101
|
Zhou H, Tang L, Shang ZF, Zhou PK, Li M. PIG3 downregulation enhances the radio sensitivity of NSCLC cells by promoting G2/M cell cycle arrest and apoptosis. RADIATION MEDICINE AND PROTECTION 2022. [DOI: 10.1016/j.radmp.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
102
|
Wang J, Jin Z, Wu G, Deng Z, Wang J, Xu B, Zhu H, Guo Y, Wen Z. Construction of a 3-mRNA hypoxia prognostic model to evaluate immune microenvironment in hepatocellular carcinoma. Medicine (Baltimore) 2022; 101:e30589. [PMID: 36181125 PMCID: PMC9524961 DOI: 10.1097/md.0000000000030589] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Hypoxia is a key factor in the development of hepatocellular carcinoma (HCC), which is the most common primary liver cancer with poor prognosis. The current study aimed to identify the potential prognostic biomarkers of the hypoxia-associated gene signature in patients with HCC, and to further explore the relationship between hypoxia and immune infiltration. METHODS After the determination of differentially expressed genes (DEGs) using the HCC transcriptome data of The Cancer Genome Atlas database and hypoxia-related gene set, the prognosis-associated genes were identified using univariate Cox regression analysis. Then, the hypoxia prognosis model was established via multivariate Cox regression analysis, with functional annotation conducted using Gene Set Enrichment Analysis. CIBERSORT was utilized to analyze the degree of tumor immune invasion, and an International Cancer Genome Consortium cohort to verify the reliability of the prognosis model. Expression levels of hypoxia-associated genes were detected by real-time quantitative polymerase chain reaction in HCC samples. RESULTS 3 genes (ENO1, SAP30, and STC2) constructed the hypoxia prognosis model. The patients were subdivided into 2 groups based on median risk score, with a high hypoxic score indicating poor prognosis of HCC. The hypoxia signature could be employed as an independent prognostic factor in HCC. In addition, the proportion of macrophages was higher in the high-risk group. CONCLUSION The hypoxia-associated signature could be a potential prognostic marker of HCC and provides a different perspective for immunotherapy of HCC.
Collapse
Affiliation(s)
- Jue Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zongrui Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guolin Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhenfeng Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jilong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Banghao Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hai Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ya Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhang Wen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Zhang Wen, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6#, Nanning, Guangxi 530021, China (e-mail: )
| |
Collapse
|
103
|
Supramolecular Hydrogel-Wrapped Gingival Mesenchymal Stem Cells in Cutaneous Radiation Injury. Cells 2022; 11:cells11193089. [PMID: 36231051 PMCID: PMC9564043 DOI: 10.3390/cells11193089] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 12/12/2022] Open
Abstract
Radiation-induced skin wound/dermatitis is one of the common side effects of radiotherapy or interventional radiobiology. Gingiva-derived mesenchymal stem cells (GMSCs) were indicated to have therapeutic potentials in skin diseases. However, stem cells are prone to spread and difficult to stay in the skin for a long time, limiting their curative effects and application. This study investigated the therapeutic efficacy of Nap-GDFDFpDY (pY-Gel) self-assembled peptide hydrogel-encapsulated GMSCs to treat 137Cs γ-radiation-induced skin wounds in mice. The effects were evaluated by skin damage score, hind limb extension measurement and histological and immunohistochemical analysis. In vivo studies showed that pY-Gel self-assembled peptide hydrogel-encapsulated GMSCs could effectively improve wound healing in irradiated skin tissues. In addition, it was found that GMSCs conditioned medium (CM) could promote the proliferation, migration and DNA damage repair ability of skin cells after irradiation in human keratinocyte cell line HaCaT and normal human dermal fibroblasts (HFF). Mechanistically, GMSCs-CM can promote the expression of epidermal growth factor receptor (EGFR), signal transducers and activators of transcription 3 (STAT3) and matrix metalloproteinases (MMPs), suggesting that activation of the EGFR/STAT3 signaling pathway may be involved in the repair of skin cells after exposure to radiations. In conclusion, pY-Gel self-assembled peptide hydrogel-encapsulated GMSCs have a beneficial therapeutic effect on radiation-induced cutaneous injury and may serve as a basis of novel cells therapeutic approach.
Collapse
|
104
|
Rajput M, Mishra D, Kumar K, Singh RP. Silibinin Radiosensitizes EGF Receptor-knockdown Prostate Cancer Cells by Attenuating DNA Repair Pathways. J Cancer Prev 2022; 27:170-181. [PMID: 36258717 PMCID: PMC9537578 DOI: 10.15430/jcp.2022.27.3.170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/27/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Emergence of radioresistance in prostate cancer (PCa) cells is a major obstacle in cancer therapy and contributes to the relapse of the disease. EGF receptor (EGFR) signaling plays an important role in the development of radioresistance. Herein, we have assessed the modulatory effects of silibinin on radiation-induced resistance via DNA repair pathways in EGFR-knockdown DU145 cells. shRNA-based silencing of EGFR was done in radioresistant human PCa DU145 cells and effects of ionizing radiation (IR) and silibinin were assessed using clonogenic and trypan blue assays. Furthermore, radiosensitizing effects of silibinin on PCa in context with EGFR were analyzed using flow cytometry, comet assay, and immunoblotting. Silibinin decreased the colony formation ability with an increased death of DU145 cells exposed to IR (5 Gray), with a concomitant decrease in Rad51 protein expression. Silibinin (25 μM) augmented the IR-induced cytotoxic effect in EGFR-knockdown PCa cells, along with induction of G2/M phase cell cycle arrest. Further, we studied homologous recombination (HR) and non-homologous end joining (NHEJ) pathways in silibinin-induced DNA double-strand breaks in EGFR-knockdown DU145 cells. Silibinin down-regulated the expression of Rad51 and DNA-dependent protein kinase proteins without any considerable effect on Ku70 and Ku80 in IR-exposed EGFR-knockdown PCa cells. The pro-survival signaling proteins, phospho-extracellular signal-regulated kinases (ERK)1/2, phospho-Akt and phospho-STAT3 were decreased by silibinin in EGFR-deficient PCa cells. These findings suggest a novel mechanism of silibinin-induced radiosensitization of PCa cells by targeting DNA repair pathways, HR and NHEJ, and suppressing the pro-survival signaling pathways, ERK1/2, Akt and STAT3, in EGFR-knockdown PCa cells.
Collapse
Affiliation(s)
- Mohit Rajput
- Cancer Biology Laboratory, School of Life Sciences, New Delhi, India
| | - Deepali Mishra
- Cancer Biology Laboratory, School of Life Sciences, New Delhi, India
| | - Kunal Kumar
- Cancer Biology Laboratory, School of Life Sciences, New Delhi, India
| | - Rana P. Singh
- Cancer Biology Laboratory, School of Life Sciences, New Delhi, India,Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India,Correspondence to Rana P. Singh, E-mail: , https://orcid.org/0000-0003-4261-7044
| |
Collapse
|
105
|
Mechanisms of microRNA action in rectal cancer radiotherapy. Chin Med J (Engl) 2022; 135:2017-2025. [PMID: 35943251 PMCID: PMC9746734 DOI: 10.1097/cm9.0000000000002139] [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] [Indexed: 02/04/2023] Open
Abstract
ABSTRACT Preoperative neoadjuvant chemoradiotherapy, combined with total mesorectal excision, has become the standard treatment for advanced localized rectal cancer (RC). However, the biological complexity and heterogeneity of tumors may contribute to cancer recurrence and metastasis in patients with radiotherapy-resistant RC. The identification of factors leading to radioresistance and markers of radiosensitivity is critical to identify responsive patients and improve radiotherapy outcomes. MicroRNAs (miRNAs) are small, endogenous, and noncoding RNAs that affect various cellular and molecular targets. miRNAs have been shown to play important roles in multiple biological processes associated with RC. In this review, we summarized the signaling pathways of miRNAs, including apoptosis, autophagy, the cell cycle, DNA damage repair, proliferation, and metastasis during radiotherapy in patients with RC. Also, we evaluated the potential role of miRNAs as radiotherapeutic biomarkers for RC.
Collapse
|
106
|
Ding K, He Y, Wei J, Fu S, Wang J, Chen Z, Zhang H, Qu Y, Liang K, Gong X, Qiu L, Chen D, Xiao B, Du H. A score of DNA damage repair pathway with the predictive ability for chemotherapy and immunotherapy is strongly associated with immune signaling pathway in pan-cancer. Front Immunol 2022; 13:943090. [PMID: 36081518 PMCID: PMC9445361 DOI: 10.3389/fimmu.2022.943090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
DNA damage repair (DDR) is critical in maintaining normal cellular function and genome integrity and is associated with cancer risk, progression, and therapeutic response. However, there is still a lack of a thorough understanding of the effects of DDR genes’ expression level in cancer progression and therapeutic resistance. Therefore, we defined a tumor-related DDR score (TR-DDR score), utilizing the expression levels of 20 genes, to quantify the tumor signature of DNA damage repair pathways in tumors and explore the possible function and mechanism for the score among different cancers. The TR-DDR score has remarkably predictive power for tumor tissues. It is a more accurate indicator for the response of chemotherapy or immunotherapy combined with the tumor-infiltrating lymphocyte (TIL) and G2M checkpoint score than the pre-existing predictors (CD8 or PD-L1). This study points out that the TR-DDR score generally has positive correlations with patients of advanced-stage, genome-instability, and cell proliferation signature, while negative correlations with inflammatory response, apoptosis, and p53 pathway signature. In the context of tumor immune response, the TR-DDR score strongly positively correlates with the number of T cells (CD4+ activated memory cells, CD8+ cells, T regs, Tfh) and macrophages M1 polarization. In addition, by difference analysis and correlation analysis, COL2A1, MAGEA4, FCRL4, and ZIC1 are screened out as the potential modulating factors for the TR-DDR score. In summary, we light on a new biomarker for DNA damage repair pathways and explore its possible mechanism to guide therapeutic strategies and drug response prediction.
Collapse
Affiliation(s)
- Ke Ding
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Youhua He
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shuying Fu
- College of Life Science, Zhaoqing University, Zhaoqing, China
| | - Jiajian Wang
- Clinical Laboratory Department of Longgang District People’s Hospital of Shenzhen & The Second Affiliated Hospital of the Chinese University of Hong Kong, Shenzhen, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Haibo Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yimo Qu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Keying Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xiaocheng Gong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Li Qiu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Dong Chen
- Fangrui Institute of Innovative Drugs, South China University of Technology, Guangzhou, China
| | - Botao Xiao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Botao Xiao, ; Hongli Du,
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Botao Xiao, ; Hongli Du,
| |
Collapse
|
107
|
DNA Repair Inhibitors Potentiate Fractionated Radiotherapy More Than Single-Dose Radiotherapy in Breast Cancer Cells. Cancers (Basel) 2022; 14:cancers14153794. [PMID: 35954456 PMCID: PMC9367425 DOI: 10.3390/cancers14153794] [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: 06/06/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 12/10/2022] Open
Abstract
Pharmacological inhibitors of DNA damage response (DDR) proteins, such as the ataxia-telangiectasia mutated (ATM) and ataxia-telangiectasia and Rad3-related (ATR) kinases and poly (ADP-ribose) polymerase (PARP), have been developed to overcome tumor radioresistance. Despite demonstrating radiosensitization preclinically, they have performed suboptimally in clinical trials, possibly due to an incomplete understanding of the influence of DDR inhibition on ionizing radiation (IR) dose fractionation and sublethal damage repair. Hence, this study aimed to evaluate the radiosensitizing ability under fractionation of ATM inhibitor AZD0156, ATR inhibitor AZD6738 and PARP inhibitor AZD2281 (olaparib), utilizing MDA-MB-231 and MCF-7 human breast cancer cells. Clonogenic assays were performed to assess cell survival and sublethal damage repair after treatment with DDR inhibitors and either single-dose or fractionated IR. Immunofluorescence microscopy was utilized to evaluate DNA double-strand break repair kinetics. Cell cycle distributions were investigated using flow cytometry. All inhibitors showed significant radiosensitization, which was significantly greater following fractionated IR than single-dose IR. They also led to more unrepaired DNA double-strand breaks at 24 h post-IR. This study provides preclinical evidence for the role of AZD0156, AZD6738 and olaparib as radiosensitizing agents. Still, it highlights the need to evaluate these drugs in fractionated settings mirroring clinical practice to optimize the trial design.
Collapse
|
108
|
Wang Y, Oda S, Suzuki MG, Mitani H, Aoki F. Cell cycle-dependent radiosensitivity in mouse zygotes. DNA Repair (Amst) 2022; 117:103370. [PMID: 35863142 DOI: 10.1016/j.dnarep.2022.103370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/22/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022]
Abstract
Mammalian zygotes are hypersensitive to radiation exposure compared with later-stage embryos and somatic cells, which may be due to an unusual DNA damage response (DDR). DNA damage checkpoints are an essential part of the DDR, allowing for faithful replication of cells. Although the DDR and radiosensitivity of somatic cells are dependent on the cell cycle phase, it remains largely unclear how the irradiation of zygotes at different phases affects cell cycle progression and preimplantation development. Here, mouse zygotes were irradiated with 10 Gy γ-rays at all four cell cycle phases. DNA damage checkpoints were activated by γ-irradiation at the G2 phase, but not at the G1, S, and M phases. The absence of DNA damage checkpoints at the G1 and M phases seems to be due to the low abundance of phosphorylated CHK2, which plays a key role in checkpoint activation in response to ionizing radiation. The cause of the inoperative S phase checkpoint may lie downstream of CHK2 activation. The inactive DNA damage checkpoints at the G1 and S phases contributed to micronucleus formation in the subsequent 2-cell stage, whereas irradiation at the M phase led to the highest incidence of chromatin bridges. The low developmental rates of embryos irradiated at the G1, S, and M phases suggest that embryos with these two types of chromatin abnormalities are prone to developmental failure. Taken together, these results suggest that the radiosensitivity of zygotes can be ascribed to a defective DDR at the G1, S, and M phases.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Shoji Oda
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Masataka G Suzuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Hiroshi Mitani
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Fugaku Aoki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.
| |
Collapse
|
109
|
Guo D, Jin J, Liu J, Wang Y, Li D, He Y. Baicalein Inhibits the Progression and Promotes Radiosensitivity of Esophageal Squamous Cell Carcinoma by Targeting HIF-1A. Drug Des Devel Ther 2022; 16:2423-2436. [PMID: 35937565 PMCID: PMC9346416 DOI: 10.2147/dddt.s370114] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/16/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To explore the mechanism of the effect of baicalein on radioresistance of esophageal cancer, and to provide ideas for the treatment of patients with poor radiotherapy effect of esophageal cancer. Methods The glycolytic rate assay kit was used to detect the changes in glycolytic metabolism in esophageal cancer cells after treatment with baicalein, and mass spectrometry was used to detect whether baicalein could affect the level of glycolysis-related metabolites in esophageal cancer cells. The binding of baicalein to the target protein was simulated by molecular docking technique, the protein expression level was detected by Western Blot, and the changes in the cell cycle were detected by flow cytometry. Results Radiation combined with baicalein could significantly inhibit the proliferation and migration of esophageal cancer cells compared with that of 6 Gy rays alone. The results of the glycolytic rate assay showed that baicalein could inhibit the glycolysis of esophageal cancer cells. Metabonomic studies showed that baicalein could affect the expression levels of glycolysis-related metabolites. The results of network pharmacology showed that baicalein could target several key glycolysis enzymes and glycolysis-related proteins, such as HIF-1A. The results of the WB experiment showed that glycolysis-related proteins and cycle-related proteins were down-regulated after baicalein treatment. Conclusion The main mechanism of baicalein inhibiting radiation resistance of esophageal cancer cells is that targeting HIF-1A protein regulates glucose metabolism and then regulates Cyclin D1/CDK4 axis to change the cell cycle.
Collapse
Affiliation(s)
- Dongli Guo
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Jing Jin
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Jianghui Liu
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Yingying Wang
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Daojuan Li
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Yutong He
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Correspondence: Yutong He, Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Zhongshan East Road Street, Changan District, Shijiazhuang, People’s Republic of China, Tel +8613903398303, Email
| |
Collapse
|
110
|
Jin K, He M, Chen B, Zhou X, Zhang C, Zhang Z, Hu D, Jiang Z, Wei Q, Qiu S, Yang L. A single-sample mRNA molecular classification of bladder cancer predicting prognosis and response to immunotherapy. Transl Androl Urol 2022; 11:943-958. [PMID: 35958899 PMCID: PMC9360513 DOI: 10.21037/tau-21-887] [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: 10/05/2021] [Accepted: 05/18/2022] [Indexed: 11/12/2022] Open
Abstract
Background As an immunogenic cancer, crosstalk between cancer cells and immune cells has been gradually recognized in bladder cancer (BC). Several studies have emphasized the clinical significance of the molecular stratification of BC without highlighting the role of the immune microenvironment. Although immunotherapy acted as a prospective treatment, more precise molecular stratification should be established to select those sensitive to immunotherapy. Methods To select specific immune genes forming subtypes indicating disparate prognoses, we performed bioinformatic analysis using BC transcriptomic profiles from six published datasets, with 408 BC samples in The Cancer Genome Atlas (TCGA) database and 295 individuals in International Cancer Genome Consortium (ICGC) database. Survival analyses were conducted using Kaplan-Meier curves, while Kruskal-Wallis tests were applied to test the differences among groups. Except for unsupervised clustering based on the differential expression of genes, we additionally performed binomial logistic regression, focusing on the mRNA level of a single sample. Results Unsupervised clustering showed that 4 clusters captured the best segmentation. After validation with survival data and simplification using binomial logistic regression, we found that cluster B and cluster D showed worse survival outcomes (P=0.012). Considering the similar survival outcomes of these two clusters, we recombined and performed another survival analysis, which also showed significant survival differences (P=0.0041). Bonding with clinical data, a greater proportion of risk factors were assigned to the worse prognosis subtype, especially showing higher grades in the subtype (P<0.001). In addition, immune cell infiltration, single nucleotide polymorphism (SNP) and copy number variation (CNV) all showed differences between clusters, indicating changes in the immune microenvironment and mutation burden. Through phenotypical analysis, we found metabolism and proliferation phenotypes associated with the immune clusters and mutually exclusive in BC, of which proliferation contributed to worse outcomes. Using the tumor immune dysfunction and exclusion (TIDE) score, a worse immunotherapy benefit was predicted in clusters B&D, defined as the worse prognosis subtype. Conclusions With this novel clustering criterion based on immune-related genes, we provide a better understanding of the immune microenvironment, further guiding the use of immunotherapy.
Collapse
Affiliation(s)
- Kun Jin
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Mingjing He
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Chen
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chichen Zhang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zilong Zhang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Hu
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongyuan Jiang
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shi Qiu
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Center of Biomedical Big Data, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Yang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
111
|
Subtil FSB, Gröbner C, Recknagel N, Parplys AC, Kohl S, Arenz A, Eberle F, Dikomey E, Engenhart-Cabillic R, Schötz U. Dual PI3K/mTOR Inhibitor NVP-BEZ235 Leads to a Synergistic Enhancement of Cisplatin and Radiation in Both HPV-Negative and -Positive HNSCC Cell Lines. Cancers (Basel) 2022; 14:cancers14133160. [PMID: 35804930 PMCID: PMC9265133 DOI: 10.3390/cancers14133160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Head and neck cancers (HNSCCs), especially in the advanced stages, are predominantly treated by radiochemotherapy, including cisplatin. The cure rates are clearly higher for HPV-positive HNSCCs when compared to HPV-negative HNSCCs. For both entities, this treatment is accompanied by serious adverse reactions, mainly due to cisplatin administration. We reported earlier that for both HPV-positive and negative HNSCC cells, the effect of radiotherapy was strongly enhanced when pretreated using the dual PI3K/mTOR inhibitor NVP-BEZ235 (BEZ235). The current study shows that for HPV-positive cells, BEZ235 will strongly enhance the effect of cisplatin alone. More important, preincubation with BEZ235 was found to alter the purely additive effect normally seen when cisplatin is combined with radiation into a strong synergistic enhancement. This tri-modal combination might allow for the enhancement of the effect of radiochemotherapy, even with reduced cisplatin. Abstract The standard of care for advanced head and neck cancers (HNSCCs) is radiochemotherapy, including cisplatin. This treatment results in a cure rate of approximately 85% for oropharyngeal HPV-positive HNSCCs, in contrast to only 50% for HPV-negative HNSCCs, and is accompanied by severe side effects for both entities. Therefore, innovative treatment modalities are required, resulting in a better outcome for HPV-negative HNSCCs, and lowering the adverse effects for both entities. The effect of the dual PI3K/mTOR inhibitor NVP-BEZ235 on a combined treatment with cisplatin and radiation was studied in six HPV-negative and six HPV-positive HNSCC cell lines. Cisplatin alone was slightly more effective in HPV-positive cells. This could be attributed to a defect in homologous recombination, as demonstrated by depleting RAD51. Solely for HPV-positive cells, pretreatment with BEZ235 resulted in enhanced cisplatin sensitivity. For the combination of cisplatin and radiation, additive effects were observed. However, when pretreated with BEZ235, this combination changed into a synergistic interaction, with a slightly stronger enhancement for HPV-positive cells. This increase could be attributed to a diminished degree of DSB repair in G1, as visualized via the detection of γH2AX/53BP1 foci. BEZ235 can be used to enhance the effect of combined treatment with cisplatin and radiation in both HPV-negative and -positive HNSCCs.
Collapse
Affiliation(s)
- Florentine S. B. Subtil
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Carolin Gröbner
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Niklas Recknagel
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Ann Christin Parplys
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Sibylla Kohl
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Andrea Arenz
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Fabian Eberle
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Ekkehard Dikomey
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
- Laboratory of Radiobiology & Experimental Radiooncology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Rita Engenhart-Cabillic
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
| | - Ulrike Schötz
- Department of Radiotherapy and Radiooncology, Philipps-University, 35043 Marburg, Germany; (F.S.B.S.); (C.G.); (N.R.); (A.C.P.); (S.K.); (A.A.); (F.E.); (E.D.); (R.E.-C.)
- Correspondence: ; Tel.: +49-6421-28-21978
| |
Collapse
|
112
|
Wu Z, Huang C, Li R, Li H, Lu H, Lin Z. PRKCI Mediates Radiosensitivity via the Hedgehog/GLI1 Pathway in Cervical Cancer. Front Oncol 2022; 12:887139. [PMID: 35785194 PMCID: PMC9243290 DOI: 10.3389/fonc.2022.887139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Insensitivity to radiotherapy accounts for the majority of therapeutic failures in cervical cancer (CC) patients who undergo radical radiotherapy. We aimed to elucidate the molecular mechanisms underlying radiosensitivity to identify methods to improve the overall 5-year survival rate. The atypical protein kinase C iota (aPKCι) gene PRKCI exhibits tumor-specific copy number amplification (CNA) in CC. We investigated how PRKCI decreases radiosensitivity in CC and assessed the interplay between PRKCI and the Hedgehog (Hh)/GLI1 pathway in the present research. Methods The biological functions of PRKCI in CC radiosensitivity were explored through immunohistochemistry, colony formation, Cell Counting Kit-8 (CCK-8), cell cycle, apoptosis assays, and xenograft models. qRT-PCR, Western blotting analysis, and immunofluorescence assays were utilized to evaluate the interplay between PRKCI and the Hh/GLI1 pathway and its mechanism in PRKCI-decreased radiosensitivity in CC. Furthermore, the effect of auranofin (AF), a selective inhibitor of PKCι, on CC cells was explored through biochemical assays in vitro and in vivo. Results We found that high PRKCI expression was responsible for decreased survival in CC. PRKCI was intimately associated with radiation-triggered alterations in proliferation, the cell cycle, apoptosis, and xenograft growth. The Hh/GLI1 pathway was activated when PRKCI expression was altered. PRKCI functions downstream of the Hh/GLI1 pathway to phosphorylate and activate the transcription factor GLI1. AF acts as a radiosensitizer and showed biological effects in vitro and in vivo. Conclusions PRKCI is a therapeutic target for regulating radiosensitivity in CC. This molecule regulates radiosensitivity by modulating GLI1 relocalization and phosphorylation in CC via the Hh/GLI1 pathway.
Collapse
Affiliation(s)
- Zhuna Wu
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, China
| | - Chunxian Huang
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruixin Li
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Li
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaiwu Lu
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Huaiwu Lu, ; Zhongqiu Lin,
| | - Zhongqiu Lin
- Department of Gynecological Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Huaiwu Lu, ; Zhongqiu Lin,
| |
Collapse
|
113
|
Liu T, Ma L, Song L, Yan B, Zhang S, Wang B, Zuo N, Sun X, Deng Y, Ren Q, Li Y, Zhou J, Liu Q, Wei L. CENPM upregulation by E5 oncoprotein of human papillomavirus promotes radiosensitivity in head and neck squamous cell carcinoma. Oral Oncol 2022; 129:105858. [DOI: 10.1016/j.oraloncology.2022.105858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/10/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022]
|
114
|
Bekker RA, Kim S, Pilon-Thomas S, Enderling H. Mathematical modeling of radiotherapy and its impact on tumor interactions with the immune system. Neoplasia 2022; 28:100796. [PMID: 35447601 PMCID: PMC9043662 DOI: 10.1016/j.neo.2022.100796] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/27/2022] [Accepted: 04/01/2022] [Indexed: 11/01/2022]
Abstract
Radiotherapy is a primary therapeutic modality widely utilized with curative intent. Traditionally tumor response was hypothesized to be due to high levels of cell death induced by irreparable DNA damage. However, the immunomodulatory aspect of radiation is now widely accepted. As such, interest into the combination of radiotherapy and immunotherapy is increasing, the synergy of which has the potential to improve tumor regression beyond that observed after either treatment alone. However, questions regarding the timing (sequential vs concurrent) and dose fractionation (hyper-, standard-, or hypo-fractionation) that result in improved anti-tumor immune responses, and thus potentially enhanced tumor inhibition, remain. Here we discuss the biological response to radiotherapy and its immunomodulatory properties before giving an overview of pre-clinical data and clinical trials concerned with answering these questions. Finally, we review published mathematical models of the impact of radiotherapy on tumor-immune interactions. Ranging from considering the impact of properties of the tumor microenvironment on the induction of anti-tumor responses, to the impact of choice of radiation site in the setting of metastatic disease, these models all have an underlying feature in common: the push towards personalized therapy.
Collapse
|
115
|
Shi M, Wan J, Wang H, Yu H. Cytokeratin 13 promotes radiotherapy sensitivity of nasopharyngeal carcinoma by downregulating the MEK/ERK pathway. IUBMB Life 2022; 74:543-553. [PMID: 35426961 DOI: 10.1002/iub.2617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/22/2022] [Accepted: 04/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Radiation therapy is the first treatment choice for nasopharyngeal carcinoma (NPC), while radiation resistance and recurrence have become the primary factors and are associated with poor prognosis in the clinical treatment of NPC patients. The purpose of the present study was to explore the sensitivity and molecular basis of cytokeratin 13 (CK13) that regulates NPC radiotherapy. METHODS HNE-3 or C666-1 cell line was used for overexpression and knockdown tests. Under radiotherapy conditions, CCK-8 assay, clone formation assay, and flow cytometry analyzed the effects of CK13 overexpression on cell proliferation, apoptosis, and cell cycle, respectively. In addition, Western blotting detected CK13-mediated downregulation of cell cycle-related genes. The mouse subcutaneous tumor-bearing experiment identified the effects of CK13 overexpression on the treatment of NPC in vivo. Further, Western blotting, CCK-8 assay, and flow cytometry investigated whether the CK13-mediated cell apoptosis involves the MEK/ERK signaling pathway. RESULTS Overexpression of CK13 significantly inhibited the survival of HNE-3 cells under radiotherapy in vitro and in vivo, and there was a substantial decrease in cyclin-dependent kinase 4 and 6 (CDK4/6) levels promoting the cell percentage number in the G2/M phase and, subsequently, the ratio of the apoptotic cells. In contrast, the knockdown of CK13 showed the opposite partial regulatory effect. Interestingly, CK13 overexpression also showed a reduction in the survival of C666-1 cells and an increased ratio of the apoptotic cells under radiotherapy treatment. Furthermore, higher levels of CK13 downregulated the MEK/ERK signaling pathway, resulting in decreased HNE-3 cell proliferation and increased apoptosis. However, ERK activators were able to rescue the process partially. CONCLUSIONS Together, these results showed that CK13 promoted the radiosensitivity of NPC cells by downregulating the MEK/ERK signaling pathway. Thus, targeting CK13 provided insights into the treatment of NPC radiotherapy.
Collapse
Affiliation(s)
- Ming Shi
- Department of Otorhinolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yunnan University, the Second People's Hospital of Yunnan Province, Yunnan Eye Hospital, Kunming, China
| | - Jia Wan
- Department of Otorhinolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yunnan University, the Second People's Hospital of Yunnan Province, Yunnan Eye Hospital, Kunming, China
| | - Huan Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yunnan University, the Second People's Hospital of Yunnan Province, Yunnan Eye Hospital, Kunming, China
| | - Hong Yu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yunnan University, the Second People's Hospital of Yunnan Province, Yunnan Eye Hospital, Kunming, China
| |
Collapse
|
116
|
ROS-Related miRNAs Regulate Immune Response and Chemoradiotherapy Sensitivity in Hepatocellular Carcinoma by Comprehensive Analysis and Experiment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4713518. [PMID: 35585886 PMCID: PMC9110211 DOI: 10.1155/2022/4713518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/09/2022] [Indexed: 11/28/2022]
Abstract
Reactive oxygen species (ROS) plays an essential role in the development of cancer. Here, we chose ROS-related miRNAs for consensus clustering analysis and ROS score construction. We find that ROS is extremely associated with prognosis, tumor immune microenvironment (TIME), gene mutations, N6-methyladenosine (m6A) methylation, and chemotherapy sensitivity in hepatocellular carcinoma (HCC). Mechanistically, ROS may affect the prognosis of HCC patients in numerous ways. Moreover, miR-210-3p and miR-106a-5p significantly increased the ROS level and stagnated cell cycle at G2/M in HCC; the results were more obvious in cells after ionizing radiation (IR). Finally, the two miRNAs suppressed cell proliferation, migration, and invasion and promoted apoptosis in huh7 and smmc7721 cells. It indicated that ROS might affect the prognosis of HCC patients through immune response and increase the sensitivity of HCC patients to radiotherapy and chemotherapy.
Collapse
|
117
|
Ma A, Biersack B, Goehringer N, Nitzsche B, Höpfner M. Novel Thienyl-Based Tyrosine Kinase Inhibitors for the Treatment of Hepatocellular Carcinoma. J Pers Med 2022; 12:jpm12050738. [PMID: 35629160 PMCID: PMC9146161 DOI: 10.3390/jpm12050738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
New medical treatments are urgently needed for advanced hepatocellular carcinoma (HCC). Recently, we showed the anticancer effects of novel thiophene-based kinase inhibitors. In this study, we further characterized the antineoplastic effects and modes of action of the two most promising inhibitors, Thio-Iva and Thio-Dam, and compared their effects with the clinically relevant multi-kinase inhibitor, sorafenib, in HCC cells. Crystal violet staining and real-time cell growth monitoring showed pronounced antiproliferative effects in Huh-7 and SNU-449 cells with IC50 values in the (sub-)micromolar range. Long-term incubation experiments revealed the reduced clonogenicity of Thio-Iva and Thio-Dam-treated HCC cells. LDH-release tests excluded cytotoxicity as an unspecific mode of action of the inhibitors, while flow cytometry analysis revealed a dose-dependent and pronounced G2/M phase cell cycle arrest and cyclin B1 suppression. Additionally, mitochondria-driven apoptosis was observed through the cytosolic increase of reactive oxygen species, a concomitant PARP cleavage, and caspase-3 induction. Both compounds were found to effectively inhibit the capillary tube formation of endothelial EA.hy926 cells in vitro, pointing towards additional antiangiogenic effects. Antiangiogenic and antineoplastic effects were confirmed in vivo by CAM assays. In summary, the thienyl-acrylonitrile derivatives, Thio-Iva and Thio-Dam, exert significant antineoplastic and antiangiogenic effects in HCC cells.
Collapse
Affiliation(s)
- Andi Ma
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
| | - Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany;
| | - Nils Goehringer
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
| | - Bianca Nitzsche
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
- Correspondence:
| | - Michael Höpfner
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany; (A.M.); (N.G.); (M.H.)
| |
Collapse
|
118
|
Fornalski KW, Adamowski Ł, Dobrzyński L, Jarmakiewicz R, Powojska A, Reszczyńska J. The radiation adaptive response and priming dose influence: the quantification of the Raper-Yonezawa effect and its three-parameter model for postradiation DNA lesions and mutations. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:221-239. [PMID: 35150289 PMCID: PMC9021059 DOI: 10.1007/s00411-022-00963-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/12/2022] [Indexed: 05/14/2023]
Abstract
The priming dose effect, called also the Raper-Yonezawa effect or simply the Yonezawa effect, is a special case of the radiation adaptive response phenomenon (radioadaptation), which refers to: (a) faster repair of direct DNA lesions (damage), and (b) DNA mutation frequency reduction after irradiation, by applying a small priming (conditioning) dose prior to the high detrimental (challenging) one. This effect is observed in many (but not all) radiobiological experiments which present the reduction of lesion, mutation or even mortality frequency of the irradiated cells or species. Additionally, the multi-parameter model created by Dr. Yonezawa and collaborators tried to explain it theoretically based on experimental data on the mortality of mice with chronic internal irradiation. The presented paper proposes a new theoretical approach to understanding and explaining the priming dose effect: it starts from the radiation adaptive response theory and moves to the three-parameter model, separately for two previously mentioned situations: creation of fast (lesions) and delayed damage (mutations). The proposed biophysical model was applied to experimental data-lesions in human lymphocytes and chromosomal inversions in mice-and was shown to be able to predict the Yonezawa effect for future investigations. It was also found that the strongest radioadaptation is correlated with the weakest cellular radiosensitivity. Additional discussions were focussed on more general situations where many small priming doses are used.
Collapse
Affiliation(s)
- Krzysztof W Fornalski
- National Centre for Nuclear Research (NCBJ), ul. A. Sołtana 7, 05-400, Otwock-Świerk, Poland.
| | - Łukasz Adamowski
- National Centre for Nuclear Research (NCBJ), ul. A. Sołtana 7, 05-400, Otwock-Świerk, Poland
| | - Ludwik Dobrzyński
- National Centre for Nuclear Research (NCBJ), ul. A. Sołtana 7, 05-400, Otwock-Świerk, Poland
| | - Rafał Jarmakiewicz
- Faculty of Physics, Warsaw University of Technology, ul. Koszykowa 75, 00-662, Warsaw, Poland
| | - Aleksandra Powojska
- Faculty of Physics, Warsaw University of Technology, ul. Koszykowa 75, 00-662, Warsaw, Poland
| | - Joanna Reszczyńska
- Department of Biophysics, Physiology and Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw (WUM), ul. T. Chałubińskiego 5, 02-004, Warsaw, Poland
| |
Collapse
|
119
|
Meattini I, Livi L, Lorito N, Becherini C, Bacci M, Visani L, Fozza A, Belgioia L, Loi M, Mangoni M, Lambertini M, Morandi A. Integrating radiation therapy with targeted treatments for breast cancer: from bench to bedside. Cancer Treat Rev 2022; 108:102417. [PMID: 35623219 DOI: 10.1016/j.ctrv.2022.102417] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/02/2022]
|
120
|
Nguyen T, Mueller S, Malbari F. Review: Neurological Complications From Therapies for Pediatric Brain Tumors. Front Oncol 2022; 12:853034. [PMID: 35480100 PMCID: PMC9035987 DOI: 10.3389/fonc.2022.853034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022] Open
Abstract
Surgery, chemotherapy and radiation have been the mainstay of pediatric brain tumor treatment over the past decades. Recently, new treatment modalities have emerged for the management of pediatric brain tumors. These therapies range from novel radiotherapy techniques and targeted immunotherapies to checkpoint inhibitors and T cell transfer therapies. These treatments are currently investigated with the goal of improving survival and decreasing morbidity. However, compared to traditional therapies, these novel modalities are not as well elucidated and similarly has the potential to cause significant short and long-term sequelae, impacting quality of life. Treatment complications are commonly mediated through direct drug toxicity or vascular, infectious, or autoimmune mechanisms, ranging from immune effector cell associated neurotoxicity syndrome with CART-cells to neuropathy with checkpoint inhibitors. Addressing treatment-induced complications is the focus of new trials, specifically improving neurocognitive outcomes. The aim of this review is to explore the pathophysiology underlying treatment related neurologic side effects, highlight associated complications, and describe the future direction of brain tumor protocols. Increasing awareness of these neurologic complications from novel therapies underscores the need for quality-of-life metrics and considerations in clinical trials to decrease associated treatment-induced morbidity.
Collapse
Affiliation(s)
- Thien Nguyen
- Department of Pediatrics, University of San Francisco, San Francisco, CA, United States
- *Correspondence: Thien Nguyen,
| | - Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of San Francisco, San Francisco, CA, United States
| | - Fatema Malbari
- Division of Neurology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
121
|
Chow B, Warkentin B, McEwen M, Huang F, Nanda K, Gamper AM, Menon G. Uncertainties Associated with Clonogenic Assays using a Cs-137 Irradiator and Ir-192 Afterloader: A Comprehensive Compilation for Radiation Researchers. Radiat Res 2022; 198:40-56. [PMID: 35391488 DOI: 10.1667/rade-21-00205.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
Abstract
Clonogenic assays are the gold standard for measuring cell clonogenic survival and enable quantification of a cell line's radiosensitivity through the calculation of the surviving fraction, the ratio of cell clusters (colonies) formed after radiation exposure compared to the number formed without exposure. Such studies regularly utilize Cs-137 irradiators. While uncertainties for specific procedural aspects have been described previously, a comprehensive review has not been completed. We therefore quantified uncertainties associated with clonogenic assays performed using a Cs-137 Shepherd irradiator, and a recently established brachytherapy afterloader in vitro radiation delivery apparatus (BAIRDA), through a series of experiments and a literature review. The clonogenic assay is subject to uncertainties that affect the determination of the surviving fraction (e.g., accuracy of the number of cells seeded, potential effects of hypothermia, and the threshold number of cells for a cluster to be identified as a colony). Furthermore, dose delivery uncertainties related to both the Cs-137 irradiator and BAIRDA were also quantified. The combined standard (k = 1) uncertainty was ± 6.0% in the surviving fraction for the Cs-137 irradiator (±6.3% for BAIRDA), up to ± 1.3% in the dose delivered by the Cs-137 irradiator, and up to ± 2.2% in the dose delivered by BAIRDA. The largest individual uncertainties were associated with the number of cells seeded on a plate (3.4%) and inter-observer variability in counting (4.1%), suggesting that effective reduction of uncertainties in the conduct of the clonogenic assay proper may provide the greatest relief on the uncertainty budget. Finally, measurable impact on experimental findings was assessed by applying this uncertainty to clonogenic assays of SW756 cells using either a Cs-137 irradiator or BAIRDA, introducing a maximum shift in the reported radiobiological parameters a/b and T1/2 of 0.3 Gy and 0.4 h, respectively, while the 95% confidence interval increased by 0.5 Gy and decreased by 0.4 h, respectively. Though the overall impact on radiobiological parameter estimation was small, the individual uncertainties could have a significant influence in other applications of in vitro experiments in radiation biology. Hence, better understanding of the uncertainties associated with both clonogenic assays and the radiation source used can improve the accuracy of experimental analysis and reproducibility of the results.
Collapse
Affiliation(s)
- Braden Chow
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Brad Warkentin
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Malcolm McEwen
- Ionizing Radiation Standards, National Research Council of Canada, Ottawa, Canada
| | - Fleur Huang
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Kareena Nanda
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Armin M Gamper
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Geetha Menon
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
122
|
Gong L, Zhang Y, Zhao J, Zhang Y, Tu K, Jiao L, Xu Q, Zhang M, Han S. All-In-One Biomimetic Nanoplatform Based on Hollow Polydopamine Nanoparticles for Synergistically Enhanced Radiotherapy of Colon Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107656. [PMID: 35150039 DOI: 10.1002/smll.202107656] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/04/2022] [Indexed: 06/14/2023]
Abstract
Even though radiotherapy is the most important therapeutic strategy for colon cancer treatment, there is an enormous demand to improve radiosensitivity in solid tumor destruction. For this purpose, a biomimetic nanoplatform based on hollow polydopamine nanoparticles (HP) with homologous targeting and pH-responsive drug release properties is designed. In this work, HP is constructed by using a chelation competition-induced polymerization strategy and then modified with the cancer cell membrane. Hollow polydopamine integrated with Pt nanoparticles (Pt@HP) has a catalase-like activity, which can be used to trigger endogenous H2 O2 into O2 , relieving hypoxia of the tumor microenvironment (TME). With mesoporous shells and large cavities, Pt@HP shows efficient apoptin100-109 (AP) and verteporfin (VP) loading to form AVPt@HP@M. Under X-ray irradiation, AVPt@HP@M exerts a radiosensitization effect via multiple strategies, including relieving hypoxia (Pt NPs), enhancing tumor apoptosis (AP), and X-ray-induced photodynamic therapy (X-PDT) (VP). Further metabonomics analysis shows that the specific mechanism of the AVPt@HP@M is through influencing purine metabolism. Without appreciable systemic toxicity, this nanoplatform highlights a new strategy for effective radiosensitization and provides a reference for treating malignant tumors.
Collapse
Affiliation(s)
- Liuyun Gong
- Department of Radiotherapy, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Jing Zhao
- Department of Radiotherapy, the First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, 215000, China
| | - Yilei Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lianying Jiao
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, 710061, China
| | - Suxia Han
- Department of Radiotherapy, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
123
|
Audisio M, Tucci M, Di Stefano RF, Parlagreco E, Ungaro A, Turco F, Audisio A, Di Prima L, Ortega C, Di Maio M, Scagliotti GV, Buttigliero C. New emerging targets in advanced urothelial carcinoma: is it the primetime for personalized medicine? Crit Rev Oncol Hematol 2022; 174:103682. [DOI: 10.1016/j.critrevonc.2022.103682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022] Open
|
124
|
Wang Y, Yu T, Han Y, He Y, Song Y, Guo L, An L, Yang C, Wang F. Phosphorylation of MAD2 at Ser195 Promotes Spindle Checkpoint Defects and Sensitizes Cancer Cells to Radiotherapy in ATM Deficient Cells. Front Cell Dev Biol 2022; 10:817831. [PMID: 35309941 PMCID: PMC8924061 DOI: 10.3389/fcell.2022.817831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
The spindle assembly checkpoint (SAC) is a critical monitoring device in mitosis for the maintenance of genomic stability. Specifically, the SAC complex comprises several factors, including Mad1, Mad2, and Bub1. Ataxia-telangiectasia mutated (ATM) kinase, the crucial regulator in DNA damage response (DDR), also plays a critical role in mitosis by regulating Mad1 dimerization and SAC. Here, we further demonstrated that ATM negatively regulates the phosphorylation of Mad2, another critical component of the SAC, which is also involved in DDR. Mechanistically, we found that phosphorylation of Mad2 is aberrantly increased in ATM-deficient cells. Point-mutation analysis further revealed that Serine 195 mainly mediated Mad2 phosphorylation upon ATM ablation. Functionally, the phosphorylation of Mad2 causes decreased DNA damage repair capacity and is related to the resistance to cancer cell radiotherapy. Altogether, this study unveils the key regulatory role of Mad2 phosphorylation in checkpoint defects and DNA damage repair in ATM-deficient cells.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianyu Yu
- Department of General Surgery, Pudong New Area Gongli Hospital Affiliated to Naval Military Medical University, Naval Military Medical University, Shanghai, China
| | - Yi Han
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Leiming Guo
- Department of R&D, Shanghai Creative Immune Therapeutics Co., Ltd, Shanghai, China
| | - Liwei An
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunying Yang
- Central Laboratory, Shanghai Putuo District People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China.,Central Laboratory, Shanghai Putuo District People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
125
|
Celora GL, Bader SB, Hammond EM, Maini PK, Pitt-Francis JM, Byrne HM. DNA-structured mathematical model of cell-cycle progression in cyclic hypoxia. J Theor Biol 2022; 545:111104. [PMID: 35337794 DOI: 10.1016/j.jtbi.2022.111104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 01/22/2023]
Abstract
New experimental data have shown how the periodic exposure of cells to low oxygen levels (i.e., cyclic hypoxia) impacts their progress through the cell-cycle. Cyclic hypoxia has been detected in tumours and linked to poor prognosis and treatment failure. While fluctuating oxygen environments can be reproduced in vitro, the range of oxygen cycles that can be tested is limited. By contrast, mathematical models can be used to predict the response to a wide range of cyclic dynamics. Accordingly, in this paper we develop a mechanistic model of the cell-cycle that can be combined with in vitro experiments, to better understand the link between cyclic hypoxia and cell-cycle dysregulation. A distinguishing feature of our model is the inclusion of impaired DNA synthesis and cell-cycle arrest due to periodic exposure to severely low oxygen levels. Our model decomposes the cell population into five compartments and a time-dependent delay accounts for the variability in the duration of the S phase which increases in severe hypoxia due to reduced rates of DNA synthesis. We calibrate our model against experimental data and show that it recapitulates the observed cell-cycle dynamics. We use the calibrated model to investigate the response of cells to oxygen cycles not yet tested experimentally. When the re-oxygenation phase is sufficiently long, our model predicts that cyclic hypoxia simply slows cell proliferation since cells spend more time in the S phase. On the contrary, cycles with short periods of re-oxygenation are predicted to lead to inhibition of proliferation, with cells arresting from the cell-cycle in the G2 phase. While model predictions on short time scales (about a day) are fairly accurate (i.e, confidence intervals are small), the predictions become more uncertain over longer periods. Hence, we use our model to inform experimental design that can lead to improved model parameter estimates and validate model predictions.
Collapse
Affiliation(s)
| | - Samuel B Bader
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Ester M Hammond
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Philip K Maini
- Mathematical Institute, University of Oxford, Oxford, UK
| | | | - Helen M Byrne
- Mathematical Institute, University of Oxford, Oxford, UK
| |
Collapse
|
126
|
Devic C, Bodgi L, Sonzogni L, Pilleul F, Ribot H, Charry CD, Le Moigne F, Paul D, Carbillet F, Munier M, Foray N. Influence of cellular models and individual factor in the biological response to chest CT scan exams. Eur Radiol Exp 2022; 6:14. [PMID: 35301607 PMCID: PMC8931147 DOI: 10.1186/s41747-022-00266-0] [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: 09/09/2021] [Accepted: 01/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background While computed tomography (CT) exams are the major cause of medical exposure to ionising radiation, there is increasing evidence that the potential radiation-induced risks must be documented. We investigated the impact of cellular models and individual factor on the deoxyribonucleic acid double-strand breaks (DSB) recognition and repair in human fibroblasts and mammary epithelial cells exposed to current chest CT scan conditions. Method Twelve human primary fibroblasts and four primary human mammary epithelial cell lines with different levels of radiosensitivity/susceptibility were exposed to a standard chest CT scan exam using adapted phantoms. Cells were exposed to a single helical irradiation (14.4 mGy) or to a topogram followed, after 1 min, by one single helical examination (1.1 mGy + 14.4 mGy). DSB signalling and repair was assessed through anti-γH2AX and anti-pATM immunofluorescence. Results Chest CT scan induced a significant number of γH2AX and pATM foci. The kinetics of both biomarkers were found strongly dependent on the individual factor. The topogram may also influence the biological response of radiosensitive/susceptible fibroblasts to irradiation. Altogether, our findings show that a chest CT scan exam may result in 2 to 3 times more unrepaired DSB in cells from radiosensitive/susceptible patients. Conclusions Both individual and tissue factors in the recognition and repair of DSB after current CT scan exams are important. Further investigations are needed to better define the radiosensitivity/susceptibility of individual humans. Supplementary Information The online version contains supplementary material available at 10.1186/s41747-022-00266-0.
Collapse
Affiliation(s)
- Clément Devic
- Institut National de la Santé et de la Recherche Médicale, U1296, « Radiations: Defense, Health and Environment », Bât Cheney A 28 Rue Laennec Centre Léon-Bérard, 69008, Lyon, France.,Fibermetrix™ SAS, 7 Allée de l'Europe, 67960, Entzheim, France
| | - Larry Bodgi
- Radiation Oncology Department, American University of Beirut Medical Center, Beirut, 1107 2020, Lebanon
| | - Laurène Sonzogni
- Institut National de la Santé et de la Recherche Médicale, U1296, « Radiations: Defense, Health and Environment », Bât Cheney A 28 Rue Laennec Centre Léon-Bérard, 69008, Lyon, France
| | - Frank Pilleul
- Département de Radiologie, Centre Léon Bérard, 28 rue Laennec, 69008, Lyon, France
| | - Hervé Ribot
- Service de Radiologie, Hôpital d'Instruction des Armées « Desgenettes », Boulevard Pinel, 69003, Lyon, France
| | - Charlotte De Charry
- Service de Radiologie, Hôpital d'Instruction des Armées « Desgenettes », Boulevard Pinel, 69003, Lyon, France
| | - François Le Moigne
- Service de Radiologie, Hôpital d'Instruction des Armées « Desgenettes », Boulevard Pinel, 69003, Lyon, France
| | - Didier Paul
- Institut National de la Santé et de la Recherche Médicale, U1296, « Radiations: Defense, Health and Environment », Bât Cheney A 28 Rue Laennec Centre Léon-Bérard, 69008, Lyon, France
| | - Fanny Carbillet
- Institut National de la Santé et de la Recherche Médicale, U1296, « Radiations: Defense, Health and Environment », Bât Cheney A 28 Rue Laennec Centre Léon-Bérard, 69008, Lyon, France.,ALARA Expertise SAS, 7 Allée de l'Europe, 67960, Entzheim, France
| | - Mélodie Munier
- Institut National de la Santé et de la Recherche Médicale, U1296, « Radiations: Defense, Health and Environment », Bât Cheney A 28 Rue Laennec Centre Léon-Bérard, 69008, Lyon, France.,Fibermetrix™ SAS, 7 Allée de l'Europe, 67960, Entzheim, France
| | - Nicolas Foray
- Institut National de la Santé et de la Recherche Médicale, U1296, « Radiations: Defense, Health and Environment », Bât Cheney A 28 Rue Laennec Centre Léon-Bérard, 69008, Lyon, France.
| |
Collapse
|
127
|
He H, Lin K, Zou C, Pan J, Fu W, Zhou Y, Lin H, Chen C, Su Y. Knockdown of Annexin A2 Enhances Radiosensitivity by Increasing G2/M-Phase Arrest, Apoptosis and Activating the p38 MAPK-HSP27 Pathway in Nasopharyngeal Carcinoma. Front Oncol 2022; 12:769544. [PMID: 35371986 PMCID: PMC8968728 DOI: 10.3389/fonc.2022.769544] [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: 09/28/2021] [Accepted: 02/21/2022] [Indexed: 11/30/2022] Open
Abstract
Annexin A2 (ANXA2) has been found to be involved in cancer proliferation, metastasis and prognosis; however, its exact role in nasopharyngeal carcinoma (NPC) radioresistance remains unknown. We found that ANXA2 expression was correlated with prognosis in NPC patients, and longer overall survival in NPC patients with low ANXA2 expression than those with high ANXA2 expression. ANXA2 knockdown increased the radiosensitivity in radioresistant NPC cells, and ANXA2 overexpression decreased the radiosensitivity in NPC cells. Knocking-down ANXA2 expression increased the irradiation-induced apoptosis of radioresistant NPC cells, and ANXA2 overexpression decreased the irradiation-induced apoptosis of NPC cells. ANXA2 knockdown induced G2/M phase arrest in NPC cells post-irradiation, and ANXA2 overexpression abrogated G2/M phase arrest in NPC cells post-irradiation. ANXA2 overexpression resulted in inhibition of the p38 MAPK-HSP27 pathway, while ANXA2 knockdown resulted in activation of the p38 MAPK-HSP27 pathway. In addition, ANXA2 knockdown increased the radiosensitivity of the xenografted tumors in nude mice. Our data demonstrate that knockdown of Annexin A2 enhanced radiosensitivity in NPC by increasing G2/M-phase arrest, apoptosis and activating the p38 MAPK-HSP27 pathway. ANXA2 may be a promising target used to overcome radioresistance in NPC.
Collapse
Affiliation(s)
- Huocong He
- Laboratory of Radiation Biology and Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Keyu Lin
- Laboratory of Radiation Biology and Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Changyan Zou
- Laboratory of Radiation Biology and Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jianru Pan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Wankai Fu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Yan Zhou
- Department of Epidemiology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Huamei Lin
- Laboratory of Radiation Biology and Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Chao Chen
- Laboratory of Radiation Biology and Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Ying Su
- Laboratory of Radiation Biology and Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Translational Cancer Medicine, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- *Correspondence: Ying Su,
| |
Collapse
|
128
|
Study of Mutagenic and Antitoxic Properties of Gentabiferon-B. MACEDONIAN VETERINARY REVIEW 2022. [DOI: 10.2478/macvetrev-2022-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The combination of immunomodulators and antibiotics in the treatment of animals with diseases of bacterial etiology is one of the effective strategies for animal therapy. The drug gentabiferon-B combines antibiotic gentamicin and species-specific (bovine) recombinant interferons -α and -γ. The study aimed to evaluate the effect of course application of gentabiferon-B on the cytogenetic stability of bone marrow cells of outbred mice after administering mitomycin C (MMC). The proportion of polychromatophilic erythrocytes in the bone marrow was assessed. There was no effect of gentabiferon-B on the frequency of polychromatophilic erythrocytes with micronuclei in both healthy animals and mice with MMC-induced cytogenetic instability. The course application of gentabiferon-B before the administration of MMC led to an increase in the proportion of polychromatophilic erythrocytes (46.03±2.61%) which was non-significantly different than the negative control group. The administration of MMC alone caused a decrease in the proportion of polychromatophilic erythrocytes to 33.33±1.83%. The antitoxic effect of gentabiferon-B led to an increase in the level of polychromatophilic erythrocytes by 38.1% compared to the group that received only MMC. Studies have shown that gentabiferon-B does not have mutagenic activity and anticlastogenic properties, however, it reduces the toxic effect of MMC. In conclusion, it is indicative that gentabiferon-B has antitoxic properties and can be safely used in animal therapy.
Collapse
|
129
|
Nong HB, Zhang YN, Bai YG, Zhang Q, Liu MF, Zhou Q, Shi ZH, Zeng GF, Zong SH. Adapalene Inhibits Prostate Cancer Cell Proliferation In Vitro and In Vivo by Inducing DNA Damage, S-phase Cell Cycle Arrest, and Apoptosis. Front Pharmacol 2022; 13:801624. [PMID: 35273495 PMCID: PMC8902295 DOI: 10.3389/fphar.2022.801624] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
Aims: Prostate cancer is a well-known aggressive malignant tumor in men with a high metastasis rate and poor prognosis. Adapalene (ADA) is a third-generation synthetic retinoid with anticancer properties. We investigated the anti-tumor activity and molecular mechanisms of ADA in the RM-1 prostate cancer cell line in vivo and in vitro. Methods: The effects of ADA on cell proliferation were estimated using the CCK-8 and colony formation assays. The wound-healing assay and the Transwell assay were employed to examine the migratory capacity and invasiveness of the cells. Flow cytometry was utilized to evaluate the cell cycle and apoptosis, and Western blotting analysis was used to assess the expression of the associated proteins. Micro-CT, histomorphological, and immunohistochemical staining were used to assess the effects of ADA on bone tissue structure and tumor growth in a mouse model of prostate cancer bone metastasis. Result: ADA dramatically inhibited cell proliferation, migration, invasiveness, and induced S-phase arrest and apoptosis. ADA also regulated the expression of S-phase associated proteins and elevated the levels of DNA damage markers, p53, and p21 after ADA treatment, suggesting that the anti-tumor effect of ADA manifests through the DNA damage/p53 pathway. Furthermore, we observed that ADA could effectively inhibited tumor growth and bone destruction in mice. Conclusion: ADA inhibited prostate cancer cell proliferation, elicited apoptosis, and arrested the cell cycle in the S-phase. ADA also slowed the rate of tumor growth and bone destruction in vitro. Overall, our results suggest that ADA may be a potential treatment against prostate cancer.
Collapse
Affiliation(s)
- Hai-Bin Nong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Ya-Nan Zhang
- Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Nanning, China
| | - Yi-Guang Bai
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.,Department of Orthopaedics, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, China
| | - Qiong Zhang
- Department of Nutrition and Food Hygiene, College of Public Hygiene of Guangxi Medical University, Nanning, China
| | - Ming-Fu Liu
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Quan Zhou
- Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Nanning, China
| | - Zhuo-Hua Shi
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China
| | - Gao-Feng Zeng
- Department of Nutrition and Food Hygiene, College of Public Hygiene of Guangxi Medical University, Nanning, China
| | - Shao-Hui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, China.,Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
130
|
Tsao M, Barnes E, Karam I, Rembielak A. Hypofractionated Radiation Therapy in Keratinocyte Carcinoma. Clin Oncol (R Coll Radiol) 2022; 34:e218-e224. [DOI: 10.1016/j.clon.2022.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/02/2022] [Accepted: 02/18/2022] [Indexed: 12/20/2022]
|
131
|
Liu G, Yang S, Liu Y, Xu Y, Qiu H, Sun J, Song J, Shi L. The adenosine-A2a receptor regulates the radioresistance of gastric cancer via PI3K-AKT-mTOR pathway. Int J Clin Oncol 2022; 27:911-920. [PMID: 35122587 DOI: 10.1007/s10147-022-02123-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/19/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Radiotherapy is a key strategy in gastric cancer (GC) treatment. However, radioresistance remains a serious concern. It is unclear whether the accumulation of adenosine A2a receptor (ADO-A2aR) is related to radioresistance in GC. In this study, the molecular role of ADO-A2aR in GC radioresistance was investigated. METHODS Colony formation assays were used to assess the role of ADO-A2aR on radioresistance. GC stem cell surface marker expression (including Nanog, OCT-4, SOX-2 and CD44) and PI3K/AKT/mTOR signaling pathway associated protein levels (including phosphorylated PI3K, phosphorylated AKT and phosphorylated mTOR) were determined via western blotting, flow cytometry and immunofluorescence. In addition, the role of ADO-A2aR on radioresistance was explored in vivo using murine xenograft models. RESULTS ADO-A2aR regulated GC cell stemness both in vitro and in vivo. This was shown to induce radioresistance in GC. ADO-A2aR was revealed to significantly induce cell cycle arrest and promote GC cell apoptosis. These activities were closely linked to activation of the PI3K/AKT/mTOR pathway. CONCLUSION This study identified that ADO enhances GC cell stemness via interaction with A2aR and subsequent activation of the PI3K/AKT/mTOR pathway. Ultimately, this resulted in radioresistance. A2aR is a potential target to improve GC radiosensitivity.
Collapse
Affiliation(s)
- Guihong Liu
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.,Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Song Yang
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.,Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Yang Liu
- Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Yumei Xu
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Hui Qiu
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Jian Sun
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.,Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Jun Song
- Departments of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China. .,Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.
| | - Linsen Shi
- Departments of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.
| |
Collapse
|
132
|
Liu J, Hormuth DA, Yang J, Yankeelov TE. A Multi-Compartment Model of Glioma Response to Fractionated Radiation Therapy Parameterized via Time-Resolved Microscopy Data. Front Oncol 2022; 12:811415. [PMID: 35186747 PMCID: PMC8855115 DOI: 10.3389/fonc.2022.811415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Conventional radiobiology models, including the linear-quadratic model, do not explicitly account for the temporal effects of radiation, thereby making it difficult to make time-resolved predictions of tumor response to fractionated radiation. To overcome this limitation, we propose and validate an experimental-computational approach that predicts the changes in cell number over time in response to fractionated radiation. METHODS We irradiated 9L and C6 glioma cells with six different fractionation schemes yielding a total dose of either 16 Gy or 20 Gy, and then observed their response via time-resolved microscopy. Phase-contrast images and Cytotox Red images (to label dead cells) were collected every 4 to 6 hours up to 330 hours post-radiation. Using 75% of the total data (i.e., 262 9L curves and 211 C6 curves), we calibrated a two-species model describing proliferative and senescent cells. We then applied the calibrated parameters to a validation dataset (the remaining 25% of the data, i.e., 91 9L curves and 74 C6 curves) to predict radiation response. Model predictions were compared to the microscopy measurements using the Pearson correlation coefficient (PCC) and the concordance correlation coefficient (CCC). RESULTS For the 9L cells, we observed PCCs and CCCs between the model predictions and validation data of (mean ± standard error) 0.96 ± 0.007 and 0.88 ± 0.013, respectively, across all fractionation schemes. For the C6 cells, we observed PCCs and CCCs between model predictions and the validation data were 0.89 ± 0.008 and 0.75 ± 0.017, respectively, across all fractionation schemes. CONCLUSION By proposing a time-resolved mathematical model of fractionated radiation response that can be experimentally verified in vitro, this study is the first to establish a framework for quantitative characterization and prediction of the dynamic radiobiological response of 9L and C6 gliomas to fractionated radiotherapy.
Collapse
Affiliation(s)
- Junyan Liu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - David A. Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Thomas E. Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, The University of Texas at Austin, Austin, TX, United States
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
133
|
Larionova I, Rakina M, Ivanyuk E, Trushchuk Y, Chernyshova A, Denisov E. Radiotherapy resistance: identifying universal biomarkers for various human cancers. J Cancer Res Clin Oncol 2022; 148:1015-1031. [PMID: 35113235 DOI: 10.1007/s00432-022-03923-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/12/2022] [Indexed: 12/16/2022]
Abstract
Radiotherapy (RT) is considered as a standard in the treatment of most solid cancers, including glioblastoma, lung, breast, rectal, prostate, colorectal, cervical, esophageal, and head and neck cancers. The main challenge in RT is tumor cell radioresistance associated with a high risk of locoregional relapse and distant metastasis. Despite significant progress in understanding mechanisms of radioresistance, its prediction and overcoming remain unresolved. This review presents the state-of-the-art for the potential universal biomarkers correlated to the radioresistance and poor outcome in different cancers. We describe radioresistance biomarkers functionally attributed to DNA repair, signal transduction, hypoxia, and angiogenesis. We also focus on high throughput genetic and proteomic studies, which revealed a set of molecular biomarkers related to radioresistance. In conclusion, we discuss biomarkers which are overlapped in most several cancers.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia.
| | - Militsa Rakina
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, 634050, Tomsk, Russia
| | - Elena Ivanyuk
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| | - Yulia Trushchuk
- Department of Gynecologic Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| | - Alena Chernyshova
- Department of Gynecologic Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| |
Collapse
|
134
|
Wang YC, Tsai SH, Chen MH, Hsieh FY, Chang YC, Tung FI, Liu TY. Mineral Nanomedicine to Enhance the Efficacy of Adjuvant Radiotherapy for Treating Osteosarcoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:5586-5597. [PMID: 35050587 DOI: 10.1021/acsami.1c21729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
It is vital to remove residual tumor cells after resection to avoid the recurrence and metastasis of osteosarcoma. In this study, a mineral nanomedicine, europium-doped calcium fluoride (CaF2:Eu) nanoparticles (NPs), is developed to enhance the efficacy of adjuvant radiotherapy (i.e., surgical resection followed by radiotherapy) for tumor cell growth and metastasis of osteosarcoma. In vitro studies show that CaF2:Eu NPs (200 μg/mL) exert osteosarcoma cell (143B)-selective toxicity and migration-inhibiting effects at a Eu dopant amount of 2.95 atomic weight percentage. These effects are further enhanced under X-ray irradiation (6 MeV, 4 Gy). Furthermore, in vivo tests show that intraosseous injection of CaF2:Eu NPs and X-ray irradiation have satisfactory therapeutic efficacy in controlling primary tumor size and inhibiting primary tumor metastasis. Overall, our results suggest that CaF2:Eu NPs with their osteosarcoma cell (143B)-selective toxicity and migration-inhibiting effects combined with radiotherapy might be nanomedicines for treating osteosarcoma after tumor resection.
Collapse
Affiliation(s)
- Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Sheng-Han Tsai
- Department of Urology, Cheng Hsin General Hospital, Taipei 112401, Taiwan
| | - Ming-Hong Chen
- Department of Neurosurgery, Taipei Municipal Wanfang Hospital, Taipei 116079, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110301, Taiwan
| | - Fu-Yu Hsieh
- Franz Biotech Incorporation, Taipei 105065, Taiwan
| | - Yuan-Chen Chang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Fu-I Tung
- Department of Orthopaedics, Yang-Ming Branch, Taipei City Hospital, Taipei 111024, Taiwan
- Department of Health and Welfare, College of City Management, University of Taipei, Taipei 111036, Taiwan
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
135
|
Radiosensitizing Effect of Celastrol by Inhibiting G2/M Phase Arrest Induced by the c-myc Gene of Human SW1353 Chondrosarcoma Cells: Network and Experimental Analyses. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1948657. [PMID: 35141331 PMCID: PMC8820907 DOI: 10.1155/2022/1948657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/30/2021] [Indexed: 11/21/2022]
Abstract
Objective Studies have unveiled that the components of Tripterygium wilfordii Hook F (TWHF) such as celastrol could attenuate apoptosis and proliferation of various tumor cells. This study is focused on the radiosensitization effect and apoptotic pathways of celastrol via the inhibition of the c-myc gene and the influence of which combined with radiotherapy on the proliferation, apoptosis, invasion, and metastasis of chondrosarcoma cells. Methods A variety of bioinformatic tools were applied to explore the expression level and prognosis of the c-myc gene in different tumor cells and chondrosarcoma cells. We used pharmacology network to analyze the components, pathways, targets, molecular functions of TWHF and explore the relevant effective components over the MYC gene. Clone formation assay, CCK-8 assay, flow cytometry, and transwell migration assay were applied to detect the effects of celastrol on the expression of c-myc gene, cell apoptosis, and cell cycle. Radiation therapy was used to observe the radiosensitization effect of celastrol on chondrosarcoma. Results This study shows that the c-myc gene is overexpressed in various tumor cells and bone tumor cells to varying degrees. Celastrol can significantly inhibit the expression of the c-myc gene, induce G2/M phase arrest through regulation of G2/M phase-related proteins, and promote SW1353 cell apoptosis through the mitochondrial signaling pathway. In addition, we also found that the use of triptorubin to inhibit c-myc gene expression in combination with radiotherapy can increase the osteosarcoma cells' apoptosis rate through the mitochondrial signaling pathway significantly. Conclusions Our study validated the radiosensitization effect of celastrol through knocking down the expression of the c-myc gene to induce G2/M phase arrest and provides a new idea for the treatment of refractory or recurrent chondrosarcoma that is not sensitive to radiotherapy.
Collapse
|
136
|
Hashimoto T, Urushihara Y, Murata Y, Fujishima Y, Hosoi Y. AMPK increases expression of ATM through transcriptional factor Sp1 and induces radioresistance under severe hypoxia in glioblastoma cell lines. Biochem Biophys Res Commun 2022; 590:82-88. [PMID: 34973534 DOI: 10.1016/j.bbrc.2021.12.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/24/2022]
Abstract
We have previously reported that severe hypoxia increases expression and activity of the DNA damage sensor ATM by activation of the key energy sensor AMPK. Here, to elucidate molecular mechanisms underlying increased expression and activity of ATM by AMPK under severe hypoxia, we investigated roles of transcriptional factors Sp1 and FoxO3a using human glioblastoma cell lines T98G and A172. Severe hypoxia increased expression of ATM, AMPKα and Sp1 but not that of FoxO3a. Knockdown of AMPKα suppressed expression of ATM and Sp1 and suppressed cellular radioresistance under severe hypoxia without affecting cell cycle distribution. Knockdown of Sp1 suppressed expression of ATM. These results suggest that increased expression and activity of AMPK under severe hypoxia induce cellular radioresistance through AMPK/Sp1/ATM pathway.
Collapse
Affiliation(s)
- Takuma Hashimoto
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yusuke Urushihara
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yasuhiko Murata
- MSD K.K., 1-13-12 Kudankita, Chiyoda-ku, Tokyo, 102-8667, Japan
| | - Yohei Fujishima
- Department of Risk Analysis and Biodosimetry, Institute of Radiation Emergency Medicine, Hirosaki University, 66-1 Hon-cho, Hirosaki-shi, Aomori, 036-8564, Japan
| | - Yoshio Hosoi
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
137
|
IGF-I and Hyaluronic Acid Mitigate the Negative Effect of Irradiation on Human Skin Keratinocytes. Cancers (Basel) 2022; 14:cancers14030588. [PMID: 35158856 PMCID: PMC8833477 DOI: 10.3390/cancers14030588] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Patients undergoing radiation therapy for the treatment of various types of cancer often experience side effects such as radiation dermatitis. A gold standard treatment is still lacking. The objective of the present study was to find novel therapeutic strategies for the regeneration and repair of damaged skin areas after irradiation. An in vitro 2D and 3D primary keratinocyte model was used to test the effect of insulin-like growth factor I (IGF-I), keratinocyte growth factor (KGF), platelet lysate (PL), hyaluronic acid (HA), and adipose-derived stem cell (ADSC) conditioned medium on the functional abilities (viability, migration) and the gene expression of irradiated keratinocytes. Hyaluronic acid and IGF-I effectively reduced the irradiation damage of primary keratinocytes by stimulating viability and migration and reducing cell apoptosis and necrosis. These findings indicate that the negative effects of irradiation on keratinocytes located in the patient’s skin can be counterbalanced with HA and IGF-I treatment. Abstract Ionizing radiation has become an integral part of modern cancer therapy regimens. Various side effects, such as radiation dermatitis, affect patients in acute and chronic forms and decrease therapy compliance significantly. In this study, primary keratinocytes were irradiated in a 2-dimensional (2D) culture as well as on a 3-dimensional (3D) collagen-elastin matrix with doses of 2 and 5 Gy. The effect of different concentrations of IGF-I, KGF, platelet lysate (PL), high and low molecular weight hyaluronic acid (H-HA, L-HA), and adipose-derived stem cell (ADSC) conditioned medium was analyzed in respect to cell viability (WST-8), wound closure (migration), and the gene expression (quantitative real-time PCR) of 2D cultures. The 3D culture was evaluated by WST-8. A mixture of H-HA and L-HA, as well as IGF-I, could significantly stimulate the keratinocyte viability and migration which were severely reduced by irradiation. The MKI67and IL6 gene expression of irradiated keratinocytes was significantly higher after H-HA/L-HA treatment. The stimulating effects of H-HA/L-HA and IGF-I were able to be confirmed in 3D culture. A positive influence on cell viability, migration, and gene expression was achieved after the treatment with H-L-HA and IGF-I. These results open the possibility of a novel therapeutic method for both the prevention and the treatment of radiation dermatitis.
Collapse
|
138
|
Ghaderi N, Jung J, Brüningk SC, Subramanian A, Nassour L, Peacock J. A Century of Fractionated Radiotherapy: How Mathematical Oncology Can Break the Rules. Int J Mol Sci 2022; 23:ijms23031316. [PMID: 35163240 PMCID: PMC8836217 DOI: 10.3390/ijms23031316] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is involved in 50% of all cancer treatments and 40% of cancer cures. Most of these treatments are delivered in fractions of equal doses of radiation (Fractional Equivalent Dosing (FED)) in days to weeks. This treatment paradigm has remained unchanged in the past century and does not account for the development of radioresistance during treatment. Even if under-optimized, deviating from a century of successful therapy delivered in FED can be difficult. One way of exploring the infinite space of fraction size and scheduling to identify optimal fractionation schedules is through mathematical oncology simulations that allow for in silico evaluation. This review article explores the evidence that current fractionation promotes the development of radioresistance, summarizes mathematical solutions to account for radioresistance, both in the curative and non-curative setting, and reviews current clinical data investigating non-FED fractionated radiotherapy.
Collapse
Affiliation(s)
- Nima Ghaderi
- Department of Biomedical Engineering, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA; (N.G.); (J.J.)
| | - Joseph Jung
- Department of Biomedical Engineering, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA; (N.G.); (J.J.)
| | - Sarah C. Brüningk
- Machine Learning & Computational Biology Lab, Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland;
- Swiss Institute for Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA;
| | - Lauren Nassour
- Department of Radiation Oncology, University of Alabama Birmingham, Birmingham, AL 35205, USA;
| | - Jeffrey Peacock
- Department of Radiation Oncology, University of Alabama Birmingham, Birmingham, AL 35205, USA;
- Correspondence:
| |
Collapse
|
139
|
Yuan J, Li X, Yu S. CDK7-dependent transcriptional addiction in bone and soft tissue sarcomas: Present and Future. Biochim Biophys Acta Rev Cancer 2022; 1877:188680. [PMID: 35051528 DOI: 10.1016/j.bbcan.2022.188680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/25/2022]
Abstract
Cancer arises from genetic alterations that invariably contribute to dysregulated transcriptional programs. These dysregulated programs establish and maintain specific cancer cell states, leading to an intensive dependence on a set of certain regulators of gene expression. The CDK7 functions as the core of transcription, and governs RNA polymerase II and the downstream oncogenes expression in cancers. CDK7 inhibition leads to reduced recruitment of super-enhancers-driven oncogenic transcription factors, and the depression of these associated oncogenes expression, which indicates the dependence of transcriptional addiction of cancers on CDK7. Given that specified oncoproteins of sarcomas commonly function at oncogenic transcription, targeting CDK7-denpendent transcriptional addiction may be of guiding significance for the treatment of sarcomas. In this review, we summarize the advances in mechanism of targeted CDK7-dependent transcriptional addiction and discuss the path ahead to potential application discovery in bone and soft tissue sarcomas, providing theoretical considerations for bio-orthogonal therapeutic strategies.
Collapse
Affiliation(s)
- Jin Yuan
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyang Li
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical sciences and Peking Union Medical College, Beijing, China.
| | - Shengji Yu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
140
|
SERPINB3 (SCCA1) inhibits cathepsin L and lysoptosis, protecting cervical cancer cells from chemoradiation. Commun Biol 2022; 5:46. [PMID: 35022555 PMCID: PMC8755728 DOI: 10.1038/s42003-021-02893-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
The endogenous lysosomal cysteine protease inhibitor SERPINB3 (squamous cell carcinoma antigen 1, SCCA1) is elevated in patients with cervical cancer and other malignancies. High serum SERPINB3 is prognostic for recurrence and death following chemoradiation therapy. Cervical cancer cells genetically lacking SERPINB3 are more sensitive to ionizing radiation (IR), suggesting this protease inhibitor plays a role in therapeutic response. Here we demonstrate that SERPINB3-deficient cells have enhanced sensitivity to IR-induced cell death. Knock out of SERPINB3 sensitizes cells to a greater extent than cisplatin, the current standard of care. IR in SERPINB3 deficient cervical carcinoma cells induces predominantly necrotic cell death, with biochemical and cellular features of lysoptosis. Rescue with wild-type SERPINB3 or a reactive site loop mutant indicates that protease inhibitory activity is required to protect cervical tumor cells from radiation-induced death. Transcriptomics analysis of primary cervix tumor samples and genetic knock out demonstrates a role for the lysosomal protease cathepsin L in radiation-induced cell death in SERPINB3 knock-out cells. These data support targeting of SERPINB3 and lysoptosis to treat radioresistant cervical cancers. Wang et al. demonstrate the cytoprotective role of SERPINB3 against radiation-induced necrosis, showing that cells lacking SERPINB3 protein both in culture and in mice are more sensitive to radiation and cisplatin-induced cell death. The authors also report that the cell death induced by radiation in SERPINB3-lacking cells is lysoptosis and implicate the lysosomal protease cathepsin L in this process.
Collapse
|
141
|
Dobešová L, Gier T, Kopečná O, Pagáčová E, Vičar T, Bestvater F, Toufar J, Bačíková A, Kopel P, Fedr R, Hildenbrand G, Falková I, Falk M, Hausmann M. Incorporation of Low Concentrations of Gold Nanoparticles: Complex Effects on Radiation Response and Fate of Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14010166. [PMID: 35057061 PMCID: PMC8781406 DOI: 10.3390/pharmaceutics14010166] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 01/27/2023] Open
Abstract
(1) Background: In oncology research, a long-standing discussion exists about pros and cons of metal nanoparticle-enhanced radiotherapy and real mechanisms behind the tumor cell response to irradiation (IR) in presence of gold nanoparticles (GNPs). A better understanding of this response is, however, necessary to develop more efficient and safety nanoparticle (NP) types designed to disturb specific processes in tumor cells. (2) Aims and Methods: We combined 3D confocal microscopy and super-resolution single molecule localization microscopy (SMLM) to analyze, at the multiscale, the early and late effects of 10 nm-GNPs on DNA double strand break (DSB) induction and repair in tumor cells exposed to different doses of photonic low-LET (linear energy transfer) radiation. The results were correlated to different aspects of short and long-term cell viability. SkBr3 breast cancer cells (selected for the highest incidence of this cancer type among all cancers in women, and because most breast tumors are treated with IR) were incubated with low concentrations of GNPs and irradiated with 60Co γ-rays or 6 MV X-rays. In numerous post-irradiation (PI) times, ranging from 0.5 to 24 h PI, the cells were spatially (3D) fixed and labeled with specific antibodies against γH2AX, 53BP1 and H3K9me3. The extent of DSB induction, multi-parametric micro- and nano-morphology of γH2AX and 53BP1 repair foci, DSB repair kinetics, persistence of unrepaired DSBs, nanoscale clustering of γH2AX and nanoscale (hetero)chromatin re-organization were measured by means of the mentioned microscopy techniques in dependence of radiation dose and GNP concentration. (3) Results: The number of γH2AX/53BP1 signals increased after IR and an additional increase was observed in GNP-treated (GNP(+)) cells compared to untreated controls. However, this phenomenon reflected slight expansion of the G2-phase cell subpopulation in irradiated GNP(+) specimens instead of enhanced DNA damage induction by GNPs. This statement is further supported by some micro- and nano-morphological parameters of γH2AX/53BP1 foci, which slightly differed for cells irradiated in absence or presence of GNPs. At the nanoscale, Ripley’s distance frequency analysis of SMLM signal coordinate matrices also revealed relaxation of heterochromatin (H3K9me3) clusters upon IR. These changes were more prominent in presence of GNPs. The slight expansion of radiosensitive G2 cells correlated with mostly insignificant but systematic decrease in post-irradiation survival of GNP(+) cells. Interestingly, low GNP concentrations accelerated DSB repair kinetics; however, the numbers of persistent γH2AX/53BP1 repair foci were slightly increased in GNP(+) cells. (4) Conclusions: Low concentrations of 10-nm GNPs enhanced the G2/M cell cycle arrest and the proportion of radiosensitive G2 cells, but not the extent of DNA damage induction. GNPs also accelerated DSB repair kinetics and slightly increased presence of unrepaired γH2AX/53BP1 foci at 24 h PI. GNP-mediated cell effects correlated with slight radiosensitization of GNP(+) specimens, significant only for the highest radiation dose tested (4 Gy).
Collapse
Affiliation(s)
- Lucie Dobešová
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
- Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
| | - Theresa Gier
- Kirchhoff Institute for Physics, Heidelberg University, 69120 Heidelberg, Germany; (T.G.); (G.H.)
| | - Olga Kopečná
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
| | - Eva Pagáčová
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
| | - Tomáš Vičar
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, 616 00 Brno, Czech Republic;
| | - Felix Bestvater
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Jiří Toufar
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
- Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
| | - Alena Bačíková
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
| | - Pavel Kopel
- Department of Inorganic Chemistry, Faculty of Science, Palacky University Olomouc, 779 00 Olomouc, Czech Republic;
| | - Radek Fedr
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
| | - Georg Hildenbrand
- Kirchhoff Institute for Physics, Heidelberg University, 69120 Heidelberg, Germany; (T.G.); (G.H.)
| | - Iva Falková
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
| | - Martin Falk
- Institute of Biophysics, The Czech Academy of Sciences, 612 65 Brno, Czech Republic; (L.D.); (O.K.); (E.P.); (J.T.); (A.B.); (R.F.); (I.F.)
- Correspondence: (M.F.); (M.H.); Tel.: +420-728-084-060 (M.F.); +49-6221-549-824 (M.H.)
| | - Michael Hausmann
- Kirchhoff Institute for Physics, Heidelberg University, 69120 Heidelberg, Germany; (T.G.); (G.H.)
- Correspondence: (M.F.); (M.H.); Tel.: +420-728-084-060 (M.F.); +49-6221-549-824 (M.H.)
| |
Collapse
|
142
|
Lou D, Zhu D, Wang Z, Zhang R, Yu Z, Gong F, Peng Y, Zeng S, Liu Y, Li A, Fan Q. Effect of GADD45G on the radioresistance of nasopharyngeal carcinoma cells. Anticancer Drugs 2022; 33:e84-e93. [PMID: 34282742 DOI: 10.1097/cad.0000000000001145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The development of radioresistance by nasopharyngeal carcinoma (NPC) cells almost always results in tumor recurrence and metastasis, making clinical treatment of the disease difficult. In this study, the mechanism of radioresistance in NPC cells was investigated. First, a gene array and quantitative reverse-transcription-PCR assays were used to screen for genes exhibiting significantly altered expression in the DNA damage signaling pathway. Based on those results, GADD45G was further studied in the context of radioresistance. A GADD45G-knockout NPC cell line (CNE-2R-KO) was constructed using CRISPR-Cas9 technology and used for a comparison of differences in radioresistance with other radiosensitive and radioresistant NPC cells, as evaluated using colony formation assays. Cell cycle changes were observed using flow cytometry. Cell proliferation and migration were measured using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide and wound healing assays, respectively. The sequencing results revealed the successful construction of the CNE-2R-KO cell line, the radiosensitivity of which was higher than that of its parent radioresistant cell line owing to the GADD45G knockout. This was likely related to the increase in the number of cells in the G1 phase and decrease in those in the S1 phase as well as the increased cell proliferation rate and decreased migratory ability. GADD45G is associated with radioresistance in NPC cells and likely has a role in the occurrence and metastasis of NPC.
Collapse
Affiliation(s)
- Dandan Lou
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Daoqi Zhu
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Zetai Wang
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Zhijian Yu
- Department of Traditional Chinese Medicine Formulae, School of Traditional Chinese Medicine, Southern Medical University
| | - Fengying Gong
- Department of Traditional Chinese Medicine, NanFang Hospital, Guangdong, Guangzhou, China
| | - Yan Peng
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Siying Zeng
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Ying Liu
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou
| | - Aiwu Li
- Department of Traditional Chinese Medicine, NanFang Hospital, Guangdong, Guangzhou, China
| | - Qin Fan
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| |
Collapse
|
143
|
Jiménez-Cortegana C, Klapp V, Bloy N, Galassi C, Sato A, Yamazaki T, Buqué A, Galluzzi L, Petroni G. Cytofluorometric assessment of cell cycle progression in irradiated cells. Methods Cell Biol 2022; 172:1-16. [DOI: 10.1016/bs.mcb.2021.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
144
|
Guo SB, Huang WJ, Tian XP. Brusatol modulates diverse cancer hallmarks and signaling pathways as a potential cancer therapeutic. ACTA MATERIA MEDICA 2022; 1. [DOI: 10.15212/amm-2022-0014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Cancer is a consequence of uncontrolled cell proliferation that is associated with cell-cycle disruption. It is a multifactorial disease that depends on the modulation of numerous oncogenic signaling pathways and targets. Although a battle against cancer has been waged for centuries, this disease remains a major cause of death worldwide. Because of the development of resistance to current anticancer drugs, substantial effort has been focused on discovering more effective agents for tumor therapy. Natural products have powerful prospects as anticancer drugs. Brusatol, a component isolated from the plant Brucea javanica, has been demonstrated to efficiently combat a wide variety of tumors. Extensive studies have indicated that brusatol exhibits anticancer effects by arresting the cell cycle; promoting apoptosis; inducing autophagy; attenuating epithelial-mesenchymal transition; inhibiting migration, invasion and angiogenesis; and increasing chemosensitivity and radiosensitivity. These effects involve various oncogenic signaling pathways, including the MAPK, NF-κB, PI3K/AKT/mTOR, JAK/STAT and Keap1/Nrf2/ARE signaling pathways. This review describes the evidence suggesting that brusatol is a promising drug candidate for cancer therapeutics.
Collapse
Affiliation(s)
- Song-Bin Guo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Wei-Juan Huang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, P.R. China
| | - Xiao-Peng Tian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| |
Collapse
|
145
|
Álvarez-Abril B, Bloy N, Galassi C, Sato A, Jiménez-Cortegana C, Klapp V, Aretz A, Guilbaud E, Buqué A, Galluzzi L, Yamazaki T. Cytofluorometric assessment of acute cell death responses driven by radiation therapy. Methods Cell Biol 2022; 172:17-36. [DOI: 10.1016/bs.mcb.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
146
|
Xiang XS, Li PC, Wang WQ, Liu L. Histone deacetylases: A novel class of therapeutic targets for pancreatic cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188676. [PMID: 35016922 DOI: 10.1016/j.bbcan.2022.188676] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is the seventh leading cause of cancer death worldwide, with a low 5-year survival rate. Novel agents are urgently necessary to treat the main pathological type, known as pancreatic ductal carcinoma (PDAC). The dysregulation of histone deacetylases (HDACs) has been identified in association with PDAC, which can be more easily targeted by small molecular inhibitors than gene mutations and may represent a therapeutic breakthrough for PDAC. However, the contributions of HDACs to PDAC remain controversial, and pharmacokinetic challenges have limited the application of HDAC inhibitors (HDACis) in PDAC. This review summarizes the mechanisms associated with success and failure of HDACis in PDAC and discusses the recent progress made in HDACi development and application, such as combination therapies designed to enhance efficacy. More precise strategies involving HDACis might eventually improve the outcomes of PDAC treatment.
Collapse
Affiliation(s)
- Xue-Song Xiang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng-Cheng Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
147
|
RT-PCR-assisted quantification of type I IFN responses in irradiated cancer cells. Methods Cell Biol 2022; 172:145-161. [DOI: 10.1016/bs.mcb.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
148
|
Ivashkevich A. The role of isoflavones in augmenting the effects of radiotherapy. Front Oncol 2022; 12:800562. [PMID: 36936272 PMCID: PMC10016616 DOI: 10.3389/fonc.2022.800562] [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: 10/23/2021] [Accepted: 08/31/2022] [Indexed: 03/05/2023] Open
Abstract
Cancer is one of the major health problems and the second cause of death worldwide behind heart disease. The traditional soy diet containing isoflavones, consumed by the Asian population in China and Japan has been identified as a protective factor from hormone-related cancers. Over the years the research focus has shifted from emphasizing the preventive effect of isoflavones from cancer initiation and promotion to their efficacy against established tumors along with chemo- and radiopotentiating effects. Studies performed in mouse models and results of clinical trials emphasize that genistein or a mixture of isoflavones, containing in traditional soy diet, could be utilized to both potentiate the response of cancer cells to radiotherapy and reduce radiation-induced toxicity in normal tissues. Currently ongoing clinical research explores a potential of another significant isoflavone, idronoxil, also known as phenoxodiol, as radiation enhancing agent. In the light of the recent clinical findings, this article reviews the accumulated evidence which support the clinically desirable interactions of soy isoflavones with radiation therapy resulting in improved tumor treatment. This review discusses important aspects of the development of isoflavones as anticancer agents, and mechanisms potentially relevant to their activity in combination with radiation therapy of cancer. It gives a critical overview of studies characterizing isoflavone targets such as topoisomerases, ENOX2/PMET, tyrosine kinases and ER receptor signaling, and cellular effects on the cell cycle, DNA damage, cell death, and immune responses.
Collapse
Affiliation(s)
- Alesia Ivashkevich
- Faculty of Engineering and Information Sciences, University of Wollongong, Wollongong, NSW, Australia
- Noxopharm, Gordon, NSW, Australia
- *Correspondence: Alesia Ivashkevich,
| |
Collapse
|
149
|
Mansoori M, Abdi Rad I, Mirzaei A, Tam KJ, Mohsen Hosseini S, Mahmodlu R, Mansouri F, Saeednejad Zanjani L, Madjd Z. Does GD2 synthase (GD2S) detect cancer stem cells in blood samples of breast carcinomas? J Appl Biomed 2021; 19:181-189. [PMID: 34907737 DOI: 10.32725/jab.2021.019] [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: 11/28/2020] [Accepted: 09/03/2021] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Cancer stem cells (CSCs) are a theorized subset of cells within the tumor that is thought to drive disease recurrence and metastatic spread. The aim of this study is to investigate mRNA and protein levels of ganglioside GD2 synthase (GD2S), in breast cancer (BC) patients. METHODS 65 PBMCs of preoperative BC patients without chemotherapy were compared to PBMCs after chemotherapy and controls. RESULTS GD2S were significantly higher in BC patients after chemotherapy compared to pre-chemotherapy at both mRNA and protein. GD2S was higher in pre-chemotherapy blood samples compared to control samples. CONCLUSIONS Higher expression of GD2S in BC samples compared to healthy control indicates the potential utility of GD2S as a marker of malignancy.
Collapse
Affiliation(s)
- Maryam Mansoori
- Iran University of Medical Sciences, Oncopathology Research Center, Tehran, Iran.,Iran University of Medical Sciences, Faculty of Advanced Technologies in Medicine, Department of Molecular Medicine, Tehran, Iran
| | - Isa Abdi Rad
- Urmia University of Medical Sciences, Cellular and Molecular Research Center, Urmia, Iran
| | - Alireza Mirzaei
- Iran University of Medical Sciences, Shafa Orthopedic Hospital, Bone and Joint Reconstruction Research Center, Tehran, Iran
| | - Kevin J Tam
- University of British Columbia, Vancouver Prostate Centre, Department of Urologic Sciences, Vancouver, Canada
| | - Seyed Mohsen Hosseini
- Omid specialty and subspecialty Hospital, Oncology and Radiotherapy Ward, Urmia, Iran
| | - Rahim Mahmodlu
- Urmia University of Medical Sciences, Faculty of Medicine, Imam Khomeini Hospital, Department of Surgery, Urmia, Iran
| | - Fatemeh Mansouri
- Urmia University of Medical Sciences, Faculty of Medicine, Department of Genetics and Immunology, Urmia, Iran
| | | | - Zahra Madjd
- Iran University of Medical Sciences, Oncopathology Research Center, Tehran, Iran.,Iran University of Medical Sciences, Faculty of Advanced Technologies in Medicine, Department of Molecular Medicine, Tehran, Iran
| |
Collapse
|
150
|
Koziorowski J, Ballinger J. Theragnostic radionuclides: a clinical perspective. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2021; 65:306-314. [PMID: 34881851 DOI: 10.23736/s1824-4785.21.03424-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The concept of theragnostics goes back to the earliest days of nuclear medicine, with [123I/131I]iodide in thyroid disease and [123I/131I]MIBG in phaeochromocytoma being examples in long-term use. However, in recent years there has been a great expansion in the application of theragnostics, beginning with [68Ga/177Lu]-labelled somatostatin peptides for evaluation and treatment of neuroendocrine tumors. We are currently seeing the rapid development of [68Ga/177Lu]PSMA theragnostics in metastatic prostate cancer. While these applications are very promising, there are a number of practicalities which must be addressed in the development and introduction of novel theragnostics. The physical half-lives of the diagnostic and therapeutic radionuclides must be appropriate for imaging and delivery of targeted cell killing, respectively. The types of radioactive emissions are critical; beta particles can traverse several millimeters but also risk damaging non-target tissues, while alpha particles deliver their energy over a much shorter path length, a few cell diameters, and must be more directly targeted. It must be practical to produce the therapeutic radionuclide and the final radiopharmaceutical and deliver them to the final user within an appropriate time-frame determined by half-life and stability. The biodistribution of the agent must demonstrate adequate accumulation and retention in the target tissue with clearance from adjacent and/or radio-sensitive normal tissues. The commercial success of recently introduced theragnostics suggests a rosy future for personalized medicine.
Collapse
|