1
|
Zhang P, Wang Z, Xu Y, Wu M. Mechanism underlying the role of the circRNA OMA1/miR-654-3p/RAF1 axis in children with inflammatory bowel disease. Cytotechnology 2025; 77:42. [PMID: 39867828 PMCID: PMC11759725 DOI: 10.1007/s10616-025-00703-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
Inflammatory bowel disease (IBD), a chronic gastrointestinal disorder, often emerges during childhood and poses significant challenges due to its adverse effects on growth, development, and psychosocial well-being. Circular RNAs (circRNAs) have been implicated in the pathogenesis of diverse diseases. However, the specific biological role and mechanisms of circRNA OMA1 in children with IBD remain largely unexplored. This study investigates the functions and mechanistic pathways of circRNA OMA1 in the progression of IBD. Quantitative real-time PCR (qRT-PCR) was employed to quantify circRNA OMA1 and miR-654-3p expression levels in the serum of children with IBD and in HT-29 cells. Downstream miRNA and mRNA targets of circRNA OMA1 were predicted using StarBase and validated via luciferase reporter assays. An in vitro IBD model was established by treating the human colonic epithelial cell line (HT-29) with 2% dextran sulfate sodium (DSS). Cell viability and apoptosis were assessed using the MTT assay and flow cytometry, respectively. Expression of the apoptosis-related protein cleaved caspase-3 was analyzed via western blotting, and proinflammatory cytokine levels (TNF-α, IL-1β, and IL-6) were measured using ELISA. The expression of circRNA OMA1 was notably lower in the serum of children with IBD and in DSS-treated HT-29 cells than in healthy controls, whereas miR-654-3p expression was upregulated. Bioinformatics analyses revealed a direct interaction between circRNA OMA1 and miR-654-3p. Overexpression of circRNA OMA1 through plasmid transfection increased circRNA OMA1 levels and suppressed miR-654-3p expression in HT-29 cells under both basal and DSS-stimulated conditions. Conversely, transfection with a miR-654-3p mimic reversed these effects. Upregulation of circRNA OMA1 ameliorated DSS-induced injury in HT-29 cells by enhancing cell viability, reducing apoptosis, and downregulating cleaved caspase-3 expression. Moreover, circRNA OMA1 overexpression inhibited the secretion of inflammatory cytokines TNF-α, IL-1β, and IL-6. However, these protective effects were partially reversed by treatment with the miR-654-3p mimic. Additionally, miR-654-3p was shown to directly target RAF1, negatively regulating its expression. The proliferation-promoting and apoptosis-suppressing effects of miR-654-3p inhibitor treatment were mitigated by RAF1-siRNA. Conclusion: Upregulation of circRNA OMA1 alleviates DSS-induced colonic cell apoptosis and inflammation by modulating the miR-654-3p/RAF1 axis. These findings suggest that circRNA OMA1 could be a promising biomarker for the diagnosis and treatment of IBD. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00703-z.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Child Health, Maternal and Child Health Hospital of Hubei Province, No. 745 Wuluo Road, Wuhan, 430070 China
| | - Zhenhui Wang
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070 China
| | - Yufen Xu
- Department of Child Health, Maternal and Child Health Hospital of Hubei Province, No. 745 Wuluo Road, Wuhan, 430070 China
| | - Meirong Wu
- Department of Child Health, Maternal and Child Health Hospital of Hubei Province, No. 745 Wuluo Road, Wuhan, 430070 China
| |
Collapse
|
2
|
Wang F, Zhang S, Chen Z, Gu X, Zhang G, Zhang H, Yuan W. N7-methyladenosine-induced SLC7A7 serves as a prognostic biomarker in pan-cancer and promotes CRC progression in colorectal cancer. Sci Rep 2024; 14:30755. [PMID: 39730571 DOI: 10.1038/s41598-024-80885-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Solute transport family 7A member 7 (SLC7A7) mutations contribute to lysinuric protein intolerance (LPI), which is the mechanism of action that has been extensively studied. In colorectal cancer (CRC), SLC7A7 appears to play a role, but the features and mechanisms are not yet well understood. Survival was analyzed using the Kaplan-Meier analysis. Enrichment analysis was performed to characterize, immune infiltration, methylation, genetic instability, and crucial pathways of SLC7A7. Afterward, functional experiments were conducted in vitro to investigate how SLC7A7 affects tumor metastasis. Mechanistically, quantitative real-time PCR (qRT-PCR), western blot (WB), and methylated RNA immunoprecipitation (me-RIP) were carried out to confirm the methylation modification of SLC7A7 and related functions. High levels of expression of SLC7A7 are predictive of a worse prognosis for CRC patients. Enrichment analysis showed that SLC7A7 was significantly enriched during EMT and could be enriched in the Wnt/β-catenin signaling pathway, immune infiltration analysis of pan-cancer showed that SLC7A7 was significantly enriched in macrophages, and methylation analysis showed that SLC7A7 methylation modification affected the prognosis of specific cancers. SLC7A7 was indicated to promote the migration and invasion of CRC cells in in vitro functional experiments. Mechanistically, SLC7A7 was observed to potentially interact with the Wnt/β-catenin signaling pathway, possibly by influencing adenomatous polyposis coli (APC) expression. Furthermore, we identified that SLC7A7 undergoes N7-methylguanosine (m7G) modification, which may regulate SLC7A7 mRNA stability, with Quaking (QKI) potentially playing a role in this process by recognizing the m7G modification. Our results indicate that SLC7A7 may promote CRC metastasis through the SLC7A7/APC/Wnt/β-catenin signaling pathway. Moreover, m7G modification might be involved in regulating SLC7A7 mRNA stability, highlighting a novel layer of regulation.
Collapse
Affiliation(s)
- Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Shiqian Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Zhuang Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hairong Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China.
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
3
|
Ercelik M, Tekin C, Gurbuz M, Tuncbilekli Y, Dogan HY, Mutlu B, Eser P, Tezcan G, Parın FN, Yildirim K, Sarihan M, Akpinar G, Kasap M, Bekar A, Kocaeli H, Taskapilioglu MO, Aksoy SA, Ozpar R, Hakyemez B, Tunca B. A new nano approach to prevent tumor growth in the local treatment of glioblastoma: Temozolomide and rutin-loaded hybrid layered composite nanofiber. Asian J Pharm Sci 2024; 19:100971. [PMID: 39640055 PMCID: PMC11617954 DOI: 10.1016/j.ajps.2024.100971] [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: 04/26/2024] [Revised: 06/14/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Total resection of glioblastoma (GB) tumors is nearly impossible, and systemic administration of temozolomide (TMZ) is often inadequate. This study presents a hybrid layered composite nanofiber mesh (LHN) designed for localized treatment in GB tumor bed. The LHN, consisting of polyvinyl alcohol and core-shell polylactic acid layers, was loaded with TMZ and rutin. In vitro analysis revealed that LHNTMZ and LHNrutin decelerated epithelial-mesenchymal transition and growth of stem-like cells, while the combination, LHNTMZ +rutin, significantly reduced sphere size compared to untreated and LHNTMZ-treated cells (P < 0.0001). In an orthotopic C6-induced GB rat model, LHNTMZ +rutin therapy demonstrated a more pronounced tumor-reducing effect than LHNTMZ alone. Tumor volume, assessed by magnetic resonance imaging, was significantly reduced in LHNTMZ +rutin-treated rats compared to untreated controls. Structural changes in tumor mitochondria, reduced membrane potential, and decreased PARP expression indicated the activation of apoptotic pathways in tumor cells, which was further confirmed by a reduction in PHH3, indicating decreased mitotic activity of tumor cells. Additionally, the local application of LHNs in the GB model mitigated aggressive tumor features without causing local tissue inflammation or adverse systemic effects. This was evidenced by a decrease in the angiogenesis marker CD31, the absence of inflammation or necrosis in H&E staining of the cerebellum, increased production of IFN-γ, decreased levels of interleukin-4 in splenic T cells, and lower serum AST levels. Our findings collectively indicate that LHNTMZ +rutin is a promising biocompatible model for the local treatment of GB.
Collapse
Affiliation(s)
- Melis Ercelik
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Cagla Tekin
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Melisa Gurbuz
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Yagmur Tuncbilekli
- Department of Neurosurgery, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Hazal Yılmaz Dogan
- Department of Metallurgical and Materials Engineering, Bursa Technical University, Bursa, Turkey
| | - Busra Mutlu
- Department of Metallurgical and Materials Engineering, Bursa Technical University, Bursa, Turkey
- Central Research Laboratory, Bursa Technical University, Bursa, Turkey
| | - Pınar Eser
- Department of Neurosurgery, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Gulcin Tezcan
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa, Turkey
| | - Fatma Nur Parın
- Department of Polymer Materials Engineering, Faculty of Engineering and Natural Sciences, Bursa Technical University, Bursa, Turkey
| | - Kenan Yildirim
- Department of Polymer Materials Engineering, Faculty of Engineering and Natural Sciences, Bursa Technical University, Bursa, Turkey
| | - Mehmet Sarihan
- Department of Medical Biology/Proteomics Laboratory, Kocaeli University, Kocaeli, Turkey
| | - Gurler Akpinar
- Department of Medical Biology/Proteomics Laboratory, Kocaeli University, Kocaeli, Turkey
| | - Murat Kasap
- Department of Medical Biology/Proteomics Laboratory, Kocaeli University, Kocaeli, Turkey
| | - Ahmet Bekar
- Department of Neurosurgery, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Hasan Kocaeli
- Department of Neurosurgery, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | | | - Secil Ak Aksoy
- Inegol Vocation School, Bursa Uludag University, Bursa, Turkey
| | - Rıfat Ozpar
- Department of Radiology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Bahattin Hakyemez
- Department of Radiology, Faculty of Medicine, Bursa Uludağ University, Bursa, Turkey
| | - Berrin Tunca
- Department of Medical Biology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
4
|
Yang GE, Kim MH, Jeong MS, Lee SY, Choi YH, Nam JK, Kim TN, Leem SH. Association between PDCD6-VNTR polymorphism and urinary cancer susceptibility. Genes Genomics 2024; 46:1281-1291. [PMID: 38850471 DOI: 10.1007/s13258-024-01523-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/08/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Programmed cell death 6 (PDCD6) is known to be involved in apoptosis and tumorigenesis. Given the reported association with urinary cancer susceptibility through SNP analysis, we further analyzed the entire genomic structure of PDCD6. METHODS Three VNTR regions (MS1-MS3) were identified through the analysis of the genomic structure of PDCD6. To investigate the association between these VNTR regions and urinary cancer susceptibility, genomic DNA was extracted from 413 cancer-free male controls, 267 bladder cancer patients, and 331 prostate cancer patients. Polymerase chain reaction (PCR) was performed to analyze the PDCD6-MS regions. Statistical analysis was performed to determine the association between specific genotypes and cancer risk. In addition, the effect of specific VNTRs on PDCD6 expression was also confirmed using a reporter vector. RESULTS Among the three VNTR regions, MS1 and MS2 exhibited monomorphism, while the MS3 region represented polymorphism, with its transmission to subsequent generations through meiosis substantiating its utility as a DNA typing marker. In a case-control study, the presence of rare alleles within PDCD6-MS3 exhibited significant associations with both bladder cancer (OR = 2.37, 95% CI: 1.33-4.95, P = 0.019) and prostate cancer (OR = 2.11, 95% CI: 1.03-4.36, P = 0.038). Furthermore, through luciferase assays, we validated the impact of the MS3 region on modulating PDCD6 expression. CONCLUSIONS This study suggests that the PDCD6-MS3 region could serve as a prognostic marker for urinary cancers, specifically bladder cancer and prostate cancer. Moreover, the subdued influence exerted by PDCD6-MS3 on the expression of PDCD6 offers another insight concerning the progression of urinary cancer.
Collapse
Affiliation(s)
- Gi-Eun Yang
- Department of Biomedical Sciences, Dong-A University, Busan, 49315, Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Korea
| | - Min-Hye Kim
- Department of Biomedical Sciences, Dong-A University, Busan, 49315, Korea
| | - Mi-So Jeong
- Department of Biomedical Sciences, Dong-A University, Busan, 49315, Korea
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Korea
| | - Sang-Yeop Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Anti-Aging Research Center, Dong-Eui University, Busan, 47227, Korea
| | - Jong-Kil Nam
- Department of Urology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Research Institute for Convergence of Biomedical Science and Technology, Yangsan, 50612, Korea
| | - Tae Nam Kim
- Department of Urology, Pusan National University Hospital, Pusan National University School of Medicine, Biomedical Research Institute and Pusan National University Hospital, Busan, 49241, Korea.
| | - Sun-Hee Leem
- Department of Biomedical Sciences, Dong-A University, Busan, 49315, Korea.
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Korea.
| |
Collapse
|
5
|
Zhu Y, Li Q. Multifaceted roles of PDCD6 both within and outside the cell. J Cell Physiol 2024; 239:e31235. [PMID: 38436472 DOI: 10.1002/jcp.31235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
Programmed cell death protein 6 (PDCD6) is an evolutionarily conserved Ca2+-binding protein. PDCD6 is involved in regulating multifaceted and pleiotropic cellular processes in different cellular compartments. For instance, nuclear PDCD6 regulates apoptosis and alternative splicing. PDCD6 is required for coat protein complex II-dependent endoplasmic reticulum-to-Golgi apparatus vesicular transport in the cytoplasm. Recent advances suggest that cytoplasmic PDCD6 is involved in the regulation of cytoskeletal dynamics and innate immune responses. Additionally, membranous PDCD6 participates in membrane repair through endosomal sorting complex required for transport complex-dependent membrane budding. Interestingly, extracellular vesicles are rich in PDCD6. Moreover, abnormal expression of PDCD6 is closely associated with many diseases, especially cancer. PDCD6 is therefore a multifaceted but pivotal protein in vivo. To gain a more comprehensive understanding of PDCD6 functions and to focus and stimulate PDCD6 research, this review summarizes key developments in its role in different subcellular compartments, processes, and pathologies.
Collapse
Affiliation(s)
- Yigao Zhu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qingchao Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
6
|
Wang P, Laster K, Jia X, Dong Z, Liu K. Targeting CRAF kinase in anti-cancer therapy: progress and opportunities. Mol Cancer 2023; 22:208. [PMID: 38111008 PMCID: PMC10726672 DOI: 10.1186/s12943-023-01903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
The RAS/mitogen-activated protein kinase (MAPK) signaling cascade is commonly dysregulated in human malignancies by processes driven by RAS or RAF oncogenes. Among the members of the RAF kinase family, CRAF plays an important role in the RAS-MAPK signaling pathway, as well as in the progression of cancer. Recent research has provided evidence implicating the role of CRAF in the physiological regulation and the resistance to BRAF inhibitors through MAPK-dependent and MAPK-independent mechanisms. Nevertheless, the effectiveness of solely targeting CRAF kinase activity remains controversial. Moreover, the kinase-independent function of CRAF may be essential for lung cancers with KRAS mutations. It is imperative to develop strategies to enhance efficacy and minimize toxicity in tumors driven by RAS or RAF oncogenes. The review investigates CRAF alterations observed in cancers and unravels the distinct roles of CRAF in cancers propelled by diverse oncogenes. This review also seeks to summarize CRAF-interacting proteins and delineate CRAF's regulation across various cancer hallmarks. Additionally, we discuss recent advances in pan-RAF inhibitors and their combination with other therapeutic approaches to improve treatment outcomes and minimize adverse effects in patients with RAF/RAS-mutant tumors. By providing a comprehensive understanding of the multifaceted role of CRAF in cancers and highlighting the latest developments in RAF inhibitor therapies, we endeavor to identify synergistic targets and elucidate resistance pathways, setting the stage for more robust and safer combination strategies for cancer treatment.
Collapse
Affiliation(s)
- Penglei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Kyle Laster
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, China-US (Henan) Hormel Cancer Institute, AMS, College of Medicine, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, 450052, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, China-US (Henan) Hormel Cancer Institute, AMS, College of Medicine, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.
- Basic Medicine Sciences Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
7
|
Hu JM, Chang YL, Hsieh CC, Huang SM. The Synergistic Cytotoxic Effects of GW5074 and Sorafenib by Impacting Mitochondrial Functions in Human Colorectal Cancer Cell Lines. Front Oncol 2022; 12:925653. [PMID: 35747833 PMCID: PMC9209736 DOI: 10.3389/fonc.2022.925653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) ranks third in the United States for incidence or mortality. Surgical resection is the primary treatment for patients at an early stage, while patients with advanced and metastatic CRC receive combined treatment with chemotherapy, radiotherapy, or targeted therapy. C-RAF plays a key role in maintaining clonogenic and tumorigenic capacity in CRC cells and it might be a potential therapeutic target for CRC. Sorafenib is a popular oral multi-kinase inhibitor, including a B-RAF inhibitor that targets the RAF-MEK-ERK pathway. Sorafenib, as a single agent, has tumor-suppressing efficacy, but its clinical application is limited due to many complex drug resistance mechanisms and side effects. GW5074 is one of the C-RAF inhibitors and has the potential to enhance the efficacy of existing cancer chemotherapies. In this study, we investigated whether the combination of sorafenib with GW5074 could reduce the dosage of sorafenib and enhance its tumor-suppressive effect in two CRC cell lines, HCT116 and LoVo cells. Our findings demonstrate that GW5074 can potentiate the cytotoxicity of sorafenib and dramatically reduce the half-maximal inhibitory concentration (IC50) dose of sorafenib from 17 and 31 µM to 0.14 and 0.01 µM in HCT116 and LoVo cells, respectively. GW5074, similar to sorafenib, suppressed the cellular proliferation and induced cellular apoptosis and cytosolic ROS, but had no further enhancement on the above-mentioned effects when combined with sorafenib. The synergistic effects of GW5074 and sorafenib were mainly found in mitochondrial functions, including ROS generation, membrane potential disruption, and fission–fusion dynamics, which were examined by using the flow cytometry analysis. In summary, the C-RAF inhibitor GW5074 might potentiate the cytotoxicity of the B-RAF inhibitor sorafenib mediated through mitochondrial dysfunctions, suggesting that GW5074 potentially serves as a sensitizer for sorafenib application to reduce the risk of drug resistance of CRC treatment. Our findings also provide novel insights on using C-RAF inhibitors combined with sorafenib, the current CRC therapeutic drug choice, in CRC treatment.
Collapse
Affiliation(s)
- Je-Ming Hu
- Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Surgery, Division of Colorectal Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Lung Chang
- Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Chih Hsieh
- School of Pharmacy and Institute of Pharmacy, National Defense Medical Center, Taipei, Taiwan
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shih-Ming Huang
- Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
- *Correspondence: Shih-Ming Huang,
| |
Collapse
|
8
|
Li M, Xu DM, Lin SB, Yang ZL, Xu TY, Yang JH, Lin ZX, Huang ZK, Yin J. Transcriptional expressions of hsa-mir-183 predicted target genes as independent indicators for prognosis in bladder urothelial carcinoma. Aging (Albany NY) 2022; 14:3782-3800. [PMID: 35503998 PMCID: PMC9134959 DOI: 10.18632/aging.204040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/22/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To uncover novel prognostic and therapeutic targets for BLCA, our study is the first to investigate the role of hsa-mir-183 and its up-regulated predicted target genes in bladder urothelial carcinoma. METHODS To address this issue, our study explored the roles of hsa-mir-183 predicted target genes in the prognosis of BLCA via UALCAN, Metascape, Kaplan-Meier plotter, Human Protein Atlas, TIMER2.0, cBioPortal and Genomics of Drug Sensitivity in Cancer databases. RESULTS High transcriptional expressions of PDCD6, GNG5, PHF6 and MAL2 were markedly relevant to favorable OS in BLCA patients, whereas SLC25A15 and PTDSS1 had opposite expression significance. Additionally, high transcriptional expression of PDCD6, GNG5, PHF6, MAL2, SLC25A15 and PTDSS1 were significantly correlated with BLCA individual cancer stages and molecular subtypes. Furthermore, high mutation rate of PDCD6, MAL2, SLC25A15 and PTDSS1 were observed. Finally, TP53 mutation of PDCD6, GNG5, PHF6, MAL2, SLC25A15 and PTDSS1 has guiding significance for drug selection in BLCA. CONCLUSIONS PDCD6, GNG5, PHF6, MAL2, SLC25A15 and PTDSS1 could be the advanced independent indicators for prognosis of BLCA patients, and TP53-mutation might be a biomarker for drug option in BLCA patients.
Collapse
Affiliation(s)
- Ming Li
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Da-Ming Xu
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Shu-Bin Lin
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zheng-Liang Yang
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Teng-Yu Xu
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jin-Huan Yang
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ze-Xin Lin
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ze-Kai Huang
- Division of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jun Yin
- Division of Hematology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
9
|
Zhuo Y, Hu W, Liao M, Chen J, Zhang J, Wang J, Zhang Y, Zhang F, Shi Y, Qian H, Li S, Sun C. Targeting TFAP2C/PDCD6 Pathway by lncRNA PP7080 Expedites Tumorigenesis and Contributes to an Immunosuppressive Tumor Microenvironment in Non‐Small Cell Lung Cancer. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yue Zhuo
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Wei Hu
- Precision Research Center for Refractory Diseases Institute for Clinical Research Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai 201620 P. R. China
| | - Mei‐Juan Liao
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Jia‐Hao Chen
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Jian Zhang
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Juan Wang
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Yu Zhang
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Fa‐Xue Zhang
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Yu‐Fan Shi
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Hui‐Ling Qian
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
| | - Shu‐Jun Li
- Department of Physical Examination Wuhan Hospital for the Prevention and Treatment of Occupational Diseases Wuhan Hubei 430022 P. R. China
| | - Cheng‐Cao Sun
- Department of Occupational and Environmental Health School of Public Health Wuhan University Wuhan Hubei 430071 P. R. China
- Department of Molecular and Cellular Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
10
|
Xu Y, Chen X, Pan S, Wang ZW, Zhu X. TM7SF2 regulates cell proliferation and apoptosis by activation of C-Raf/ERK pathway in cervical cancer. Cell Death Discov 2021; 7:299. [PMID: 34667152 PMCID: PMC8526692 DOI: 10.1038/s41420-021-00689-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/14/2021] [Accepted: 09/29/2021] [Indexed: 01/09/2023] Open
Abstract
Transmembrane 7 superfamily member 2 (TM7SF2) coding an enzyme involved in cholesterol metabolism has been found to be differentially expressed in kinds of tissues. Nevertheless, the role of TM7SF2 in the regulation of growth and progression among various cancers is unclear. In this study, the immunohistochemistry (IHC) assay, real-time RT-PCR and western blotting analysis were used to determine the TM7SF2 expression in cervical cancer tissues. Next, we used multiple methods to determine the ability of cell proliferation, migration, invasion, apoptosis, and cell cycle in cervical cancer cells after TM7SF2 modulation, such as CCK8 assay, colony formation assay, Transwell assay, wound healing assay, and flow cytometry. Our results revealed that upregulation of TM7SF2 facilitated cell proliferation and metastasis, suppressed cell apoptosis and prevented G0/G1 phase arrests in C33A and SiHa cells. Consistently, the opposite effects were observed after TM7SF2 knockout in cervical cancer cells. Further, we found that TM7SF2 participated in promoting tumorigenesis and progression via activation of C-Raf/ERK pathway in cervical cancer, which can be partly reversed by Raf inhibitor LY3009120. Moreover, TM7SF2 overexpression contributed to enhancement of xenograft tumor growth in vivo. Our findings indicated that TM7SF2 plays a vital role in tumor promotion by involving in C-Raf/ERK activation. Therefore, TM7SF2 could serve as a therapeutic target in future cervical cancer treatment.
Collapse
Affiliation(s)
- Yichi Xu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xin Chen
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shuya Pan
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhi-Wei Wang
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.
| | - Xueqiong Zhu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.
| |
Collapse
|
11
|
Yang G, Li G, Du X, Zhou W, Zou X, Liu Y, Lv H, Li Z. Down-regulation of IGHG1 enhances Protoporphyrin IX accumulation and inhibits hemin biosynthesis in colorectal cancer by suppressing the MEK-FECH axis. Open Life Sci 2021; 16:930-936. [PMID: 34553073 PMCID: PMC8422984 DOI: 10.1515/biol-2021-0098] [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: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 11/25/2022] Open
Abstract
Immunoglobulin γ-1 heavy chain constant region (IGHG1) is a functional isoform of immunoglobulins and plays an important role in the cytolytic activity of immune effector cells. Dysregulated IGHG1 was implicated in the occurrence and development of various tumors. Protoporphyrin IX (PpIX) is an endogenous fluorophore and is used in photodynamic therapy, which induces the generation of reactive oxygen species to initiate the death of tumor cells. However, the roles of IGHG1 in the colorectal cancer cell proliferation and PpIX accumulation have not been reported yet. Data from qRT-PCR and western blot analysis showed that IGHG1 was up-regulated in the colorectal cancer cells. Colorectal cancer cells were then transfected with shRNA targeting IGHG1 to down-regulate IGHG1 and conducted with Cell Counting Kit 8 (CCK8) and colony formation assays. Results demonstrated that shRNA-mediated down-regulation of IGHG1 decreased cell viability of colorectal cancer and suppressed cell proliferation. Moreover, PpIX accumulation was promoted and the hemin content was decreased by the silence of IGHG1. Interference of IGHG1 reduced the phosphorylated extracellular signal-regulated kinase (ERK) and ferrochelatase (FECH) expression, resulting in retarded cell proliferation in an MEK-FECH axis-dependent pathway.
Collapse
Affiliation(s)
- Guangjian Yang
- Department of Pathology, The First People’s Hospital of Longquanyi District of Chengdu, Chengdu, Sichuan, 610100, China
| | - Gang Li
- Department of Anorectal, The First People’s Hospital of Longquanyi District of Chengdu, No. 201, Group 3, Chengdu, Sichuan, 610100, China
| | - Xuemei Du
- Department of Pathology, The First People’s Hospital of Longquanyi District of Chengdu, Chengdu, Sichuan, 610100, China
| | - Wenting Zhou
- Department of Pathology, The First People’s Hospital of Longquanyi District of Chengdu, Chengdu, Sichuan, 610100, China
| | - Xiaohong Zou
- Department of Pathology, The First People’s Hospital of Longquanyi District of Chengdu, Chengdu, Sichuan, 610100, China
| | - Yuanfu Liu
- Department of Pathology, The First People’s Hospital of Longquanyi District of Chengdu, Chengdu, Sichuan, 610100, China
| | - Hong Lv
- Department of Pathology, The First People’s Hospital of Longquanyi District of Chengdu, Chengdu, Sichuan, 610100, China
| | - Zhenjiang Li
- Department of Research and Development, Sichuan Haosidelifu Science and Technology Ltd, Chengdu, Sichuan, 610041, China
| |
Collapse
|
12
|
Zhu G, Xia H, Tang Q, Bi F. An epithelial-mesenchymal transition-related 5-gene signature predicting the prognosis of hepatocellular carcinoma patients. Cancer Cell Int 2021; 21:166. [PMID: 33712026 PMCID: PMC7953549 DOI: 10.1186/s12935-021-01864-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background Tumor metastasis is one of the leading reasons of the dismal prognosis of hepatocellular carcinoma (HCC). Epithelial-mesenchymal transition (EMT) is closely associated with tumor metastasis including HCC. The purpose of this study is to construct and validate an EMT-related gene signature for predicting the prognosis of HCC patients. Methods Gene expression data of HCC patients was downloaded from The Cancer Genome Atlas (TCGA) database. Gene set enrichment analysis (GSEA) was performed to found the EMT-related gene sets which were obviously distinct between normal samples and paired HCC samples. Cox regression analysis was used to develop an EMT-related prognostic signature, and the performance of the signature was evaluated by Kaplan–Meier curves and time-dependent receiver operating characteristic (ROC) curves. A nomogram incorporating the independent predictors was established. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression levels of the hub genes in HCC cell lines, and the role of PDCD6 in the metastasis of HCC was determined by functional experiments. Results An EMT-related 5-gene signature (PDCD6, TCOF1, TRIM28, EZH2 and FAM83D) was constructed using univariate and multivariate Cox regression analysis. Based on the signature, the HCC patients were classified into high- and low-risk groups, and patients in high-risk group had a poor prognosis. Time-dependent ROC and Cox regression analyses suggested that the signature could predict HCC prognosis exactly and independently. The predictive capacity of the signature was also validated in two external cohorts. GSEA results showed that many cancer-related signaling pathways such as PI3K/Akt/mTOR pathway and TGF-β/SMAD pathway were enriched in high-risk group. The result of qRT-PCR revealed that PDCD6, TCOF1 and FAM83D were highly expressed in HCC cancer cells. Among them, PDCD6 were found to promote cell migration and invasion. Conclusion The EMT-related 5-gene signature can serve as a promising prognostic biomarker for HCC patients and may provide a novel mechanism of HCC metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01864-5.
Collapse
Affiliation(s)
- Gongmin Zhu
- Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, No.37 guoxue lane, Chengdu, 610041, Sichuan Province, China
| | - Hongwei Xia
- Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, No.37 guoxue lane, Chengdu, 610041, Sichuan Province, China
| | - Qiulin Tang
- Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, No.37 guoxue lane, Chengdu, 610041, Sichuan Province, China
| | - Feng Bi
- Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, No.37 guoxue lane, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
13
|
miRNA as promising theragnostic biomarkers for predicting radioresistance in cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2020; 157:103183. [PMID: 33310279 DOI: 10.1016/j.critrevonc.2020.103183] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 02/08/2023] Open
Abstract
Radioresistance remains as an obstacle in cancer treatment. This systematic review and meta-analysis aimed to evaluate the association between the expression of miRNAs and responses to radiotherapy and the prognosis of different tumors. In total, 77 miRNAs in 19 cancer types were studied, in which 24 miRNAs were upregulated and 58 miRNAs were downregulated in cancer patients. Five miRNAs were differentially expressed. Moreover, 75 miRNAs were found to be related to radioresistance, while 5 were observed to be related to radiosensitivity. The pooled HR and 95 % confidence interval for the combined studies was 1.135 (0.819-1.574; P-value = 0.4). The HR values of the subgroup analysis for miR-21 (HR = 2.344; 95 % CI: 1.927-2.850; P-value = 0.000), nasopharyngeal carcinoma (HR = 0.448; 95 % CI: 0.265-0.760; P = 0.003) and breast cancer (HR = 1.131; 95 % CI: 0.311-4.109; P = .85) were obtained. Our results highlighted that across the published literature, miRNAs can modulate tumor radioresistance or sensitivity by affecting radiation-related signaling pathways. It seems that miRNAs could be considered as a theragnostic biomarker to predict and monitor clinical response to radiotherapy. Thus, the prediction of radioresistance in malignant patients will improve radiotherapy outcomes and radiotherapeutic resistance.
Collapse
|