1
|
Chen J, Cui J, Jiao B, Zheng Z, Yu H, Wang H, Zhang G, Lai S, Gan Z, Yu Q. Biomimetic Nanosensitizer Potentiates Efficient Glioblastoma Gene-Radiotherapy through Synergistic Hypoxia Mitigation and PLK1 Silencing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58241-58261. [PMID: 39287499 DOI: 10.1021/acsami.4c11566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Postoperative radiotherapy currently stands as the cornerstone of glioblastoma (GBM) treatment. Nevertheless, low-dose radiotherapy has been proven ineffective for GBM, due to hypoxia in the GBM microenvironment, which renders the resistance to radiation-induced cell death. Moreover, the overexpression of the PLK1 gene in glioma cells enhances GBM proliferation, invasion, metastasis, and resistance to radiation. This study introduced a hybrid membrane-camouflaged biomimetic lipid nanosensitizer (CNL@miPA), which efficiently encapsulated gold nanoclusters (PA) and miR-593-5p by a chimeric membrane derived from lipids, cancer cells, and natural killer cells. CNL@miPA exhibited exceptional blood-brain barrier and tumor tissue penetration, effectively ameliorating hypoxia and synergizing with radiotherapy. By enabling prolonged miRNA circulation in the bloodstream and achieving high enrichment at the tumor site, CNL@miPA significantly suppressed tumor growth in combination treatment, thereby significantly extending the survival period of treated mice. Overall, the developed biomimetic nanosensitizer represented an efficient and multifunctional targeted delivery system, offering a novel strategy for gene-radiotherapy of GBM.
Collapse
Affiliation(s)
- Jiawei Chen
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajunzi Cui
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Binbin Jiao
- Department of Urology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Ziyan Zheng
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Haiwang Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hanbing Wang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Guan Zhang
- Department of Urology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shicong Lai
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
- The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
2
|
Yang WL, Zhang WF, Wang Y, Lou Y, Cai Y, Zhu J. Origin recognition complex 6 overexpression promotes growth of glioma cells. Cell Death Dis 2024; 15:485. [PMID: 38971772 PMCID: PMC11227543 DOI: 10.1038/s41419-024-06764-w] [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: 01/05/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 07/08/2024]
Abstract
The discovery of novel oncotargets for glioma is of immense significance. We here explored the expression patterns, biological functions, and underlying mechanisms associated with ORC6 (origin recognition complex 6) in glioma. Through the bioinformatics analyses, we found a significant increase in ORC6 expression within human glioma tissues, correlating with poorer overall survival, higher tumor grade, and wild-type isocitrate dehydrogenase status. Additionally, ORC6 overexpression is detected in glioma tissues obtained from locally-treated patients and across various primary/established glioma cells. Further bioinformatics scrutiny revealed that genes co-expressed with ORC6 are enriched in multiple signaling cascades linked to cancer. In primary and immortalized (A172) glioma cells, depleting ORC6 using specific shRNA or Cas9-sgRNA knockout (KO) significantly decreased cell viability and proliferation, disrupted cell cycle progression and mobility, and triggered apoptosis. Conversely, enhancing ORC6 expression via a lentiviral construct augmented malignant behaviors in human glioma cells. ORC6 emerged as a crucial regulator for the expression of key oncogenic genes, including Cyclin A2, Cyclin B2, and DNA topoisomerase II (TOP2A), within glioma cells. Silencing or KO of ORC6 reduced the mRNA and protein levels of these genes, while overexpression of ORC6 increased their expression in primary glioma cells. Bioinformatics analyses further identified RBPJ as a potential transcription factor of ORC6. RBPJ shRNA decreased ORC6 expression in primary glioma cells, while its overexpression increased it. Additionally, significantly enhanced binding between the RBPJ protein and the proposed ORC6 promoter region was detected in glioma tissues and cells. In vivo experiments demonstrated a significant reduction in the growth of patient-derived glioma xenografts in the mouse brain subsequent to ORC6 KO. ORC6 depletion, inhibited proliferation, decreased expression of Cyclin A2/B2/TOP2A, and increased apoptosis were detected within these ORC6 KO intracranial glioma xenografts. Altogether, RBPJ-driven ORC6 overexpression promotes glioma cell growth, underscoring its significance as a promising therapeutic target.
Collapse
Affiliation(s)
- Wen-Lei Yang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Feng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yin Wang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yue Lou
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Cai
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Zhu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Pang SG, Zhang X, Li ZX, He LF, Chen F, Liu ML, Huang YZ, Mo JM, Luo KL, Xiao JJ, Zhu F. TOPK Inhibition Enhances the Sensitivity of Colorectal Cancer Cells to Radiotherapy by Reducing the DNA Damage Response. Curr Med Sci 2024; 44:545-553. [PMID: 38900386 DOI: 10.1007/s11596-024-2884-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/22/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE Abnormal expression of T-lymphokine-activated killer cell-originated protein kinase (TOPK) was reported to be closely related to the resistance of prostate cancer to radiotherapy and to targeted drug resistance in lung cancer. However, the role of TOPK inhibition in enhancing radiosensitivity of colorectal cancer (CRC) cells is unclear. This study aimed to evaluate the radiosensitization of TOPK knockdown in CRC cells. METHODS The expression of TOPK was detected in CRC tissues by immunohistochemistry, and the effect of TOPK knockdown was detected in CRC cells by Western blotting. CCK-8 and clonogenic assays were used to detect the growth and clonogenic ability of CRC cells after TOPK knockdown combined with radiotherapy in CRC cells. Furthermore, proteomic analysis showed that the phosphorylation of TOPK downstream proteins changed after radiotherapy. DNA damage was detected by the comet assay. Changes in the DNA damage response signaling pathway were analyzed by Western blotting, and apoptosis was detected by flow cytometry. RESULTS The expression of TOPK was significantly greater in CRC tissues at grades 2-4 than in those at grade 1. After irradiation, CRC cells with genetically silenced TOPK had shorter comet tails and reduced expression levels of DNA damage response-associated proteins, including phospho-cyclin-dependent kinase 1 (p-CDK1), phospho-ataxia telangiectasia-mutated (p-ATM), poly ADP-ribose polymerase (PARP), and meiotic recombination 11 homolog 1 (MRE11). CONCLUSIONS TOPK was overexpressed in patients with moderately to poorly differentiated CRC. Moreover, TOPK knockdown significantly enhanced the radiosensitivity of CRC cells by reducing the DNA damage response.
Collapse
Affiliation(s)
- Shi-Gui Pang
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Xin Zhang
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Zhao-Xin Li
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Li-Fei He
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Feng Chen
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Ming-Long Liu
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Ying-Ze Huang
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Jian-Mei Mo
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Kong-Lan Luo
- Department of Oncology, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Juan-Juan Xiao
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, 475000, China.
| | - Feng Zhu
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, 475000, China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
5
|
Naik A, Lattab B, Qasem H, Decock J. Cancer testis antigens: Emerging therapeutic targets leveraging genomic instability in cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200768. [PMID: 38596293 PMCID: PMC10876628 DOI: 10.1016/j.omton.2024.200768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cancer care has witnessed remarkable progress in recent decades, with a wide array of targeted therapies and immune-based interventions being added to the traditional treatment options such as surgery, chemotherapy, and radiotherapy. However, despite these advancements, the challenge of achieving high tumor specificity while minimizing adverse side effects continues to dictate the benefit-risk balance of cancer therapy, guiding clinical decision making. As such, the targeting of cancer testis antigens (CTAs) offers exciting new opportunities for therapeutic intervention of cancer since they display highly tumor specific expression patterns, natural immunogenicity and play pivotal roles in various biological processes that are critical for tumor cellular fitness. In this review, we delve deeper into how CTAs contribute to the regulation and maintenance of genomic integrity in cancer, and how these mechanisms can be exploited to specifically target and eradicate tumor cells. We review the current clinical trials targeting aforementioned CTAs, highlight promising pre-clinical data and discuss current challenges and future perspectives for future development of CTA-based strategies that exploit tumor genomic instability.
Collapse
Affiliation(s)
- Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Boucif Lattab
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Hanan Qasem
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
6
|
Mao P, Wang T, Du CW, Yu X, Wang MD. CXCL5 promotes tumorigenesis and angiogenesis of glioblastoma via JAK-STAT/NF-κb signaling pathways. Mol Biol Rep 2023; 50:8015-8023. [PMID: 37541997 DOI: 10.1007/s11033-023-08671-3] [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: 03/14/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND The tumor microenvironment contains chemokines that play a crucial role in various processes, such as tumorigenesis, inflammation, and therapy resistance, in different types of cancer. CXCL5 is a significant chemokine that has been shown to promote tumor proliferation, invasion, angiogenesis, and therapy resistance when overexpressed in various types of cancer. This research aims to investigate the impact of CXCL5 on the biological functions of glioblastoma (GBM). METHODS The TCGA GBM and GEO databases were utilized to perform transcriptome microarray analysis and oncogenic signaling pathway analysis of CXCL5 in GBM. Validation of CXCL5 expression was performed using RT-qPCR and Western Blot. The impact of CXCL5 on cell proliferation, tumorigenesis, and angiogenesis in GBM was assessed through various methods, including cell proliferation assay, cloning assay, intracranial xenograft tumor models, and tube formation assay. Clinical prognosis was evaluated in 59 samples of gliomas with varying degrees of malignancy (grades 2, 3, and 4) and the TCGA GBM database, based on CXCL5 expression levels. The activities of the JAK-STAT and NF-κB signaling pathways were detected using Western Blot. RESULTS The expression of CXCL5 was highly enriched in GBM. Moreover, the inhibition of CXCL5 showed a significant efficacy in suppressing cellular proliferation and angiogenesis, resulting in extended survival rates in xenograft mouse models in comparison to the control group. Notably, pretreatment with dapsone exhibited a reversal of the impact of CXCL5 on the formation of colonies and tubes in GBM cells. Elevated expression of CXCL5 was correlated with poor outcomes in GBM patients. Furthermore, the overexpression of CXCL5 has been associated with the activation of JAK-STAT and NF-κB signaling pathways. CONCLUSIONS CXCL5 plays an important role in tumorigenesis and angiogenesis, indicating the potential for novel therapies targeting CXCL5 in GBM.
Collapse
Affiliation(s)
- Ping Mao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Tuo Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Chang-Wang Du
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xiao Yu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Mao-De Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
7
|
Tang X, Wang Z, Xie Y, Liu Y, Yang K, Li T, Shen H, Zhao M, Jin J, Xiao H, Liu H, Gu N. Radiation-Triggered Selenium-Engineered Mesoporous Silica Nanocapsules for RNAi Therapy in Radiotherapy-Resistant Glioblastoma. ACS NANO 2023; 17:4062-4076. [PMID: 36759969 DOI: 10.1021/acsnano.3c00269] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Radiotherapy-resistant glioblastoma (rrGBM) remains a significant clinical challenge because of high infiltrative growth characterized by activation of antiapoptotic signal transduction. Herein, we describe an efficiently biodegradable selenium-engineered mesoporous silica nanocapsule, initiated by high-energy X-ray irradiation and employed for at-site RNA interference (RNAi) to inhibit rrGBM invasion and achieve maximum therapeutic benefit. Our radiation-triggered RNAi nanocapsule showed high physiological stability, good blood-brain barrier transcytosis, and potent rrGBM accumulation. An intratumoral RNAi nanocapsule permitted low-dose X-ray radiation-triggered dissociation for cofilin-1 knockdown, inhibiting rrGBM infiltration. More importantly, tumor suppression was further amplified by electron-affinity aminoimidazole products converted from metronidazole polymers under X-ray radiation-exacerbated hypoxia, which sensitized cell apoptosis to ionizing radiation by fixing reactive oxygen species-induced DNA lesions. In vivo experiments confirmed that our RNAi nanocapsule reduced tumor growth and invasion, prolonging survival in an orthotopic rrGBM model. Generally, we present a promising radiosensitizer that would effectively improve rrGBM-patient outcomes with low-dose X-ray irradiation.
Collapse
Affiliation(s)
- Xianglong Tang
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Zhen Wang
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Yandong Xie
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Yuyang Liu
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Taiping Li
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Hong Shen
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Juan Jin
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, The Affiliated Brain Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Hongyi Liu
- Department of Neurosurgery, The Affiliated Brain Hospital with Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing 210029, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
8
|
Zhang L, Liu B, Su J. CCNB2 as a potential biomarker of bladder cancer via the high throughput technology. Medicine (Baltimore) 2023; 102:e32825. [PMID: 36820589 PMCID: PMC9907924 DOI: 10.1097/md.0000000000032825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
Bladder cancer and oral squamous cell carcinoma (OSCC) seriously affect people's health. However, the relationship between bladder cancer and OSCC remains unclear. Got GSE138206, GSE146483, GSE184616, and bladder cancer datasets GSE65635, GSE100926 from Gene Expression Omnibus database. Weighted gene co-expression network analysis was used to identify the significant module. Functional enrichment analysis was performed via the Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes. Furthermore, the Gene Set Enrichment Analysis was also used to complete the enrichment analysis. Comparative Toxicogenomics Database found most relevant diseases to core genes. TargetScan is used to forecast analysis of microRNA and target genes. In Gene Ontology analysis, differentially expressed genes were mostly concentrated in cell differentiation, extrallular region, structural molecule activity, and actin binding. In Kyoto Encyclopedia of Genes and Genomes analysis, the differentially expressed genes were mainly enriched in PI3K-Akt signaling pathway, pathway in cancer, and extracellular matrix-receptor interaction. Seven hub genes (cyclin B2 [CCNB2], TK1, CDC20, PCNA, CKS1B, CDCA5, MCM4) were obtained. Hub genes (CCNB2, CDC20) are highly expressed in OSCC and bladder cancer samples. CCNB2 was one common oncogene of bladder cancer and OSCC.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Urology Surgery, Fuxing Hospital Affiliated to Capital Medical University, Xicheng District, Beijing, China
- * Correspondence: Lei Zhang, Department of Urology Surgery, Fuxing Hospital Affiliated to Capital Medical University, No. 20 Fuxingmenwai Dajie, Xicheng District, Beijing 100038, China (e-mail: )
| | - Bin Liu
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, Hebei, PR China
| | - Jianzhi Su
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, Hebei, PR China
| |
Collapse
|
9
|
Li F, Liu C, Nong W, Lin L, Ge Y, Luo B, Xiao S, Zhang Q, Xie X. Identification of potential biomarkers in cancer testis antigens for glioblastoma. Am J Transl Res 2023; 15:799-816. [PMID: 36915736 PMCID: PMC10006807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/16/2022] [Indexed: 03/16/2023]
Abstract
OBJECTIVE To screen and validate cancer testis antigens (CTAs) as potential biomarkers and explore their molecular mechanisms in glioblastoma (GBM). METHODS Ribonucleic acid sequencing (RNA-seq) and bioinformatics analyses were utilized to screen the highly expressed CTAs in GBM. Correlation analysis was used to identify potential biomarkers associated with tumor purity and prognosis. Immunohistochemistry was applied for detection of protein expression. Protein-protein interaction (PPI) network construction, functional enrichment analysis, and binding domain prediction were performed to investigate the underlying molecular mechanisms of GBM. RESULTS A total of 8 highly expressed CTAs were identified in GBM. One of them was PDZ-binding kinase (PBK). PBK messenger RNA (mRNA) was most highly expressed in GBM and associated with tumor purity and prognosis, PBK protein expression was also significantly increased in GBM tissues and correlated with p53 expression. Functional enrichment analysis revealed that the PBK related genes were predominantly enriched in cell cycle pathway with 38 genes enriched. The proteins encoding by these 38 genes were performed by binding domain prediction analysis, which demonstrated 15 proteins interacting with PBK. Most of these proteins were up regulated in GBM. CONCLUSION PBK is highly expressed in GBM. It may serve as a potential biomarker for GBM targeting therapy and the cell cycle modulator by interacting with certain key molecules of cell cycle in GBM.
Collapse
Affiliation(s)
- Feng Li
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China
| | - Chang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University Nanning, Guangxi, China.,Postdoctoral Research Station, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China
| | - Weixia Nong
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Lina Lin
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China
| | - Yingying Ge
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Shaowen Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University Nanning, Guangxi, China
| | - Qingmei Zhang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| |
Collapse
|
10
|
Tan B, Zhang J, Wang W, Ma H, Yang Y. Tumor-suppressive E3 ubiquitin ligase CHIP inhibits the PBK/ERK axis to repress stem cell properties and radioresistance in non-small cell lung cancer. Apoptosis 2022; 28:397-413. [PMID: 36436119 DOI: 10.1007/s10495-022-01789-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/28/2022]
Abstract
Recently, radioresistant cancer cells surviving radiotherapy have been suggested to show more aggressive phenotypes than parental cells, and the underlying mechanisms may be associated with cancer stem cells. This study provided novel mechanistic insights for E3 ubiquitin ligase CHIP in stem cell properties and radioresistance of non-small cell lung cancer (NSCLC). After bioinformatic prediction for key genes involved, NSCLC tissues and cells were collected to measure the expression of CHIP and PBK. E3 ubiquitin ligase CHIP was poorly expressed, while PBK was highly expressed in NSCLC tissues and cells. CHIP reduced the protein stability of PBK through the ubiquitin-protease pathway to repress the activation of ERK pathway. Based on the gain- or loss-of-function experiments, it was noted that restoration of CHIP curtailed stem cell properties and radioresistance in NSCLC, as manifested by inhibited sphere formation and cell proliferation, decreased number of CD133+CD44+ cells and expression of OCT4, SOX2, and NANOG, as well as facilitated apoptosis of NSCLC cells. Besides, in vivo animal experiments further confirmed that CHIP restrained tumorigenic ability and improved radiosensitivity of NSCLC cells by inhibiting PBK/ERK axis. Collectively, CHIP suppressed stem cell properties and radioresistance of NSCLC cells by inhibiting PBK/ERK axis, therefore offering a potential therapeutic target for enhancing efficacy of radiotherapy.
Collapse
Affiliation(s)
- Bo Tan
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China.
| | - Jingwei Zhang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China
| | - Wen Wang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China
| | - Haibo Ma
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yuanyuan Yang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui, Zhengzhou, 450008, Henan, China
| |
Collapse
|
11
|
In Silico Analysis of the Correlation of KIF2C with Prognosis and Immune Infiltration in Glioma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6320828. [PMID: 35387222 PMCID: PMC8977321 DOI: 10.1155/2022/6320828] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
Glioblastoma (GBM) is one of the most commonly pivotal malignant caners. Numerous reports have revealed the crucial roles of immune infiltration in the initiation and progression of GBM. In this study, we first identified differentially expressed genes (DEGs) in the progression of GBM using CGGA databases. Totally, 156 upregulated DEGs and 251 downregulated DEGs were revealed. By constructing a protein-protein interaction network, KIF2C was identified as a hub gene in GBM. Further analysis revealed an evidently positive association existing in KIF2C expression and the advanced stages of gliomas. Higher expression of KIF2C was in WHO grade IV samples relative to that in grade III and grade II samples. In addition, our results showed that KIF2C was higher in IDH1 wild-type samples than IDH1 mutant glioma samples, in 1p/19q noncodel samples than 1p/19q code glioma samples, and in recurrent samples than primary glioma samples. Moreover, our results showed that higher expression of KIF2C correlated with shorter survival time in both primary and recurrent gliomas and could act as a potential biomarker for the prognosis of GBM. Further analysis demonstrated that higher expression of KIF2C was related to higher levels of endothelial cell, T cell CD8+ naïve, common lymphoid progenitor, T cell CD4+ Th2, T cell CD4+ Th2, macrophage, macrophage M1, T cell CD4+ memory, and T cell CD4+ effector memory, but was related to lower levels of NK cell, B cell plasma, T cell CD4+ Th1, T cell regulatory (Tregs), neutrophil, and T cell NK. We thought this study could provide potential biomarkers for the prediction of prognosis and immune infiltration of gliomas.
Collapse
|
12
|
Guha A, Waris S, Nabors LB, Filippova N, Gorospe M, Kwan T, King PH. The versatile role of HuR in Glioblastoma and its potential as a therapeutic target for a multi-pronged attack. Adv Drug Deliv Rev 2022; 181:114082. [PMID: 34923029 PMCID: PMC8916685 DOI: 10.1016/j.addr.2021.114082] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/15/2021] [Accepted: 12/12/2021] [Indexed: 02/03/2023]
Abstract
Glioblastoma (GBM) is a malignant and aggressive brain tumor with a median survival of ∼15 months. Resistance to treatment arises from the extensive cellular and molecular heterogeneity in the three major components: glioma tumor cells, glioma stem cells, and tumor-associated microglia and macrophages. Within this triad, there is a complex network of intrinsic and secreted factors that promote classic hallmarks of cancer, including angiogenesis, resistance to cell death, proliferation, and immune evasion. A regulatory node connecting these diverse pathways is at the posttranscriptional level as mRNAs encoding many of the key drivers contain adenine- and uridine rich elements (ARE) in the 3' untranslated region. Human antigen R (HuR) binds to ARE-bearing mRNAs and is a major positive regulator at this level. This review focuses on basic concepts of ARE-mediated RNA regulation and how targeting HuR with small molecule inhibitors represents a plausible strategy for a multi-pronged therapeutic attack on GBM.
Collapse
Affiliation(s)
- Abhishek Guha
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Saboora Waris
- Shaheed Zulfiqar Ali Bhutto Medical University, PIMS, G-8, Islamabad, Pakistan
| | - Louis B Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Natalia Filippova
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, United States
| | - Thaddaeus Kwan
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Peter H King
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, United States.
| |
Collapse
|
13
|
Teodoro A, Gonçalves FJ, Oliveira H, Marques S. Venom of Viperidae: A Perspective of its Antibacterial and Antitumor
Potential. Curr Drug Targets 2022; 23:126-144. [DOI: 10.2174/1389450122666210811164517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022]
Abstract
:
The emergence of multi-drug resistant bacteria and limitations on cancer treatment represent
two important challenges in modern medicine. Biological compounds have been explored with
a particular focus on venoms. Although they can be lethal or cause considerable damage to humans,
venom is also a source rich in components with high therapeutic potential.
:
Viperidae family is one of the most emblematic venomous snake families and several studies highlighted
the antibacterial and antitumor potential of viper toxins. According to the literature, these
activities are mainly associated to five protein families - svLAAO, Disintegrins, PLA2, SVMPs and
C-type lectins- that act through different mechanisms leading to the inhibition of the growth of bacteria,
as well as, cytotoxic effects and inhibition of metastasis process. In this review, we provide
an overview of the venom toxins produced by species belonging to the Viperidae family, exploring
their roles during the envenoming and their pharmacological properties, in order to demonstrate its
antibacterial and antitumor potential.
Collapse
Affiliation(s)
- André Teodoro
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Fernando J.M. Gonçalves
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
- CESAM- Centre for Environmental and
Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Helena Oliveira
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
- CESAM- Centre for Environmental and
Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Sérgio Marques
- Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
- CESAM- Centre for Environmental and
Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
14
|
Lee DH, Jeong YJ, Won JY, Sim HI, Park Y, Jin HS. PBK/TOPK Is a Favorable Prognostic Biomarker Correlated with Antitumor Immunity in Colon Cancers. Biomedicines 2022; 10:biomedicines10020299. [PMID: 35203508 PMCID: PMC8869639 DOI: 10.3390/biomedicines10020299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitor therapy has proven efficacy in a subset of colon cancer patients featuring a deficient DNA mismatch repair system or a high microsatellite instability profile. However, there is high demand for more effective biomarkers to expand the colon cancer population responding to ICI therapy. PBK/TOPK, a serine/threonine kinase, plays a role in cell cycle regulation and mitotic progression. Here, we investigated the correlation between PBK/TOPK expression and tumor immunity and its prognostic value in colon cancer. Based on large-scale bioinformatics analysis, we discovered that elevated PBK/TOPK expression predicted a favorable outcome in patients with colon cancer and was positively associated with immune infiltration levels of CD8+ T cells, CD4+ T cells, natural killer cells, and M1 macrophages. In contrast, a negative correlation was found between PBK/TOPK expression and immune suppressor cells, including regulatory T cells and M2 macrophages. Furthermore, the expression of PBK/TOPK was correlated with the expression of T-cell cytotoxicity genes in colon cancer. Additionally, high PBK/TOPK expression was associated with mutations in DNA damage repair genes, and thus with increased tumor mutation and neoantigen burden. These findings suggest that PBK/TOPK may serve as a prognostic and predictive biomarker for immunotherapy in colon cancer.
Collapse
Affiliation(s)
- Dong-Hee Lee
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (D.-H.L.); (Y.-J.J.); (J.-Y.W.)
| | - Yu-Jeong Jeong
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (D.-H.L.); (Y.-J.J.); (J.-Y.W.)
| | - Ju-Young Won
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (D.-H.L.); (Y.-J.J.); (J.-Y.W.)
| | - Hye-In Sim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Yoon Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Correspondence: (Y.P.); (H.-S.J.)
| | - Hyung-Seung Jin
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (D.-H.L.); (Y.-J.J.); (J.-Y.W.)
- Correspondence: (Y.P.); (H.-S.J.)
| |
Collapse
|
15
|
Deng R, Li J, Zhao H, Zou Z, Man J, Cao J, Yang L. Identification of potential biomarkers associated with immune infiltration in papillary renal cell carcinoma. J Clin Lab Anal 2021; 35:e24022. [PMID: 34606125 PMCID: PMC8605132 DOI: 10.1002/jcla.24022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/06/2021] [Accepted: 09/12/2021] [Indexed: 12/27/2022] Open
Abstract
Background Immunotherapeutic approaches have recently emerged as effective treatment regimens against various types of cancer. However, the immune‐mediated mechanisms surrounding papillary renal cell carcinoma (pRCC) remain unclear. This study aimed to investigate the tumor microenvironment (TME) and identify the potential immune‐related biomarkers for pRCC. Methods The CIBERSORT algorithm was used to calculate the abundance ratio of immune cells in each pRCC samples. Univariate Cox analysis was used to select the prognostic‐related tumor‐infiltrating immune cells (TIICs). Multivariate Cox regression analysis was performed to develop a signature based on the selected prognostic‐related TIICs. Then, these pRCC samples were divided into low‐ and high‐risk groups according to the obtained signature. Analyses using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were performed to investigate the biological function of the DEGs (differentially expressed genes) between the high‐ and low‐risk groups. The hub genes were identified using a weighted gene co‐expression network analysis (WGCNA) and a protein‐protein interaction (PPI) analysis. The hub genes were subsequently validated by multiple clinical traits and databases. Results According to our analyses, nine immune cells play a vital role in the TME of pRCC. Our analyses also obtained nine potential immune‐related biomarkers for pRCC, including TOP2A, BUB1B, BUB1, TPX2, PBK, CEP55, ASPM, RRM2, and CENPF. Conclusion In this study, our data revealed the crucial TIICs and potential immune‐related biomarkers for pRCC and provided compelling insights into the pathogenesis and potential therapeutic targets for pRCC.
Collapse
Affiliation(s)
- Ran Deng
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| | - Jianpeng Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| | - Hong Zhao
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| | - Zhirui Zou
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| | - Jiangwei Man
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| | - Jinlong Cao
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| | - Li Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Gansu Province for Urological Diseases, Lanzhou, China.,Clinical Center of Gansu Province for Nephron-urology, Lanzhou, China
| |
Collapse
|
16
|
Wang Y, Zhang H, Wang M, He J, Guo H, Li L, Wang J. CCNB2/SASP/Cathepsin B & PGE2 Axis Induce Cell Senescence Mediated Malignant Transformation. Int J Biol Sci 2021; 17:3538-3553. [PMID: 34512164 PMCID: PMC8416730 DOI: 10.7150/ijbs.63430] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Glioma is the most frequent and aggressive adult brain tumor with maximum mortality. However, the gene alteration and mechanism underlying malignant transformation of glioma remain largely unknown. We aimed to find key factors regulating tumor progression and malignant transformation of glioma. Here we compared the gene expression profiles of 693 glioma patients by HGG vs. LGG model, and identified a key factor CCNB2 for malignant transformation in glioma. CCNB2 induced a senescence-associated secretory phenotype (SASP) of glioma cells, and the malignant progression, such as invasion and excessive proliferation was mediated by secreting SASP cytokines, Cathepsin B and PGE2. These findings demonstrated a previously undiscovered link between senescence, CCNB2/SASP/Cathepsin B & PGE2 axis and malignant transformation in glioma. This might provide novel insights on developing new therapeutic regimens for abrogating aggressiveness of glioma.
Collapse
Affiliation(s)
- Ying Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China
| | - Hanbing Zhang
- Department of Neurosurgery, Shanghai Deji Hospital, Qingdao University, Shanghai 200331, China
| | - Minglei Wang
- Department of Neurosurgery, PuTuo District People's Hospital, Shanghai 200060, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Hui Guo
- Department of Neurosurgery, Shanghai Deji Hospital, Qingdao University, Shanghai 200331, China
| | - Lihua Li
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China
| | - Jie Wang
- Department of Neurosurgery, Shanghai Deji Hospital, Qingdao University, Shanghai 200331, China
| |
Collapse
|
17
|
Cyclin B2 (CCNB2) Stimulates the Proliferation of Triple-Negative Breast Cancer (TNBC) Cells In Vitro and In Vivo. DISEASE MARKERS 2021; 2021:5511041. [PMID: 34354775 PMCID: PMC8331305 DOI: 10.1155/2021/5511041] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer. Currently, targeting therapy makes great advances for the treatment of TNBC, whereas more effective therapeutic targets are urgently needed. Cyclin B2 (CCNB2), which belongs to B-type cyclins, is known as a cell cycle regulator. CCNB2 is synthesized at G1 phase in cancer cells and downregulated at anaphase. The defects of CCNB2 led to the abnormal cell cycle and tumorigenesis. Though there are wide effects of CCNB2 on multiple types of tumors, the potential role of CCNB2 in TNBC progression is still unclear. Herein, we found that CCNB2 was highly expressed in human TNBC tissues and correlated with the prognosis and clinical pathological features including tumor size (p = 0.022∗) and pTNM stage (p = 0.021∗) of patients with TNBC. CCNB2 could promote the proliferation of TNBC cells in vitro and in mice. Our findings therefore confirmed the involvement of CCNB2 in TNBC progression and provided the evidence that CCNB2 could serve as a promising molecular target of TNBC.
Collapse
|
18
|
Huang H, Lee MH, Liu K, Dong Z, Ryoo Z, Kim MO. PBK/TOPK: An Effective Drug Target with Diverse Therapeutic Potential. Cancers (Basel) 2021; 13:cancers13092232. [PMID: 34066486 PMCID: PMC8124186 DOI: 10.3390/cancers13092232] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Cancer is a major public health problem worldwide, and addressing its morbidity, mortality, and prevalence is the first step towards appropriate control measures. Over the past several decades, many pharmacologists have worked to identify anti-cancer targets and drug development strategies. Within this timeframe, many natural compounds have been developed to inhibit cancer growth by targeting kinases, such as AKT, AURKA, and TOPK. Kinase assays and computer modeling are considered to be effective and powerful tools for target screening, as they can predict physical interactions between small molecules and their bio-molecular targets. In the present review, we summarize the inhibitors and compounds that target TOPK and describe its role in cancer progression. The extensive body of research that has investigated the contribution of TOPK to cancer suggests that it may be a promising target for cancer therapy. Abstract T-lymphokine-activated killer cell-originated protein kinase (TOPK, also known as PDZ-binding kinase or PBK) plays a crucial role in cell cycle regulation and mitotic progression. Abnormal overexpression or activation of TOPK has been observed in many cancers, including colorectal cancer, triple-negative breast cancer, and melanoma, and it is associated with increased development, dissemination, and poor clinical outcomes and prognosis in cancer. Moreover, TOPK phosphorylates p38, JNK, ERK, and AKT, which are involved in many cellular functions, and participates in the activation of multiple signaling pathways related to MAPK, PI3K/PTEN/AKT, and NOTCH1; thus, the direct or indirect interactions of TOPK make it a highly attractive yet elusive target for cancer therapy. Small molecule inhibitors targeting TOPK have shown great therapeutic potential in the treatment of cancer both in vitro and in vivo, even in combination with chemotherapy or radiotherapy. Therefore, targeting TOPK could be an important approach for cancer prevention and therapy. Thus, the purpose of the present review was to consider and analyze the role of TOPK as a drug target in cancer therapy and describe the recent findings related to its role in tumor development. Moreover, this review provides an overview of the current progress in the discovery and development of TOPK inhibitors, considering future clinical applications.
Collapse
Affiliation(s)
- Hai Huang
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea;
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju, Jeollanamdo 58245, Korea;
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou 450001, China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou 450001, China
| | - Zeayoung Ryoo
- School of Life Science, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (Z.R.); (M.O.K.); Tel.: +82-54-530-1234 (M.O.K.)
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea;
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
- Correspondence: (Z.R.); (M.O.K.); Tel.: +82-54-530-1234 (M.O.K.)
| |
Collapse
|
19
|
Zhou J, Guo H, Liu L, Hao S, Guo Z, Zhang F, Gao Y, Wang Z, Zhang W. Construction of co-expression modules related to survival by WGCNA and identification of potential prognostic biomarkers in glioblastoma. J Cell Mol Med 2021; 25:1633-1644. [PMID: 33449451 PMCID: PMC7875936 DOI: 10.1111/jcmm.16264] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/29/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumour with poor prognosis. The potential pathogenesis and therapeutic target are still need to be explored. Herein, TCGA expression profile data and clinical information were downloaded, and the WGCNA was conducted. Hub genes which closely related to poor prognosis of GBM were obtained. Further, the relationship between the genes of interest and prognosis of GBM, and immune microenvironment were analysed. Patients from TCGA were divided into high‐ and low‐risk group. WGCNA was applied to the high‐ and low‐risk group and the black module with the lowest preservation was identified which could distinguish the prognosis level of these two groups. The top 10 hub genes which were closely related to poor prognosis of patients were obtained. GO analysis showed the biological process of these genes mainly enriched in: Cell cycle, Progesterone‐mediated oocyte maturation and Oocyte meiosis. CDCA5 and CDCA8 were screened out as the genes of interest. We found that their expression levels were closely related to overall survival. The difference analysis resulted from the TCGA database proved both CDCA5 and CDCA8 were highly expressed in GBM. After transfection of U87‐MG cells with small interfering RNA, it revealed that knockdown of the CDCA5 and CDCA8 could influence the biological behaviours of proliferation, clonogenicity and apoptosis of GBM cells. Then, single‐gene analysis was performed. CDCA5 and CDCA8 both had good correlations with genes that regulate cell cycle in the p53 signalling pathway. Moreover, it revealed that high amplification of CDCA5 was correlated with CD8+ T cells while CDCA8 with CD4+ T cells in GBM. These results might provide new molecular targets and intervention strategy for GBM.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China.,Shanxi University of Chinese Medicine, Taiyuan, China
| | - Hao Guo
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Likun Liu
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Shulan Hao
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Zhi Guo
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Fupeng Zhang
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Yu Gao
- Department of Oncology, Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Zhi Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Weiwei Zhang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, China
| |
Collapse
|
20
|
Ou A, Yung WKA, Majd N. Molecular Mechanisms of Treatment Resistance in Glioblastoma. Int J Mol Sci 2020; 22:E351. [PMID: 33396284 PMCID: PMC7794986 DOI: 10.3390/ijms22010351] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults and is almost invariably fatal. Despite our growing understanding of the various mechanisms underlying treatment failure, the standard-of-care therapy has not changed over the last two decades, signifying a great unmet need. The challenges of treating glioblastoma are many and include inadequate drug or agent delivery across the blood-brain barrier, abundant intra- and intertumoral heterogeneity, redundant signaling pathways, and an immunosuppressive microenvironment. Here, we review the innate and adaptive molecular mechanisms underlying glioblastoma's treatment resistance, emphasizing the intrinsic challenges therapeutic interventions must overcome-namely, the blood-brain barrier, tumoral heterogeneity, and microenvironment-and the mechanisms of resistance to conventional treatments, targeted therapy, and immunotherapy.
Collapse
Affiliation(s)
| | - W. K. Alfred Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 431, Houston, TX 77030, USA;
| | - Nazanin Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 431, Houston, TX 77030, USA;
| |
Collapse
|