1
|
Benbelkacem M, Moulai N, Chader H, Ouahioune W, Bourouba M. Dichloroacetate and chloroquine in combination with arsenite suppress ROS-induced oral squamous cell carcinoma (OSCC) development and improve BALB/c mice survival. Free Radic Biol Med 2024; 227:593-607. [PMID: 39694118 DOI: 10.1016/j.freeradbiomed.2024.12.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/10/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a disabling tumor with poor response to chemotherapy. Here, we sought to explore a new chemotherapeutic approach based on a combined induction of cytotoxic ROS and targeting of autophagy and aerobic glycolysis as central contributors to OSCC carcinogenesis and chemoresistance. To this end, tongue OSCC was generated in BALB/c mice using 4NQO. Treatment of mouse-derived OSSC explants with NaAsO2 resulted in a strong inhibition of MTT activity and Bcl-2 and Ki-67 expression. The addition of chloroquine (CQ) and dichloroacetate (DCA) to arsenite, resulted in additive inhibitory effects on Bcl-2 and Ki-67 expression. Whereas NaAsO2 alone inhibited aerobic glycolysis (LDHA), it also alleviated autophagy (LC3B) and ROS levels (MDA). DCA improved NaAsO2-dependent inhibition of aerobic glycolysis. CQ addition to arsenite, suppressed autophagy without affecting the Warburg effect. NaAsO2 combination with CQ and DCA improved the oxidative status balance by boosting anti-oxidative CAT and SOD and controlling pro-oxidant MDA activity. The administration of the combo to 4NQO-mice resulted in a significant survival advantage over the control group (90 % vs. 35 % survival at week 32, p < 0.02; HR (log-rank) = 0.166, CI 95 % 0.03-0.73). This effect was accompanied by a significant increase in mice's mean body weight (p < 0.009). Contrarily to the control, administration of the combo resulted in the absence of progression towards severe dysplasia and OSCC and an overrepresentation of low/mild dysplasia events (100 %). Interestingly, signs of hepatocellular and renal toxicity following combo administration were limited in comparison to control. Taken together, these results suggest that NaAsO2 combined with CQ and DCA may constitute an interesting alternative to eliminating chemo-resistant OSSC tumors by inhibiting aerobic glycolysis and autophagy and controlling ROS generation. In vivo, the drugs may provide a survival advantage by inhibiting tumor development.
Collapse
Affiliation(s)
- Mounia Benbelkacem
- Laboratory of Cellular and Molecular Biology (LBCM), Team Biotechnology and System Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Bab-Ezzouar, Algiers, Algeria
| | - Nabila Moulai
- Laboratory of Anatomopathology, Frantz Fanon Hospital, Blida: University of Blida, Faculty of Medicine, Algeria
| | - Henni Chader
- Laboratory of Pharmaco-toxicology, National Agency of Pharmaceutical Products, Algiers, Algeria; Faculty of Pharmacy, University of Algiers 1, Algeria
| | - Wahiba Ouahioune
- Laboratory of Anatomopathology, Frantz Fanon Hospital, Blida: University of Blida, Faculty of Medicine, Algeria
| | - Mehdi Bourouba
- Laboratory of Cellular and Molecular Biology (LBCM), Team Biotechnology and System Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Bab-Ezzouar, Algiers, Algeria.
| |
Collapse
|
2
|
Hashemi M, Khoushab S, Aghmiuni MH, Anaraki SN, Alimohammadi M, Taheriazam A, Farahani N, Entezari M. Non-coding RNAs in oral cancer: Emerging biomarkers and therapeutic frontier. Heliyon 2024; 10:e40096. [PMID: 39583806 PMCID: PMC11582460 DOI: 10.1016/j.heliyon.2024.e40096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Around the world, oral cancer (OC) is a major public health problem, resulting in a significant number of deaths each year. Early detection and treatment are crucial for improving patient outcomes. Recent progress in DNA sequencing and transcriptome profiling has revealed extensive non-coding RNAs (ncRNAs) transcription, underscoring their regulatory importance. NcRNAs influence genomic transcription and translation and molecular signaling pathways, making them valuable for various clinical applications. Combining spatial transcriptomics (ST) and spatial metabolomics (SM) with single-cell RNA sequencing provides deeper insights into tumor microenvironments, enhancing diagnostic and therapeutic precision for OC. Additionally, the exploration of salivary biomarkers offers a non-invasive diagnostic avenue. This article explores the potential of ncRNAs as diagnostic and therapeutic tools for OC.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Hobabi Aghmiuni
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeid Nemati Anaraki
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Operative, Faculty of Dentistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University,Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Saadh MJ, Ehymayed HM, Alazzawi TS, Fahdil AA, Athab ZH, Yarmukhamedov B, Al-Anbari HHA, Shallal MM, Alsaikhan F, Farhood B. Role of circRNAs in regulating cell death in cancer: a comprehensive review. Cell Biochem Biophys 2024:10.1007/s12013-024-01492-6. [PMID: 39243349 DOI: 10.1007/s12013-024-01492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Despite multiple diagnostic and therapeutic advances, including surgery, radiation therapy, and chemotherapy, cancer preserved its spot as a global health concern. Prompt cancer diagnosis, treatment, and prognosis depend on the discovery of new biomarkers and therapeutic strategies. Circular RNAs (circRNAs) are considered as a stable, conserved, abundant, and varied group of RNA molecules that perform multiple roles such as gene regulation. There is evidence that circRNAs interact with RNA-binding proteins, especially capturing miRNAs. An extensive amount of research has presented the substantial contribution of circRNAs in various types of cancer. To fully understand the linkage between circRNAs and cancer growth as a consequence of various cell death processes, including autophagy, ferroptosis, and apoptosis, more research is necessary. The expression of circRNAs could be controlled to limit the occurrence and growth of cancer, providing a more encouraging method of cancer treatment. Consequently, it is critical to understand how circRNAs affect various forms of cancer cell death and evaluate whether circRNAs could be used as targets to induce tumor death and increase the efficacy of chemotherapy. The current study aims to review and comprehend the effects that circular RNAs exert on cell apoptosis, autophagy, and ferroptosis in cancer to investigate potential cancer treatment targets.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of dentist, National University of Science and Technology, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Bekhzod Yarmukhamedov
- Department of Surgical Dentistry and Dental Implantology, Tashkent State Dental Institute, Tashkent, Uzbekistan
- Department of Scientific affairs, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Li J, Li W, Li L, Wang W, Zhang M, Tang X. Induction of Peroxiredoxin 1 by Hypoxia Promotes Cellular Autophagy and Cell Proliferation in Oral Leukoplakia via HIF-1α/BNIP3 Pathway. J Mol Histol 2024; 55:403-413. [PMID: 38758520 DOI: 10.1007/s10735-024-10197-2] [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: 12/17/2023] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Hypoxia is a key trigger in the transformation of oral leukoplakia into oral cancer. However, it is still too early to determine the role of hypoxia in the development of oral leukoplakia. Prx1, an antioxidant protein, upregulated by hypoxia, regulates cellular autophagy in leukoplakia. This study aimed to understand the mechanisms by which hypoxia induces Prx1 expression during autophagy in oral leukoplakia. We used an experimental model of tongue epithelial hyperplasia induced by 4-nitroquinoline-1-oxide (4NQO) and dysplastic oral keratinocytes. Prx1 knockdown DOK cells, Leuk-1 cells and control cells were harvested, and cell proliferation was assayed using the Cell Counting Kit-8. Several hypoxia and autophagy-related proteins were examined using quantitative real-time polymerase chain reaction, immunohistochemistry, immunofluorescence, and western blotting in cells and mouse tongue tissues. In addition, the ultrastructure of the cells was observed by transmission electron microscopy. Hypoxia induces cell proliferation, autophagic vesicles and the expression of Prx1, BNIP3, LC3II/I and Beclin-1 in DOK and Leuk-1 cells. However, these effects were all attenuated by Prx1 knockdown. Histologically, 4NQO induced epithelial hyperplasia in the tongue mucosa. The expression of proliferation marker PCNA, autophagy-related proteins LC3B and Beclin-1, as well as HIF-1α/BNIP3 was significantly lower in the tongue tissues of Prx1flox/flox:Cre+ mice compared with Prx1flox/flox mice. In Prx1flox/flox:Cre+ mice, an increased expression of HIF-1α/BNIP3, LC3B and Beclin-1 was detected in epithelial hyperplasia tongue tissues compared to normal tissues. The current study suggests that Prx1 may promotes cell proliferation and autophagy in oral leukoplakia cells via the HIF-1α/BNIP3 pathway.
Collapse
Affiliation(s)
- Jing Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Wenjing Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Lingyu Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Wenchao Wang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Min Zhang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China.
| | - Xiaofei Tang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China.
| |
Collapse
|
5
|
Li N, Zuo R, He Y, Gong W, Wang Y, Chen L, Luo Y, Zhang C, Liu Z, Chen P, Guo H. PD-L1 induces autophagy and primary resistance to EGFR-TKIs in EGFR-mutant lung adenocarcinoma via the MAPK signaling pathway. Cell Death Dis 2024; 15:555. [PMID: 39090096 PMCID: PMC11294607 DOI: 10.1038/s41419-024-06945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Resistance to epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) is a significant cause of treatment failure and cancer recurrence in non-small cell lung cancer (NSCLC). Approximately 30% of patients with EGFR-activating mutations exhibit primary resistance to EGFR-TKIs. However, the potential mechanisms of primary resistance to EGFR-TKIs remain poorly understood. Recent studies have shown that increased expression of programmed death ligand-1 (PD-L1) is associated with EGFR-TKIs resistance. Therefore, the present study aimed to investigate the mechanism of PD-L1 in primary resistance to EGFR-TKIs in EGFR-mutant lung adenocarcinoma (LUAD) cells. We found that PD-L1 was associated with poor prognosis in patients with EGFR-mutant LUAD, while the combination of EGFR-TKIs with chemotherapy could improve its therapeutic efficacy. In vitro and in vivo experiments revealed that PD-L1 promoted the proliferation and autophagy and inhibited the apoptosis of LUAD cells. Mechanistic studies demonstrated that upregulation of PD-L1 was critical in inducing autophagy through the mitogen-activated protein kinase (MAPK) signaling pathway, which was beneficial for tumor progression and the development of gefitinib resistance. Furthermore, we found that gefitinib combined with pemetrexed could synergistically enhance antitumor efficacy in PD-L1-overexpression LUAD cells. Overall, our study demonstrated that PD-L1 contributed to primary resistance to EGFR-TKIs in EGFR-mutant LUAD cells, which may be mediated by inducing autophagy via the MAPK signaling pathway. These findings not only help improve the prognosis of patients with EGFR-mutant LUAD but also provide a reference for the research of other cancer types.
Collapse
Affiliation(s)
- Na Li
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Ran Zuo
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yuchao He
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Wenchen Gong
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yu Wang
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Liwei Chen
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yi Luo
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Cuicui Zhang
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Zhiyong Liu
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Peng Chen
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Hua Guo
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
6
|
Yuan J, Li Q, Sun Y, Wang Y, Li Y, You Z, Ni A, Zong Y, Ma H, Chen J. Multi-tissue transcriptome profiling linked the association between tissue-specific circRNAs and the heterosis for feed intake and efficiency in chicken. Poult Sci 2024; 103:103783. [PMID: 38713987 PMCID: PMC11091503 DOI: 10.1016/j.psj.2024.103783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/09/2024] Open
Abstract
Heterosis has been widely utilized in chickens. The nonadditive inheritance of genes contributes to this biological phenomenon. However, the role of circRNAs played in the heterosis is poorly determined. In this study, we observed divergent heterosis for residual feed intake (RFI) between 2 crossbreds derived from a reciprocal cross between White Leghorns and Beijing You chickens. Then, circRNA landscape for 120 samples covering the hypothalamus, liver, duodenum mucosa and ovary were profiled to elucidate the regulatory mechanisms of heterosis. We detected that a small proportion of circRNAs (7.83-20.35%) were additively and non-additively expressed, in which non-additivity was a major inheritance of circRNAs in the crossbreds. Tissue-specific expression of circRNAs was prevalent across 4 tissues. Weighted gene co-expression network analysis revealed circRNA-mRNA co-expression modules associated with feed intake and RFI in the hypothalamus and liver, and the co-expressed genes were enriched in oxidative phosphorylation pathway. We further identified 8 nonadditive circRNAs highly correlated with 16 nonadditive genes regulating negative heterosis for RFI in the 2 tissues. Circ-ITSN2 was validated in the liver tissue for its significantly positive correlation with PGPEP1L. Moreover, the bioinformatic analysis indicated that candidate circRNAs might be functioned by binding the microRNAs and interacting with the RNA binding proteins. The integration of multi-tissue transcriptome firstly linked the association between tissue-specific circRNAs and the heterosis for feed intake and efficiency in chicken, which provide novel insights into the molecular mechanism underlying heterosis for feed efficiency. The validated circRNAs can act as potential biomarkers for predicting RFI and its heterosis.
Collapse
Affiliation(s)
- Jingwei Yuan
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qin Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanyan Sun
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yuanmei Wang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yunlei Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhangjing You
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Aixin Ni
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yunhe Zong
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hui Ma
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jilan Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
7
|
Deng W, Fu J, Lin S, Wen Q, Fu L, Chen X. Hsa_circRNA_101036 aggravates hypoxic-induced endoplasmic reticulum stress via the miR-21-3p/TMTC1 axis in oral squamous cell carcinoma. Heliyon 2024; 10:e32969. [PMID: 38994041 PMCID: PMC11238011 DOI: 10.1016/j.heliyon.2024.e32969] [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: 07/25/2023] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Objective Circular RNAs (circRNAs) have been identified as potential biomarkers and therapeutic targets for various types of cancer, including Oral squamous cell carcinoma (OSCC). Hsa_circRNA_101036 was found to function as a cancer suppressor gene in OSCC; however, the underlying regulatory mechanism remains unclear. We investigated the role of hsa_circRNA_101036 in OSCC development and progression and explored its potential as a therapeutic target. Methods We performed a bioinformatics analysis and used experimental approaches to investigate the regulatory mechanism of hsa_circRNA_101036. The database StarBase v.2.0 was used to predict potential target-miRNAs of hsa_circRNA_101036. The levels of hsa_circRNA_101036, miR-21-3p, and TMTC2 expression in samples of OSCC cancer tissue (n = 15) and adjacent tissue (n = 15) were determined. We also examined the effects of hsa_circRNA_101036 overexpression on OSCC cell lines by using cell viability, migration, and invasion assays. The proportions of apoptotic cells and the reactive oxygen species (ROS) levels were analyzed by flow cytometry. We also investigated how hsa_circRNA_101036 overexpression affected the levels of miR-21-3p and TMTC2, and endoplasmic reticulum (ER) stress in OSCC cells. Results The levels of hsa_circRNA_101036 and TMTC2 expression were significantly lower, while miR-21-3p expression was higher in tumor tissues and OSCC cells when compared to adjacent tissues and normal oral fibroblasts, respectively. The levels of HIF-1α and miR-21-3p expression were significantly increased under conditions of hypoxia, while the levels of hsa_circRNA_101036 and TMTC2 were decreased. The expression levels of proteins associated with ER stress, the proportions of apoptotic cells, and the levels of ROS were all increased by hypoxia stimulation. In addition, overexpression of hsa_circRNA_101036, but not mutant hsa_circRNA_101036, was found to enhance the effect of hypoxia on HSC3 and OECM-1 cells. Hsa_circRNA_101036 overexpression suppressed tumor growth and induced ER stress. Finally, knockdown of miR-21-3p had the same effect as overexpression of hsa_circRNA_101036. Conclusion Our findings suggest that hsa_circRNA_101036 plays a critical role in the development and progression of OSCC. Overexpression of hsa_circRNA_101036 aggravated ER stress, and increased cell apoptosis and ROS production in OSCC under hypoxic conditions. Hsa_circRNA_101036 up-regulated TMTC2 expression by sponging miR-21-3p in OSCC.
Collapse
Affiliation(s)
- Wei Deng
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Juan Fu
- Department of Infectious Diseases, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Shigeng Lin
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Qitao Wen
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Liangbin Fu
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xiaoze Chen
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
8
|
Hao Z, Liu X, He H, Wei Z, Shu X, Wang J, Sun B, Zhou H, Wang J, Niu Y, Hu Z, Hu S, Liu Y, Fu Z. CYP2E1 deficit mediates cholic acid-induced malignant growth in hepatocellular carcinoma cells. Mol Med 2024; 30:79. [PMID: 38844847 PMCID: PMC11157842 DOI: 10.1186/s10020-024-00844-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Increased level of serum cholic acid (CA) is often accompanied with decreased CYP2E1 expression in hepatocellular carcinoma (HCC) patients. However, the roles of CA and CYP2E1 in hepatocarcinogenesis have not been elucidated. This study aimed to investigate the roles and the underlying mechanisms of CYP2E1 and CA in HCC cell growth. METHODS The proteomic analysis of liver tumors from DEN-induced male SD rats with CA administration was used to reveal the changes of protein expression in the CA treated group. The growth of CA-treated HCC cells was examined by colony formation assays. Autophagic flux was assessed with immunofluorescence and confocal microscopy. Western blot analysis was used to examine the expression of CYP2E1, mTOR, AKT, p62, and LC3II/I. A xenograft tumor model in nude mice was used to examine the role of CYP2E1 in CA-induced hepatocellular carcinogenesis. The samples from HCC patients were used to evaluate the clinical value of CYP2E1 expression. RESULTS CA treatment significantly increased the growth of HCC cells and promoted xenograft tumors accompanied by a decrease of CYP2E1 expression. Further studies revealed that both in vitro and in vivo, upregulated CYP2E1 expression inhibited the growth of HCC cells, blocked autophagic flux, decreased AKT phosphorylation, and increased mTOR phosphorylation. CYP2E1 was involved in CA-activated autophagy through the AKT/mTOR signaling. Finally, decreased CYP2E1 expression was observed in the tumor tissues of HCC patients and its expression level in tumors was negatively correlated with the serum level of total bile acids (TBA) and gamma-glutamyltransferase (GGT). CONCLUSIONS CYP2E1 downregulation contributes to CA-induced HCC development presumably through autophagy regulation. Thus, CYP2E1 may serve as a potential target for HCC drug development.
Collapse
Affiliation(s)
- Zhiwei Hao
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xuemin Liu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Huanhuan He
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Zhixuan Wei
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xiji Shu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Jianzhi Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Binlian Sun
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Hongyan Zhou
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Jiucheng Wang
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Ying Niu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Zhiyong Hu
- Department of Pathology, Renmin Hospital of Huangpi District of Jianghan University, Wuhan, 430399, China
| | - Shaobo Hu
- Liver transplant center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yuchen Liu
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China.
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China.
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.
- Liver transplant center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhengqi Fu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China.
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China.
| |
Collapse
|
9
|
Umapathy VR, Natarajan PM, Swamikannu B. Molecular and Therapeutic Roles of Non-Coding RNAs in Oral Cancer-A Review. Molecules 2024; 29:2402. [PMID: 38792263 PMCID: PMC11123887 DOI: 10.3390/molecules29102402] [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: 03/15/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Oral cancer (OC) is among the most common malignancies in the world. Despite advances in therapy, the worst-case scenario for OC remains metastasis, with a 50% survival rate. Therefore, it is critical to comprehend the pathophysiology of the condition and to create diagnostic and treatment plans for OC. The development of high-throughput genome sequencing has revealed that over 90% of the human genome encodes non-coding transcripts, or transcripts that do not code for any proteins. This paper describes the function of these different kinds of non-coding RNAs (ncRNAs) in OC as well as their intriguing therapeutic potential. The onset and development of OC, as well as treatment resistance, are linked to dysregulated ncRNA expression. These ncRNAs' potentially significant roles in diagnosis and prognosis have been suggested by their differing expression in blood or saliva. We have outlined every promising feature of ncRNAs in the treatment of OC in this study.
Collapse
Affiliation(s)
- Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Dr. M.G.R. Educational and Research Institute, Thai Moogambigai Dental College and Hospital, Chennai 600107, Tamil Nadu, India
| | - Prabhu Manickam Natarajan
- Department of Clinical Sciences, Centre of Medical and Bio-Allied Health Sciences and Research Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Bhuminathan Swamikannu
- Department of Prosthodontics, Sree Balaji Dental College and Hospital, Pallikaranai, BIHER, Chennai 600100, Tamil Nadu, India;
| |
Collapse
|
10
|
Samare-Najaf M, Samareh A, Savardashtaki A, Khajehyar N, Tajbakhsh A, Vakili S, Moghadam D, Rastegar S, Mohsenizadeh M, Jahromi BN, Vafadar A, Zarei R. Non-apoptotic cell death programs in cervical cancer with an emphasis on ferroptosis. Crit Rev Oncol Hematol 2024; 194:104249. [PMID: 38145831 DOI: 10.1016/j.critrevonc.2023.104249] [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: 08/30/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Cervical cancer, a pernicious gynecological malignancy, causes the mortality of hundreds of thousands of females worldwide. Despite a considerable decline in mortality, the surging incidence rate among younger women has raised serious concerns. Immortality is the most important characteristic of tumor cells, hence the carcinogenesis of cervical cancer cells pivotally requires compromising with cell death mechanisms. METHODS The current study comprehensively reviewed the mechanisms of non-apoptotic cell death programs to provide possible disease management strategies. RESULTS Comprehensive evidence has stated that focusing on necroptosis, pyroptosis, and autophagy for disease management is associated with significant limitations such as insufficient understanding, contradictory functions, dependence on disease stage, and complexity of intracellular pathways. However, ferroptosis represents a predictable role in cervix carcinogenesis, and ferroptosis-related genes demonstrate a remarkable correlation with patient survival and clinical outcomes. CONCLUSION Ferroptosis may be an appropriate option for disease management strategies from predicting prognosis to treatment.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran.
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nastaran Khajehyar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Delaram Moghadam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Rastegar
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Mohsenizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | | | - Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Zarei
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Sun ZY, Yang CL, Huang LJ, Mo ZC, Zhang KN, Fan WH, Wang KY, Wu F, Wang JG, Meng FL, Zhao Z, Jiang T. circRNADisease v2.0: an updated resource for high-quality experimentally supported circRNA-disease associations. Nucleic Acids Res 2024; 52:D1193-D1200. [PMID: 37897359 PMCID: PMC10767896 DOI: 10.1093/nar/gkad949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/30/2023] Open
Abstract
circRNADisease v2.0 is an enhanced and reliable database that offers experimentally verified relationships between circular RNAs (circRNAs) and various diseases. It is accessible at http://cgga.org.cn/circRNADisease/ or http://cgga.org.cn:9091/circRNADisease/. The database currently includes 6998 circRNA-disease entries across multiple species, representing a remarkable 19.77-fold increase compared to the previous version. This expansion consists of a substantial rise in the number of circRNAs (from 330 to 4246), types of diseases (from 48 to 330) and covered species (from human only to 12 species). Furthermore, a new section has been introduced in the database, which collects information on circRNA-associated factors (genes, proteins and microRNAs), molecular mechanisms (molecular pathways), biological functions (proliferation, migration, invasion, etc.), tumor and/or cell line and/or patient-derived xenograft (PDX) details, and prognostic evidence in diseases. In addition, we identified 7 159 865 relationships between mutations and circRNAs among 30 TCGA cancer types. Due to notable enhancements and extensive data expansions, the circRNADisease 2.0 database has become an invaluable asset for both clinical practice and fundamental research. It enables researchers to develop a more comprehensive understanding of how circRNAs impact complex diseases.
Collapse
Affiliation(s)
- Zhi-Yan Sun
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Chang-Lin Yang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Li-Jie Huang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Zong-Chao Mo
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Shenzhen 518045, China
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Ke-Nan Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Wen-Hua Fan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Kuan-Yu Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Fan Wu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Ji-Guang Wang
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Shenzhen 518045, China
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Fan-Lin Meng
- Marketing and Management Department, CapitalBio Technology, Beijing 101111, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China
| | - Zheng Zhao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Shenzhen 518045, China
- Division of Life Science, Department of Chemical and Biological Engineering, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
12
|
Yang X, Lou C, Zhang Q, Liu G, Ding Y, Zhang Q, Ye C. Hypoxia-induced circRTN4IP1 promotes progression and glycolysis of hepatocellular carcinoma cells. Funct Integr Genomics 2023; 23:339. [PMID: 37982910 DOI: 10.1007/s10142-023-01256-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/21/2023]
Abstract
Hypoxia is one of the hallmarks of solid tumors, especially in hepatocellular carcinoma (HCC). CircRNAs are reported to be tightly connected to hypoxia and also have essential roles in cancer progression. However, many circRNAs implicated in hypoxia-mediated HCC progression are still unclear and require further exploration. In this study, a hypoxia cell model was structured by exposing cells to hypoxia conditions (1% O2) and normoxia conditions (21% O2) as a control. The effects of hypoxia and normoxia on cell viability, migration, invasion, and glycolysis were examined. The expressions of circRNARTN4IP1 under hypoxia were identified. Finally, molecular mechanisms and biological function of circRTN4IP1 were explored. We confirmed that hypoxia treatment facilitated capacities of proliferation, migration, invasion, and glycolysis in tumor cells. Hypoxia induced a significant increase expression of circRTN4IP1 in cells. Functionally, knockdown of circRTN4IP1 inhibited cell malignant progression and glycolysis under hypoxia HCC cells. Mechanistically, HIF1A targeted the promoter region of circRTN4IP1 and positively regulated the expression of circRTN4IP1. In addition, circRTN4IP1 targeted miR-532-5p/G6PC3 axis. In short, hypoxia induced activation of the HIF1A/circRTN4IP1/miR-532-5p/G6PC3 signaling axis, which promoted proliferation, migration, invasion, and glycolysis of HCC cells. This study may reveal a possible mechanism driving the progression of hypoxia HCC, so as to find potential effective candidates for targeting hypoxia microenvironment therapy.
Collapse
Affiliation(s)
- Xijing Yang
- Department of Biotherapy, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), No. 700, North Moyu Road, Jiading District, Shanghai, 201805, China
| | - Cheng Lou
- Department of Oncology, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, 201805, China
| | - Qing Zhang
- Clinical Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guofang Liu
- Department of Biotherapy, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), No. 700, North Moyu Road, Jiading District, Shanghai, 201805, China
| | - Yongmei Ding
- Department of Biotherapy, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), No. 700, North Moyu Road, Jiading District, Shanghai, 201805, China
| | - Qian Zhang
- Department of Biotherapy, Third Affiliated Hospital of Naval Medical University (Eastern Hepatobiliary Surgery Hospital), No. 700, North Moyu Road, Jiading District, Shanghai, 201805, China.
| | - Chun Ye
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, No. 389, Xincun Road, Putuo District, Shanghai, 200065, China.
| |
Collapse
|
13
|
Wenhao R, Yali C, Shaoming L, Jingjing Z, Ling G, Keqian Z. circAP1M2 activates ATG9A-associated autophagy by inhibiting miR-1249-3p to promote cisplatin resistance in oral squamous cell carcinoma. J Cell Physiol 2023; 238:2612-2624. [PMID: 37661341 DOI: 10.1002/jcp.31116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
Cisplatin (CDDP) is the first-line chemotherapeutic agent for oral squamous cell carcinoma (OSCC). Susceptibility to drug resistance during treatment is a significant challenge in enhancing the therapeutic efficacy of OSCC. Autophagy is an essential element to guarantee the cancer cells' survival under chemo-stress conditions. We established a cisplatin-resistant OSCC cell line (CAL27/CDDP) and showed that circAP1M2 is a remarkably upregulated circular RNA in OSCC. Knockdown of circAP1M2 contributes to reversing cisplatin chemoresistance in vivo, while enhanced autophagic activity in cisplatin-resistant OSCC cells contributes to chemoresistance. Mechanistically, we showed that circAP1M2 induces autophagy-associated cisplatin resistance via the miR-1249-3p-ATG9A axis in OSCC cells. This study provides insights into the specific influence of a newly identified circular RNA circAP1M2 in OSCC regarding drug abuse and the treatment of a broad range of cancers that can benefit from cisplatin.
Collapse
Affiliation(s)
- Ren Wenhao
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng Yali
- Department of Stomatology, Huaian Hospital of Huaian City, Huaian, Jiangsu, China
| | - Li Shaoming
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Stomatology of Qingdao University, Qingdao, Shandong, China
| | - Zheng Jingjing
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Gao Ling
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhi Keqian
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
14
|
Zhu M, Chen D, Ruan C, Yang P, Zhu J, Zhang R, Li Y. CircRNAs: A Promising Star for Treatment and Prognosis in Oral Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:14194. [PMID: 37762497 PMCID: PMC10532269 DOI: 10.3390/ijms241814194] [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: 08/15/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
CircRNAs are a class of endogenous long non-coding RNAs with a single-stranded circular structure. Most circRNAs are relatively stable, highly conserved, and specifically expressed in tissue during the cell and developmental stages. Many circRNAs have been discovered in OSCC. OSCC is one of the most severe and frequent forms of head and neck cancer today, with a poor prognosis and low overall survival rate. Due to its prevalence, OSCC is a global health concern, characterized by genetic and epigenomic changes. However, the mechanism remains vague. With the advancement of biotechnology, a large number of circRNAs have been discovered in mammalian cells. CircRNAs are dysregulated in OSCC tissues and thus associated with the clinicopathological characteristics and prognosis of OSCC patients. Research studies have demonstrated that circRNAs can serve as biomarkers for OSCC diagnosis and treatment. Here, we summarized the properties, functions, and biogenesis of circRNAs, focusing on the progress of current research on circRNAs in OSCC.
Collapse
Affiliation(s)
| | | | | | | | | | - Rongxin Zhang
- Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (M.Z.); (D.C.); (C.R.); (J.Z.)
| | - Yan Li
- Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (M.Z.); (D.C.); (C.R.); (J.Z.)
| |
Collapse
|
15
|
Dey S, Biswas B, Manoj Appadan A, Shah J, Pal JK, Basu S, Sur S. Non-Coding RNAs in Oral Cancer: Emerging Roles and Clinical Applications. Cancers (Basel) 2023; 15:3752. [PMID: 37568568 PMCID: PMC10417002 DOI: 10.3390/cancers15153752] [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: 06/07/2023] [Revised: 06/29/2023] [Accepted: 07/12/2023] [Indexed: 08/13/2023] Open
Abstract
Oral cancer (OC) is among the most prevalent cancers in the world. Certain geographical areas are disproportionately affected by OC cases due to the regional differences in dietary habits, tobacco and alcohol consumption. However, conventional therapeutic methods do not yield satisfying treatment outcomes. Thus, there is an urgent need to understand the disease process and to develop diagnostic and therapeutic strategies for OC. In this review, we discuss the role of various types of ncRNAs in OC, and their promising clinical implications as prognostic or diagnostic markers and therapeutic targets. MicroRNA (miRNA), long ncRNA (lncRNA), circular RNA (circRNA), PIWI-interacting RNA (piRNA), and small nucleolar RNA (snoRNA) are the major ncRNA types whose involvement in OC are emerging. Dysregulated expression of ncRNAs, particularly miRNAs, lncRNAs, and circRNAs, are linked with the initiation, progression, as well as therapy resistance of OC via modulation in a series of cellular pathways through epigenetic, transcriptional, post-transcriptional, and translational modifications. Differential expressions of miRNAs and lncRNAs in blood, saliva or extracellular vesicles have indicated potential diagnostic and prognostic importance. In this review, we have summarized all the promising aspects of ncRNAs in the management of OC.
Collapse
Affiliation(s)
| | | | | | | | | | - Soumya Basu
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth (DPU), Pimpri 411033, India; (S.D.)
| | - Subhayan Sur
- Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth (DPU), Pimpri 411033, India; (S.D.)
| |
Collapse
|
16
|
Cui L, Huang C, Zhou D. Overexpression of circCDR1as drives oral squamous cell carcinoma progression. Oral Dis 2023; 29:957-967. [PMID: 34817912 DOI: 10.1111/odi.14085] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/29/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) mediate the progression of human cancers, including oral squamous cell carcinoma (OSCC). The aim of this study was to investigate the functions of circRNA CDR1 Antisense RNA (circCDR1as) in OSCC. Moreover, the relationships among circCDR1as, microRNA-876-5p (miR-876-5p) and Solute Carrier Family 7 Member 11 (SLC7A11) in OSCC development were explored. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to determine the expression of circCDR1as, miR-876-5p, and SLC7A11. Cell Counting kit-8 assay, cell colony formation assay, and 5-ethynyl-2'-deoxyuridine (EDU) assay were used to assess cell proliferation. Transwell assay was adopted for cell migration and invasion. RESULTS CircCDR1as level was aberrantly elevated in OSCC tissues and cells. Overexpression of circCDR1as promoted autophagy, cell cycle, proliferation, and metastasis and repressed apoptosis in OSCC cells. CircCDR1as directly targeted miR-876-5p and miR-876-5p interacted with SLC7A11. MiR-876-5p overexpression reversed the effects of circCDR1as elevation on OSCC cell autophagy, cell cycle, growth, motility, and apoptosis. Inhibition of miR-876-5p aggravated the malignant behaviors of OSCC cells, while SLC7A11 silencing ameliorated the impacts. In addition, circCDR1as knockdown blocked tumor growth in vivo. CONCLUSION CircCDR1as acted as an oncogene in OSCC progression through elevating SLC7A11 by targeting miR-876-5p.
Collapse
Affiliation(s)
- Lingling Cui
- Department of Stomatology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chao Huang
- Medical Administration Division, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Dun Zhou
- Hyperbaric Oxygen Treatment Room, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
17
|
Sun Q, Zhu E. Molecular mechanism and diagnostic marker investigation of endoplasmic reticulum stress on periodontitis. BMC Oral Health 2023; 23:135. [PMID: 36894919 PMCID: PMC9996967 DOI: 10.1186/s12903-023-02822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
PURPOSE The aim of this study was to reveal the biological function of endoplasmic reticulum stress (ERS)-related genes (ERSGs) in periodontitis, and provide potential ERS diagnostic markers for clinical therapy of periodontitis. METHODS The differentially expressed ERSGs (DE-ERSGs) were reveled based on periodontitis-related microarray dataset in Gene Expression Omnibus (GEO) database and 295 ERS in previous study, followed by a protein-protein interaction network construction. Then, the subtypes of periodontitis were explored, followed by validation with immune cell infiltration and gene set enrichment. Two machine learning algorithms were used to reveal potential ERS diagnostic markers of periodontitis. The diagnostic effect, target drug and immune correlation of these markers were further evaluated. Finally, a microRNA(miRNA)-gene interaction network was constructed. RESULTS A total of 34 DE-ERSGs were revealed between periodontitis samples and control, followed by two subtypes investigated. There was a significant difference of ERS score, immune infiltration and Hallmark enrichment between two subtypes. Then, totally 7 ERS diagnostic markers including FCGR2B, XBP1, EDEM2, ATP2A3, ERLEC1, HYOU1 and YOD1 were explored, and the v the time-dependent ROC analysis showed a reliable result. In addition, a drug-gene network was constructed with 4 up-regulated ERS diagnostic markers and 24 drugs. Finally, based on 32 interactions, 5 diagnostic markers and 20 miRNAs, a miRNA-target network was constructed. CONCLUSIONS Up-regulated miR-671-5p might take part in the progression of periodontitis via stimulating the expression of ATP2A3. ERSGs including XBP1 and FCGR2B might be novel diagnostic marker for periodontitis.
Collapse
Affiliation(s)
- Qianqian Sun
- Department of Stomatology, Sunshine Union Hospital, Yingqian Road, High-tech Zone, Weifang, 261000, Shandong, China
| | - Enqiang Zhu
- Department of Stomatology, Sunshine Union Hospital, Yingqian Road, High-tech Zone, Weifang, 261000, Shandong, China.
| |
Collapse
|
18
|
Zhao L, Wu X, Zhang Z, Fang L, Yang B, Li Y. ELF1 suppresses autophagy to reduce cisplatin resistance via the miR-152-3p/NCAM1/ERK axis in lung cancer cells. Cancer Sci 2023. [PMID: 36846943 DOI: 10.1111/cas.15770] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 02/12/2023] [Accepted: 02/23/2023] [Indexed: 03/01/2023] Open
Abstract
Resistance to chemotherapeutic drugs limits the efficacy of chemotherapy in non-small cell lung cancer (NSCLC). Autophagy is an essential mechanism which involves in drug resistance. Our previous research has revealed that miR-152-3p represses NSCLC progression. However, the mechanism of miR-152-3p in autophagy-mediated chemoresistance in NSCLC remains unclear. Cisplatin-resistant cell lines (A549/DDP and H446/DDP) were transfected with related vectors and subjected to cisplatin, autophagy inhibitor, activator, or extracellular signal-regulated kinase (ERK) activator. Flow cytometry, CCK8 and colony formation assays were performed for testing apoptosis and cell viability. The related RNAs or proteins were detected by qRT-PCR or Western blot. Chromatin immunoprecipitation, luciferase reporter assay or RNA immunoprecipitation were used for validating the interaction between miR-152-3p and ELF1 or NCAM1. Co-IP verified the binding between NCAM1 and ERK. The role of miR-152-3p in cisplatin resistance of NSCLC was also validated in vivo. The results showed that miR-152-3p and ELF1 were decreased in NSCLC tissues. miR-152-3p reversed cisplatin resistance by inhibiting autophagy through NCAM1. NCAM1 promoted autophagy through the ERK pathway and facilitated cisplatin resistance. ELF1 positively regulated miR-152-3p level by directly interacting with miR-152-3p promoter. miR-152-3p targeted NCAM1 to regulate NCAM1 level and then affected the binding of NCAM1 to ERK1/2. ELF1 inhibited autophagy and reversed cisplatin resistance through miR-152-3p/NCAM1. miR-152-3p inhibited autophagy and cisplatin resistance of xenograft tumor in mice. In conclusion, our study revealed that ELF1 inhibited autophagy to attenuate cisplatin resistance through the miR-152-3p/NCAM1/ERK pathway in H446/DDP and A549/DDP cells, suggesting a potential novel treatment strategy for NSCLC.
Collapse
Affiliation(s)
- Lifeng Zhao
- Departments of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xiangsheng Wu
- School of Clinical Medicine, Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Zhiwen Zhang
- School of Clinical Medicine, Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Lini Fang
- Departments of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Bo Yang
- Departments of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yepeng Li
- Departments of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
19
|
Xue Y, Jiang X, Wang J, Zong Y, Yuan Z, Miao S, Mao X. Effect of regulatory cell death on the occurrence and development of head and neck squamous cell carcinoma. Biomark Res 2023; 11:2. [PMID: 36600313 PMCID: PMC9814270 DOI: 10.1186/s40364-022-00433-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/08/2022] [Indexed: 01/06/2023] Open
Abstract
Head and neck cancer is a malignant tumour with a high mortality rate characterized by late diagnosis, high recurrence and metastasis rates, and poor prognosis. Head and neck squamous cell carcinoma (HNSCC) is the most common type of head and neck cancer. Various factors are involved in the occurrence and development of HNSCC, including external inflammatory stimuli and oncogenic viral infections. In recent years, studies on the regulation of cell death have provided new insights into the biology and therapeutic response of HNSCC, such as apoptosis, necroptosis, pyroptosis, autophagy, ferroptosis, and recently the newly discovered cuproptosis. We explored how various cell deaths act as a unique defence mechanism against cancer emergence and how they can be exploited to inhibit tumorigenesis and progression, thus introducing regulatory cell death (RCD) as a novel strategy for tumour therapy. In contrast to accidental cell death, RCD is controlled by specific signal transduction pathways, including TP53 signalling, KRAS signalling, NOTCH signalling, hypoxia signalling, and metabolic reprogramming. In this review, we describe the molecular mechanisms of nonapoptotic RCD and its relationship to HNSCC and discuss the crosstalk between relevant signalling pathways in HNSCC cells. We also highlight novel approaches to tumour elimination through RCD.
Collapse
Affiliation(s)
- Yuting Xue
- grid.412651.50000 0004 1808 3502Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xuejiao Jiang
- grid.24696.3f0000 0004 0369 153XBeijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Junrong Wang
- grid.412651.50000 0004 1808 3502Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuxuan Zong
- Department of Breast Surgery, The First of hospital of Qiqihar, Qiqihar, China
| | - Zhennan Yuan
- grid.412651.50000 0004 1808 3502Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Susheng Miao
- grid.412651.50000 0004 1808 3502Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xionghui Mao
- grid.412651.50000 0004 1808 3502Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
20
|
Ghafouri-Fard S, Askari A, Hussen BM, Rasul MF, Hatamian S, Taheri M, Kiani A. A review on the role of miR-671 in human disorders. Front Mol Biosci 2022; 9:1077968. [PMID: 36545507 PMCID: PMC9760869 DOI: 10.3389/fmolb.2022.1077968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
miR-671 is encoded by a gene on 7q36.1 and contributes to the pathogenesis of a variety of disorders, including diverse types of cancers, atherosclerosis, ischemic stroke, liver fibrosis, osteoarthritis, Parkinson's disease, rheumatoid arthritis, acute myocardial infarction and Crohn's disease. In the context of cancer, different studies have revealed opposite roles for this miRNA. In brief, it has been shown to be down-regulated in pancreatic ductal carcinoma, ovarian cancer, gastric cancer, osteosarcoma, esophageal squamous cell carcinoma and myelodysplastic syndromes. Yet, miR-671 has been up-regulated in glioma, colorectal cancer, prostate cancer and hepatocellular carcinoma. Studies in breast, lung and renal cell carcinoma have reported inconsistent results. The current review aims at summarization of the role of miR-671 in these disorders focusing on its target mRNA in each context and dysregulated signaling pathways. We also provide a summary of the role of this miRNA as a prognostic factor in malignancies.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arian Askari
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, Cihan University-Erbil, Kurdistan Region, Iraq,Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Iraq
| | - Sevak Hatamian
- Department of Anesthesia, Shahid Madani Hospital, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Institute of Human Genetics, Jena University Hospital, Jena, Germany,*Correspondence: Mohammad Taheri, ; Arda Kiani,
| | - Arda Kiani
- Tracheal Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran,*Correspondence: Mohammad Taheri, ; Arda Kiani,
| |
Collapse
|
21
|
Han X, Tian R, Wang C, Li Y, Song X. CircRNAs: Roles in regulating head and neck squamous cell carcinoma. Front Oncol 2022; 12:1026073. [PMID: 36483049 PMCID: PMC9723173 DOI: 10.3389/fonc.2022.1026073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/31/2022] [Indexed: 09/15/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), the most common head and neck malignant tumor, with only monotherapy, is characterized by poor prognosis, and low 5-year survival rate. Due to the lack of therapeutic targets, the targeted drugs for HNSCC are rare. Therefore, exploring the regulation mechanism of HNSCC and identifying effective therapeutic targets will be beneficial to its treatment of. Circular RNA (CircRNA) is a class of RNA molecules with a circular structure, which is widely expressed in human body. CircRNAs regulate gene expression by exerting the function as a miRNA sponge, thereby mediating the occurrence and development of HNSCC cell proliferation, apoptosis, migration, invasion, and other processes. In addition, circRNAs are also involved in the regulation of tumor sensitivity to chemical drugs and other biological functions. In this review, we systematically listed the functions of circRNAs and explored the regulatory mechanisms of circRNAs in HNSCC from the aspects of tumor growth, cell death, angiogenesis, tumor invasion and metastasis, tumor stem cell regulation, tumor drug resistance, immune escape, and tumor microenvironment. It will assist us in discovering new diagnostic markers and therapeutic targets, while encourage new ideas for the diagnosis and treatment of HNSCC.
Collapse
Affiliation(s)
- Xiao Han
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Ruxian Tian
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Cai Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Yumei Li
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Otorhinolaryngology Head and Neck Surgery, Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| |
Collapse
|
22
|
Jin Z, Sun X, Wang Y, Zhou C, Yang H, Zhou S. Regulation of autophagy fires up the cold tumor microenvironment to improve cancer immunotherapy. Front Immunol 2022; 13:1018903. [PMID: 36300110 PMCID: PMC9589261 DOI: 10.3389/fimmu.2022.1018903] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells, have revolutionized the treatment of patients with advanced and metastatic tumors resistant to traditional therapies. However, the immunosuppressed tumor microenvironment (TME) results in a weak response to immunotherapy. Therefore, to realize the full potential of immunotherapy and obstacle barriers, it is essential to explore how to convert cold TME to hot TME. Autophagy is a crucial cellular process that preserves cellular stability in the cellular components of the TME, contributing to the characterization of the immunosuppressive TME. Targeted autophagy ignites immunosuppressive TME by influencing antigen release, antigen presentation, antigen recognition, and immune cell trafficking, thereby enhancing the effectiveness of cancer immunotherapy and overcoming resistance to immunotherapy. In this review, we summarize the characteristics and components of TME, explore the mechanisms and functions of autophagy in the characterization and regulation of TME, and discuss autophagy-based therapies as adjuvant enhancers of immunotherapy to improve the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Zhicheng Jin
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Xuefeng Sun
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Yaoyao Wang
- Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, China
| | - Chao Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
- *Correspondence: Suna Zhou, ; HaihuaYang,
| | - Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China
- Department of Radiation Oncology, Xi’an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, China
- *Correspondence: Suna Zhou, ; HaihuaYang,
| |
Collapse
|
23
|
Gou Q, Zheng LL, Huang H. Unravelling the roles of Autophagy in OSCC: A renewed perspective from mechanisms to potential applications. Front Pharmacol 2022; 13:994643. [PMID: 36263139 PMCID: PMC9574005 DOI: 10.3389/fphar.2022.994643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/02/2022] [Indexed: 11/15/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is associated with a low survival rate and a high disability rate, making it a serious health burden, particularly in Southeast Asian countries. Therefore, improvements in the diagnosis, treatment, and prognosis prediction of OSCC are highly warranted. Autophagy has a significant impact on cancer development. Studies on autophagy in various human cancers have made outstanding contributions; however, the relationship between autophagy and OSCC remains to be explored. This review highlights the roles of autophagy in OSCC and discusses the relationship between autophagy and Epithelial–mesenchymal transition. Considering the lack of OSCC biomarkers, we focus on the studies involving OSCC-related bioinformatics analysis and molecular targets. Based on some classical targets, we summarize several key autophagy-related biomarkers with a considerable potential for clinical application, which may become the hotspot of OSCC research. In conclusion, we elaborate on the interrelationship between autophagy and OSCC and highlight the shortcomings of current studies to provide insights into the potential clinical strategies.
Collapse
Affiliation(s)
- Qiutong Gou
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Ling-Li Zheng
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- *Correspondence: Ling-Li Zheng, ; Haixia Huang,
| | - Haixia Huang
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
- *Correspondence: Ling-Li Zheng, ; Haixia Huang,
| |
Collapse
|
24
|
Yang Y, Li Y, Yuan H, Liu X, Ren Y, Gao C, Jiao T, Cai Y, Zhao S. Characterization of circRNA–miRNA–mRNA networks regulating oxygen utilization in type II alveolar epithelial cells of Tibetan pigs. Front Mol Biosci 2022; 9:854250. [PMID: 36213124 PMCID: PMC9532862 DOI: 10.3389/fmolb.2022.854250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Understanding the signaling pathway regulatory mechanisms in type II alveolar epithelial (ATII) cells, the progenitor cells responsible for proliferating and regenerating type I alveolar epithelial (ATI) and ATII cells, in Tibetan pigs is beneficial for exploring methods of preventing and repairing cellular damage during hypoxia. We simulated a hypoxic environment (2% O2) for culture ATII cells of Tibetan pigs and Landrace pigs, with cells cultured under normoxic conditions (21% O2) as a control group, and performed integrated analysis of circular RNA (circRNA)–microRNA (miRNA)–messenger RNA (mRNA) regulatory axes by whole-transcriptome sequencing. Functional enrichment analysis indicated that the source genes of the differential expressed circRNAs (DEcircRNAs) were primarily involved in cell proliferation, cellular processes, and cell killing. A series of DEcircRNAs were derived from inhibitors of apoptosis proteins and led to a key autonomous effect as modulators of cell repair in Tibetan pigs under hypoxia. The significant higher expression of COL5A1 in TL groups may inhibited apoptosis of ATII cells in Tibetan pigs under lower oxygen concentration, and may lead their better survive in the hypoxia environment. In addition, a competing endogenous RNA (ceRNA) network of functional interactions was constructed that included novel_circ_000898-ssc-miR-199a-5p-CAV1 and novel_circ_000898-ssc-miR-378-BMP2, based on the node genes ssc-miR-199a-5p and ssc-miR-378, which may regulate multiple miRNAs and mRNAs that mediate endoplasmic reticulum (ER) stress-induced apoptosis and inflammation and attenuate hypoxia-induced injury in ATII cells under hypoxic conditions. These results broaden our knowledge of circRNAs, miRNAs, and mRNAs associated with hypoxia and provide new insights into the hypoxic response of ATII cells in Tibetan pigs.
Collapse
Affiliation(s)
- Yanan Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yongqing Li
- Xinjiang Academy of Animal Sciences, Ürümqi, Xinjiang, China
| | - Haonan Yuan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xuanbo Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yue Ren
- Academy of Agriculture and Animal Husbandry Sciences, Institute of Animal Husbandry and Veterinary Medicine, Lhasa, China
| | - Caixia Gao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ting Jiao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- College of Grassland Science, Gansu Agricultural University, Lanzhou, China
| | - Yuan Cai
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- *Correspondence: Shengguo Zhao,
| |
Collapse
|
25
|
Long Noncoding RNAs and Circular RNAs Regulate AKT and Its Effectors to Control Cell Functions of Cancer Cells. Cells 2022; 11:cells11192940. [PMID: 36230902 PMCID: PMC9563963 DOI: 10.3390/cells11192940] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/06/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
AKT serine-threonine kinase (AKT) and its effectors are essential for maintaining cell proliferation, apoptosis, autophagy, endoplasmic reticulum (ER) stress, mitochondrial morphogenesis (fission/fusion), ferroptosis, necroptosis, DNA damage response (damage and repair), senescence, and migration of cancer cells. Several lncRNAs and circRNAs also regulate the expression of these functions by numerous pathways. However, the impact on cell functions by lncRNAs and circRNAs regulating AKT and its effectors is poorly understood. This review provides comprehensive information about the relationship of lncRNAs and circRNAs with AKT on the cell functions of cancer cells. the roles of several lncRNAs and circRNAs acting on AKT effectors, such as FOXO, mTORC1/2, S6K1/2, 4EBP1, SREBP, and HIF are explored. To further validate the relationship between AKT, AKT effectors, lncRNAs, and circRNAs, more predicted AKT- and AKT effector-targeting lncRNAs and circRNAs were retrieved from the LncTarD and circBase databases. Consistently, using an in-depth literature survey, these AKT- and AKT effector-targeting database lncRNAs and circRNAs were related to cell functions. Therefore, some lncRNAs and circRNAs can regulate several cell functions through modulating AKT and AKT effectors. This review provides insights into a comprehensive network of AKT and AKT effectors connecting to lncRNAs and circRNAs in the regulation of cancer cell functions.
Collapse
|
26
|
Yuan Y, Zhang X, Fan X, Peng Y, Jin Z. The emerging roles of circular RNA-mediated autophagy in tumorigenesis and cancer progression. Cell Death Dis 2022; 8:385. [PMID: 36104321 PMCID: PMC9474543 DOI: 10.1038/s41420-022-01172-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022]
Abstract
AbstractCircular RNA (circRNA) is characterized by a specific covalently closed ring structure. The back-splicing of precursor mRNA is the main way of circRNA generation, and various cis/trans-acting elements are involved in regulating the process. circRNAs exhibit multiple biological functions, including serving as sponges of microRNAs, interacting with proteins to regulate their stabilities and abilities, and acting as templates for protein translation. Autophagy participates in many physiological and pathological processes, especially it plays a vital role in tumorigenesis and carcinoma progression. Increasing numbers of evidences have revealed that circRNAs are implicated in regulating autophagy during tumor development. Until now, the roles of autophagy-associated circRNAs in carcinoma progression and their molecular mechanisms remain unclear. Here, the emerging regulatory roles and mechanisms of circRNAs in autophagy were summarized. Furtherly, the effects of autophagy-associated circRNAs on cancer development were described. We also prospected the potential of autophagy-associated circRNAs as novel therapeutic targets of tumors and as biomarkers for cancer diagnosis and prognosis.
Collapse
|
27
|
Zhou X, Lin J, Wang F, Chen X, Zhang Y, Hu Z, Jin X. Circular RNA-regulated autophagy is involved in cancer progression. Front Cell Dev Biol 2022; 10:961983. [PMID: 36187468 PMCID: PMC9515439 DOI: 10.3389/fcell.2022.961983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/03/2022] [Indexed: 12/05/2022] Open
Abstract
Circular RNAs (circRNAs) are a sort of long, non-coding RNA molecules with a covalently closed continuous ring structure without 5'-3' polarity and poly-A tail. The modulative role of circRNAs in malignant diseases has been elucidated by many studies in recent years via bioinformatics and high-throughput sequencing technologies. Generally, circRNA affects the proliferative, invasive, and migrative capacity of malignant cells via various mechanisms, exhibiting great potential as novel biomarkers in the diagnoses or treatments of malignancies. Meanwhile, autophagy preserves cellular homeostasis, serving as a vital molecular process in tumor progression. Mounting studies have demonstrated that autophagy can not only contribute to cancer cell survival but can also induce autophagic cell death in specific conditions. A growing number of research studies have indicated that there existed abundant associations between circRNAs and autophagy. Herein, we systemically reviewed and discussed recent studies on this topic in different malignancies and concluded that the circRNA–autophagy axis played crucial roles in the proliferation, metastasis, invasion, and drug or radiation resistance of different tumor cells.
Collapse
|
28
|
Li S, Gao L, Liu J, Guo C, Zheng J, Zhi K, Ren W. The microRNA-10b-Bim axis promotes cancer progression through activating autophagy in oral squamous cell carcinoma. Cell Death Dis 2022; 8:373. [PMID: 36008375 PMCID: PMC9411559 DOI: 10.1038/s41420-022-01168-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022]
Abstract
Autophagy is related to many cellular mechanisms and dysregulation of autophagy involves the pathological process in cancer. miR-10b activates autophagy, which promotes invasion and migration of OSCC. Its functional role in the mechanism of OSCC to autophagy remains to be unclear. Overexpression of miR-10b was followed by enhanced OSCC invasion and migration and activated autophagic protein, such as LC3II/ATG5. MiR-10b attracted Bim directly according to the Bio-informatics analyses and double luciferases reporter assays. Functional experiments further revealed that miR-10b could promote invasion and migration in vitro. In addition, miR-10b induced autophagy via inhibiting Bim in invasion and migration of OSCC. Notably, animal experiments confirmed that miR-10b-Bim promoted proliferation and autophagy in OSCC. In addition, this study provides a theoretical support for regulating the mechanism of OSCC by inducing autophagy with miR-10b-Bim as a target.
Collapse
Affiliation(s)
- Shaoming Li
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, No.1677 Wutaishan Road, Qingdao, 266555, China.,School of Stomatology of Qingdao University, Qingdao, 266003, China
| | - Ling Gao
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, No.1677 Wutaishan Road, Qingdao, 266555, China.,Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
| | - Jiacheng Liu
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, No.1677 Wutaishan Road, Qingdao, 266555, China.,School of Stomatology of Qingdao University, Qingdao, 266003, China
| | - Chao Guo
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, No.1677 Wutaishan Road, Qingdao, 266555, China.,School of Stomatology of Qingdao University, Qingdao, 266003, China
| | - Jingjing Zheng
- Department of Endodontics, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, No.1677 Wutaishan Road, Qingdao, 266555, China. .,Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
| | - Wenhao Ren
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, No.1677 Wutaishan Road, Qingdao, 266555, China. .,Key Lab of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266555, China.
| |
Collapse
|
29
|
Hypoxia-responsive circRNAs: A novel but important participant in non-coding RNAs ushered toward tumor hypoxia. Cell Death Dis 2022; 13:666. [PMID: 35915091 PMCID: PMC9343381 DOI: 10.1038/s41419-022-05114-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 01/21/2023]
Abstract
Given the rapid developments in RNA-seq technologies and bioinformatic analyses, circular RNAs (circRNAs) have gradually become recognized as a novel class of endogenous RNAs, characterized by covalent loop structures lacking free terminals, which perform multiple biological functions in cancer genesis, progression and metastasis. Hypoxia, a common feature of the tumor microenvironments, profoundly affects several fundamental adaptive responses of tumor cells by regulating the coding and non-coding transcriptomes and renders cancer's phenotypes more aggressive. Recently, hypoxia-responsive circRNAs have been recognized as a novel player in hypoxia-induced non-coding RNA transcriptomics to modulate the hypoxic responses and promote the progression and metastasis of hypoxic tumors. Moreover, via extracellular vesicles-exosomes, these hypoxia-responsive circRNAs could transmit hypoxia responses from cancer cells to the cells of surrounding matrices, even more distant cells of other organs. Here, we have summarized what is known about hypoxia-responsive circRNAs, with a focus on their interaction with hypoxia-inducible factors (HIFs), regulation of hypoxic responses and relevance with malignant carcinoma's clinical features, which will offer novel insights on the non-coding RNAs' regulation of cancer cells under hypoxic stress and might aid the identification of new theranostic targets and define new therapeutic strategies for those cancer patients with resistance to radiochemotherapy, because of the ubiquity of tumoral hypoxia.
Collapse
|
30
|
Sun R, Zhou Y, Cai Y, Shui C, Wang X, Zhu J. circ_0000045 promotes proliferation, migration, and invasion of head and neck squamous cell carcinomas via regulating HSP70 and MAPK pathway. BMC Cancer 2022; 22:799. [PMID: 35854245 PMCID: PMC9297571 DOI: 10.1186/s12885-022-09880-y] [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: 09/01/2021] [Accepted: 07/05/2022] [Indexed: 02/08/2023] Open
Abstract
Objective Head and neck squamous cell carcinoma (HNSCC) is one severe malignancy driven by complex cellular and signaling mechanisms. However, the roles of circular RNAs (circRNAs) in HNSCC’s development remains poorly understood. Therefore, this study investigated the functions of differentially expressed circRNAs in regulating HNSCC cell functions. Methods Differentially expressed circRNAs were characterized through RNA sequencing in HNSCC tissues. CircRNA’s identity was then confirmed using RT-PCR and Sanger’s sequencing. Next, expression levels of circRNA and mRNA were detected by qRT-PCR, after which protein abundances were measured by Western blotting. Subsequently, the proliferation, migration, and invasion of HNSCC cells was assessed by MTS, wound healing, and Transwell system, respectively, followed by identification of circRNA-binding proteins in HNSCC cells by circRNA pull-down, coupled with mass spectrometry. Results Great alterations in circRNA profiles were detected in HNSCC tissues, including the elevated expression of circ_0000045. As observed, silencing of circ_0000045 effectively repressed the proliferation, migration, and invasion of HNSCC cell lines (FaDu and SCC-9). Contrarily, circ_0000045’s overexpression promoted the proliferation, migration, and invasion in FaDu and SCC-9 cells. Results also showed that circ_0000045 was associated with multiple RNA-binding proteins in HNSCC cells, such as HSP70. Moreover, circ_0000045 knockdown enhanced HSP70 expression and inhibited JNK2 and P38’s expression in HNSCC cells, which were oppositely regulated by circ_0000045’s overexpression. Conclusion The high expression of circ_0000045; therefore, promoted cell proliferation, migration, and invasion during HNSCC’s development through regulating HSP70 protein and mitogen-activated protein kinase signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09880-y.
Collapse
Affiliation(s)
- Ronghao Sun
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute,University of Electronic Science and Technology of China, No.55, 4th section of Southern Renmin Road, Chengdu, Sichuan, 610041, China. .,Department of Thyroid and Parathyroid Surgery, West China Hospital, No. 37, Guoxue Alley, Chengdu, Sichuan, 610041, China.
| | - Yuqiu Zhou
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute,University of Electronic Science and Technology of China, No.55, 4th section of Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Yongcong Cai
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute,University of Electronic Science and Technology of China, No.55, 4th section of Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Chunyan Shui
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute,University of Electronic Science and Technology of China, No.55, 4th section of Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Xu Wang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute,University of Electronic Science and Technology of China, No.55, 4th section of Southern Renmin Road, Chengdu, Sichuan, 610041, China
| | - Jingqiang Zhu
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute,University of Electronic Science and Technology of China, No.55, 4th section of Southern Renmin Road, Chengdu, Sichuan, 610041, China. .,Department of Thyroid and Parathyroid Surgery, West China Hospital, No. 37, Guoxue Alley, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
31
|
Erfanparast L, Taghizadieh M, Shekarchi AA. Non-Coding RNAs and Oral Cancer: Small Molecules With Big Functions. Front Oncol 2022; 12:914593. [PMID: 35898889 PMCID: PMC9309727 DOI: 10.3389/fonc.2022.914593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/16/2022] [Indexed: 12/24/2022] Open
Abstract
Oral cancer remains a major public concern with considerable socioeconomic impact in the world. Despite substantial advancements have been made in treating oral cancer, the five-year survival rate for oral cancer remained undesirable, and the molecular mechanisms underlying OSCC carcinogenesis have not been fully understood. Noncoding RNAs (ncRNAs) include transfer RNAs (tRNAs), as well as small RNAs such as microRNAs, and the long ncRNAs such as HOTAIR are a large segment of the transcriptome that do not have apparent protein-coding roles, but they have been verified to play important roles in diverse biological processes, including cancer cell development. Cell death, such as apoptosis, necrosis, and autophagy, plays a vital role in the progression of cancer. A better understanding of the regulatory relationships between ncRNAs and these various types of cancer cell death is therefore urgently required. The occurrence and development of oral cancer can be controlled by increasing or decreasing the expression of ncRNAs, a method which confers broad prospects for oral cancer treatment. Therefore, it is urgent for us to understand the influence of ncRNAs on the development of different modes of oral tumor death, and to evaluate whether ncRNAs have the potential to be used as biological targets for inducing cell death and recurrence of chemotherapy. The purpose of this review is to describe the impact of ncRNAs on cell apoptosis and autophagy in oral cancer in order to explore potential targets for oral cancer therapy.
Collapse
Affiliation(s)
- Leila Erfanparast
- Department of Pediatric Dentistry, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Mohammad Taghizadieh,
| | - Ali Akbar Shekarchi
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Han Y, Xia L, Wang X, Xiong H, Zeng L, Wang Z, Zhang T, Xia K, Hu X, Su T. Study on the expression and function of chordin like 1 in oral squamous cell carcinoma. Oral Dis 2022. [PMID: 35510812 DOI: 10.1111/odi.14240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the role and related mechanism of chordin like 1 (CHRDL1) in oral squamous cell carcinoma (OSCC). METHODS The expressions of CHRDL1 were analyzed in both mRNA and protein levels by bioinformatics analysis, immunohistochemistry, and fluorescence in situ hybridization in OSCC. Survival analysis was used to determine the relationship between CHRDL1 and prognosis. In addition, enrichment analysis was used to suggest signal pathways involved in CHRDL1. Besides, the relationships between CHRDL1 and miRNAs, hypoxia, and immune infiltration were explored. RESULTS The mRNA level of CHRDL1 in OSCC was significantly lower than that in normal tissues, while the protein level was significantly higher than that in normal tissues. The high mRNA levels of CHRDL1 suggested a poor prognosis in patients with OSCC. The enrichment results showed that CHRDL1 might be involved in the Calcium signaling pathway, dilated cardiomyopathy, and focal adhesion. 7 immune cells were positively correlated with CHRDL1, while Tgd was negatively correlated with CHRDL1. In addition, we also found that hsa-miR-455-3p directly targeted CHRDL1 and reduced the mRNA levels of CHRDL1. CONCLUSION CHRDL1 plays a vital role in promoting cancer in OSCC and is downregulated at the mRNA levels by hsa-miR-455-3p.
Collapse
Affiliation(s)
- Ying Han
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Lu Xia
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Xiaomeng Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Haofeng Xiong
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, China
| | - Liujun Zeng
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, China
| | - Zijia Wang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, China
| | - Tianyi Zhang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, China
| | - Kun Xia
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Xin Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, China
| | - Tong Su
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya hospital of Central South University, Changsha, Hunan, 410008, China.,Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
33
|
Hsu PC, Hsu CC, Hsia YJ, Kuo CY. Chrysophanol Suppresses Cell Growth via mTOR/PPAR-α Regulation and ROS Accumulation in Cultured Human Tongue Squamous Carcinoma SAS Cells. Curr Issues Mol Biol 2022; 44:1528-1538. [PMID: 35723362 PMCID: PMC9164035 DOI: 10.3390/cimb44040104] [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: 02/23/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Oral cancer, a type of head and neck cancer, can pose a significant risk of death unless diagnosed and treated early. Alternative treatments are urgently needed owing to the high mortality rate, limitations of conventional treatments, and many complications. The anthraquinone compound chrysophanol acts as a tumor suppressor on some types of cancer cells. To date, it has not been clarified how chrysophanol affects human tongue squamous carcinoma. This study was aimed to examine the effects of chrysophanol on oral cancer treatment. The results show that chrysophanol caused cell death, reduced the expression of the mammalian target of rapamycin (mTOR)/peroxisome proliferator-activated receptor-alpha (PPAR-α), and increased reactive oxygen species (ROS) production. We also used two ion chelators, deferoxamine (DFO) and liproxstatin-1 (Lipro), to further determine whether chrysophanol inhibits cell growth and regulates mTOR/PPAR-α expression and ROS production, both of which are involved in iron homeostasis. The results show that DFO and Lipro reversed the increase in cell death, downregulation of mTOR/PPAR-α, and decrease in ROS accumulation. In conclusion, chrysophanol inhibits the growth of oral squamous cell carcinoma cells by modulating mTOR/PPAR-α and by causing ROS accumulation.
Collapse
Affiliation(s)
- Po-Chih Hsu
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (P.-C.H.); (C.-C.H.); (Y.-J.H.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Chia-Chen Hsu
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (P.-C.H.); (C.-C.H.); (Y.-J.H.)
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Yi-Jan Hsia
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (P.-C.H.); (C.-C.H.); (Y.-J.H.)
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Correspondence: ; Fax: +886-2-6628-9009
| |
Collapse
|
34
|
The World of Oral Cancer and Its Risk Factors Viewed from the Aspect of MicroRNA Expression Patterns. Genes (Basel) 2022; 13:genes13040594. [PMID: 35456400 PMCID: PMC9027895 DOI: 10.3390/genes13040594] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
Oral cancer is one of the leading causes of death worldwide, with a reported 5-year survival rate of around 50% after treatment. Epigenetic modifications are considered to have a key role in oral carcinogenesis due to histone modifications, aberrant DNA methylation, and altered expression of miRNAs. MicroRNAs (miRNAs) are small non-coding RNAs that have a key role in cancer development by regulating signaling pathways involved in carcinogenesis. MiRNA deregulation identified in oral cancer has led to the idea of using them as potential biomarkers for early diagnosis, prognosis, and the development of novel therapeutic strategies. In recent years, a key role has been observed for risk factors in preventing and treating this malignancy. The purpose of this review is to summarize the recent knowledge about the altered mechanisms of oral cancer due to risk factors and the role of miRNAs in these mechanisms.
Collapse
|
35
|
Yang Z, Huang C, Wen X, Liu W, Huang X, Li Y, Zang J, Weng Z, Lu D, Tsang CK, Li K, Xu A. Circular RNA circ-FoxO3 attenuates blood-brain barrier damage by inducing autophagy during ischemia/reperfusion. Mol Ther 2022; 30:1275-1287. [PMID: 34763084 PMCID: PMC8899525 DOI: 10.1016/j.ymthe.2021.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/23/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023] Open
Abstract
Blood-brain barrier (BBB) damage can be a result of central nervous system (CNS) diseases and may be a cause of CNS deterioration. However, there are still many unknowns regarding effective and targeted therapies for maintaining BBB integrity during ischemia/reperfusion (I/R) injury. In this study, we demonstrate that the circular RNA of FoxO3 (circ-FoxO3) promotes autophagy via mTORC1 inhibition to attenuate BBB collapse under I/R. Upregulation of circ-FoxO3 and autophagic flux were detected in brain microvessel endothelial cells in patients with hemorrhagic transformation and in mice models with middle cerebral artery occlusion/reperfusion. In vivo and in vitro studies indicated that circ-FoxO3 alleviated BBB damage principally by autophagy activation. Mechanistically, we found that circ-FoxO3 inhibited mTORC1 activity mainly by sequestering mTOR and E2F1, thus promoting autophagy to clear cytotoxic aggregates for improving BBB integrity. These results demonstrate that circ-FoxO3 plays a novel role in protecting against BBB damage, and that circ-FoxO3 may be a promising therapeutic target for neurological disorders associated with BBB damage.
Collapse
Affiliation(s)
- Zhenguo Yang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China,Affiliated Hospital of Guangdong Medical University, 57 South Renmin Ave, Zhanjiang 524001, China
| | - Cheng Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Xueyi Wen
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Wenlin Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China,Affiliated Hospital of Guangdong Medical University, 57 South Renmin Ave, Zhanjiang 524001, China
| | - Xiaoxiong Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Yufeng Li
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Jiankun Zang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Zean Weng
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Dan Lu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Chi Kwan Tsang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China
| | - Keshen Li
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China.
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, & Clinical Neuroscience Institute of Jinan University, 613 West Huangpu Ave, Guangzhou 510632, China.
| |
Collapse
|
36
|
Lv S, Qian Z, Li J, Piao S, Li J. Identification and Validation of a Hypoxia-Immune-Based Prognostic mRNA Signature for Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5286251. [PMID: 35178089 PMCID: PMC8844353 DOI: 10.1155/2022/5286251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/22/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a commonly encountered head and neck malignancy. Increasing evidence shows that there are abnormal immune response and chronic cell hypoxia in the development of OSCC. However, there is a lack of a reliable hypoxia-immune-based gene signature that may serve to accurately prognosticate OSCC. METHODS The mRNA expression data of OSCC patients were extracted from the TCGA and GEO databases. Hypoxia status was identified using the t-distributed Stochastic Neighbor Embedding (t-SNE) algorithm. Both ESTIMATE and single-sample gene-set enrichment analysis (ssGSEA) were used for further evaluation of immune status. The DEGs in different hypoxia and immune status were determined, and univariate Cox regression was used to identify significantly prognostic genes. A machine learning method, least absolute shrinkage and selection operator (LASSO) Cox regression analysis, allowed us to construct prognostic gene signature to predict the overall survival (OS) of OSCC patients. RESULTS A total of 773 DEGs were identified between hypoxia high and low groups. According to immune cell infiltration, patients were divided into immune high, medium, and low groups and immune-associated DEGs were identified. A total of 193 overlapped DEGs in both immune and hypoxia status were identified. With the univariate and LASSO Cox regression model, eight signature mRNAs (FAM122C, RNF157, RANBP17, SOWAHA, KIAA1211, RIPPLY2, INSL3, and DNAH1) were selected for further calculation of their respective risk scores. The risk score showed a significant association with age and perineural and lymphovascular invasion. In the GEO validation cohort, a better OS was observed in patients from the low-risk group in comparison with those in the high-risk group. High-risk patients also demonstrated different immune infiltration characteristics from the low-risk group and the low-risk group showed potentially better immunotherapy efficacy in contrast to high-risk ones. CONCLUSION The hypoxia-immune-based gene signature has prognostic potential in OSCC.
Collapse
Affiliation(s)
- Shaohua Lv
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
- Stomatology School, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang, China
| | - Zhipeng Qian
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jianhao Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Songlin Piao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Jichen Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
- Stomatology School, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang, China
| |
Collapse
|
37
|
Zhang Y, Zhang Y, Ai B, Gong J, Li Y, Yu S, Cai X, Zhang L. GTF2E2 is a novel biomarker for recurrence after surgery and promotes progression of esophageal squamous cell carcinoma via miR-139-5p/GTF2E2/FUS axis. Oncogene 2022; 41:782-796. [PMID: 34853466 PMCID: PMC8816730 DOI: 10.1038/s41388-021-02122-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most lethal gastrointestinal malignancies with high mortality. Recurrence develops within only a few years after curative resection and perioperative adjuvant therapy in 30-50% of these patients. Therefore, it is essential to identify postoperative recurrence biomarkers to facilitate selecting the following surveillance and therapeutic strategies. The general transcription factor IIE subunit beta (GTF2E2) is crucial for physiological and pathological functions, but its roles in the aggression and recurrence of ESCC remain ambiguous. In this study, we found that GTF2E2 was highly expressed in ESCC samples, and elevated GTF2E2 expression predicted early recurrence after surgery for ESCC patients. High expression of GTF2E2 associated with more aggressive clinic features and poor prognosis. GTF2E2 promoted the proliferation and mobility of ESCC cells in vitro and in vivo. We further revealed that miR-139-5p repressed GTF2E2 expression by downregulating its mRNA through binding with Argonaute 2 (Ago2). Rescue assays suggested that miR-139-5p affected GTF2E2-mediated ESCC progression. Moreover, GTF2E2 positively interacted with FUS promoter and regulated FUS expression, and the phenotype changes caused by GTF2E2 manipulation were recovered by rescuing FUS expression in ESCC cells. Additionally, we demonstrated that GTF2E2 promotes ESCC cells progression via activation of the AKT/ERK/mTOR pathway. In conclusion, GTF2E2 may serve as a novel biomarker for recurrence after surgery and a potential therapeutic target for ESCC patients, and it promotes ESCC progression via miR-139-5p/GTF2E2/FUS axis.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Disease Progression
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/pathology
- Esophageal Neoplasms/surgery
- Esophageal Neoplasms/metabolism
- Esophageal Squamous Cell Carcinoma/genetics
- Esophageal Squamous Cell Carcinoma/pathology
- Esophageal Squamous Cell Carcinoma/surgery
- Esophageal Squamous Cell Carcinoma/metabolism
- Gene Expression Regulation, Neoplastic
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/metabolism
- Prognosis
- Transcription Factors, TFII/genetics
- Transcription Factors, TFII/metabolism
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yuxin Zhang
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Bo Ai
- Thoracic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Juejun Gong
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yichen Li
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Shiying Yu
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Xiuyu Cai
- Department of VIP Inpatient, Sun Yet-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No.651 Dongfeng Road East, Guangzhou, Guangdong, 510060, P. R. China.
| | - Li Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
38
|
Aghajani Mir M, Dinmohammadi H, Moudi E, Motamed N, Daraei A. Clinical values of expression signature of circCDR1AS and circHIAT1 in prostate cancer: Two circRNAs with regulatory function in androgen receptor (AR) and PI3K/AKT signaling pathways. J Clin Lab Anal 2022; 36:e24220. [PMID: 35007362 PMCID: PMC8841177 DOI: 10.1002/jcla.24220] [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: 09/08/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Background Prostate cancer (PCa) is a genetically heterogeneous disease with highly molecular aberrations. It has been revealed that a newly discovered class of non‐coding RNAs called circular RNAs (circRNAs) play key roles in dictating tumor behaviors and phenotypes of the prostate tumors. In the current study, our aim was to determine the expression profiles of circHIAT1 and circCDR1AS in PCa compared with benign prostatic hyperplasia (BPH) tissues, as well as their clinicopathological relevance. Methods The 50 prostate tissues including 25 PCa tissues and 25 BPH samples were collected for analyzing the expression levels of target circRNAs by quantitative real‐time PCR (qRT‐PCR). Results The results revealed that expression of circCDR1AS was significantly elevated in PCa compared with the BPH (p < 0.05). We also observed that PCa patients over the age of 60 had a higher expression of the circCDR1AS than patients under the age of 60 (p = 0.017). Moreover, a lower expression level of circHIAT1 was found in the PCa than BPH tissues (p < 0.05), and finally, the findings indicated that the area under the curve (AUC) of circCDR1AS was 0.848, with 92% sensitivity and 76% specificity, as well as an AUC of 0.828, with the 80% sensitivity and 76% specificity for circHIAT1. Conclusion These observations suggest that the abnormal expression of circCDR1AS and circHIAT1 can be regarded as two different types of molecular pathology with potential biomarker values for PCa, although further studies are needed.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Department of Genetics and Molecular Medicine, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Dinmohammadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Emadoddin Moudi
- Department of Urology, Babol University of Medical Sciences, Babol, Iran
| | - Nima Motamed
- The Faculty Member of the Department of Social Medical, Social Determinants of Health Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
39
|
Lin Q, Shi Y, Liu Z, Mehrpour M, Hamaï A, Gong C. Non-coding RNAs as new autophagy regulators in cancer progression. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166293. [PMID: 34688868 DOI: 10.1016/j.bbadis.2021.166293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/17/2021] [Accepted: 10/10/2021] [Indexed: 12/09/2022]
Abstract
Recent advances highlight that non-coding RNAs (ncRNAs) are emerging as fundamental regulators in various physiological as well as pathological processes by regulating macro-autophagy. Studies have disclosed that macro-autophagy, which is a highly conserved process involving cellular nutrients, components, and recycling of organelles, can be either selective or non-selective and ncRNAs show their regulation on selective autophagy as well as non-selective autophagy. The abnormal expression of ncRNAs will result in the impairment of autophagy and contribute to carcinogenesis and cancer progression by regulating both selective autophagy as well as non-selective autophagy. This review focuses on the regulatory roles of ncRNAs in autophagy and their involvement in cancer which may provide valuable therapeutic targets for cancer management.
Collapse
Affiliation(s)
- Qun Lin
- Breast Tumor Center, 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 510120, China; Bioland Laboratory, 510005 Guangzhou, China
| | - Yu Shi
- Breast Tumor Center, 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 510120, China; Bioland Laboratory, 510005 Guangzhou, China
| | - Zihao Liu
- Breast Tumor Center, 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 510120, China; Bioland Laboratory, 510005 Guangzhou, China
| | - Maryam Mehrpour
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, 75993, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75993 Paris, France
| | - Ahmed Hamaï
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, 75993, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75993 Paris, France
| | - Chang Gong
- Breast Tumor Center, 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 510120, China; Bioland Laboratory, 510005 Guangzhou, China.
| |
Collapse
|
40
|
Abd El-Aziz YS, Leck LYW, Jansson PJ, Sahni S. Emerging Role of Autophagy in the Development and Progression of Oral Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:6152. [PMID: 34944772 PMCID: PMC8699656 DOI: 10.3390/cancers13246152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a cellular catabolic process, which is characterized by degradation of damaged proteins and organelles needed to supply the cell with essential nutrients. At basal levels, autophagy is important to maintain cellular homeostasis and development. It is also a stress responsive process that allows the cells to survive when subjected to stressful conditions such as nutrient deprivation. Autophagy has been implicated in many pathologies including cancer. It is well established that autophagy plays a dual role in different cancer types. There is emerging role of autophagy in oral squamous cell carcinoma (OSCC) development and progression. This review will focus on the role played by autophagy in relation to different aspects of cancer progression and discuss recent studies exploring the role of autophagy in OSCC. It will further discuss potential therapeutic approaches to target autophagy in OSCC.
Collapse
Affiliation(s)
- Yomna S. Abd El-Aziz
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Lionel Y. W. Leck
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Patric J. Jansson
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
| |
Collapse
|
41
|
Wang J, Zhang X, Zhang J, Chen S, Zhu J, Wang X. Long noncoding RNA CRART16 confers 5-FU resistance in colorectal cancer cells by sponging miR-193b-5p. Cancer Cell Int 2021; 21:638. [PMID: 34844630 PMCID: PMC8628471 DOI: 10.1186/s12935-021-02353-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background The emergence of chemoresistance to 5-fluorouracil (5-FU)-based chemotherapy is the main cause of treatment failure in advanced and metastatic colorectal cancer (CRC) patients. Long noncoding RNAs (lncRNAs) have been reported to be involved in 5-FU resistance. Previously, we first detected that lncRNA cetuximab resistance-associated RNA transcript 16 (CRART16) could contribute to cetuximab resistance by upregulating V-Erb-B2 erythroblastic leukemia viral oncogene homologue 3 (ERBB3) expression by sponging miR-371a-5p in CRC cells. The current study aimed to explore the role of CRART16 in acquired 5-FU resistance in CRC cells and its possible mechanism. Methods Quantitative real-time PCR (RT-qPCR) was used to measure the expression levels of CRART16 in a 5-FU-resistant CRC cell subline (SW620/5-FU) and the parent cell line. Lentivirus transduction was performed to establish SW620 and Caco-2 cells stably overexpressing CRART16. Cell Counting Kit-8 (CCK-8) assays and colony formation assays were applied to measure cell chemosensitivity to 5-FU. Flow cytometric and immunofluorescence staining were adopted to assess cell apoptosis induced by 5-FU. The dual-luciferase reporter assay was used to validate the direct interactions between CRART16 and miR-193b-5p and between miR-193b-5p and high-mobility group AT-hook-2 (HMGA2). The expression levels of HMGA2, apoptosis-associated proteins and p-ERK were examined by western blotting. The statistical differences within any two groups were used Student’s t test. Results CRART16 was upregulated in SW620/5-FU cells. Overexpression of CRART16 reduced the sensitivity of CRC cells to 5-FU by attenuating apoptosis. In addition, CRART16 promoted 5-FU resistance by suppressing the expression of miR-193b-5p. Furthermore, CRART16 modulated the expression of HMGA2 by inhibiting miR-193b-5p and activated the MAPK signaling pathway. Conclusions CRART16 confers 5-FU resistance in CRC cells through the CRART16/miR-193b-5p/HMGA2/MAPK pathway.
Collapse
Affiliation(s)
- Jingui Wang
- Department of General Surgery, Peking University First Hospital, NO. 8 Xishiku Street, Xicheng, Beijing, 100034, People's Republic of China
| | - Xiaoqian Zhang
- Department of General Surgery, Peking University First Hospital, NO. 8 Xishiku Street, Xicheng, Beijing, 100034, People's Republic of China.,Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, No. 17, Panjiayuan Nanli, Chaoyang, Beijing, 100021, People's Republic of China
| | - Junling Zhang
- Department of General Surgery, Peking University First Hospital, NO. 8 Xishiku Street, Xicheng, Beijing, 100034, People's Republic of China
| | - Shangwen Chen
- Department of General Surgery, Peking University First Hospital, NO. 8 Xishiku Street, Xicheng, Beijing, 100034, People's Republic of China
| | - Jing Zhu
- Department of General Surgery, Peking University First Hospital, NO. 8 Xishiku Street, Xicheng, Beijing, 100034, People's Republic of China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, NO. 8 Xishiku Street, Xicheng, Beijing, 100034, People's Republic of China.
| |
Collapse
|
42
|
Yin X, Li W, Zhang J, Zhao W, Cai H, Zhang C, Liu Z, Guo Y, Wang J. AMPK-Mediated Metabolic Switching Is High Effective for Phytochemical Levo-Tetrahydropalmatine (l-THP) to Reduce Hepatocellular Carcinoma Tumor Growth. Metabolites 2021; 11:metabo11120811. [PMID: 34940569 PMCID: PMC8703446 DOI: 10.3390/metabo11120811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Targeting cancer cell metabolism has been an attractive approach for cancer treatment. However, the role of metabolic alternation in cancer is still unknown whether it functions as a tumor promoter or suppressor. Applying the cancer gene-metabolism integrative network model, we predict adenosine monophosphate-activated protein kinase (AMPK) to function as a central hub of metabolic landscape switching in specific liver cancer subtypes. For the first time, we demonstrate that the phytochemical levo-tetrahydropalmatine (l-THP), a Corydalis yanhusuo-derived clinical drug, as an AMPK activator via autophagy-mediated metabolic switching could kill the hepatocellular carcinoma HepG2 cells. Mechanistically, l-THP promotes the autophagic response by activating the AMPK-mTOR-ULK1 and the ROS-JNK-ATG cascades and impairing the ERK/AKT signaling. All these processes ultimately synergize to induce the decreased mitochondrial oxidative phosphorylation (OXPHOS) and mitochondrial damage. Notably, silencing AMPK significantly inhibits the autophagic flux and recovers the decreased OXPHOS metabolism, which results in HepG2 resistance to l-THP treatment. More importantly, l-THP potently reduces the growth of xenograft HepG2 tumor in nude mice without affecting other organs. From this perspective, our findings support the conclusion that metabolic change is an alternative approach to influence the development of HCC.
Collapse
Affiliation(s)
- Xunzhe Yin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (H.C.); (C.Z.)
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (W.L.); (J.Z.); (W.Z.)
| | - Wenbo Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (W.L.); (J.Z.); (W.Z.)
| | - Jiaxin Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (W.L.); (J.Z.); (W.Z.)
| | - Wenjing Zhao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (W.L.); (J.Z.); (W.Z.)
| | - Huaxing Cai
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (H.C.); (C.Z.)
| | - Chi Zhang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (H.C.); (C.Z.)
| | - Zuojia Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; (W.L.); (J.Z.); (W.Z.)
- Correspondence: (Z.L.); (Y.G.)
| | - Yan Guo
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (H.C.); (C.Z.)
- Correspondence: (Z.L.); (Y.G.)
| | - Jin Wang
- Department of Chemistry and Physics, Stony Brook University, Stony Brook, NY 11794-3400, USA;
| |
Collapse
|
43
|
Zou C, Li X, Lv X, Wu S, Song J, Tang Z, Luo H, Wei H, Ai Y. Circular RNA Mitochondrial Translation Optimization 1 homologue (CircMTO1) induced by Zinc Finger Protein 460 (ZNF460) promotes oral squamous cell carcinoma progression through the microRNA miR-320a / Alpha Thalassemia/Mental Retardation, X-linked (ATRX) axis. Bioengineered 2021; 12:9585-9597. [PMID: 34738503 PMCID: PMC8810001 DOI: 10.1080/21655979.2021.1997699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common cancer types of head and neck cancer, accounting for 95% of all cases. However, the mechanisms underlying the pathogenesis of OSCC remain unclear. Circular RNA (CircRNA) has been extensively studied in the past decades and is a promising direction for the development of OSCC therapeutic targets. In this study, we aimed to investigate the role of circMTO1 in OSCC progression. First, we validated the characterization and expression of circMTO1 in OSCC. It was found that circMTO1 was upregulated in OSCC tumor tissues and cells. Subsequently, we conducted biological experiments. It was found that circMTO1 knockdown inhibited OSCC cell proliferation, migration, and invasion. Furthermore, we conducted a series of experiments to elucidate the underlying mechanisms. A novel circMTO1/miR-320a/ATRX axis was identified. Our results suggest that circMTO1 modulates ATRX expression to accelerate OSCC progression by sponging miR-320a. Moreover, we found that circMTO1 expression in OSCC was transcriptionally regulated by Zinc Finger Protein 460 (ZNF460). Our study showed a novel ZNF460/circMTO1/miR-320a/ATRX signaling in OSCC development.
Collapse
Affiliation(s)
- Chen Zou
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xia Li
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xiaozhi Lv
- Department of Oral and Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Siyuan Wu
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jing Song
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Zhe Tang
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Hailing Luo
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Haigang Wei
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Yilong Ai
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| |
Collapse
|
44
|
Non-coding RNA-mediated autophagy in cancer: A protumor or antitumor factor? Biochim Biophys Acta Rev Cancer 2021; 1876:188642. [PMID: 34715268 DOI: 10.1016/j.bbcan.2021.188642] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022]
Abstract
Autophagy, usually referred to as macroautophagy, is a cytoprotective behavior that helps cells, especially cancer cells, escape crises. However, the role of autophagy in cancer remains controversial. The induction of autophagy is favorable for tumor growth, as it can degrade damaged cell components accumulated during nutrient deficiency, chemotherapy, or other stresses in a timely manner. Whereas the antitumor effect of autophagy might be closely related to its crosstalk with metabolism, immunomodulation, and other pathways. Recent studies have verified that lncRNAs and circRNAs modulate autophagy in carcinogenesis, cancer cells proliferation, apoptosis, metastasis, and chemoresistance via multiple mechanisms. A comprehensive understanding of the regulatory relationships between ncRNAs and autophagy in cancer might resolve chemoresistance and also offer intervention strategies for cancer therapy. This review systematically displays the regulatory effects of lncRNAs and circRNAs on autophagy in the contexts of cancer initiation, progression, and resistance to chemo- or radiotherapy and provides a novel insight into cancer therapy.
Collapse
|
45
|
Lai Q, Wang M, Hu C, Tang Y, Li Y, Hao S. Circular RNA regulates the onset and progression of cancer through the mitogen-activated protein kinase signaling pathway. Oncol Lett 2021; 22:817. [PMID: 34671431 PMCID: PMC8503804 DOI: 10.3892/ol.2021.13078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/27/2021] [Indexed: 01/04/2023] Open
Abstract
The rapid increase in cancer morbidity and mortality worldwide is a major challenge for public health providers. Therefore, there is an urgent need to explore the molecular mechanism of tumorigenesis and identify potential diagnostic biomarkers and therapeutic methods. Circular RNA (circRNA) is characterized by a stable structure and tissue-specific expression; these features are useful in medical research and clinical applications. In recent years, with the development of high-throughput sequencing technology, the potential use of circRNA in cancer prognosis and treatment has been extensively explored. Abnormal circRNA expression interferes with specific signaling pathways such as the MAPK pathway; this phenomenon may provide potential diagnostic biomarkers and new therapeutic targets. The present article discusses the research progress on the regulatory roles of MAPK/ERK pathway-related circRNA molecules in the development and progression of different types of tumors. This review may provide insight into the development of circRNA-based cancer management strategies.
Collapse
Affiliation(s)
- Qun Lai
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Min Wang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Chunmei Hu
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yan Tang
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yarong Li
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Shuhong Hao
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
46
|
Meng X, Lou QY, Yang WY, Wang YR, Chen R, Wang L, Xu T, Zhang L. The role of non-coding RNAs in drug resistance of oral squamous cell carcinoma and therapeutic potential. Cancer Commun (Lond) 2021; 41:981-1006. [PMID: 34289530 PMCID: PMC8504146 DOI: 10.1002/cac2.12194] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC), the eighth most prevalent cancer in the world, arises from the interaction of multiple factors including tobacco, alcohol consumption, and betel quid. Chemotherapeutic agents such as cisplatin, 5-fluorouracil, and paclitaxel have now become the first-line options for OSCC patients. Nevertheless, most OSCC patients eventually acquire drug resistance, leading to poor prognosis. With the discovery and identification of non-coding RNAs (ncRNAs), the functions of dysregulated ncRNAs in OSCC development and drug resistance are gradually being widely recognized. The mechanisms of drug resistance of OSCC are intricate and involve drug efflux, epithelial-mesenchymal transition, DNA damage repair, and autophagy. At present, strategies to explore the reversal of drug resistance of OSCC need to be urgently developed. Nano-delivery and self-cellular drug delivery platforms are considered as effective strategies to overcome drug resistance due to their tumor targeting, controlled release, and consistent pharmacokinetic profiles. In particular, the combined application of new technologies (including CRISPR systems) opened up new horizons for the treatment of drug resistance of OSCC. Hence, this review explored emerging regulatory functions of ncRNAs in drug resistance of OSCC, elucidated multiple ncRNA-meditated mechanisms of drug resistance of OSCC, and discussed the potential value of drug delivery platforms using nanoparticles and self-cells as carriers in drug resistance of OSCC.
Collapse
Affiliation(s)
- Xiang Meng
- Key Lab. of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Hefei, Anhui, 230032, P. R. China
| | - Qiu-Yue Lou
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Wen-Ying Yang
- Key Lab. of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Hefei, Anhui, 230032, P. R. China
| | - Yue-Rong Wang
- Key Lab. of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Hefei, Anhui, 230032, P. R. China
| | - Ran Chen
- School of Stomatology, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Lu Wang
- Key Lab. of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Hefei, Anhui, 230032, P. R. China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230032, P. R. China
- School of Pharmacy, Anhui Key Lab. of Bioactivity of Natural Products, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Lei Zhang
- Key Lab. of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Hefei, Anhui, 230032, P. R. China
- Department of Periodontology, Anhui Stomatology Hospital affiliated to Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| |
Collapse
|
47
|
Wu K, Mao YY, Chen Q, Zhang B, Zhang S, Wu HJ, Li Y. Hypoxia-induced ROS promotes mitochondrial fission and cisplatin chemosensitivity via HIF-1α/Mff regulation in head and neck squamous cell carcinoma. Cell Oncol (Dordr) 2021; 44:1167-1181. [PMID: 34460078 DOI: 10.1007/s13402-021-00629-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 10/20/2022] Open
Abstract
PURPOSE Chemotherapy based on cisplatin (CDDP) has been established as the treatment of choice for head and neck squamous cell carcinoma (HNSCC). Malignant tumors respond to microenvironmental alterations through a dynamic balance between mitochondrial fission and fusion. HNSCCs are known to exhibit hypoxic conditions, yet the respective effects and underlying mechanisms of hypoxia on chemosensitivity and mitochondrial dynamics remain to be resolved. METHODS The effect of hypoxia on the chemosensitivity of HNCC cells was determined by flow cytometry. Mitochondrial fission factor (Mff) expression was assessed by RT-PCR and Western blotting in hypoxic HNSCC cells, and further verified in primary CDDP-sensitive and CDDP-resistant HSNCC samples. The biological function of Mff was evaluated by loss of function and gain of function analyses, both in vitro and in vivo. RESULTS We found that hypoxia promoted mitochondrial fission and CDDP sensitivity in HNSCC cells. Importantly, Mff was found to be correlated with chemosensitivity in primary clinical samples under hypoxic conditions. Hypoxia-inducible factor 1α (HIF-1α) was found to markedly increase Mff transcription and to directly bind to Mff. Hypoxia enhanced the release of reactive oxygen species (ROS) and upregulated the expression of Mff via HIF-1α in HNSCC cells. ROS depletion in HNSCC cells attenuated HIF-1α expression, Mff expression and mitochondrial fission. Moreover, Mff knockdown led to suppression of hypoxia-induced mitochondrial fission and to decreased CDDP chemosensitivity in vivo and in vitro. CONCLUSIONS Our findings indicate that hypoxia-induced release of ROS can promote mitochondrial fission and CDDP chemosensitivity via HIF1α/Mff regulation in HNSCC cells, indicating that Mff may serve as a biomarker to predict neoadjuvant chemosensitivity in HNSCC patients and as a target for overcoming chemoresistance.
Collapse
Affiliation(s)
- Kun Wu
- Department of Stomatology, Second Xiangya Hospital, Central South University, Renmin road, No. 139, Changsha, 410011, Hunan, China
| | - Yuan-Yuan Mao
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi Chen
- Department of Stomatology Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bolin Zhang
- Department of Stomatology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Zhang
- Department of Stomatology, Second Xiangya Hospital, Central South University, Renmin road, No. 139, Changsha, 410011, Hunan, China.
| | - Han-Jiang Wu
- Department of Stomatology, Second Xiangya Hospital, Central South University, Renmin road, No. 139, Changsha, 410011, Hunan, China.
| | - Yan Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Songjia Road, Shanghai, 200025, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
48
|
Zhi Y, Gao L, Wang B, Ren W, Liang KX, Zhi K. Ferroptosis Holds Novel Promise in Treatment of Cancer Mediated by Non-coding RNAs. Front Cell Dev Biol 2021; 9:686906. [PMID: 34235152 PMCID: PMC8255676 DOI: 10.3389/fcell.2021.686906] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/21/2021] [Indexed: 12/17/2022] Open
Abstract
Ferroptosis is a newly identified form of regulated cell death that is associated with iron metabolism and oxidative stress. As a physiological mechanism, ferroptosis selectively removes cancer cells by regulating the expression of vital chemical molecules. Current findings on regulation of ferroptosis have largely focused on the function of non-coding RNAs (ncRNAs), especially microRNAs (miRNAs), in mediating ferroptotic cell death, while the sponging effect of circular RNAs (circRNAs) has not been widely studied. In this review, we discuss the molecular regulation of ferroptosis and highlight the value of circRNAs in controlling ferroptosis and carcinogenesis. Herein, we deliberate future role of this emerging form of regulated cell death in cancer therapeutics and predict the progression and prognosis of oncogenesis in future clinical therapy.
Collapse
Affiliation(s)
- Yuan Zhi
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ling Gao
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Baisheng Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kristina Xiao Liang
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine (K1), University of Bergen, Bergen, Norway
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Shang Y, Jiang YL, Ye LJ, Chen LN, Ke Y. Resveratrol acts via melanoma-associated antigen A12 (MAGEA12)/protein kinase B (Akt) signaling to inhibit the proliferation of oral squamous cell carcinoma cells. Bioengineered 2021; 12:2253-2262. [PMID: 34085601 PMCID: PMC8806796 DOI: 10.1080/21655979.2021.1934242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The present study examined how resveratrol affects cell growth and MAGEA12/Akt signaling pathway in OSCC cells. Cal-27 cells were transiently transfected with a plasmid encoding MAGEA12, and the effects of overexpression were assessed in terms of cell viability, colony formation and the epithelial–mesenchymal transition. Cal-27 cells and MAGEA12-overexpressing cells were treated with resveratrol, then the cell viability and colony formation were also assessed by CCK8 assay and microscope, respectively. Levels of MAGEA12, p-Akt, Akt, Cyclin D1, and CDK14 genes and these proteins were analyzed using quantitative reverse-transcription polymerase-chain reaction and western blot. In the present research, we first generated and transiently transfected MAGEA12 plasmid into Cal-27 cells. Our results suggested that overexpressing MAGEA12 led to an increase in levels of phospho-Akt, which was associated with increased cell viability, colony formation. Moreover, overexpressing MAGEA12 also resulted in the up-regulation of Cyclin D1 and CDK14, indicating MAGEA12 induces the cell proliferation of Cal-27 cells. In addition, these effects were partially reversed by inhibiting Akt. Furthermore, resveratrol could inhibit the proliferation and colony in Cal-27 cells and decrease the expressions of MAGEA12 and p-Akt depending on the time and concentration. These effects were also partially reversed by MAGEA12 overexpression and Akt activation. In summary, resveratrol may suppress the growth of OSCC cells by inactivating MAGEA12/Akt signaling. These findings suggest that resveratrol may be a therapeutic drug for OSCC in clinical.
Collapse
Affiliation(s)
- Yu Shang
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, P.R.C
| | - Yu-Ling Jiang
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, P.R.C
| | - Li-Jun Ye
- Department of Stomatology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, P.R.C
| | - Li-Na Chen
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, P.R.C
| | - Yue Ke
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, P.R.C
| |
Collapse
|
50
|
The P2X7 Receptor in the Maintenance of Cancer Stem Cells, Chemoresistance and Metastasis. Stem Cell Rev Rep 2021; 16:288-300. [PMID: 31813120 DOI: 10.1007/s12015-019-09936-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metastasis is the worst prognosis predictor in the clinical course of cancer development. Features of metastatic cancer cells include migratory ability, low degree of differentiation, self-renewal and proliferation potentials, as well as resistance to therapies. Metastatic cells do not present all of the necessary characteristics at once. Indeed, they have a unique phenotypic plasticity, allowing the acquisition of features that make them successful in all steps of metastasis. Cancer stem cells (CSC), the most undifferentiated cells in the tumor mass, display highest metastatic potential and resistance to radio- and chemotherapy. Growing tumors exhibit marked upregulation of P2X7 receptor expression and secrete ATP. Since the P2X7 receptor plays an important role in the maintenance of undifferentiated state of pluripotent cells, its importance on cell fate regulation in the tumor mass is suggested. Considering the extensive crosstalk between CSCs, epithelial-mesenchymal transition, drug resistance and metastasis, current knowledge implicating P2X7 receptor function in these phenomena and new avenues for therapeutic strategies to control metastasis are reviewed.
Collapse
|