1
|
Yu J, Cai W, Zhou T, Men B, Chen S, Tu D, Guo W, Wang J, Zhao F, Wang Y. CEACAM1 increased the lymphangiogenesis through miR-423-5p and NF- kB in Non-Small Cell Lung Cancer. Biochem Biophys Rep 2024; 40:101833. [PMID: 39398537 PMCID: PMC11470192 DOI: 10.1016/j.bbrep.2024.101833] [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: 07/01/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Background Lung cancer causes significant mortality, with invasion and metastasis being the main features that cause most cancer deaths. Lymph node metastasis is the primary metastatic route in non-small cell carcinoma (NSCLC) and influences the staging and prognosis of NSCLC. Cumulative studies have reported that Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is involved in the progression of various cancers. However, few studies have discussed the function of CEACAM1 in lymphangiogenesis in NSCLC. Here, we examined how CEACAM1 influences lymphangiogenesis in NSCLC. Methods A total of 30 primary squamous cell carcinoma (LUSC) patients diagnosed with LN metastasis were prospectively selected. LUSC tumor tissues, para-cancerous tissues, and positive lymph node tissues were harvested. The expression and subcellular location of CEACAM1, CD31, and LVYE1 in clinical samples were detected by immunohistochemistry. Next, the CEACAM1 and hsa-miR-423-5p expressions were detected by qPCR. The protein expression of lymphangiogenesis-associated proteins and critical cytokines of the NF-κB pathway in HDLECs was detected by Western blot. A tube formation assay was performed to detect the lymphangiogenesis in different groups. The interaction between CEACAM1 and hsa-miR-423-5p was verified using a dual luciferase assay. Results CEACAM1 was found to be a potential gene associated with lung cancer prognosis. It was positively correlated with angiogenesis and lymphangiogenesis. Then, we detected the function of CEACAM1 in lymphangiogenesis and found that CEACAM1 promoted lymphangiogenesis. hsa-miR-423-5p overexpression inhibited lymphangiogenesis via targeting CEACAM1. Finally, we observed that CEACAM1 can activate the NF-κB pathway and, therefore, promote lymphangiogenesis. Conclusion We found that CEACAM1 enhanced lymphangiogenesis in NSCLC via NF-kB activation and was repressed by miR-423-5p. This suggests the value of CEACAM1 as a new therapeutic marker in NSCLC.
Collapse
Affiliation(s)
- Jie Yu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wenke Cai
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Tao Zhou
- Department of Respiration, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Bo Men
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Shunqiong Chen
- Department of Respiration, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Dong Tu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wei Guo
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Jicui Wang
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Feipeng Zhao
- Department of Plastic and Maxillofacial Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
2
|
Simiene J, Kunigenas L, Prokarenkaite R, Dabkeviciene D, Strainiene E, Stankevicius V, Cicenas S, Suziedelis K. Prognostic Value of miR-10a-3p in Non-Small Cell Lung Cancer Patients. Onco Targets Ther 2024; 17:1017-1032. [PMID: 39559728 PMCID: PMC11572442 DOI: 10.2147/ott.s475644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/20/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose Poor lung cancer patients' outcomes and survival rates demand the discovery of new biomarkers for the specific, significant, and less invasive detection of non-small cell lung cancer (NSCLC) progression. The present study aimed to investigate the potential of miRNA expression as biomarkers in NSCLC utilizing a preclinical cell culture setup based on screening of miRNAs in NSCLC cells grown in 3D cell culture. Patients and Methods The study was performed using lung cancer cell lines, varying in different levels of aggressiveness: NCI-H1299, A549, Calu-1, and NCI-H23, as well as noncancerous bronchial epithelial cell line HBEC3, which were grown in 3D cell culture. Total RNA from all cell lines was extracted and small RNA libraries were prepared and sequenced using the Illumina NGS platform. The expression of 8 differentially expressed miRNAs was further validated in 89 paired tissue specimens and plasma samples obtained from NSCLC patients. Statistical analysis was performed to determine whether miRNA expression and clinicopathological characteristics of NSCLC patients could be considered as independent factors significantly influencing PFS or OS. Results Differentially expressed miRNAs, including let-7d-3p, miR-10a-3p, miR-28-3p, miR-28-5p, miR-100-3p, miR-182-5p, miR-190a-5p, and miR-340-5p, were identified through next-generation sequencing in NSCLC cell lines with varying levels of aggressiveness. Validation of patient samples, including tumor and plasma specimens, revealed that out of the 8 investigated miRNAs, only plasma miR-10a-3p showed a significant increase, which was associated with significantly extended progression-free survival (PFS) (p=0.009). Furthermore, miR-10a-3p in plasma emerged as a statistically significant prognostic variable for NSCLC patients' PFS (HR: 0.5, 95% CI: 0.3-0.9, p=0.029). Conclusion Our findings of screening miRNA expression patterns in NSCLC cells grown in 3D cell culture indicated that the expression level of circulating miR-10a-3p has the potential as a novel non-invasive biomarker to reflect the short-term prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Julija Simiene
- Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, LT-10223, Lithuania
| | - Linas Kunigenas
- Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, LT-10223, Lithuania
| | - Rimvile Prokarenkaite
- Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, LT-10223, Lithuania
| | - Daiva Dabkeviciene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, LT-10223, Lithuania
- Biobank, National Cancer Institute, Vilnius, LT-08406, Lithuania
| | - Egle Strainiene
- Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
- Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, LT-10223, Lithuania
| | - Vaidotas Stankevicius
- Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, LT-10223, Lithuania
| | - Saulius Cicenas
- Department of Thoracic Surgery and Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
| | - Kestutis Suziedelis
- Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, LT-08406, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, LT-10223, Lithuania
| |
Collapse
|
3
|
Cui X, Lei X, Huang T, Mao X, Shen Z, Yang X, Peng W, Yu J, Zhang S, Huo P. Follicular fluid-derived extracellular vesicles miR-34a-5p regulates granulosa cell glycolysis in polycystic ovary syndrome by targeting LDHA. J Ovarian Res 2024; 17:223. [PMID: 39538292 PMCID: PMC11562512 DOI: 10.1186/s13048-024-01542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is highly prevalent in women of reproductive age worldwide, exhibits highly heterogeneous clinical presentation and biochemical parameters, and has no cure. This study aimed to investigate the role of miR-34a-5p in PCOS, its effect on glycolysis in granulosa cells (GCs), and its potential contribution to follicular dysregulation. METHODS Herein, Follicular follicular fluid (FF) samples were collected from six patients with PCOS and six healthy controls undergoing in vitro fertilization-embryo transfer. The isolated extracellular vesicles were characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Additionally, miRNA sequencing was performed to identify differentially expressed microRNAs, and their functions were analyzed through transcriptomics. The In vitro effects of miR-34a-5p on glycolysis, cell proliferation, and apoptosis were assessed in human ovarian granulosa tumour cell line (KGN cells). Targets of miR-34a-5p were identified by dual-luciferase reporter assays, and quantitative real-time polymerase chain reaction and western blotting were performed to determine gene and protein expression. RESULTS The level of miR-34a-5p in FF-derived extracellular vesicles derived from patients with PCOS was significantly higher than that of the control group. Transcriptomic analysis highlighted miR-34a-5p as a key regulator of glycolysis and apoptosis. Furthermore, in vitro analysis showed that miR-34a-5p targeted lactate dehydrogenase A (LDHA), inhibited glycolysis, reduced energy supply to GCs, and promoted apoptosis of KGN cells. Conversely, miR-34a-5p inhibition restored glycolysis function and cell viability. CONCLUSION The findings of this study show that miR-34a-5p mediates GC apoptosis in PCOS by targeting LDHA and inhibiting glycolysis, suggesting its crucial role in PCOS pathophysiology, and offering potential therapeutic targets for improving follicular development and fertility outcomes in patients with PCOS. Further research is needed to explore the clinical implications of miR-34a-5p and its use as a biomarker for early diagnosis and prognosis of PCOS.
Collapse
Affiliation(s)
- Xueying Cui
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Xiaocan Lei
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, 421001, Hunan, China
| | - Tao Huang
- The Second Affiliated Hospital of Guilin Medical University, Guilin, 541100, Guangxi, China
| | - Xueyan Mao
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiwei Shen
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, 421001, Hunan, China
| | - Xiuli Yang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Wanting Peng
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Jingjing Yu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541004, Guangxi, China.
| | - Peng Huo
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541004, Guangxi, China.
| |
Collapse
|
4
|
Yang X, Zhang D, Li M, Shao Y, Zhang X, Xue Y. P4HA1: an important target for treating fibrosis related diseases and cancer. Front Pharmacol 2024; 15:1493420. [PMID: 39568592 PMCID: PMC11576223 DOI: 10.3389/fphar.2024.1493420] [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: 09/09/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Fibrosis is significantly associated with a wide variety of diseases and is involved in their progression. Fibrosis activated under the influence of different combinations of factors is considered a double-edged sword. Although there has been much research on organ fibrosis in recent years, a variety of organ fibrosis diseases and cancers are not well controlled in terms of prevention, treatment, and prognosis. Clinical studies still lack exploration and discovery of effective targets for the pathogenesis of organ fibrosis. Prolyl 4-hydroxylase subunit alpha 1 (P4HA1) is a protein kinase and the synthesis and secretion of collagen are related to the sustained activation of P4HA1. As further studies are being conducted, the potential role of P4HA1 in the development of fibrosis-associated diseases and cancer is becoming clear. Consequently, we conducted a systematic review and discussion on the role of P4HA1 in the pathogenesis of various fibrosis-related diseases and cancers. We reviewed the possible strategies of P4HA1 in the diagnosis and treatment of fibrosis-related diseases and cancers, and analyzed its potential relevance as a biomarker in the diagnosis and treatment of fibrosis-related diseases and cancer.
Collapse
Affiliation(s)
- Xuewen Yang
- Department of Pharmacology, Institute of Pharmacokinetics and Liver Molecular Pharmacology, Baotou Medical College, Baotou, China
| | - Dong Zhang
- Department of Pharmacology, Institute of Pharmacokinetics and Liver Molecular Pharmacology, Baotou Medical College, Baotou, China
| | - Mengmeng Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yingchun Shao
- The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Xiyang Zhang
- Faculty of Basic Medicine, Chongqing Three Gorges Medical College, Chongqing, China
| | - Yongzhi Xue
- Department of Pharmacology, Institute of Pharmacokinetics and Liver Molecular Pharmacology, Baotou Medical College, Baotou, China
| |
Collapse
|
5
|
Abida, Altamimi ASA, Ghaboura N, Balaraman AK, Rajput P, Bansal P, Rawat S, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H, Deb PK. Therapeutic Potential of lncRNAs in Regulating Disulfidptosis for Cancer Treatment. Pathol Res Pract 2024; 263:155657. [PMID: 39437641 DOI: 10.1016/j.prp.2024.155657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Non-coding RNAs (lncRNAs) play critical roles in various cellular processes, including a novel form of regulated cell death known as disulfidptosis, characterized by accumulating protein disulfide bonds and severe endoplasmic reticulum stress. This review highlights the therapeutic potential of lncRNAs in regulating disulfidptosis for cancer treatment, emphasizing their influence on key pathway components such as GPX4, SLC7A11, and PDIA family members. Recent studies have demonstrated that targeting specific lncRNAs can sensitize cancer cells to disulfidptosis, offering a promising approach to cancer therapy. The regulation of disulfidptosis by lncRNAs involves various signaling pathways, including oxidative stress, ER stress, and calcium signaling. This review also discusses the molecular mechanisms underlying lncRNA regulation of disulfidptosis, the challenges of developing lncRNA-based therapies, and the future potential of this rapidly advancing field in cancer research.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice, Pharmacy Program, Batterjee Medical College, PO Box 6231, Jeddah 21442, Saudi Arabia
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Pranchal Rajput
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institue of Technology (BIT), Mesra, Ranchi, Jharkhand 835215, India
| |
Collapse
|
6
|
Thangavelu L, Goyal A, Afzal M, Moglad E, Rawat S, Kazmi I, Alzarea SI, Almalki WH, Rani R, Madhubabu P, Rajput P, Bansal P. Pyroptosis in lung cancer: The emerging role of non-coding RNAs. Pathol Res Pract 2024; 263:155619. [PMID: 39357188 DOI: 10.1016/j.prp.2024.155619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Lung cancer remains an intractable malignancy worldwide, prompting novel therapeutic modalities. Pyroptosis, a lethal form of programmed cell death featured by inflammation, has been involved in cancer progression and treatment response. Simultaneously, non-coding RNA has been shown to have important roles in coordinating pattern formation and oncogenic pathways, including long non-coding RNA (lncRNAs), microRNA (miRNAs), circular RNA (circRNAs), and small interfering RNA (siRNAs). Recent studies have revealed that ncRNAs can promote or inhibit pyroptosis by interacting with key molecular players such as NLRP3, GSDMD, and various transcription factors. This dual role of ncRNAs offers a unique therapeutic potential to manipulate pyroptosis pathways, providing opportunities for innovative cancer treatments. In this review, we integrate current research findings to propose novel strategies for leveraging ncRNA-mediated pyroptosis as a therapeutic intervention in lung cancer. We explore the potential of ncRNAs as biomarkers for predicting patient response to treatment and as targets for overcoming resistance to conventional therapies.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Sushama Rawat
- Graphic Era (Deemed to be University), Clement Town, 248002, Dehradun, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Richa Rani
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab 140413, India
| | | | - Pranchal Rajput
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India
| | - Pooja Bansal
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali 140307, Punjab, India
| |
Collapse
|
7
|
Jalilian S, Bastani MN. From virus to cancer: Epstein-Barr virus miRNA connection in Burkitt's lymphoma. Infect Agent Cancer 2024; 19:54. [PMID: 39425210 PMCID: PMC11487968 DOI: 10.1186/s13027-024-00615-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
In Burkitt's lymphoma (BL), Epstein-Barr virus-encoded microRNAs (EBV miRNAs) are emerging as crucial regulatory agents that impact cellular and viral gene regulation. This review investigates the multifaceted functions of EBV miRNAs in the pathogenesis of Burkitt lymphoma. EBV miRNAs regulate several cellular processes that are essential for BL development, such as apoptosis, immune evasion, and cellular proliferation. These small, non-coding RNAs target both viral and host mRNAs, finely adjusting the cellular environment to favor oncogenesis. Prominent miRNAs, such as BART (BamHI-A rightward transcript) and BHRF1 (BamHI fragment H rightward open reading frame 1), are emphasized for their roles in tumor growth and immune regulation. For example, BART miRNAs prevent apoptosis by suppressing pro-apoptotic proteins, whereas BHRF1 miRNAs promote viral latency and immunological evasion. Understanding the intricate connections among EBV miRNAs and their targets illuminates BL pathogenesis and suggests novel treatment approaches. Targeting EBV miRNAs or their specific pathways offers a feasible option for developing innovative therapies that aim to disrupt the carcinogenic processes initiated by these viral components. future studies should focus on precisely mapping miRNA‒target networks and developing miRNA-based diagnostic and therapeutic tools. This comprehensive article highlights the importance of EBV miRNAs in Burkitt lymphoma, indicating their potential as biomarkers and targets for innovative treatment strategies.
Collapse
Affiliation(s)
- Shahram Jalilian
- Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 15794 - 61357, Iran
| | - Mohammad-Navid Bastani
- Department of Virology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 15794 - 61357, Iran.
| |
Collapse
|
8
|
Yuan F, Hu Y, Lei Y, Jin L. Recent progress in microRNA research for prostate cancer. Discov Oncol 2024; 15:480. [PMID: 39331237 PMCID: PMC11436510 DOI: 10.1007/s12672-024-01376-4] [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: 05/31/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
In recent years, prostate cancer (PCa) has seen an increasing prevalence, particularly among middle-aged and older men, positioning it as a significant health concern. Current PCa screening predominantly utilizes prostate-specific antigen (PSA) testing, digital rectal examination (DRE), and the Gleason scoring system. However, these diagnostic methods can sometimes be imprecise. Research has identified that specific microRNAs (miRNAs) exhibit altered expression levels in PCa patients, suggesting their potential as biomarkers for both diagnosis and prognosis. Furthermore, advancements in integrating miRNAs with traditional Chinese medicine (TCM) have shown promising results in PCa treatment, potentially serving as micro-markers for TCM syndrome differentiation and treatment effectiveness. Recent developments in anti-cancer therapies that target miRNAs have also been implemented in clinical settings, laying the groundwork for personalized and precise treatment strategies for PCa. This review aims to summarize the expression patterns of miRNAs in PCa patients and explore their roles in the diagnosis, treatment, and prognosis of the disease.
Collapse
Affiliation(s)
- Fan Yuan
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, People's Republic of China
| | - Yue Hu
- Health Management (Physical Examination) Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, People's Republic of China
| | - Yanmei Lei
- Department of Nuclear Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, People's Republic of China.
| | - Lingna Jin
- Health Management (Physical Examination) Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, People's Republic of China.
| |
Collapse
|
9
|
Su Z, Lu C, Zhang F, Liu H, Li M, Qiao M, Zou X, Luo D, Li H, He M, Se H, Jing J, Wang X, Yang H, Yang H. Cancer-associated fibroblasts-secreted exosomal miR-92a-3p promotes tumor growth and stemness in hepatocellular carcinoma through activation of Wnt/β-catenin signaling pathway by suppressing AXIN1. J Cell Physiol 2024; 239:e31344. [PMID: 38949237 DOI: 10.1002/jcp.31344] [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: 01/14/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are a major cellular component in the tumor microenvironment and have been shown to exhibit protumorigenic effects in hepatocellular carcinoma (HCC). This study aimed to delve into the mechanisms underlying the tumor-promoting effects of CAFs in HCC. Small RNA sequencing was conducted to screen differential expressed microRNAs in exosomes derived from CAFs and normal fibroblasts (NFs). The miR-92a-3p expression was then measured using reverse transcriptase quantitative real-time PCR in CAFs, NFs, CAFs-derived exosomes (CAFs-Exo), and NF-derived exosomes (NFs-Exo). Compared to NFs or NF-Exo, CAFs and CAFs-Exo significantly promoted HCC cell proliferation, migration, and stemness. Additionally, compared to NFs or NF-Exo, miR-92a-3p level was notably higher in CAFs and CAFs-Exo, respectively. Exosomal miR-92a-3p was found to enhance HCC cell proliferation, migration, and stemness. Meanwhile, AXIN1 was targeted by miR-92a-3p. Exosomal miR-92a-3p could activate β-catenin/CD44 signaling in HCC cells by inhibiting AXIN1 messenger RNA. Furthermore, in vivo studies verified that exosomal miR-92a-3p notably promoted tumor growth and stemness through targeting AXIN1/β-catenin axis. Collectively, CAFs secreted exosomal miR-92a-3p was capable of promoting growth and stemness in HCC through activation of Wnt/β-catenin signaling pathway by suppressing AXIN1. Therefore, targeting CAFs-derived miR-92a-3p may be a potential strategy for treating HCC.
Collapse
Affiliation(s)
- Zenong Su
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Department of Graduate School, Baotou Medical College, Baotou, Inner Mongolia Autonomous Region, China
| | - Chao Lu
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Department of Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Feifei Zhang
- Department of Nuclear Medicine, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Huan Liu
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Department of Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Meiqing Li
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Meng Qiao
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiaohong Zou
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Danyang Luo
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haojing Li
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Min He
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Han Se
- Department of Graduate School, Baotou Medical College, Baotou, Inner Mongolia Autonomous Region, China
| | - Jing Jing
- Department of Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiangcheng Wang
- Department of Nuclear Medicine, Shenzhen People's Hospital, Shenzhen, Guangzhou, China
| | - Hao Yang
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Hong Yang
- Department of Oncology, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- Institute of Cancer, Inner Mongolia People's Hospital, People's Hospital of Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
10
|
Liu S, Ruan Y, Chen X, He B, Chen Q. miR-137: a potential therapeutic target for lung cancer. Front Cell Dev Biol 2024; 12:1427724. [PMID: 39247624 PMCID: PMC11377224 DOI: 10.3389/fcell.2024.1427724] [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: 05/04/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Lung cancer is a prevalent malignancy and the leading cause of cancer-related deaths, posing a significant threat to human health. Despite advancements in treatment, the prognosis for lung cancer patients remains poor due to late diagnosis, cancer recurrence, and drug resistance. Epigenetic research, particularly in microRNAs, has introduced a new avenue for cancer prevention and treatment. MicroRNAs, including miR-137, play a vital role in tumor development by regulating various cellular processes. MiR-137 has garnered attention for its tumor-suppressive properties, with studies showing its potential in inhibiting cancer progression. In lung cancer, miR-137 is of particular interest, with numerous reports exploring its role and mechanisms. A comprehensive review is necessary to consolidate current evidence. This review highlights recent studies on miR-137 in lung cancer, covering cell proliferation, migration, apoptosis, drug resistance, and therapy, emphasizing its potential as a biomarker and therapeutic target for lung cancer treatment and prognosis.
Collapse
Affiliation(s)
- Shuanshuan Liu
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Yanyun Ruan
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Xu Chen
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bao He
- Department of Neurosurgery, The First People's hospital of Kunshan, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu, China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
11
|
Hu Y, Wu X, Tan X, Zhang J. Hsa_circRNA_007630 knockdown delays colon cancer progression by modulation of ferroptosis via miR-506-3p/AURKA axis. J Biochem Mol Toxicol 2024; 38:e23771. [PMID: 39015057 DOI: 10.1002/jbt.23771] [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: 02/04/2024] [Revised: 06/06/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Colon cancer contributes to high mortality rates internationally that has seriously endangered human health. Aurora kinase A (AURKA) served as a key molecule in colon cancer. However, its role of AURKA on regulating ferroptosis in colon cancer and their possible interactions with miRNAs and circRNAs remain still elusive. Comprehensive bioinformatics analysis after RNA-sequencing was conducted to determine the differentially expressed genes (DEGs), ferroptosis-related DEGs and hub genes. The direct relationship between miR-506-3p and hsa_circRNA_007630 or AURKA was predicted, then verified by dual luciferase reporter and quantitative real-time polymerase chain reaction. The rescue experiments were conducted by cotransfection with si-hsa_circRNA_007630, miR-506-3p inhibitor or pcDNA-AURKA in HT29 cells. Erastin was used to induce ferroptosis in HT29 cells and validated by detecting levels of intracellular Fe2+, lipid reactive oxygen species, glutathione, malondialdehyde and ferroptosis markers expression. We screened a total of 331 DEGs, 26 ferroptosis-related genes, among which 3 hub genes were identified through PPI network analysis. Therein, AURKA expression was elevated in colon cancer cells. Moreover, AURKA was targeted by miR-506-3p, and hsa_circRNA_007630 operated as miR-506-3p sponge. The effect of hsa_circRNA_007630 depletion on the inhibiting malignant phenotypes of HT29 cells was rescued by inhibition of miR-506-3p or AURKA overexpression. Additionally, AURKA reduced erastin-induced ferroptosis in HT29 cells. Depletion of circRNA_007630 exerts as a suppressive role in colon cancer through a novel miR-506-3p/AURKA pathway related to ferroptosis, and might become a novel marker for colon cancer.
Collapse
Affiliation(s)
- Ying Hu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiongjian Wu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiaobin Tan
- Department of Clinical Laboratory, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Jingzhi Zhang
- Department of Gastroenterology, Ganzhou People's Hospital (Ganzhou Hospital Affiliated to Nanchang University), Ganzhou City, China
| |
Collapse
|
12
|
Siddique R, Gupta G, Mgm J, Kumar A, Kaur H, Ariffin IA, Pramanik A, Almalki WH, Ali H, Shahwan M, Patel N, Murari K, Mishra R, Thapa R, Bhat AA. Targeting notch-related lncRNAs in cancer: Insights into molecular regulation and therapeutic potential. Pathol Res Pract 2024; 257:155282. [PMID: 38608371 DOI: 10.1016/j.prp.2024.155282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024]
Abstract
Cancer is a group of diseases marked by unchecked cell proliferation and the ability for the disease to metastasize to different body areas. Enhancements in treatment and early detection are crucial for improved outcomes. LncRNAs are RNA molecules that encode proteins and have a length of more than 200 nucleotides. LncRNAs are crucial for chromatin architecture, gene regulation, and other cellular activities that impact both normal growth & pathological processes, even though they are unable to code for proteins. LncRNAs have emerged as significant regulators in the study of cancer biology, with a focus on their intricate function in the Notch signaling pathway. The imbalance of this pathway is often linked to a variety of malignancies. Notch signaling is essential for cellular functions like proliferation, differentiation, and death. The cellular response is shaped by these lncRNAs through their modulation of essential Notch pathway constituents such as receptors, ligands, and downstream effectors around it. Furthermore, a variety of cancer types exhibit irregular expression of Notch-related lncRNAs, underscoring their potential use as therapeutic targets and diagnostic markers. Gaining an understanding of the molecular processes behind the interaction between the Notch pathway and lncRNAs will help you better understand the intricate regulatory networks that control the development of cancer. This can open up new possibilities for individualized treatment plans and focused therapeutic interventions. The intricate relationships between lncRNAs & the Notch pathway in cancer are examined in this review.
Collapse
Affiliation(s)
- Raihan Siddique
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Johar Mgm
- Management and Science University (MSU), Shah Alam, Selangor 40100 MSU, Malaysia
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand- 831001, India
| | - I A Ariffin
- Management and Science University (MSU), Shah Alam, Selangor 40100 MSU, Malaysia
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Divison of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Neeraj Patel
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Krishna Murari
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Riya Mishra
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India.
| |
Collapse
|
13
|
Chen S, Wang H, Guo M, Zhao X, Yang J, Chen L, Zhao J, Chen C, Zhou Y, Xu L. Promoter A1312C mutation leads to microRNA-7 downregulation in human non-small cell lung cancer. Cell Signal 2024; 117:111095. [PMID: 38346527 DOI: 10.1016/j.cellsig.2024.111095] [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: 01/02/2024] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
MicroRNA-7 (miRNA-7, miR-7) is a unique class of tumor suppressors, plays an important role in various physiological and pathological processes including human non-small cell lung cancer (NSCLC). In previous works, we revealed that miR-7 could regulate the growth and metastasis of human NSCLC cells. However, the mechanism of dysregulated miR-7 expression in NSCLC remains to be further elucidated. In this study, based on clinical sample analysis, we found that the downregulated expression of miR-7 was dominantly attributed to the decreased level of pri-miR-7-2 in human NSCLC. Furthermore, there were four site mutations in the miR-7-2 promoter sequence. Notably, among these four sites, mutation at -1312 locus (A → C, termed as A1312C mutation) was dominate, and A1312C mutation further led to decreased expression of miR-7 in human NSCLC cells, accompanied with elevated transduction of NDUFA4/ERK/AKT signaling pathway. Mechanistically, homeobox A5 (HOXA5) is the key transcription factors regulating miR-7 expression in NSCLC. A1312C mutation impairs HOXA5 binding, thereby reducing the transcriptional activity of miR-7-2 promoter, resulting in downregulation of miR-7 expression. Together, these data may provide new insights into the dysregulation of specific miRNA expression in NSCLC and ultimately prove to be helpful in the diagnostic, prognostic, and therapeutic strategies against NSCLC.
Collapse
Affiliation(s)
- Shipeng Chen
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Hui Wang
- The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Xu Zhao
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Jing Yang
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Longqing Chen
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China.
| | - Ya Zhou
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Medical Physics, Zunyi Medical University, Zunyi 563000, Guizhou, China.
| | - Lin Xu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi 563000, China; Department of Immunology, Zunyi Medical University, Zunyi 563000, Guizhou, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
14
|
Biswas U, Roy R, Ghosh S, Chakrabarti G. The interplay between autophagy and apoptosis: its implication in lung cancer and therapeutics. Cancer Lett 2024; 585:216662. [PMID: 38309614 DOI: 10.1016/j.canlet.2024.216662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/02/2024] [Accepted: 01/17/2024] [Indexed: 02/05/2024]
Abstract
Maintaining cellular homeostasis relies on the interplay between apoptosis and autophagy, and disruption in either of these processes can contribute to the development of cancer. Autophagy can hinder the apoptotic process, and when autophagy is inhibited in such instances, it can enhance the rate of apoptosis. However, evidence suggests that excessive autophagy can also lead to apoptotic cell death. Also, excess autophagy can cause excessive digestion of cellular organelles, causing autophagic cell death. Targeting autophagy in non-small cell lung cancer (NSCLC), the most common form of lung cancer, can be very tricky due to the dual nature of autophagy. According to genetic analysis, various mutations in p53 and EGFR, G:C to A:T transversions seem responsible for the development of lung cancer in smokers and non-smokers. These events trigger cytoprotective autophagy or induce apoptotic cell death through different but interconnected signalling pathways. Lung cancer being the leading cause of death worldwide, calls for more attention to disease prognosis and new therapeutics in the market. However, molecules responsible for autophagy to apoptosis transition are yet to be studied elaborately. Also, the role of effector caspases during this shift needs to be elucidated in future. To comprehend how therapeutics operate through the modulation of autophagy and apoptosis and to target such pathways, it is crucial to emphasize these intricate connections. Many therapeutics discussed in this review targeting both apoptosis and autophagy have shown promising results in vitro and in vivo, however, few have crossed the hurdles of clinical trial. Nevertheless, the quest for safer and better efficacious agents is still alive, with the sole aim to develop novel cancer chemotherapeutic(s).
Collapse
Affiliation(s)
- Urmita Biswas
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, India
| | - Ranita Roy
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, India
| | - Swarnasree Ghosh
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, India
| | - Gopal Chakrabarti
- Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, India.
| |
Collapse
|
15
|
Rodrigues L, Da Cruz Paula A, Soares P, Vinagre J. Unraveling the Significance of DGCR8 and miRNAs in Thyroid Carcinoma. Cells 2024; 13:561. [PMID: 38607000 PMCID: PMC11011343 DOI: 10.3390/cells13070561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024] Open
Abstract
MicroRNAs (miRNAs) act as negative regulators for protein-coding gene expression impacting cell proliferation, differentiation, and survival. These miRNAs are frequently dysregulated in cancer and constitute classes of blood-based biomarkers useful for cancer detection and prognosis definition. In thyroid cancer (TC), the miRNA biogenesis pathway plays a pivotal role in thyroid gland formation, ensuring proper follicle development and hormone production. Several alterations in the miRNA biogenesis genes are reported as a causality for miRNA dysregulation. Mutations in microprocessor component genes are linked to an increased risk of developing TC; in particular, a recurrent mutation affecting DGCR8, the E518K. In this review, we explore these novel findings and resume the current state-of-the-art in miRNAs in thyroid carcinomas.
Collapse
Affiliation(s)
- Lia Rodrigues
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto (i3S), Rua Alfredo Allen, 4200-135 Porto, Portugal; (L.R.); (A.D.C.P.); (P.S.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, 4200-135 Porto, Portugal
- Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Escola Superior de Saúde do Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Arnaud Da Cruz Paula
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto (i3S), Rua Alfredo Allen, 4200-135 Porto, Portugal; (L.R.); (A.D.C.P.); (P.S.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, 4200-135 Porto, Portugal
| | - Paula Soares
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto (i3S), Rua Alfredo Allen, 4200-135 Porto, Portugal; (L.R.); (A.D.C.P.); (P.S.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, 4200-135 Porto, Portugal
- Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - João Vinagre
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto (i3S), Rua Alfredo Allen, 4200-135 Porto, Portugal; (L.R.); (A.D.C.P.); (P.S.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, 4200-135 Porto, Portugal
- Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
16
|
Deng Y, Ding H, Zhang Y, Feng X, Ye Q, Tian R, Xu Y, He Q, Fu Q, Li R. TP53 mitigates cisplatin resistance in non-small cell lung cancer by mediating the effects of resistant cell-derived exosome mir-424-5p. Heliyon 2024; 10:e26853. [PMID: 38439876 PMCID: PMC10909722 DOI: 10.1016/j.heliyon.2024.e26853] [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: 11/28/2023] [Revised: 01/21/2024] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
Background Cisplatin (DDP) is the principal agent used for chemotherapy in patients with non-small cell lung cancer (NSCLC). Nevertheless, DDP resistance is an essential cause for a worse prognosis of patient. Therefore, this study proposes to discover features of miR-424-5p in DDP resistance of NSCLC. Method After exogenous modulation of miR-424-5p expression, A549 cell activity was measured using CCK-8 and flow cytometry. A549/DDP and A549/DDP-associated subcutaneous tumor model were constructed to investigate the effect of miR-424-5p on DDP resistance in NSCLC in vivo. TargetScan and JASPAR databases predicted the potential molecular mechanism of miR-424-5p. A549-and A549/DDP-derived exosomes were isolated and characterized using a transmission electron microscope and nanoparticle tracking analysis. Result Overexpression of miR-424-5p facilitated proliferation and DDP resistance in A549 cells, and knockdown of miR-424-5p did the opposite. Knockdown of miR-424-5p enhanced DDP restriction on tumor weight and volume. Moreover, SOCS5 and SOCS56 (SOCS5/6) were downstream targets of miR-424-5p. miR-424-5p down-regulated SOCS5/6 expression to activate JAK2/STAT3 and PI3K/AKT pathways. Notably, tumor protein p53 (TP53) is a transcription factor for the miR-424-5p host gene, as confirmed by the dual-luciferase reporter gene. Cellular and animal experiments indicated that TP53 limited the regulatory function of miR-424-5p on NSCLC growth, DDP resistance, and related molecules. Interestingly, miR-424-5p was markedly enriched in A549/DDP cell-derived exosomes than in A549 cell-derived exosomes, and TP53 down-regulated miR-424-5p expression in A549/DDP cell-derived exosomes. Conclusion DDP-resistant cell-derived exosome miR-424-5p contributes to NSCLC growth and DDP resistance by targeting SOCS5 and SOCS6 to activate JAK2/STAT3 and PI3K/AKT pathways, which are blocked by TP53.
Collapse
Affiliation(s)
- Yan Deng
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Hao Ding
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Yanhua Zhang
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Xudong Feng
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Qing Ye
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Rui Tian
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Yuchuan Xu
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Qingqing He
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Qiaofen Fu
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| | - Rongqing Li
- Department of Radiotherapy, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, 650032, China
| |
Collapse
|
17
|
Sun G, Ni K, Shen J, Liu D, Wang H. microRNA-486-5p Regulates DNA Damage Inhibition and Cisplatin Resistance in Lung Adenocarcinoma by Targeting AURKB. Crit Rev Eukaryot Gene Expr 2024; 34:13-23. [PMID: 38505869 DOI: 10.1615/critreveukaryotgeneexpr.v34.i4.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Lung adenocarcinoma (LUAD) severely affects human health, and cisplatin (DDP) resistance is the main obstacle in LUAD treatment, the mechanism of which is unknown. Bioinformatics methods were utilized to predict expression and related pathways of AURKB in LUAD tissues, as well as the upstream regulated microRNAs. qRT-PCR assayed expression of AURKB and microRNA-486-5p. RIP and dual-luciferase experiments verified the binding and interaction between the two genes. CCK-8 was used to detect cell proliferation ability and IC50 values. Flow cytometry was utilized to assess the cell cycle. Comet assay and western blot tested DNA damage and γ-H2AX protein expression, respectively. In LUAD, AURKB was upregulated, but microRNA-486-5p was downregulated. The targeted relationship between the two was confirmed by RIP and dual-luciferase experiments. Cell experiments showed that AURKB knock-down inhibited cell proliferation, reduced IC50 values, induced cell cycle arrest, and caused DNA damage. The rescue experiment presented that high expression of microRNA-486-5p could weaken the impact of AURKB overexpression on LUAD cell behavior and DDP resistance. microRNA-486-5p regulated DNA damage to inhibit DDP resistance in LUAD by targeting AURKB, implying that microRNA-486-5p/AURKB axis may be a possible therapeutic target for DDP resistance in LUAD patients.
Collapse
Affiliation(s)
- Gaozhong Sun
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Kewei Ni
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Jian Shen
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Dongdong Liu
- Department of Thoracic Surgery, Cancer Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | | |
Collapse
|
18
|
Pagoni M, Cava C, Sideris DC, Avgeris M, Zoumpourlis V, Michalopoulos I, Drakoulis N. miRNA-Based Technologies in Cancer Therapy. J Pers Med 2023; 13:1586. [PMID: 38003902 PMCID: PMC10672431 DOI: 10.3390/jpm13111586] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
The discovery of therapeutic miRNAs is one of the most exciting challenges for pharmaceutical companies. Since the first miRNA was discovered in 1993, our knowledge of miRNA biology has grown considerably. Many studies have demonstrated that miRNA expression is dysregulated in many diseases, making them appealing tools for novel therapeutic approaches. This review aims to discuss miRNA biogenesis and function, as well as highlight strategies for delivering miRNA agents, presenting viral, non-viral, and exosomic delivery as therapeutic approaches for different cancer types. We also consider the therapeutic role of microRNA-mediated drug repurposing in cancer therapy.
Collapse
Affiliation(s)
- Maria Pagoni
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Claudia Cava
- Department of Science, Technology and Society, University School for Advanced Studies IUSS Pavia, 27100 Pavia, Italy;
| | - Diamantis C. Sideris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece;
| | - Margaritis Avgeris
- Laboratory of Clinical Biochemistry—Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, “P. & A. Kyriakou” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassilios Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece;
| | - Ioannis Michalopoulos
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15701 Athens, Greece
| |
Collapse
|
19
|
Hsieh YT, Chen YC, Chou YC, Kuo PY, Yen YT, Tsai HW, Wang CR. Long noncoding RNA SNHG16 regulates TLR4-mediated autophagy and NETosis formation in alveolar hemorrhage associated with systemic lupus erythematosus. J Biomed Sci 2023; 30:78. [PMID: 37700342 PMCID: PMC10496234 DOI: 10.1186/s12929-023-00969-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Dysregulated long noncoding RNA (lncRNA) expression with increased apoptosis has been demonstrated in systemic lupus erythematosus (SLE) patients with alveolar hemorrhage (AH). SNHG16, a lncRNA, can enhance pulmonary inflammation by sponging microRNAs, and upregulate toll-like receptor 4 (TLR4) expression via stabilizing its mRNAs. TRAF6, a TLR4 downstream signal transducer, can induce autophagy and NETosis formation. In this study, we investigated whether SNHG16 could regulate TLR4-mediated autophagy and NETosis formation in SLE-associated AH. METHODS Expression of SNHG16, TLR4 and TRAF6 and cell death processes were examined in lung tissues and peripheral blood (PB) leukocytes from AH patients associated with SLE and other autoimmune diseases, and in the lungs and spleen from a pristane-induced C57BL/6 mouse AH model. SNHG16-overexpressed or -silenced alveolar and myelocytic cells were stimulated with lipopolysaccharide (LPS), a TLR4 agonist, for analyzing autophagy and NETosis, respectively. Pristane-injected mice received the intra-pulmonary delivery of lentivirus (LV)-SNHG16 for overexpression and prophylactic/therapeutic infusion of short hairpin RNA (shRNA) targeting SNHG16 to evaluate the effects on AH. Renal SNHG16 expression was also examined in lupus nephritis (LN) patients and a pristane-induced BALB/c mouse LN model. RESULTS Up-regulated SNHG16, TLR4 and TRAF6 expression with increased autophagy and NETosis was demonstrated in the SLE-AH lungs. In such patients, up-regulated SNHG16, TLR4 and TRAF6 expression was found in PB mononuclear cells with increased autophagy and in PB neutrophils with increased NETosis. There were up-regulated TLR4 expression and increased LPS-induced autophagy and NETosis in SNHG16-overexpressed cells, while down-regulated TLR4 expression and decreased LPS-induced autophagy and NETosis in SNHG16-silenced cells. Pristane-injected lung tissues had up-regulated SNHG16, TLR4/TRAF6 levels and increased in situ autophagy and NETosis formation. Intra-pulmonary LV-SNHG16 delivery enhanced AH through up-regulating TLR4/TRAF6 expression with increased cell death processes, while intra-pulmonary prophylactic and early therapeutic sh-SNHG16 delivery suppressed AH by down-regulating TLR4/TRAF6 expression with reduced such processes. In addition, there was decreased renal SNHG16 expression in LN patients and mice. CONCLUSIONS Our results demonstrate that lncRNA SNHG16 regulates TLR4-mediated autophagy and NETosis formation in the human and mouse AH lungs, and provide a therapeutic potential of intra-pulmonary delivery of shRNA targeting SNHG16 in this SLE-related lethal manifestation.
Collapse
Affiliation(s)
- Yu-Tung Hsieh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Cheng Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Pin-Yu Kuo
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ting Yen
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Wen Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chrong-Reen Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
20
|
Bache M, Kadler F, Struck O, Medenwald D, Ostheimer C, Güttler A, Keßler J, Kappler M, Riemann A, Thews O, Seliger B, Vordermark D. Correlation between Circulating miR-16, miR-29a, miR-144 and miR-150, and the Radiotherapy Response and Survival of Non-Small-Cell Lung Cancer Patients. Int J Mol Sci 2023; 24:12835. [PMID: 37629015 PMCID: PMC10454434 DOI: 10.3390/ijms241612835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Despite the success of current therapy concepts, patients with advanced non-small-cell lung cancer (NSCLC) still have a very poor prognosis. Therefore, biological markers are urgently needed, which allow the assessment of prognosis, or prediction of the success of therapy or resistance in this disease. Circulating microRNAs (miRs) have potential as biomarkers for the prognosis and prediction of response to therapy in cancer patients. Based on recent evidence that circulating miR-16, miR-29a, miR-144 and miR-150 can be regulated by ionizing radiation, the concentration of these four miRs was assessed in the plasma of NSCLC patients at different time points of radiotherapy by digital droplet PCR (ddPCR). Furthermore, their impact on patients' prognosis was evaluated. The mean plasma levels of miR-16, miR-29a, miR-144 and miR-150 significantly differed intra- and inter-individually, and during therapy in NSCLC patients, but showed a strong positive correlation. The individual plasma levels of miR-16, miR-29a and miR-144 had prognostic value in NSCLC patients during or at the end of radiotherapy in Cox's regression models. NSCLC patients with low levels of these three miRs at the end of radiotherapy had the worst prognosis. However, miR-150 plasma levels and treatment-dependent changes were not predictive. In conclusion, circulating miR-16, miR-29a and miR-144, but not miR-150, have a prognostic value in NSCLC patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Matthias Bache
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| | - Frauke Kadler
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| | - Olivia Struck
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
- Department of Radiology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany
| | - Daniel Medenwald
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| | - Christian Ostheimer
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| | - Antje Güttler
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| | - Jacqueline Keßler
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| | - Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany;
| | - Anne Riemann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112 Halle, Germany; (A.R.); (O.T.)
| | - Oliver Thews
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06112 Halle, Germany; (A.R.); (O.T.)
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 16, 06112 Halle, Germany;
- Institute for Translational Immunology, Brandenburg Medical School “Theodor Fontane”, 14770 Brandenburg, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
| | - Dirk Vordermark
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany; (F.K.); (O.S.); (D.M.); (A.G.); (J.K.); (D.V.)
| |
Collapse
|