fraction of Moraea sisyrinchium bulb against glioblastoma cells. Biomed Pharmacother 2024; 170:115931. [PMID: 38016363 DOI: 10.1016/j.biopha.2023.115931] [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: 07/24/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
INTRODUCTION Glioblastoma is a common malignant brain tumor, with limited therapeutic options. In our previous study, the Moraea sisyrinchium plant showed cytotoxicity against glioblastoma and hepatocellular carcinoma cells. Among different parts of this plant (flower, stem, and bulb), the bulb showed better anticancer potential. The present work aimed to test the anticancer activity of different fractions of the bulb extract, to determine its phytochemicals, and to study its mechanism action on glioblastoma. METHODS The bulb extract was partitioned into different fractions using immiscible solvents. The U87 glioblastoma cells were incubated with the obtained fractions. Then, the cell proliferation assay (MTT), cell migration test (scratch), cell cycle analysis (propidium iodide staining), apoptosis/necrosis assay (annexin V/propidium iodide staining), and real-time PCR (PTEN, Akt, mTOR, BAX and BCL-2 genes) were performed. Phytochemicals were determined using liquid chromatography-mass spectroscopy. RESULTS The chloroform fraction showed more antiproliferative effect than n-hexane, ethyl acetate, and n-butanol fractions. Also, chloroform fraction induced cell cycle arrest, increased apoptosis, and inhibited cell migration ability (P < 0.05). The expression of PTEN, mTOR, and BAX genes was significantly up-regulated, while the expression of Akt and Bcl-2 showed down-regulation. The phytochemicals identified in the chloroform fraction were mainly xanthones, phytosterols, and isoflavones. CONCLUSION The chloroform fraction of Moraea sisyrinchium bulb inhibits the proliferation and migration of glioblastoma cells by inducing cell cycle arrest and apoptosis by upregulation of the PTEN gene and Bax/Bcl-2 ratio. The identified compounds in the chloroform fraction are potential candidates for further investigation as anticancer agents against glioblastoma.
Collapse
Affiliation(s)
- Elham Einafshar
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Mobasheri
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maede Hasanpour
- Department of Pharmacognosy and Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayeh Rashidi
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Ghorbani
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Liu M, Mobet Y, Shen H. S-Adenosylmethionine Inhibits the Proliferation of Retinoblastoma Cell Y79, Induces Apoptosis and Cell Cycle Arrest of Y79 Cells by Inhibiting the Wnt2/β-Catenin Pathway. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0020. [PMID: 39362212 DOI: 10.2478/aite-2024-0020] [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: 04/24/2024] [Accepted: 08/05/2024] [Indexed: 10/05/2024]
Abstract
Retinoblastoma is one of the most common primary intraocular malignancies in young children. Traditional treatment methods such as chemotherapy often come with significant adverse effects, such as hearing loss, cognitive impairment, and vision loss. Therefore, there is an urgent need to explore a novel therapeutic drug that is both effective and safe. S-adenosylmethionine (SAM) is a natural compound known to exhibit anti-proliferative effects in various cancer cell lines. However, to date, no studies investigated the effects of SAM on retinoblastoma cells and its potential mechanisms of action. Therefore, this study aims to investigate the impact of SAM on retinoblastoma cells and explore its possible mechanisms of action, with the hope of providing new insights into the treatment of this disease. The optimal concentration of SAM was determined using the Cell Counting Kit-8 assay. The effect of SAM on retinoblastoma proliferation was assessed using the 5-ethynyl-2'-deoxyuridine cell proliferation assay. Y79 cells were subjected to hematoxylin and eosin stain and electron microscopy to observe any morphological changes induced by SAM. The stages of SAM's action on the retinoblastoma cell cycle and its apoptotic effects were measured using flow cytometry. The apoptotic effect of SAM on retinoblastoma was further confirmed using the TUNEL assay. Differential expression of related genes was detected through RT-PCR. In vivo subcutaneous tumor formation in nude mice and immunohistochemistry were employed to validate the effect of SAM on retinoblastoma-related phenotypes. Western blotting was conducted to investigate whether SAM modulated retinoblastoma-related phenotypes via the Wnt2/β-catenin pathway. SAM arrested the cell cycle of retinoblastoma at the G1 phase, induced apoptosis of retinoblastoma cells through the Wnt2/β-catenin pathway, and affected their morphology and even ultrastructure. In addition, in vitro and in vivo experiments demonstrated that SAM had an oncogenic effect on retinoblastoma. In this study, we verify in vitro and in vivo whether SAM inhibits the proliferation of retinoblastoma cell Y7, induces apoptosis and cell cycle arrest of Y79 cells by inhibiting the Wnt2/β-catenin pathway, and affects the morphology and structure of retinoblastoma cell Y79.
Collapse
Affiliation(s)
- Mushi Liu
- Medical College, South China University of Technology, Guangzhou, China
| | - Youchaou Mobet
- Bai Sheng Biological Products Co., Ltd, Guangzhou, China
| | - Hong Shen
- Medical College, South China University of Technology, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, China
| |
Collapse
|
15
|
Chen S, Wang K, Wang Q. Mannose: A Promising Player in Clinical and Biomedical Applications. Curr Drug Deliv 2024; 21:1435-1444. [PMID: 38310442 DOI: 10.2174/0115672018275954231220101637] [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: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 02/05/2024]
Abstract
Mannose, an isomer of glucose, exhibits a distinct molecular structure with the same formula but a different atom arrangement, contributing to its specific biological functions. Widely distributed in body fluids and tissues, particularly in the nervous system, skin, testes, and retinas, mannose plays a crucial role as a direct precursor for glycoprotein synthesis. Glycoproteins, essential for immune regulation and glycosylation processes, underscore the significance of mannose in these physiological activities. The clinical and biomedical applications of mannose are diverse, encompassing its anti-inflammatory properties, potential to inhibit bacterial infections, role in metabolism regulation, and suggested involvement in alleviating diabetes and obesity. Additionally, mannose shows promise in antitumor effects, immune modulation, and the construction of drug carriers, indicating a broad spectrum of therapeutic potential. The article aims to present a comprehensive review of mannose, focusing on its molecular structure, metabolic pathways, and clinical and biomedical applications, and also to emphasize its status as a promising therapeutic agent.
Collapse
Affiliation(s)
- Sijing Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kana Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- The Department of Gynecologic Oncology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Xiao X, Li X, Wang Y, Pan C, Zhang P, Gu G, Li T, Jiang Z, Zhang Y, Zhang L. Classification of Brainstem Gliomas Based on Tumor Microenvironment Status. Cancers (Basel) 2023; 15:4224. [PMID: 37686499 PMCID: PMC10487167 DOI: 10.3390/cancers15174224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
The inter-tumor heterogeneity of the tumor microenvironment (TME) and how it correlates with clinical profiles and biological characteristics in brainstem gliomas (BSGs) remain unknown, dampening the development of novel therapeutics against BSGs. The TME status was determined with a list of pan-cancer conserved gene expression signatures using a single-sample gene set enrichment analysis (ssGSEA) and was subsequently clustered via consensus clustering. BSGs exhibited a high inter-tumor TME heterogeneity and were classified into four clusters: "immune-enriched, fibrotic", "immune-enriched, non-fibrotic", "fibrotic", and "depleted". The "fibrotic" cluster had a higher proportion of diffuse intrinsic pontine gliomas (p = 0.041), and "PA-like" tumors were more likely to be "immune-enriched, fibrotic" (p = 0.044). The four TME clusters exhibited distinct overall survival (p < 0.001) and independently impacted BSG outcomes. A four-gene panel as well as a radiomics approach were constructed to identify the TME clusters and achieved high accuracy for determining the classification. Together, BSGs exhibited high inter-tumor heterogeneity in the TME and were classified into four clusters with distinct clinical outcomes and tumor biological properties. The TME classification was accurately identified using a four-gene panel that can potentially be examined with the immunohistochemical method and a non-invasive radiomics method, facilitating its clinical application.
Collapse
Affiliation(s)
- Xiong Xiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Xiaoou Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Tian Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Zhuang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Yang Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (X.X.); (X.L.); (Y.W.); (C.P.); (P.Z.); (G.G.); (T.L.); (Z.J.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
17
|
Gan HK, Parakh S, Osellame LD, Cher L, Uccellini A, Hafeez U, Menon S, Scott AM. Antibody drug conjugates for glioblastoma: current progress towards clinical use. Expert Opin Biol Ther 2023; 23:1089-1102. [PMID: 37955063 DOI: 10.1080/14712598.2023.2282729] [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: 08/20/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Antibody drug conjugates (ADCs) are now a proven therapeutic class for many cancers, combining highly specific targeting with the potency of high effective payloads. This review summarizes the experience with ADCs in brain tumors and examines future paths for their use in these tumors. AREAS COVERED This review will cover all the key classes of ADCs which have been tested in primary brain tumors, including commentary on the major trials to date. The efficacy of these trials, as well as their limitations, will put in context of the overall landscape of drug development in brain tumors. Importantly, this review will summarize key learnings and insights from these trials that help provide the basis for rational ways in which these drugs can be effectively and appropriate developed for patients with primary brain tumors. EXPERT OPINION ADC development in brain tumors has occurred in two major phases to date. Key learnings from previous trials provide a strong rationale for the continued development of these drugs for primary brain tumors. However, the unique biology of these tumors requires development strategies specifically tailored to maximize their optimal development.
Collapse
Affiliation(s)
- Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Sagun Parakh
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Laura D Osellame
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Biochemistry and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Heidelberg, Melbourne, Victoria, Australia
| | - Lawrence Cher
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
| | | | - Umbreen Hafeez
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
| | - Siddharth Menon
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Andrew M Scott
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
18
|
Pećina-Šlaus N, Aničić S, Bukovac A, Kafka A. Wnt Signaling Inhibitors and Their Promising Role in Tumor Treatment. Int J Mol Sci 2023; 24:ijms24076733. [PMID: 37047705 PMCID: PMC10095594 DOI: 10.3390/ijms24076733] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
In a continuous search for the improvement of antitumor therapies, the inhibition of the Wnt signaling pathway has been recognized as a promising target. The altered functioning of the Wnt signaling in human tumors points to the strategy of the inhibition of its activity that would impact the clinical outcomes and survival of patients. Because the Wnt pathway is often mutated or epigenetically altered in tumors, which promotes its activation, inhibitors of Wnt signaling are being intensively investigated. It has been shown that knocking down specific components of the Wnt pathway has inhibitory effects on tumor growth in vivo and in vitro. Thus, similar effects are expected from the application of Wnt inhibitors. In the last decades, molecules acting as inhibitors on the pathway’s specific molecular levels have been identified and characterized. This review will discuss the inhibitors of the canonical Wnt pathway, summarize knowledge on their effectiveness as therapeutics, and debate their side effects. The role of the components frequently mutated in various tumors that are principal targets for Wnt inhibitors is also going to be brought to the reader’s attention. Some of the molecules identified as Wnt pathway inhibitors have reached early stages of clinical trials, and some have only just been discovered. All things considered, inhibition of the Wnt signaling pathway shows potential for the development of future therapies.
Collapse
Affiliation(s)
- Nives Pećina-Šlaus
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| | - Sara Aničić
- Department of Physiology and Immunology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Anja Bukovac
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| | - Anja Kafka
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| |
Collapse
|
19
|
Zhao X, Richardson DR. The role of the NDRG1 in the pathogenesis and treatment of breast cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188871. [PMID: 36841367 DOI: 10.1016/j.bbcan.2023.188871] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
Breast cancer (BC) is the leading cause of cancer death in women. This disease is heterogeneous, with clinical subtypes being estrogen receptor-α (ER-α) positive, having human epidermal growth factor receptor 2 (HER2) overexpression, or being triple-negative for ER-α, progesterone receptor, and HER2 (TNBC). The ER-α positive and HER2 overexpressing tumors can be treated with agents targeting these proteins, including tamoxifen and pertuzumab, respectively. Despite these treatments, resistance and metastasis are problematic, while TNBC is challenging to treat due to the lack of suitable targets. Many studies examining BC and other tumors indicate a role for N-myc downstream-regulated gene-1 (NDRG1) as a metastasis suppressor. The ability of NDRG1 to inhibit metastasis is due, in part, to the inhibition of the initial step in metastasis, namely the epithelial-to-mesenchymal transition. Paradoxically, there are also reports of NDRG1 playing a pro-oncogenic role in BC pathogenesis. The oncogenic effects of NDRG1 in BC have been reported to relate to lipid metabolism or the mTOR signaling pathway. The molecular mechanism(s) of how NDRG1 regulates the activity of multiple signaling pathways remains unclear. Therapeutic strategies that up-regulate NDRG1 have been developed and include agents of the di-2-pyridylketone thiosemicarbazone class. These compounds target oncogenic drivers in BC cells, suppressing the expression of multiple key hormone receptors including ER-α, progesterone receptor, androgen receptor, and prolactin receptor, and can also overcome tamoxifen resistance. Considering the varying role of NDRG1 in BC pathogenesis, further studies are required to examine what subset of BC patients would benefit from pharmacopeia that up-regulate NDRG1.
Collapse
Affiliation(s)
- Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
20
|
Yang RH, Qin J, Cao JL, Zhang MZ, Li YY, Wang MQ, Fang D, Xie SQ. Dickkopf-1 drives tumor immune evasion by inducing PD-L1 expression in hepatocellular carcinoma. Biochem Pharmacol 2023; 208:115378. [PMID: 36513141 DOI: 10.1016/j.bcp.2022.115378] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Understanding the mechanisms regulating PD-L1 expression in hepatocellular carcinoma (HCC) is important to improve the response rate to PD-1/PD-L1 blockade therapy. Here, we show that DKK1 expression is positively associated with PD-L1 expression and inversely correlated with CD8+ T cell infiltration in human HCC tumor specimens. In a subcutaneous xenograft tumor model, overexpression of DKK1 significantly promotes tumor growth, tumoral PD-L1 expression, but reduces tumoral CD8+ T cell infiltration; whereas knockdown of DKK1 has opposite effects. Moreover, enforced expression of DKK1 dramatically promotes PD-L1 expression, Akt activation, β-catenin phosphorylation and total protein expression in HCC cells. By contrast, knockdown of DKK1 inhibits all, relative to controls. In addition, CKAP4 depletion, Akt inhibition, or β-catenin depletion remarkably abrogates DKK1 overexpression-induced transcriptional expression of PD-L1 in HCC cells. Reconstituted expression of the active Akt1 largely increased PD-L1 transcriptional expression in HCC cells. Similarly, expression of WT β-catenin, but not the phosphorylation-defective β-catenin S552A mutant, significantly promotes PD-L1 expression. Correlation analysis of human HCC tumor specimens further revealed that DKK1 and PD-L1 expression were positively correlated with p-β-catenin expression. Together, our findings revealed that DKK1 promotes PD-L1 expression through the activation of Akt/β-catenin signaling, providing a potential strategy to enhance the clinical efficacy of PD-1/PD-L1 blockade therapy in HCC patients.
Collapse
Affiliation(s)
- Ruo-Han Yang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China; The Academy for Advanced Interdisplinary Studies, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Jia Qin
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Jin-Lan Cao
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Ming-Zhu Zhang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Ying-Ying Li
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Meng-Qing Wang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China
| | - Dong Fang
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China; The Academy for Advanced Interdisplinary Studies, Henan University, N. Jinming Ave, Kaifeng 475004, China; Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Kaifeng 475004, China.
| | - Song-Qiang Xie
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng 475004, China; The Academy for Advanced Interdisplinary Studies, Henan University, N. Jinming Ave, Kaifeng 475004, China.
| |
Collapse
|
21
|
EZH2 interacts with HP1BP3 to epigenetically activate WNT7B that promotes temozolomide resistance in glioblastoma. Oncogene 2023; 42:461-470. [PMID: 36517590 DOI: 10.1038/s41388-022-02570-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/04/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor in adults and harbors a subpopulation of glioma stem cells (GSCs). Enhancer of Zeste Homolog 2 (EZH2), a histone lysine methyltransferase, deeply involves in the stemness maintenance of GSC. However, the precise mechanism and therapeutic potential remain elusive. We postulated that the interactome of EZH2 in GSC is unique. Therefore, we performed proteomic and transcriptomic research to unveil the oncogenic mechanism of EZH2. Immunoprecipitation and mass spectrometry were used to identify proteins that co-precipitate with EZH2. We show that EZH2 binds to heterochromatin protein 1 binding protein 3 (HP1BP3) in GSCs and impairs the methylation of H3K9. Overexpression of HP1BP3 enhances the proliferation, self-renewal and temozolomide (TMZ) resistance of GBM cells. Furthermore, EZH2 and HP1BP3 co-activate WNT7B expression thereby increasing TMZ resistance and stemness of GBM cells. Importantly, inhibition of WNT7B autocrine via LGK974 effectively reverses the TMZ resistance. Our work clarifies a new oncogenic mechanism of EZH2 by which it interacts with HP1BP3 and epigenetically activates WNT7B thereby promoting TMZ resistance in GSCs. Our results provide a rationale for targeting WNT/β-catenin pathway as a promising strategy to overcome TMZ resistance in GSCs.
Collapse
|
22
|
Yang J, Xu Y, Fu Z, Chen J, Fan W, Wu X. Progress in research and development of temozolomide brain-targeted preparations: a review. J Drug Target 2023; 31:119-133. [PMID: 36039767 DOI: 10.1080/1061186x.2022.2119243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gliomas are a heterogeneous group of brain tumours with high malignancy, for which surgical resection remains the mainstay of treatment at present. However, the overall prognosis of gliomas remains poor because of their aggressiveness and high recurrence. Temozolomide (TMZ) has anti-proliferative and cytotoxic effects and is indicated for glioblastoma multiforme and recurrent mesenchymal astrocytoma. However, TMZ is disadvantaged by low efficacy and drug resistance, and therefore it is necessary to enhance the brain drug concentration of TMZ to improve its effectiveness and reduce the toxic and adverse effects from systemic administration. There have been many nano-formulations developed for the delivery of TMZ to gliomas that overcome the limitations of TMZ penetration to tumours and increase brain targeting. In this paper, we review the research progress of TMZ nano-formulations, and also discuss challenges and opportunities in the research and development of drug delivery systems, hoping that the data and information summarised herein could provide assistance for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Jiefen Yang
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Youfa Xu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Zhiqin Fu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wei Fan
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| |
Collapse
|
23
|
Effects of a monoclonal antibody against (pro)renin receptor on gliomagenesis. Sci Rep 2023; 13:808. [PMID: 36646875 PMCID: PMC9842725 DOI: 10.1038/s41598-023-28133-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Glioblastoma is characterized by a strong self-renewal potential and poor differentiated state. We have reported previously that the (pro)renin receptor [(P)RR] is a potential target for glioma therapy by silencing the (P)RR gene. Here, we have examined the effects of a monoclonal antibody against (P)RR on gliomagenesis. Human glioma cell lines (U251MG and U87MG) and a glioma stem cell line (MGG23) were used for the in vitro study. The expressions of the Wnt/β-catenin signaling pathway (Wnt signaling pathway) components and stemness markers were measured by Western blotting. The effects of the (P)RR antibody on cell proliferation, sphere formation, apoptosis and migration were also examined. Subcutaneous xenografts were also examined in nude mice. Treatment with the (P)RR antibody reduced expression of Wnt signaling pathway components and stemness markers. Furthermore, the (P)RR antibody reduced cell proliferation and decreased sphere formation significantly. The treatment also suppressed migration and induced apoptosis. In a subcutaneous xenograft model, systemic administration of the (P)RR antibody reduced tumor volume significantly. These data show that treatment with the (P)RR antibody is a potential therapeutic strategy for treating glioblastoma.
Collapse
|
24
|
Xu X, Liang Y, Gareev I, Liang Y, Liu R, Wang N, Yang G. LncRNA as potential biomarker and therapeutic target in glioma. Mol Biol Rep 2023; 50:841-851. [PMID: 36331751 DOI: 10.1007/s11033-022-08056-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 03/22/2022] [Indexed: 11/06/2022]
Abstract
Glioma is the most frequent type of malignant tumor in the central nervous system, accounting for about 80% of primary malignant brain tumors, usually with a poor prognosis. A number of studies have been conducted on the molecular abnormalities in glioma to further understand its pathogenesis, and it has been found that lncRNAs (long non-coding RNA) play a key role in angiogenesis, tumor growth, infiltration and metastasis of glioma. Since specific lncRNAs have an aberrant expression in brain tissue, cerebrospinal fluid as well as peripheral circulation of glioma patients, they are considered to be potential biomarkers. This review focuses on the biological characteristics of lncRNA and its value as a biomarker for glioma diagnosis and prognosis. Moreover, in view of the role of lncRNAs in glioma proliferation and chemoradiotherapy resistance, we discussed the feasibility for lncRNAs as therapeutic targets. Finally, the persisting deficiencies and future prospects of using lncRNAs as clinical biomarkers and therapeutic targets were concluded.
Collapse
Affiliation(s)
- Xun Xu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, 150001, Heilongjiang, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Yuan Liang
- Department of Neurosurgery, Xuzhou Third People's Hospital, Xuzhou, China
| | - Ilgiz Gareev
- Bashkir State Medical University, Ufa, Russia, 450008
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, 150001, Heilongjiang, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Rui Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, 150001, Heilongjiang, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Ning Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, 150001, Heilongjiang, China.
- Institute of Brain Science, Harbin Medical University, Harbin, China.
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23, Nangang District, Harbin, 150001, Heilongjiang, China.
- Institute of Brain Science, Harbin Medical University, Harbin, China.
| |
Collapse
|
25
|
Elevation of spermine remodels immunosuppressive microenvironment through driving the modification of PD-L1 in hepatocellular carcinoma. Cell Commun Signal 2022; 20:175. [DOI: 10.1186/s12964-022-00981-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Spermine is frequently elevated in tumor tissues and body fluids of cancer patients and is critical for cancer cell proliferation, migration and invasion. However, the immune functions of spermine in hepatocellular carcinoma progression remains unknown. In the present study, we aimed to elucidate immunosuppressive role of spermine in hepatocellular carcinoma and to explore the underlying mechanism.
Methods
Whole-blood spermine concentration was measured using HPLC. Human primary HCC tissues were collected to examine the expression of CaSR, p-Akt, β-catenin, STT3A, PD-L1, and CD8. Mouse model of tumorigenesis and lung metastasis were established to evaluate the effects of spermine on hepatocellular carcinoma. Western blotting, immunofluorescence, real time PCR, digital Ca2+ imaging, and chromatin immunoprecipitation assay were used to investigate the underlying mechanisms by which spermine regulates PD-L1 expression and glycosylation in hepatocellular carcinoma cells.
Results
Blood spermine concentration in the HCC patient group was significantly higher than that in the normal population group. Spermine could facilitate tumor progression through inducing PD-L1 expression and decreasing the CD8+ T cell infiltration in HCC. Mechanistically, spermine activates calcium-sensing receptor (CaSR) to trigger Ca2+ entry and thereby promote Akt-dependent β-catenin stabilization and nuclear translocation. Nuclear β-catenin induced by spermine then activates transcriptional expression of PD-L1 and N-glycosyltransferase STT3A, while STT3A in turn increases the stability of PD-L1 through inducing PD-L1 protein N-glycosylation in HCC cells.
Conclusions
This study reveals the crucial function of spermine in establishing immune privilege by increasing the expression and N-glycosylation of PD-L1, providing a potential strategy for the treatment of hepatocellular carcinoma.
Collapse
|
26
|
Sun J, Wang J, Li M, Li S, Li H, Lu Y, Li F, Xin T, Jin F. circTOP2A functions as a ceRNA to promote glioma progression by upregulating RPN2. Cancer Sci 2022; 114:490-503. [PMID: 36227125 PMCID: PMC9899613 DOI: 10.1111/cas.15612] [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: 04/30/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022] Open
Abstract
Competing endogenous RNA (ceRNA)-mediated signaling pathway dysregulation provides great insight into comprehensively understanding the molecular mechanism and combined targeted therapy for glioblastoma. circRNA is characterized by high stability, tissue/developmental stage-specific expression and abundance in brain and plays significant roles in the initiation and progression of cancer. Our previous published data have demonstrated that RPN2 was significantly upregulated in glioma and promoted tumor progression via the activation of the Wnt/β-catenin pathway. Furthermore, we proved that miR-422a regulated the Wnt/β-catenin signaling pathway by directly targeting RPN2. In this study, based on the glioblastoma microarray profiles, we identified the upstream circTOP2A, which completely bound to miR-422a and was co-expressed with the RPN2. circTOP2A was significantly overexpressed in glioma and conferred a poor prognosis. circTOP2A could regulate RPN2 expression by sponging miR-422a, verified by western blot, dual-luciferase reporter gene assay, and RNA pull-down assay. Functional assays including CCK8, transwell and FITC-annexin V were performed to explore the RPN2-mediated role of the circTOP2A effect on the glioma malignant phenotype. Additionally, TOP/FOP and immunofluorescence analysis were used to confirm that sh-circTOP2A could suppress the Wnt/β-catenin pathway partly through RPN2. Finally, a tumor xenograft model was applied to validate the biological function of circTOP2A in vivo. Taken together, our findings reveal the critical role of circTOP2A in promoting glioma proliferation and invasion via a ceRNA mechanism and provide an exploitable biomarker and therapeutic target for glioma patients.
Collapse
Affiliation(s)
- Jikui Sun
- Department of NeurosurgeryAffiliated Hospital of Jining Medical University, & Shandong Provincial Key Laboratory of Stem Cells and Neuro‐oncologyJiningChina,Shandong University of Traditional Chinese MedicineJinanChina,Shandong Medicine and Health Key Laboratory of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Jinhuan Wang
- Tianjin Cerebral Vascular and Neural Degenerative Disease Key Laboratory, Department of NeurosurgeryTianjin Neurosurgical Institute, Tianjin Huanhu HospitalTianjinChina
| | - Meng Li
- Shandong Medicine and Health Key Laboratory of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Shengjie Li
- Shandong Medicine and Health Key Laboratory of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Hanyun Li
- Cheeloo College of MedicineShandong UniversityJinanChina
| | - Yan Lu
- Department of NeurosurgeryAffiliated Hospital of Jining Medical University, & Shandong Provincial Key Laboratory of Stem Cells and Neuro‐oncologyJiningChina,Medical Research CenterAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Feng Li
- Shandong Medicine and Health Key Laboratory of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Tao Xin
- Shandong University of Traditional Chinese MedicineJinanChina,Shandong Medicine and Health Key Laboratory of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Feng Jin
- Department of NeurosurgeryAffiliated Hospital of Jining Medical University, & Shandong Provincial Key Laboratory of Stem Cells and Neuro‐oncologyJiningChina
| |
Collapse
|
27
|
Wang W, Zhao Z, Han S, Wu D. miR-637 Prevents Glioblastoma Progression by Interrupting ZEB2/WNT/β-catenin Cascades. Cell Mol Neurobiol 2022; 42:2321-2335. [PMID: 34047878 PMCID: PMC11421589 DOI: 10.1007/s10571-021-01107-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
Glioblastomas (GBMs) are the most frequent primary malignancies in the central nervous system. Aberrant activation of WNT/β-catenin signaling pathways is critical for GBM malignancy. However, the regulation of WNT/β-catenin signaling cascades remains unclear. Presently, we observed the increased expression of ZEB2 and the decreased expression of miR-637 in GBM. The expression of miR-637 was negatively correlated with ZEB2 expression. miR-637 overexpression overcame the ZEB2-enhanced cell proliferation and G1/S phase transition. Besides, miR-637 suppressed the canonical WNT/β-catenin pathways by targeting WNT7A directly. Gain- and loss-of-function experiments with U251 mice demonstrated that miR-637 inhibited cell proliferation and arrested the G1/S phase transition, leading to tumor growth suppression. The collective findings suggest that ZEB2 and WNT/β-catenin cascades merge at miR-637, and the ectopic expression of miR-637 disturbs ZEB2/WNT/β-catenin-mediated GBM growth. The findings provide new clues for improving β-catenin-targeted therapy against GBM.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Zilong Zhao
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Shuai Han
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Di Wu
- Department of Tumor Biotherapy and Cancer Research, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China.
| |
Collapse
|
28
|
Hattori Y, Kurozumi K, Otani Y, Uneda A, Tsuboi N, Makino K, Hirano S, Fujii K, Tomita Y, Oka T, Matsumoto Y, Shimazu Y, Michiue H, Kumon H, Date I. Combination of Ad-SGE-REIC and bevacizumab modulates glioma progression by suppressing tumor invasion and angiogenesis. PLoS One 2022; 17:e0273242. [PMID: 36006934 PMCID: PMC9409598 DOI: 10.1371/journal.pone.0273242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
Reduced expression in immortalized cells/Dickkopf-3 (REIC/Dkk-3) is a tumor suppressor and its overexpression has been shown to exert anti-tumor effects as a therapeutic target gene in many human cancers. Recently, we demonstrated the anti-glioma effects of an adenoviral vector carrying REIC/Dkk-3 with the super gene expression system (Ad-SGE-REIC). Anti-vascular endothelial growth factor treatments such as bevacizumab have demonstrated convincing therapeutic advantage in patients with glioblastoma. However, bevacizumab did not improve overall survival in patients with newly diagnosed glioblastoma. In this study, we examined the effects of Ad-SGE-REIC on glioma treated with bevacizumab. Ad-SGE-REIC treatment resulted in a significant reduction in the number of invasion cells treated with bevacizumab. Western blot analyses revealed the increased expression of several endoplasmic reticulum stress markers in cells treated with both bevacizumab and Ad-SGE-REIC, as well as decreased β-catenin protein levels. In malignant glioma mouse models, overall survival was extended in the combination therapy group. These results suggest that the combination therapy of Ad-SGE-REIC and bevacizumab exerts anti-glioma effects by suppressing the angiogenesis and invasion of tumors. Combined Ad-SGE-REIC and bevacizumab might be a promising strategy for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Yasuhiko Hattori
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Shizuoka, Japan
- * E-mail:
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsuhito Uneda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nobushige Tsuboi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keigo Makino
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuichiro Hirano
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Fujii
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tetsuo Oka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuji Matsumoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yosuke Shimazu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Michiue
- Neutron Therapy Research Center, Okayama University, Okayama, Japan
| | - Hiromi Kumon
- Innovation Center Okayama for Nanobio-targeted Therapy, Okayama University, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
29
|
Current Opportunities for Targeting Dysregulated Neurodevelopmental Signaling Pathways in Glioblastoma. Cells 2022; 11:cells11162530. [PMID: 36010607 PMCID: PMC9406959 DOI: 10.3390/cells11162530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most common and highly lethal type of brain tumor, with poor survival despite advances in understanding its complexity. After current standard therapeutic treatment, including tumor resection, radiotherapy and concomitant chemotherapy with temozolomide, the median overall survival of patients with this type of tumor is less than 15 months. Thus, there is an urgent need for new insights into GBM molecular characteristics and progress in targeted therapy in order to improve clinical outcomes. The literature data revealed that a number of different signaling pathways are dysregulated in GBM. In this review, we intended to summarize and discuss current literature data and therapeutic modalities focused on targeting dysregulated signaling pathways in GBM. A better understanding of opportunities for targeting signaling pathways that influences malignant behavior of GBM cells might open the way for the development of novel GBM-targeted therapies.
Collapse
|
30
|
Zheng ZQ, Yuan GQ, Kang NL, Nie QQ, Zhang GG, Wang Z. Chromobox 7/8 serve as independent indicators for glioblastoma via promoting proliferation and invasion of glioma cells. Front Neurol 2022; 13:912039. [PMID: 36034290 PMCID: PMC9403790 DOI: 10.3389/fneur.2022.912039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background The chromobox family, a critical component of epigenetic regulators, participates in the tumorigenesis and progression of many malignancies. However, the roles of the CBX family members (CBXs) in glioblastoma (GBM) remain unclear. Methods The mRNA expression of CBXs was analyzed in tissues and cell lines by Oncomine and Cancer Cell Line Encyclopedia (CCLE). The differential expression of CBXs at the mRNA level was explored in The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases with the “beeswarm” R package. The protein expression of CBXs in GBM was further examined on Human Protein Atlas (HPA). The correlations between CBXs and IDH mutation and between CBXs and GBM subtypes were investigated in the TCGA portal and CGGA database with the “survminer” R package. The alteration of CBXs and their prognostic value were further determined via the cBioPortal and CGGA database with the “survival” R package. The univariate and multivariate analyses were performed to screen out the independent prognostic roles of CBXs in the CGGA database. Cytoscape was used to visualize the functions and related pathways of CBXs in GBM. U251 and U87 glioma cells with gene intervention were used to validate the role of CBX7/8 in tumor proliferation and invasion. Proliferation/invasion-related markers were conducted by Western blot and immunostaining. Results CBXs presented significantly differential expressions in pan-cancers. CBX2/3/5/8 were upregulated, whereas CBX6/7 were downregulated at mRNA level in GBM of TCGA and CGGA databases. Similarly, high expression of CBX2/3/5 and low expression of CBX6/8 were further confirmed at the protein level in the HPA. CBX2/6/7 were positively correlated with IDH mutation and CBX1/2/4/5/8 were closely related to GBM subtypes. CBX7 and CBX8 presented the independent prognostic factors for GBM patient survival. GO and KEGG analyses indicated that CBXs were closely related to the histone H3-K36, PcG protein complex, ATPase, and Wnt pathway. The overexpression of CBX7 and underexpression of CBX8 significantly inhibited the proliferation and invasion of glioma cells in vivo and in vitro. Conclusion Our results suggested that CBX7 and CBX8 served as independent prognostic indicators that promoted the proliferation and invasion of glioma cells, providing a promising strategy for diagnosing and treating GBM.
Collapse
Affiliation(s)
- Zong-Qing Zheng
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gui-Qiang Yuan
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, Changshu Second People's Hospital, Suzhou, China
| | - Na-Ling Kang
- Liver Center, The First Affiliated Hospital, Fujian Medical University, Fujian, China
| | - Qian-Qian Nie
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guo-Guo Zhang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Zhong Wang
| |
Collapse
|
31
|
Circadian Clock Genes Act as Diagnostic and Prognostic Biomarkers of Glioma: Clinic Implications for Chronotherapy. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9774879. [PMID: 35832846 PMCID: PMC9273445 DOI: 10.1155/2022/9774879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/26/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022]
Abstract
Gliomas are the most common primary intracranial tumors and closely related to circadian clock. Due to the high mortality and morbidity of gliomas, exploring novel diagnostic and early prognostic markers is necessary. Circadian clock genes (CCGs) play important roles in regulating the daily oscillation of biological processes and the development of tumor. Therefore, we explored the influences that the oscillations of circadian clock genes (CCGs) on diagnosis and prognosis of gliomas using bioinformatics. In this work, we systematically analyzed the rhythmic expression of CCGs in brain and found that some CCGs had strong rhythmic expression; the expression levels were significantly different between day and night. Four CCGs (ARNTL, NPAS2, CRY2, and DBP) with rhythmic expression were not only identified as differentially expressed genes but also had significant independent prognostic ability in the overall survival of glioma patients and were highly correlated with glioma prognosis in COX analysis. Besides, we found that CCG-based predictive model demonstrated higher predictive accuracy than that of the traditional grade-based model; this new prediction model can greatly improve the accuracy of glioma prognosis. Importantly, based on the four CCGs’ circadian oscillations, we revealed that patients sampled at night had higher predictive ability. This may help detect glioma as early as possible, leading to early cancer intervention. In addition, we explored the mechanism of CCGs affecting the prognosis of glioma. CCGs regulated the cell cycle, DNA damage, Wnt, mTOR, and MAPK signaling pathways. In addition, it also affects prognosis through gene coexpression and immune infiltration. Importantly, ARNTL can rhythmically modulated the cellular sensitivity to clinic drugs, temozolomide. The optimal point of temozolomide administration should be when ARNTL expression is highest, that is, the effect is better at night. In summary, our study provided a basis for optimizing clinical dosing regimens and chronotherapy for glioma. The four key CCGs can serve as potential diagnostic and prognostic biomarkers for glioma patients, and ARNTL also has obvious advantages in the direction of glioma chronotherapy.
Collapse
|
32
|
Wang J, Quan Y, Lv J, Gong S, Ren P. Inhibition of FAM83D displays antitumor effects in glioblastoma via down-regulation of the AKT/Wnt/β-catenin pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:1343-1356. [PMID: 35150198 DOI: 10.1002/tox.23488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 12/01/2021] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Up-regulation of family with sequence similarity 83 member D (FAM83D) has been acknowledged as a vital contributor for the carcinogenesis of numerous cancers. The relevance of FAM83D in glioblastoma (GBM), however, is not well understood. This current work aimed to determine the possible roles and mechanisms of FAM83D in GBM. By analyzing The Cancer Genome Atlas (TCGA) data, we found dramatic increases in FAM83D expression in GBM tissue. We also observed elevated levels of FAM83D in the clinical specimens of GBM. In vitro data showed that silencing FAM83D resulted in remarkable antitumor effects via inhibiting the proliferation, invasion and epithelial-mesenchymal transition of GBM cells. Moreover, the knockdown of FAM83D improved sensitivity to the chemotherapy drug temozolomide. In-depth mechanism research revealed that the silencing of FAM83D strikingly decreased the phosphorylation levels of AKT and glycogen synthase kinase-3β, and prohibited activation of the Wnt/β-catenin pathway. The suppression of AKT abolished FAM83D-mediated activation of the Wnt/β-catenin pathway. The re-expression of β-catenin reversed FAM83D-silencing-induced antitumor effects in GBM cells. In addition, GBM cells with FAM83D silencing exhibited reduced tumorigenic potential in vivo. Overall, the data from this work show that the inhibition of FAM83D displays antitumor effects in GBM via down-regulation of the AKT/Wnt/β-catenin pathway and propose FAM83D as a new therapeutic target for GBM.
Collapse
Affiliation(s)
- Jubo Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Quan
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shouping Gong
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pengyu Ren
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
Daisy Precilla S, Biswas I, Kuduvalli SS, Anitha TS. Crosstalk between PI3K/AKT/mTOR and WNT/β-Catenin signaling in GBM - Could combination therapy checkmate the collusion? Cell Signal 2022; 95:110350. [PMID: 35525406 DOI: 10.1016/j.cellsig.2022.110350] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/11/2022] [Accepted: 04/30/2022] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme is one of the calamitous primary glial brain tumors with extensive heterogeneity at cellular and molecular levels. While maximal surgical resection trailed by radio and chemotherapy employing temozolomide remains the gold-standard treatment for malignant glioma patients, the overall prognosis remains dismal and there exists an unmet need for effective therapeutic strategies. In this context, we hypothesize that proper understanding of signaling pathways responsible for glioblastoma multiforme proliferation would be the first trump card while searching for novel targeted therapies. Among the pathways aberrantly activated, PI3K/AKT/mTOR is the most significant pathway, that is clinically implicated in malignancies such as high-grade glioma. Further, the WNT/β-Catenin cascade is well-implicated in several malignancies, while its role in regulating glioma pathogenesis has only emerged recently. Nevertheless, oncogenic activation of both these pathways is a frequent event in malignant glioma that facilitates tumor proliferation, stemness and chemo-resistance. Recently, it has been reported that the cross-talk of PI3K/AKT/mTOR pathway with multiple signaling pathways could promote glioma progression and reduce the sensitivity of glioma cells to the standard therapy. However, very few studies had focused on the relationship between PI3K/AKT/mTOR and WNT/β-Catenin pathways in glioblastoma multiforme. Interestingly, in homeostatic and pathologic circumstances, both these pathways depict fine modulation and are connected at multiple levels by upstream and downstream effectors. Thus, gaining deep insights on the collusion between these pathways would help in discovering unique therapeutic targets for glioblastoma multiforme management. Hence, the current review aims to address, "the importance of inter-play between PI3K/AKT/mTOR and WNT/β-Catenin pathways", and put forward, "the possibility of combinatorially targeting them", for glioblastoma multiforme treatment enhancement.
Collapse
Affiliation(s)
- S Daisy Precilla
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Indrani Biswas
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Shreyas S Kuduvalli
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - T S Anitha
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India.
| |
Collapse
|
34
|
Li J, Ma J, Zhou L, Huang S, Sun J, Chen L, Lu Z. Green Synthesized Bismuth Oxide Nanoparticles Using Aqueous Rhizome Extract of Curcuma longa Mitigate the Proliferation of Human U87 Glioblastoma Cells by Regulation of the Wnt/ β-Catenin Signaling Pathway. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The low biocompatibility of inorganic nanoparticles (NPs) is a main concern in their wide applications in the biomedical field. Therefore, the green synthesis of NPs from plant extracts can provide safe NPs for biomedical applications. The present study was aimed to assess the anticancer
activity of bismuth oxide (Bi2O3) NPs fabricated using aqueous plant extracts from the rhizome of Curcuma longa (C. longa). Characterization of green Bi2O3 NPs was done using TEM, DLS, and X-ray diffraction analyses. Selective anticancer
activity of green Bi2O3 NPs against human glioblastoma (U87) cells was assessed using MTT, GSH, MDA, ROS, apoptosis, and caspase-3 assays. Also, qPCR analysis was done to explore the expression of β-catenin, cyclin D1, and c-myc at mRNA level as the important
genes of the Wnt/β-catenin signaling pathway. The results showed that the green Bi2O3 NPs have a crystalline nature with a size of around 30 nm with good colloidal stability attributed to potential bio-fabrication of Bi2O3 NPs. Cellular
study indicated that green Bi2O3 NPs triggered selective anticancer activity against U87 cells through reduction of GSH level and increase of MDA level, ROS level, Annexin+ cells, and caspase-3 activity. Also, it was found that IC50 concentration
of biosynthesized Bi2O3 NPs (20 μg/mL) resulted in a significant downregulation in the expression of β-catenin, cyclin D1, and c-myc genes involved in the Wnt/β-catenin signaling pathway. This study concludes that green Bi2O3
NPs bio-fabricated from rhizome of C. longa show potential selective anticancer activity.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Junfeng Ma
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Liang Zhou
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Shan Huang
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Jiahua Sun
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Lin Chen
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| | - Zhengrong Lu
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 20040, China
| |
Collapse
|
35
|
Fei YQ, Shi RT, Zhou YF, Wu JZ, Song Z. Mannose inhibits proliferation and promotes apoptosis to enhance sensitivity of glioma cells to temozolomide through Wnt/β-catenin signaling pathway. Neurochem Int 2022; 157:105348. [DOI: 10.1016/j.neuint.2022.105348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 03/10/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
|
36
|
Cheng J, Zhang R, Yan M, Li Y. Circular RNA hsa_circ_0000277 promotes tumor progression and DDP resistance in esophageal squamous cell carcinoma. BMC Cancer 2022; 22:238. [PMID: 35241028 PMCID: PMC8895546 DOI: 10.1186/s12885-022-09241-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/25/2022] [Indexed: 12/24/2022] Open
Abstract
Background Circular RNAs (circRNAs) are well-known regulators of cancer progression and chemoresistance in various types of cancers. This study was performed to investigate the function of hsa_circ_0000277 in esophageal squamous cell carcinoma (ESCC). Methods RNA levels were analyzed via the reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8 (CCK-8) assay was applied to determine cell proliferation and half maximal inhibitory concentration (IC50) of cisplatin (DDP). Colony formation ability was evaluated by colony formation assay. Cell cycle and apoptosis were measured using flow cytometry. RNA immunoprecipitation (RIP), pull-down assay and dual-luciferase reporter assays were performed for target interaction analysis. The protein levels were determined through western blot. Xenograft models were established for researching hsa_circ_0000277 function in vivo. Results Hsa_circ_0000277 expression was increased in ESCC cells and tissues, and it had important clinical significance. Downregulation of hsa_circ_0000277 repressed ESCC cell proliferation, colony formation, cell cycle, and DDP resistance. Hsa_circ_0000277 acted as a microRNA-873-5p (miR-873-5p) sponge and Sry-related high-mobility group box 4 (SOX4) was validated as a target of miR-873-5p. Moreover, hsa_circ_0000277/miR-873-5p axis and miR-873-5p/SOX4 axis regulated ESCC cell progression and DDP resistance. Hsa_circ_0000277/miR-873-5p axis activated SOX4/Wnt/β-catenin signaling pathway. Hsa_circ_0000277 facilitated tumorigenesis and DDP resistance by miR-873-5p/SOX4 axis in vivo. Conclusion These findings unraveled that hsa_circ_0000277 promoted ESCC progression and DDP resistance via miR-873-5p/SOX4/Wnt/β-catenin axis, showing a specific molecular mechanism of carcinogenesis and chemoresistance in ESCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09241-9.
Collapse
Affiliation(s)
- Jiwei Cheng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Zhengzhou, 450008, Henan Province, China
| | - Ruixiang Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Zhengzhou, 450008, Henan Province, China
| | - Ming Yan
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Zhengzhou, 450008, Henan Province, China
| | - Yin Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Zhengzhou, 450008, Henan Province, China.
| |
Collapse
|
37
|
Wu Q, Yin X, Zhao W, Xu W, Chen L. Downregulation of SFRP2 facilitates cancer stemness and radioresistance of glioma cells via activating Wnt/β-catenin signaling. PLoS One 2021; 16:e0260864. [PMID: 34852024 PMCID: PMC8635357 DOI: 10.1371/journal.pone.0260864] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/17/2021] [Indexed: 12/21/2022] Open
Abstract
Secreted frizzled-related protein 2 (SFRP2) is a glycoprotein with frizzled-like cysteine-rich domain that binds with Wnt ligands or frizzled receptors to regulate Wnt signaling. SFRP2 is frequently hypermethylated in glioma patients, and analysis of TCGA data indicates that SFRP2 is one of the most downregulated genes in radiotherapy treated glioma patients. In the present study, we aimed to explore the potential function of SFRP2 in tumorigenesis and radioresistance of glioma. The RNA sequencing data of TCGA glioma samples were downloaded and analyzed. SFRP2 expression in 166 glioma patients was evaluated by qRT-PCR. The potential functions of SFRP2 in glioma were evaluated by loss-of-function assays and gain-of-function assays in glioma cell lines. We found that SFRP2 was downregulated in radiotherapy-treated glioma patients, and low SFRP2 expression was correlated with advanced tumor stage and poor prognosis. CRISP/Cas9-meidated SFRP2 knockdown promoted soft agar colony formation, cancer stemness and radioresistance of glioma cells, while enforced SFRP2 expression exhibited opposite effects. Moreover, Wnt/β-catenin signaling was activated in radiotherapy treated glioma patients. SFRP2 knockdown activated Wnt/β-catenin signaling in glioma cell lines, while overexpression of SFRP2 inhibited Wnt/β-catenin activation. Besides, pharmacological inhibition of Wnt/β-catenin signaling by XAV-939 abrogated the effects of SFRP2 knockdown on cancer stemness and radioresistance of glioma cells. Our data for the first time demonstrated a role of SFRP2 in radioresistance of glioma cells, and suggested that inhibition of Wnt/β-catenin signaling might be a potential strategy for increasing radiosensitivity of glioma patients.
Collapse
Affiliation(s)
- Quansheng Wu
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xiaofeng Yin
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Wenbo Zhao
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Wenli Xu
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Laizhao Chen
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
- * E-mail:
| |
Collapse
|
38
|
Xie H, Lv S, Wang Z, Yuan X. E2F transcription factor 1 elevates cyclin D1 expression by suppressing transcription of microRNA-107 to augment progression of glioma. Brain Behav 2021; 11:e2399. [PMID: 34758200 PMCID: PMC8671784 DOI: 10.1002/brb3.2399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Dysregulation of microRNAs has been frequently implicated in the progression of human diseases, including glioma. This study aims to explore the interaction between E2F transcription factor 1 (E2F1) and miR-107 in the progression of glioma. METHODS Expression of miR-107 in glioma tissues and cells was examined. Putative binding sites between E2F1 and the promoter region of miR-107, and between miR-107 and cyclin D1 (CCND1) mRNA were predicted via bioinformatic systems and validated via chromatin immunoprecipitation and luciferase reporter gene assays. Altered expression of miR-107, E2F1, and CCND1 was introduced in A172 and T98G cells to examine their roles in cell growth and the activity of the Wnt/β-catenin signaling. In vivo experiments were performed by injecting cells in nude mice. RESULTS miR-107 was poorly expressed, whereas E2F1 and CCND1 were highly expressed in glioma tissues and cells. E2F1 bound to the promoter region of miR-107 to induce transcriptional repression, and miR-107 directly bound to CCND1 mRNA to reduce its expression. Overexpression of miR-107 reduced proliferation, migration and invasion, and augmented apoptosis of glioma cells, and it reduced activity of the Wnt/β-catenin pathway. The anti-tumorigenic roles of miR-107 were blocked by E2F1 or CCND1 overexpression. Similar results were reproduced in vivo where miR-107 overexpression or E2F1 inhibition blocked tumor growth in nude mice. CONCLUSION This study suggested that E2F1 reduces miR-107 transcription to induce CCND1 upregulation, which leads to progression of glioma via Wnt/β-catenin signaling activation.
Collapse
Affiliation(s)
- Huan Xie
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, P. R. China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Zhaozhen Wang
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, P. R. China
| | - Xinzhang Yuan
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, P. R. China
| |
Collapse
|
39
|
Mannino F, Pallio G, Corsaro R, Minutoli L, Altavilla D, Vermiglio G, Allegra A, Eid AH, Bitto A, Squadrito F, Irrera N. Beta-Caryophyllene Exhibits Anti-Proliferative Effects through Apoptosis Induction and Cell Cycle Modulation in Multiple Myeloma Cells. Cancers (Basel) 2021; 13:5741. [PMID: 34830893 PMCID: PMC8616110 DOI: 10.3390/cancers13225741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
Cannabinoid receptors, which are widely distributed in the body, have been considered as possible pharmacological targets for the management of several tumors. Cannabinoid type 2 receptors (CB2Rs) belong to the G protein-coupled receptor family and are mainly expressed in hematopoietic and immune cells, such as B-cells, T-cells, and macrophages; thus, CB2R activation might be useful for treating cancers affecting plasma cells, such as multiple myeloma (MM). Previous studies have shown that CB2R stimulation may have anti-proliferative effects; therefore, the purpose of the present study was to explore the antitumor effect of beta-caryophyllene (BCP), a CB2R agonist, in an in vitro model of MM. Dexamethasone-resistant (MM.1R) and sensitive (MM.1S) human multiple myeloma cell lines were used in this study. Cells were treated with different concentrations of BCP for 24 h, and a group of cells was pre-incubated with AM630, a specific CB2R antagonist. BCP treatment reduced cell proliferation through CB2R stimulation; notably, BCP considerably increased the pro-apoptotic protein Bax and decreased the anti-apoptotic molecule Bcl-2. Furthermore, an increase in caspase 3 protein levels was detected following BCP incubation, thus demonstrating its anti-proliferative effect through apoptosis activation. In addition, BCP regulated AKT, Wnt1, and beta-catenin expression, showing that CB2R stimulation may decrease cancer cell proliferation by modulating Wnt/β-catenin signaling. These effects were counteracted by AM630 co-incubation, thus confirming that BCP's mechanism of action is mainly related to CB2R modulation. A decrease in β-catenin regulated the impaired cell cycle and especially promoted cyclin D1 and CDK 4/6 reduction. Taken together, these data revealed that BCP might have significant and effective anti-cancer and anti-proliferative effects in MM cells by activating apoptosis, modulating different molecular pathways, and downregulating the cell cycle.
Collapse
Affiliation(s)
- Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| | - Giovanni Pallio
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| | - Roberta Corsaro
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| | - Domenica Altavilla
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (D.A.); (G.V.)
| | - Giovanna Vermiglio
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (D.A.); (G.V.)
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy;
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, 2713 Doha, Qatar
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria Gazzi, 98125 Messina, Italy; (F.M.); (G.P.); (R.C.); (L.M.); (N.I.)
| |
Collapse
|
40
|
Soares LC, Al-Dalahmah O, Hillis J, Young CC, Asbed I, Sakaguchi M, O’Neill E, Szele FG. Novel Galectin-3 Roles in Neurogenesis, Inflammation and Neurological Diseases. Cells 2021; 10:3047. [PMID: 34831271 PMCID: PMC8618878 DOI: 10.3390/cells10113047] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022] Open
Abstract
Galectin-3 (Gal-3) is an evolutionarily conserved and multifunctional protein that drives inflammation in disease. Gal-3's role in the central nervous system has been less studied than in the immune system. However, recent studies show it exacerbates Alzheimer's disease and is upregulated in a large variety of brain injuries, while loss of Gal-3 function can diminish symptoms of neurodegenerative diseases such as Alzheimer's. Several novel molecular pathways for Gal-3 were recently uncovered. It is a natural ligand for TREM2 (triggering receptor expressed on myeloid cells), TLR4 (Toll-like receptor 4), and IR (insulin receptor). Gal-3 regulates a number of pathways including stimulation of bone morphogenetic protein (BMP) signaling and modulating Wnt signalling in a context-dependent manner. Gal-3 typically acts in pathology but is now known to affect subventricular zone (SVZ) neurogenesis and gliogenesis in the healthy brain. Despite its myriad interactors, Gal-3 has surprisingly specific and important functions in regulating SVZ neurogenesis in disease. Gal-1, a similar lectin often co-expressed with Gal-3, also has profound effects on brain pathology and adult neurogenesis. Remarkably, Gal-3's carbohydrate recognition domain bears structural similarity to the SARS-CoV-2 virus spike protein necessary for cell entry. Gal-3 can be targeted pharmacologically and is a valid target for several diseases involving brain inflammation. The wealth of molecular pathways now known further suggest its modulation could be therapeutically useful.
Collapse
Affiliation(s)
- Luana C. Soares
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3QX, UK; (L.C.S.); (I.A.)
- Department of Oncology, University of Oxford, Oxford OX1 3QX, UK;
| | - Osama Al-Dalahmah
- Irving Medical Center, Columbia University, New York, NY 10032, USA;
| | - James Hillis
- Massachusets General Hospital, Harvard Medical School, 15 Parkman Street, Boston, MA 02114, USA;
| | - Christopher C. Young
- Department of Neurological Surgery, University of Washington, 325 Ninth Avenue, Seattle, WA 98104, USA;
| | - Isaiah Asbed
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3QX, UK; (L.C.S.); (I.A.)
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford OX1 3QX, UK;
| | - Francis G. Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3QX, UK; (L.C.S.); (I.A.)
| |
Collapse
|
41
|
Kashani B, Zandi Z, Kaveh V, Pourbagheri-Sigaroodi A, Ghaffari SH, Bashash D. Small molecules with huge impacts: the role of miRNA-regulated PI3K pathway in human malignancies. Mol Biol Rep 2021; 48:8045-8059. [PMID: 34689281 DOI: 10.1007/s11033-021-06739-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/15/2021] [Indexed: 11/28/2022]
Abstract
Along with evolution, a considerable number of signaling cascades have evolved within cells to meet their multifaceted needs. Among transmitting molecules, phosphoinositide 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR) have teamed up to build a signaling axis that effectively regulates various cellular processes including cell proliferation and migration. Given the extensive output of the PI3K/Akt/mTOR signaling axis, its aberrancy could subsequently lead to the formation of a wide range of human cancers spanning from hematologic malignancies to different types of solid tumors. Despite the high frequency of the PI3K pathway over-activation in most malignancies, mutations in the DNA sequence are not equally common. Such incompatibility sheds light on the possible effects of post-translational modification mechanisms that may take control of this pathway, some of the most important ones of which are through microRNAs (miRNAs or miRs). The present review is designed to take off the veil from the regulatory role of these small non-coding RNAs on the PI3K/Akt/mTOR signaling axis in carcinogenesis.
Collapse
Affiliation(s)
- Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Kaveh
- Department of Medical Oncology and Hematology, Iran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Ma S, Duan L, Dong H, Ma X, Guo X, Liu J, Li G, Yu Y, Xu Y, Yuan G, Zhao X, Tian G, Zhai S, Pan Y, Zhang Y. OLFML2A Downregulation Inhibits Glioma Proliferation Through Suppression of Wnt/β-Catenin Signaling. Front Oncol 2021; 11:717917. [PMID: 34650914 PMCID: PMC8506028 DOI: 10.3389/fonc.2021.717917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Glioma is a highly heterogeneous and lethal tumor with an extremely poor prognosis. Through analysis of TCGA data, we identified that OLFML2A is a key promotor of gliomagenesis. However, the molecular function of OLFML2A and its underlying mechanism of action in glioma remain unclear. In this study, we found that OLFML2A expression was significantly upregulated in glioma specimens and positively correlated with pathological grades in glioma patients. Moreover, Kaplan–Meier survival analysis of TCGA data revealed that glioma patients with higher OLFML2A expression had shorter overall survival. Importantly, OLFML2A knockdown in glioma cells inhibited cell proliferation and promoted apoptosis. Mechanistically, OLFML2A downregulation inhibits Wnt/β-catenin signaling by upregulating amyloid precursor protein (APP) expression and reducing stabilized β-catenin levels, leading to the repression of MYC, CD44, and CSKN2A2 expression. Furthermore, OLFML2A downregulation suppressed the growth of transplanted glioma subcutaneously and intracranially by inhibiting Wnt/β-catenin pathway-dependent cell proliferation. By uncovering the oncogenic effects in human and rodent gliomas, our data support OLFML2A as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Shize Ma
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Lei Duan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Huateng Dong
- Department of Pediatric Neurology, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, China
| | - Xiaodong Ma
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Xinyu Guo
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Jianli Liu
- Second Clinical School, Lanzhou University, Lanzhou, China.,Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Guoqiang Li
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yue Yu
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yanlong Xu
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Guoqiang Yuan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Xingkun Zhao
- Second Clinical School, Lanzhou University, Lanzhou, China
| | - Guopeng Tian
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Shijia Zhai
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yawen Pan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| | - Yinian Zhang
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.,Second Clinical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
43
|
Liu T, Hu J, Han B, Tan S, Jia W, Xin Y. A positive feedback loop of lncRNA-RMRP/ZNRF3 axis and Wnt/β-catenin signaling regulates the progression and temozolomide resistance in glioma. Cell Death Dis 2021; 12:952. [PMID: 34657141 PMCID: PMC8520527 DOI: 10.1038/s41419-021-04245-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/31/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
Drug resistance strikingly limits the therapeutic effect of temozolomide (TMZ) (a common drug for glioma). Long non-coding RNA (lncRNA) RMRP has been found to be implicated in glioma progression. However, the effect of RMRP on TMZ resistance along with related molecular mechanisms is poorly defined in glioma. In the present study, RMRP, ZNRF3, and IGF2BP3 were screened out by bioinformatics analysis. The expression levels of lncRNAs and mRNAs were measured by RT-qPCR assay. Protein levels of genes were detected by western blot and immunofluorescence assays. ZNRF3 mRNA stability was analyzed using Actinomycin D assay. Cell proliferative ability and survival rate were determined by CCK-8 assay. Cell apoptotic pattern was estimated by flow cytometry. The effect of RMRP knockdown on the growth of TMZ-treated glioma xenograft tumors was explored in vivo. The relationships of IGF2BP3, RMRP, and ZNRF3 were explored by bioinformatics prediction analysis, RNA immunoprecipitation, luciferase, and RNA pull-down, and chromatin immunoprecipitation assays. The results showed that RMRP was highly expressed in glioma. RMRP knockdown curbed cell proliferation, facilitated cell apoptosis and reduced TMZ resistance in glioma cells, and hindered the growth of TMZ-treated glioma xenograft tumors. RMRP exerted its functions by down-regulating ZNRF3 in glioma cells. IGF2BP3 interacted with RMRP and ZNRF3 mRNA. IGF2BP3 knockdown weakened the interaction of Argonaute 2 (Ago2) and ZNRF3. RMRP reduced ZNRF3 expression and mRNA stability by IGF2BP3. RMRP knockdown inhibited β-catenin expression by up-regulating ZNRF3. The inhibition of Wnt/β-catenin signaling pathway by XAV-939 weakened RMRP-mediated TMZ resistance in glioma cells. β-catenin promoted RMRP expression by TCF4 in glioma cells. In conclusion, RMRP/ZNRF3 axis and Wnt/β-catenin signaling formed a positive feedback loop to regulate TMZ resistance in glioma. The sustained activation of Wnt/β-catenin signaling by RMRP might contribute to the better management of cancers.
Collapse
Affiliation(s)
- Tie Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Jie Hu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Bo Han
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Shishan Tan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Wenqing Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yu Xin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
44
|
Targeting miRNAs with anesthetics in cancer: Current understanding and future perspectives. Biomed Pharmacother 2021; 144:112309. [PMID: 34653761 DOI: 10.1016/j.biopha.2021.112309] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
Anesthetics are extensively used during cancer surgeries. The progression of cancer can be influenced by perioperative events such as exposure to general or local anesthesia. However, whether they inhibit cancer or act as a causative factor for metastasis and exert deleterious effects on cancer growth differs based on the type of cancer and the therapy administration. Recent experimental data suggested that many of the most commonly used anesthetics in surgical oncology, whether general or local agents, can alter gene expression and cause epigenetic changes via modulating miRNAs. miRNAs are single-stranded non-coding RNAs that regulate gene expression at various levels, and their dysregulation contributes to the pathogenesis of cancers. However, anesthetics via regulating miRNAs can concurrently target several effectors of cellular signaling pathways involved in cell differentiation, proliferation, and viability. This review summarized the current research about the effects of different anesthetics in regulating cancer, with a particular emphasis on the role of miRNAs. A significant number of studies conducted in this area of research illuminate the effects of anesthetics on the regulation of miRNA expression; therefore, we hope that a thorough understanding of the underlying mechanisms involved in the regulation of miRNA in the context of anesthesia-induced cancer regulation could help to define optimal anesthetic regimens and provide better perspectives for further studies.
Collapse
|
45
|
Abstract
ABSTRACT Gliomas and glioblastoma comprise the majority of brain malignancies and are difficult to treat despite standard of care and advances in immunotherapy. The challenges of controlling glioma growth and recurrence involve the uniquely immunosuppressive tumor microenvironment and systemic blunting of immune responses. In addition to highlighting key features of glioma and glioblastoma composition and immunogenicity, this review presents several future directions for immunotherapy, such as vaccines and synergistic combination treatment regimens, to better combat these tumors.
Collapse
|
46
|
Hu M, Li Z, Qiu J, Zhang R, Feng J, Hu G, Ren J. CKS2 (CDC28 protein kinase regulatory subunit 2) is a prognostic biomarker in lower grade glioma: a study based on bioinformatic analysis and immunohistochemistry. Bioengineered 2021; 12:5996-6009. [PMID: 34494924 PMCID: PMC8806895 DOI: 10.1080/21655979.2021.1972197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gliomas account for the highest cases of primary brain malignancies. Whereas previous studies have demonstrated the roles of CDC28 Protein Kinase Regulatory Subunit 2 (CKS2) in various cancer types, its functions in lower grade gliomas (LGGs) remain elusive. This study aimed to profile the expression and functions of CKS2 in LGG. Multiple online databases such as The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), Gene Expression Profiling Interactive Analysis 2nd edition (GEPIA2), Tumor Immune Estimation Resource 2nd edition (TIMER2.0) as well as Gene Expression Omnibus (GEO) were used in this study. Immunohistochemistry (IHC) was performed to evaluate CKS2 protein expression. Our data demonstrated upregulation of CKS2 in LGG tissues at both mRNA and protein level, especially in grade III gliomas. Similarly, there was increased expression of CKS2 in isocitrate dehydrogenase 1 (IDH1) wildtype gliomas. In addition, increased DNA copy number and DNA hypomethylation might be associated with the upregulation of the CKS2 in LGG. Using the Kaplan–Meier survival analysis and the Cox regression analysis, CKS2 was shown to be independently associated with poor prognosis of LGG patients. Receiver operating characteristic (ROC) analysis revealed that CKS2 could effectively predict the 1-, 3- and 5-year survival rates of LGG patients. Enrichment analyses revealed that CKS2 was mainly involved in the regulation of the cell cycle in LGG. Taken together, our study demonstrated that CKS2 might be a candidate prognostic biomarker for LGG and could predict the survival rates of LGG patients. Abbreviations: LGG: lower grade glioma; CKS2: CDC28 protein kinase regulatory subunit 2; TCGA: The Cancer Genome Atlas; CGGA: the Chinese Glioma Genome Atlas; GEO: Gene Expression Omnibus; GEPIA: Gene Expression Profiling Interactive Analysis; TIMER: Tumor Immune Estimation Resource; IHC: immunohistochemistry; qRT-PCR: quantitative real-time polymerase chain reaction; PBS: phosphate buffered saline; DAB: diaminobenzidine tetrachloride; OS: overall survival; CAN: copy number alteration; IDH: Isocitrate dehydrogenase; GSEA: Gene Set Enrichment Analysis; DEG: differentially expressed gene; KEGG: Kyoto encyclopedia of genes and genomes; GO: Gene ontology; BP: biological process; CC: cellular component; MF: molecular function; NES: normalized enrichment score; NOM: nominal; FDR: false discovery rate
Collapse
Affiliation(s)
- Menglong Hu
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zongkuo Li
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jinhuan Qiu
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Zhang
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junkai Feng
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Guiming Hu
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingli Ren
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Liu H, Li Z, Sun H. MiR-493-5p inhibits the malignant development of gliomas via suppressing E2F3-mediated dysfunctions of P53 and PI3K/AKT pathways. Clin Transl Oncol 2021; 24:363-370. [PMID: 34460057 DOI: 10.1007/s12094-021-02698-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Gliomas is a major challenge of current medical system, and thousands of people are struggling in the pain of this disease worldwide. In the last decade, the functions of miRNAs have been revealed by many studies, and the intervention on miRNA dysfunctions has been thought as a promising way to counter cancer. MiR-493-5p has been identified as a tumor inhibitor to suppress the progressions of several tumors while its role in gliomas remains unknown. Hence, the study investigated the expression levels of miR-493-5p in glioma tissues and cell lines. METHODS CCK-8 assay, transwell assay and flow cytometry assay were used to observe the effects of miR-493-5p on tumor cells. The downstream targets of miR-493-5p were also searched and verified with online databases and dual-luciferase reporter assay. Moreover, the activities of P53 and PI3K/AKT pathways were also explored by western blot to illustrate the regulation mechanism of miR-493-5p on glioma development. RESULTS The results showed that miR-493-5p was significantly downregulated in pathological tissues and glioma cell lines, and the increased miR-493-5p effectively inhibited the malignant behavior and promoted the apoptosis of glioma cells. CONCLUSIONS E2F3 was confirmed as a target of miR-493-5p, and the effects of miR-493-5p on the phenotype of glioma cells could be partly reversed by E2F3. Besides, it was also found that miR-493-5p could effectively suppress the expression of E2F3 and then improve the dysfunctions of the P53 and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Hong Liu
- Department of Oncology, Binzhou Center Hospital, Shandong, 251700, China
| | - Zhen Li
- Department of Neurosurgery, LinYi People's Hospital, Shandong, China
| | - Hu Sun
- Department of Neurosurgery, Zibo Central Hospital, No. 54 West Communist Youth League Road, Shandong, 255000, China.
| |
Collapse
|
48
|
He Y, Chen Y, Tong Y, Long W, Liu Q. Identification of a circRNA-miRNA-mRNA regulatory network for exploring novel therapeutic options for glioma. PeerJ 2021; 9:e11894. [PMID: 34434651 PMCID: PMC8351580 DOI: 10.7717/peerj.11894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
Background Glioma is the most common brain neoplasm with a poor prognosis. Circular RNA (circRNA) and their associated competing endogenous RNA (ceRNA) network play critical roles in the pathogenesis of glioma. However, the alteration of the circRNA-miRNA-mRNA regulatory network and its correlation with glioma therapy haven't been systematically analyzed. Methods With GEO, GEPIA2, circBank, CSCD, CircInteractome, mirWalk 2.0, and mirDIP 4.1, we constructed a circRNA-miRNA-mRNA network in glioma. LASSO regression and multivariate Cox regression analysis established a hub mRNA signature to assess the prognosis. GSVA was used to estimate the immune infiltration level. Potential anti-glioma drugs were forecasted using the cMap database and evaluated with GSEA using GEO data. Results A ceRNA network of seven circRNAs (hsa_circ_0030788/0034182/0000227/ 0018086/0000229/0036592/0002765), 15 miRNAs(hsa-miR-1200/1205/1248/ 1303/3925-5p/5693/581/586/599/607/640/647/6867-5p/767-3p/935), and 46 mRNAs (including 11 hub genes of ARHGAP11A, DRP2, HNRNPA3, IGFBP5, IP6K2, KLF10, KPNA4, NRP2, PAIP1, RCN1, and SEMA5A) was constructed. Functional enrichment showed they influenced majority of the hallmarks of tumors. Eleven hub genes were proven to be decent prognostic signatures for glioma in both TCGA and CGGA datasets. Forty-six LASSO regression significant genes were closely related to immune infiltration. Finally, five compounds (fulvestrant, tanespimycin, mifepristone, tretinoin, and harman) were predicted as potential treatments for glioma. Among them, mifepristone and tretinoin were proven to inhibit the cell cycle and DNA repair in glioma. Conclusion This study highlights the potential pathogenesis of the circRNA-miRNA-mRNA regulatory network and identifies novel therapeutic options for glioma.
Collapse
Affiliation(s)
- Yi He
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yihong Chen
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yuxin Tong
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Wenyong Long
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Qing Liu
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| |
Collapse
|
49
|
Feng F, Zhao Z, Zhou Y, Cheng Y, Wu X, Heng X. CUX1 Facilitates the Development of Oncogenic Properties Via Activating Wnt/β-Catenin Signaling Pathway in Glioma. Front Mol Biosci 2021; 8:705008. [PMID: 34422906 PMCID: PMC8377541 DOI: 10.3389/fmolb.2021.705008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Homeobox cut like 1 (CUX1), which often presents aberrated expression in many cancer cells, exerts a crucial role in tumorigenesis. Evidence describing CUX1 in gliomagenesis is scarce, and the effects of CUX1 on the Wnt/β-catenin pathway have not been reported. Our study aimed to explore the biological functions and molecular mechanisms involved in CUX1 activity in glioma. Methods: Datasets for bioinformatics analysis were obtained from the GEO, TCGA, CGGA, GTEX and CCLE databases. qRT-PCR, western blotting (WB), and immunohistochemistry (IHC) assays were used to investigate the expression patterns of CUX1 among glioma and brain tissues. CUX1 knockdown and overexpression vectors were transfected into glioma cell lines, the CCK-8, clone formation assay, wound healing, Transwell assay, and flow cytometry were performed to detect changes in cell viability, invasiveness, and the cell cycle. WB and immunofluorescence (IF) assays were used to explore changes in cell cycle-related and Wnt/β-catenin signaling protein levels. Results: Overexpression of CUX1 was identified in glioma tissues, and especially in glioblastoma (GBM), when compared to normal controls and correlated with poor prognosis. In comparison with untreated cells, TJ905 glioma cells overexpressing CUX1 showed higher proliferation and invasion abilities and S phase cell-cycle arrest, while the knockdown of CUX1 suppressed cell invasive ability and induced G1 phase arrest. Active Wnt/β-catenin signaling was enriched and clustered in a CUX1-associated GSEA/GSVA analysis. IF and WB assays indicated that CUX1 regulated the distribution of Axin2/β-catenin in glioma cells and regulated the expression of proteins downstream of the Wnt/β-catenin signaling pathway, suggesting that CUX1 served as an upstream positive regulator of the Wnt/β-catenin pathway. Finally, the knockdown of Axin2 or β-catenin could reverse the tumor-promoting effects caused by CUX1 overexpression, suggesting that CUX1 induced gliomagenesis and malignant phenotype by activating the Wnt/β-catenin signaling pathway. Conclusion: Our data suggested that the transcription factor CUX1 could be a novel therapeutic target for glioma with gene therapy.
Collapse
Affiliation(s)
- Fan Feng
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Zongqing Zhao
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Yunfei Zhou
- Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Yanhao Cheng
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Xiujie Wu
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Xueyuan Heng
- Institute of Brain Science and Brain-Like Intelligence, Linyi People's Hospital, Linyi, China.,Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| |
Collapse
|
50
|
Behrooz AB, Vazifehmand R, Tajudin AA, Masarudin MJ, Sekawi Z, Masomian M, Syahir A. Tailoring drug co-delivery nanosystem for mitigating U-87 stem cells drug resistance. Drug Deliv Transl Res 2021; 12:1253-1269. [PMID: 34405338 DOI: 10.1007/s13346-021-01017-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 12/17/2022]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent form of brain tumor, which generally has a poor prognosis. According to consensus, recurrence of the tumor and chemotherapy resistance acquisition are the two distinguishing features of GBM originated from glioblastoma stem cells (GSCs). To eliminate these obstacles inherent in GBM chemotherapy, targeting GSCs through a smart drug delivery system has come to the front position of GBM therapeutics. In this study, B19 aptamer (Apt)-conjugated polyamidoamine (PAMAM) G4C12 dendrimer nanoparticles (NPs), called Apt-NPs, were formulated for the co-delivery of paclitaxel (PTX) and temozolomide (TMZ) to U-87 stem cells. These drugs were loaded using a double emulsification solvent evaporation method. As a result, drug-loaded Apt-NPs significantly inhibited the tumor growth of U-87 stem cells, by the initiation of apoptosis via the downregulation of autophagic and multidrug resistance (MDR) genes. Additionally, by their downregulation by qPCR of CD133, CD44, SOX2, and the canonical Wnt/β-catenin pathway, cell proliferation has substantially decreased. Altogether, the results demonstrate that this intelligent drug co-delivery system is capable of effectively transferring PTX and TMZ to U-87 stem cells and without any toxic effect on Apt-NPs alone to U-87 stem cells. Furthermore, the designed dendrimer-based pharmaceutical system along with single-stranded B19 aptamer might be utilized as a new therapeutic strategy for the treatment of U-87 stem cells drug resistance in the GBM.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Reza Vazifehmand
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Human Genetic, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Asilah Ahmad Tajudin
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mas Jaffri Masarudin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Malihe Masomian
- Centre of Virus and Vaccine Research, School of Medical and Life Science, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia. .,MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|