1
|
Cheng Y, Feng Y, Zhao J, Li L, Dong H. Near-Infrared Light-Triggered Cascade Nanosystems for Spatiotemporally Controlled Gene-Silencing and Gas Synergistic Cancer Therapy. Angew Chem Int Ed Engl 2025; 64:e202422375. [PMID: 39663885 DOI: 10.1002/anie.202422375] [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: 11/18/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
Despite the tremendous potential of DNAzymes in gene therapy, achieving gene regulation with spatiotemporal precision and satisfactory efficacy remains a key challenge. Herein, we developed a near-infrared light (NIR)-triggered cascade effect system that enables precise spatiotemporal gene silencing and gas-synergistic cancer therapy. This nanoplatform was constructed using an enzyme-activatable DNAzyme, a nitric oxide (NO) precursor, and upconversion nanoparticles (UCNPs). The UCNPs act as light converters, facilitating the precursor to produce NO under NIR light irradiation, while the generated NO not only participates directly in gas therapy but also induces the upregulation of cytoplasmic APE1 level, resulting in enhanced DNAzyme activation and spatially controlled gene silencing. By leveraging this mechanism, cascade effects mediated gas-gene synergistic therapy is achieved through the combination of NIR light-controlled NO release and APE1-activatable gene therapy. We demonstrated that this nanoplatform enables enhanced anti-tumor effects both in vitro and in vivo. This strategy represents a critical step forward in the quest for highly targeted and efficient cancer treatments.
Collapse
Affiliation(s)
- Yaru Cheng
- Department Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Shen-zhen Key Laboratory for Nano-Biosensing Technology, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Youming Feng
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Shen-zhen Key Laboratory for Nano-Biosensing Technology, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Jian Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Haifeng Dong
- Marshall Laboratory of Biomedical Engineering, Precision Medicine and Health Research Institute, Shen-zhen Key Laboratory for Nano-Biosensing Technology, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| |
Collapse
|
2
|
Hu M, Zhang Y, Zhang P, Liu K, Zhang M, Li L, Yu Z, Zhang X, Zhang W, Xu Y. Targeting APE1: Advancements in the Diagnosis and Treatment of Tumors. Protein Pept Lett 2025; 32:18-33. [PMID: 39648425 DOI: 10.2174/0109298665338519241114103223] [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: 07/11/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 12/10/2024]
Abstract
With the emergence of the precision medicine era, targeting specific proteins has emerged as a pivotal breakthrough in tumor diagnosis and treatment. Apurinic/apyrimidinic Endonuclease 1 (APE1) is a multifunctional protein that plays a crucial role in DNA repair and cellular redox regulation. This article comprehensively explores the fundamental mechanisms of APE1 as a multifunctional enzyme in biology, with particular emphasis on its potential significance in disease diagnosis and strategies for tumor treatment. Firstly, this article meticulously analyzes the intricate biological functions of APE1 at a molecular level, establishing a solid theoretical foundation for subsequent research endeavors. In terms of diagnostic applications, the presence of APE1 can be detected in patient serum samples, biopsy tissues, and through cellular in situ testing. The precise detection methods enable changes in APE1 levels to serve as reliable biomarkers for predicting tumor occurrence, progression, and patient prognosis. Moreover, this article focuses on elucidating the potential role of APE1 in tumor treatment by exploring various inhibitors, including nucleic acid-based inhibitors and small molecule drug inhibitors categories, and revealing their unique advantages in disrupting DNA repair function and modulating oxidative-reduction activity. Finally, the article provides an outlook on future research directions for APE1 while acknowledging major technical difficulties and clinical challenges that need to be overcome despite its immense potential as a target for tumor therapy.
Collapse
Affiliation(s)
- Minghui Hu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Yingyu Zhang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Kangbo Liu
- Henan Institute for Drug and Medical Device Inspection (Henan Vaccine Issuance Center), Zhengzhou, 450018, China
| | - Mengxin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Lifeng Li
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Zhidan Yu
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Xianwei Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Wancun Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Ying Xu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| |
Collapse
|
3
|
Velmurugan S, Ganesan K, Rajasundaram A, Thangam C, Cyril R, Subbaraj GK. Nanoparticles and the Vaginal Microbiota: Diagnostic and Therapeutic Innovations in Human Papilloma Virus-associated Cervical Cancer - A Systematic Review. Niger Postgrad Med J 2025; 32:1-13. [PMID: 40091465 DOI: 10.4103/npmj.npmj_265_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 03/19/2025]
Abstract
Cervical cancer (CC) remains a significant global health challenge, with the interplay between microbiome and cancer progression becoming increasingly evident. A comprehensive literature search was conducted across multiple databases, including Embase, NCBI, Google Scholar, Elsevier, Science Direct and PubMed, utilising the specific keywords such as 'cervical cancer', 'cervical microbiome', 'vaginal microbiome', 'Human papillomavirus', 'nanoparticles', 'drug delivery' and 'dysbiosis'. Studies published over the past 15 years were reviewed. A total of 400 articles were identified and 65 research articles were finally included. This systematic approach identified relevant studies, enabling an in-depth analysis of microbial species, such as Parabacteroides, Escherichia, Shigella, Gardnerella and Fusobacterium which are recognised as the potential biomarkers for CC diagnosis and progression. Dysbiosis is characterised by a reduction in helpful bacteria and a proliferation of harmful ones. It is linked to chronic inflammation and human papillomavirus infection, both of which facilitate the CC progression. Advanced nanotechnology presents innovative therapeutic options for CC treatment, enhancing drug delivery systems and targeting tumour cells more effectively. Moreover, incorporating nanocarriers into treatment regimens aims to improve the bioavailability and efficacy of existing therapies, potentially transforming the clinical approaches to CC management. This review highlights the dual role of the microbiome as both a diagnostic and prognostic biomarker and elucidates the potential of nanotechnology in optimising treatment strategies, advocating for further research on microbial interactions in cancer pathology.
Collapse
Affiliation(s)
- Saranya Velmurugan
- Medical Genetics Division, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Karthikeyan Ganesan
- Department of Pharmacology, Vinayaka Mission's Kirupananda Variyar Medical College and Hospitals, Vinayaka Mission's Research Foundation (DU), Salem, Tamil Nadu, India
| | - Archana Rajasundaram
- Department of Anatomy, Sree Balaji Medical College and Hospital, Chennai, Tamil Nadu, India
| | - C Thangam
- Department of Pharmacology, KSR Institute of Dental Science and Research, Tiruchengode, Tamil Nadu, India
| | - Rozario Cyril
- Department of Pharmacology, JKK Nattraja Dental College and Hospital, Namakkal, Tamil Nadu, India
| | - Gowtham Kumar Subbaraj
- Medical Genetics Division, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
4
|
Zhu L, Zhang Y, Ding J, Xin W, Fan GC, Song ZL, Zhang P, Luo X. Intelligent DNA Nanosystem for Broad-Spectrum Oncological Typing and Therapy. ACS Sens 2024; 9:6655-6666. [PMID: 39622787 DOI: 10.1021/acssensors.4c02246] [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] [Indexed: 12/28/2024]
Abstract
The occurrence of multiple primary cancers in individual patients underscores the need for diagnostic and therapeutic techniques with augmented cancer-targeting selectivity and broad-spectrum antitumor effects. To address this, we develop a quadruple-input-triggered OR-AND-AND logic gated oncological nanosystem (OAA). This system employs four cancer-related markers (EpCAM, MUC1, APE1, and miR-21) to generate three distinct fluorescence signals, enabling precise differentiation of various cancer cell lines (MCF-7, HepG2, and HeLa) from normal cells (MCF-10A). Additionally, the OAA system integrates photodynamic therapy (PDT) and gene silencing strategies, allowing selective activation of Ce6 release, miR-21 gene silencing, and VEGFR2 mRNA gene silencing through the OR-AND-AND logic gating mechanism in a cancer-specific manner. This synergetic therapeutic approach induces significant apoptosis in multiple cancer cell lines while sparing normal cells, demonstrating improved cancer-targeting specificity and broad-spectrum versatility. This intelligent platform precisely types and treats diverse cancer cells, powering the future exploration of advanced diagnostic and therapeutic strategies to combat highly heterogeneous diseases.
Collapse
Affiliation(s)
- Lina Zhu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yuxi Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jiani Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Wenjuan Xin
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Gao-Chao Fan
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Zhi-Ling Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453000, China
| | - Peisen Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
5
|
Hu X, Zhou P, Peng X, Ouyang Y, Li D, Wu X, Yang L. PXD101 inhibits malignant progression and radioresistance of glioblastoma by upregulating GADD45A. J Transl Med 2024; 22:1047. [PMID: 39568000 PMCID: PMC11577825 DOI: 10.1186/s12967-024-05874-5] [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: 07/03/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Histone deacetylase inhibitors (HDACis) have shown a significant antitumor effect in clinical studies, and PXD101 is a novel HDACi which can cross the blood-brain barrier. In this study, we showed that PXD101 could significantly inhibit the proliferation and invasion of glioblastoma (GBM) cells, while promoting their apoptosis and radiosensitivity. Furthermore, it was found that PXD101 exerted its antitumor function by upregulating the expression of the growth arrest and DNA damage inducible protein α (GADD45A). Mechanistically, PXD101 promoted the transcription of GADD45A by directly acetylating the histones H3 and H4, and GADD45A enhanced apoptosis and radiosensitivity through the activation of P38 in the GBM cells. In vivo experiments also showed that PXD101 combined with radiotherapy could significantly inhibit the growth of GBM. This study provides experimental evidence for application of the novel HDACi PXD101 in the treatment of GBM, as well as new molecular markers and potential intervention targets that may be used in preventing GBM malignant progression and radioresistance.
Collapse
Affiliation(s)
- Xiaohong Hu
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 110, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, 410078, China
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, 410012, China
| | - Peijun Zhou
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 110, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Xingzhi Peng
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 110, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Yiting Ouyang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 110, Changsha, 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Dan Li
- Institute of Molecular Medicine and Oncology, College of Biology, Hunan University, Changsha, 410012, China
| | - Xia Wu
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, 410078, China.
- Department of Pathology, The Second Xiangya Hospital, Central South University, Renmin Middle Road 174, Changsha, 410011, China.
| | - Lifang Yang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 110, Changsha, 410078, China.
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha, 410078, China.
| |
Collapse
|
6
|
Zhang Y, Tian J. Strategies, Challenges, and Prospects of Nanoparticles in Gynecological Malignancies. ACS OMEGA 2024; 9:37459-37504. [PMID: 39281920 PMCID: PMC11391544 DOI: 10.1021/acsomega.4c04573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Abstract
Gynecologic cancers are a significant health issue for women globally. Early detection and successful treatment of these tumors are crucial for the survival of female patients. Conventional therapies are often ineffective and harsh, particularly in advanced stages, necessitating the exploration of new therapy options. Nanotechnology offers a novel approach to biomedicine. A novel biosensor utilizing bionanotechnology can be employed for early tumor identification and therapy due to the distinctive physical and chemical characteristics of nanoparticles. Nanoparticles have been rapidly applied in the field of gynecologic malignancies, leading to significant advancements in recent years. This study highlights the significance of nanoparticles in treating gynecological cancers. It focuses on using nanoparticles for precise diagnosis and continuous monitoring of the disease, innovative imaging, and analytic methods, as well as multifunctional drug delivery systems and targeted therapies. This review examines several nanocarrier systems, such as dendrimers, liposomes, nanocapsules, and nanomicelles, for gynecological malignancies. The review also examines the enhanced therapeutic potential and targeted delivery of ligand-functionalized nanoformulations for gynecological cancers compared to nonfunctionalized anoformulations. In conclusion, the text also discusses the constraints and future exploration prospects of nanoparticles in chemotherapeutics. Nanotechnology will offer precise methods for diagnosing and treating gynecological cancers.
Collapse
Affiliation(s)
- Yingfeng Zhang
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jing Tian
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| |
Collapse
|
7
|
Lu L, Li F, Gao Y, Kang S, Li J, Guo J. Microbiome in radiotherapy: an emerging approach to enhance treatment efficacy and reduce tissue injury. Mol Med 2024; 30:105. [PMID: 39030525 PMCID: PMC11264922 DOI: 10.1186/s10020-024-00873-0] [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: 11/06/2023] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Radiotherapy is a widely used cancer treatment that utilizes powerful radiation to destroy cancer cells and shrink tumors. While radiation can be beneficial, it can also harm the healthy tissues surrounding the tumor. Recent research indicates that the microbiota, the collection of microorganisms in our body, may play a role in influencing the effectiveness and side effects of radiation therapy. Studies have shown that specific species of bacteria living in the stomach can influence the immune system's response to radiation, potentially increasing the effectiveness of treatment. Additionally, the microbiota may contribute to adverse effects like radiation-induced diarrhea. A potential strategy to enhance radiotherapy outcomes and capitalize on the microbiome involves using probiotics. Probiotics are living microorganisms that offer health benefits when consumed in sufficient quantities. Several studies have indicated that probiotics have the potential to alter the composition of the gut microbiota, resulting in an enhanced immune response to radiation therapy and consequently improving the efficacy of the treatment. It is important to note that radiation can disrupt the natural balance of gut bacteria, resulting in increased intestinal permeability and inflammatory conditions. These disruptions can lead to adverse effects such as diarrhea and damage to the intestinal lining. The emerging field of radiotherapy microbiome research offers a promising avenue for optimizing cancer treatment outcomes. This paper aims to provide an overview of the human microbiome and its role in augmenting radiation effectiveness while minimizing damage.
Collapse
Affiliation(s)
- Lina Lu
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China.
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China.
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China.
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China.
| | - Fengxiao Li
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Shuhe Kang
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jia Li
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jinwang Guo
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
8
|
Malfatti MC, Bellina A, Antoniali G, Tell G. Revisiting Two Decades of Research Focused on Targeting APE1 for Cancer Therapy: The Pros and Cons. Cells 2023; 12:1895. [PMID: 37508559 PMCID: PMC10378182 DOI: 10.3390/cells12141895] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
APE1 is an essential endodeoxyribonuclease of the base excision repair pathway that maintains genome stability. It was identified as a pivotal factor favoring tumor progression and chemoresistance through the control of gene expression by a redox-based mechanism. APE1 is overexpressed and serum-secreted in different cancers, representing a prognostic and predictive factor and a promising non-invasive biomarker. Strategies directly targeting APE1 functions led to the identification of inhibitors showing potential therapeutic value, some of which are currently in clinical trials. Interestingly, evidence indicates novel roles of APE1 in RNA metabolism that are still not fully understood, including its activity in processing damaged RNA in chemoresistant phenotypes, regulating onco-miRNA maturation, and oxidized RNA decay. Recent data point out a control role for APE1 in the expression and sorting of onco-miRNAs within secreted extracellular vesicles. This review is focused on giving a portrait of the pros and cons of the last two decades of research aiming at the identification of inhibitors of the redox or DNA-repair functions of APE1 for the definition of novel targeted therapies for cancer. We will discuss the new perspectives in cancer therapy emerging from the unexpected finding of the APE1 role in miRNA processing for personalized therapy.
Collapse
Affiliation(s)
- Matilde Clarissa Malfatti
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Alessia Bellina
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Giulia Antoniali
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
9
|
Li Y, Zhao H, Li N, Yuan C, Dong N, Wen J, Li Z, Wang Q, Wang L, Mao H. BBOX1-AS1 mediates trophoblast cells dysfunction via regulating hnRNPK/GADD45A axis†. Biol Reprod 2023; 108:408-422. [PMID: 36617174 DOI: 10.1093/biolre/ioad002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023] Open
Abstract
Recurrent pregnancy loss (RPL) is a common pathological problem during pregnancy, and its clinical etiology is complex and unclear. Dysfunction of trophoblasts may cause a series of pregnancy complications, including preeclampsia, fetal growth restriction, and RPL. Recently, lncRNAs have been found to be closely related to the occurrence and regulation of pregnancy-related diseases, but few studies have focused on their role in RPL. In this study, we identified a novel lncRNA BBOX1-AS1 that was significantly upregulated in villous tissues and serum of RPL patients. Functionally, BBOX1-AS1 inhibited proliferation, migration, invasion, tube formation and promoted apoptosis of trophoblast cells. Mechanistically, overexpression of BBOX1-AS1 activated the p38 and JNK MAPK signaling pathways by upregulating GADD45A expression. Further studies indicated that BBOX1-AS1 could increase the stability of GADD45A mRNA by binding hnRNPK and ultimately cause abnormal trophoblast function. Collectively, our study highlights that the BBOX1-AS1/hnRNPK/GADD45A axis plays an important role in trophoblast-induced RPL and that BBOX1-AS1 may serve as a potential target for the diagnosis of RPL.
Collapse
Affiliation(s)
- Yali Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Hui Zhao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Ning Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Chao Yuan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Nana Dong
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jin Wen
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Zihui Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lina Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Haiting Mao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
DNA Damage Response in Cancer Therapy and Resistance: Challenges and Opportunities. Int J Mol Sci 2022; 23:ijms232314672. [PMID: 36499000 PMCID: PMC9735783 DOI: 10.3390/ijms232314672] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Resistance to chemo- and radiotherapy is a common event among cancer patients and a reason why new cancer therapies and therapeutic strategies need to be in continuous investigation and development. DNA damage response (DDR) comprises several pathways that eliminate DNA damage to maintain genomic stability and integrity, but different types of cancers are associated with DDR machinery defects. Many improvements have been made in recent years, providing several drugs and therapeutic strategies for cancer patients, including those targeting the DDR pathways. Currently, poly (ADP-ribose) polymerase inhibitors (PARP inhibitors) are the DDR inhibitors (DDRi) approved for several cancers, including breast, ovarian, pancreatic, and prostate cancer. However, PARPi resistance is a growing issue in clinical settings that increases disease relapse and aggravate patients' prognosis. Additionally, resistance to other DDRi is also being found and investigated. The resistance mechanisms to DDRi include reversion mutations, epigenetic modification, stabilization of the replication fork, and increased drug efflux. This review highlights the DDR pathways in cancer therapy, its role in the resistance to conventional treatments, and its exploitation for anticancer treatment. Biomarkers of treatment response, combination strategies with other anticancer agents, resistance mechanisms, and liabilities of treatment with DDR inhibitors are also discussed.
Collapse
|
11
|
The DeltaN p63 Promotes EMT and Metastasis in Bladder Cancer by the PTEN/AKT Signalling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9566055. [PMID: 35463095 PMCID: PMC9019423 DOI: 10.1155/2022/9566055] [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: 01/08/2022] [Revised: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 11/20/2022]
Abstract
Bladder cancer is a common tumour of the urinary system, and more than 90% is urothelial carcinoma. Therefore, it is important for discovering the key target genes and molecules of bladder tumour cell metastasis and invasion. Our research initially explored the regulation of deltaN p63 on the progression and metastasis of bladder cancer and found that deltaN p63 can influence the occurrence of EMT through PTEN and ultimately regulate the growth and metastasis of bladder cancer. In summary, this study identified a new EMT regulator, deltaN p63, further revealed the mechanism of the invasion and metastasis of bladder cancer cells, and provided a theoretical basis for finding new target molecules and drugs to treat bladder cancer. In conclusion, this study will further reveal the mechanism of tumour cell invasion and metastasis and provide a theoretical basis for cancer treatment to find new target molecules and drugs.
Collapse
|
12
|
Inhibition of APE1 Expression Enhances the Antitumor Activity of Olaparib in Triple-Negative Breast Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6048017. [PMID: 35463096 PMCID: PMC9020940 DOI: 10.1155/2022/6048017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that is prone to recurrence and metastasis. Because of the lack of expression of estrogen receptor (ER) and progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) in TNBC, treatment methods are greatly limited. In this study, the proliferation inhibition and apoptosis-inducing effects of PARP1 inhibitors in TNBC breast cancer cells and in vivo xenograft animal models were examined to investigate the molecular role of APE1 in PARP1-targeted therapy. In TNBC patients, the expression of APE1 and PARP1 were positively correlated, and high expression of APE1 and PARP1 was associated with poor survival of TNBC. Our results indicated that knockdown APE1 could increase the sensitivity of olaparib in the treatment of TNBC. In conclusion, the results of this study will not only clarify the molecular role of APE1 in PARP1-targeted therapy for TNBC but also provide a theoretical basis for the future clinical application of targeting APE1 and PARP1 in the treatment of refractory TNBC.
Collapse
|
13
|
Song Q, Wen J, Li W, Xue J, Zhang Y, Liu H, Han J, Ning T, Lu Z. HSP90 promotes radioresistance of cervical cancer cells via reducing FBXO6 mediated CD147 polyubiquitination. Cancer Sci 2022; 113:1463-1474. [PMID: 35043518 PMCID: PMC8990293 DOI: 10.1111/cas.15269] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
HSP90 inhibition might be a promising strategy to overcome the radioresistance of some cancers. In the current study, we further explored the mechanisms of HSP90 in regulating the radiosensitivity of cervical cancer cells. Bioinformatic analysis was performed based on data from TCGA‐CESC. Cellular and molecular studies were conducted using CaSki and SiHa and the derived radioresistant (RR) subclones. Through a proteomics screen, we identified HSP90 chaperones (both HSP90α and HSP90β) as CD147‐binding partners supporting its stabilization. Targeting HSP90 sensitized CaSki‐RR and SiHa‐RR cancer cells to irradiation partially through CD147 destabilization. Mechanistically, HSP90 interacts with FBXO6 and reduces FBXO6‐mediated proteasomal degradation of CD147. Enforced FBXO6 overexpression also sensitized CaSki‐RR and SiHa‐RR cancer cells to irradiation. These effects were enhanced using 17‐AAG treatment but were weakened by CD147 overexpression. Survival analysis further confirmed the association between high FBXO6 expression and favorable progression‐free survival among patients with cervical cancer. In conclusion, this study showed that HSP90 promotes radioresistance of cervical cancer cells partially via reducing FBXO6 mediated CD147 polyubiquitination. These findings help to explain why HSP90 inhibitor exerts radio‐sensitizing effects in cervical cancer.
Collapse
Affiliation(s)
- Qi Song
- Senior Department of Obstetrics and Gynecology the Seventh Medical Center of PLA General Hospital Beijing China
| | - Juyi Wen
- Senior Department of Oncology the Fifth Medical Center of PLA General Hospital Beijing China
| | - Weiping Li
- Senior Department of Obstetrics and Gynecology the Seventh Medical Center of PLA General Hospital Beijing China
| | - Janxin Xue
- Department of Thoracic Oncology Cancer Center and State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Yufei Zhang
- Senior Department of Oncology the Fifth Medical Center of PLA General Hospital Beijing China
| | - Hongyan Liu
- Senior Department of Oncology the Fifth Medical Center of PLA General Hospital Beijing China
| | - Jixia Han
- Senior Department of Oncology the Fifth Medical Center of PLA General Hospital Beijing China
| | - Tao Ning
- Tianjin Medical University Cancer Institute and Hospital Tianjin China
| | - Zejun Lu
- Senior Department of Oncology the Fifth Medical Center of PLA General Hospital Beijing China
| |
Collapse
|
14
|
Carlos-Reyes A, Muñiz-Lino MA, Romero-Garcia S, López-Camarillo C, Hernández-de la Cruz ON. Biological Adaptations of Tumor Cells to Radiation Therapy. Front Oncol 2021; 11:718636. [PMID: 34900673 PMCID: PMC8652287 DOI: 10.3389/fonc.2021.718636] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Radiation therapy has been used worldwide for many decades as a therapeutic regimen for the treatment of different types of cancer. Just over 50% of cancer patients are treated with radiotherapy alone or with other types of antitumor therapy. Radiation can induce different types of cell damage: directly, it can induce DNA single- and double-strand breaks; indirectly, it can induce the formation of free radicals, which can interact with different components of cells, including the genome, promoting structural alterations. During treatment, radiosensitive tumor cells decrease their rate of cell proliferation through cell cycle arrest stimulated by DNA damage. Then, DNA repair mechanisms are turned on to alleviate the damage, but cell death mechanisms are activated if damage persists and cannot be repaired. Interestingly, some cells can evade apoptosis because genome damage triggers the cellular overactivation of some DNA repair pathways. Additionally, some surviving cells exposed to radiation may have alterations in the expression of tumor suppressor genes and oncogenes, enhancing different hallmarks of cancer, such as migration, invasion, and metastasis. The activation of these genetic pathways and other epigenetic and structural cellular changes in the irradiated cells and extracellular factors, such as the tumor microenvironment, is crucial in developing tumor radioresistance. The tumor microenvironment is largely responsible for the poor efficacy of antitumor therapy, tumor relapse, and poor prognosis observed in some patients. In this review, we describe strategies that tumor cells use to respond to radiation stress, adapt, and proliferate after radiotherapy, promoting the appearance of tumor radioresistance. Also, we discuss the clinical impact of radioresistance in patient outcomes. Knowledge of such cellular strategies could help the development of new clinical interventions, increasing the radiosensitization of tumor cells, improving the effectiveness of these therapies, and increasing the survival of patients.
Collapse
Affiliation(s)
- Angeles Carlos-Reyes
- Department of Chronic-Degenerative Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Marcos A. Muñiz-Lino
- Laboratorio de Patología y Medicina Bucal, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City, Mexico
| | - Susana Romero-Garcia
- Department of Chronic-Degenerative Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico, Mexico City
| | | |
Collapse
|
15
|
Qing Y, Li Q, Zhao LY, Shi P, Shan JL, Zhang W. LncRNA-PANDAR regulates the progression of thyroid carcinoma by targeting miR-637/KLK4. J Cancer 2021; 12:5879-5887. [PMID: 34476001 PMCID: PMC8408101 DOI: 10.7150/jca.55181] [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: 10/29/2020] [Accepted: 07/08/2021] [Indexed: 11/05/2022] Open
Abstract
Thyroid gland carcinoma (TC) originates from follicular or parafollicular thyroid cells and is one of the most common endocrine organ malignancies. To explore the molecular mechanism by which long-chain non-coding RNAs regulate the growth and metastasis of thyroid gland carcinoma, in this study we focused on long non-coding RNAs (lncRNAs) that have been reported to be involved in tumorigenesis. We identified Promoter Region of CDKN 1A antisense DNA damage-activated RNA (PANDAR), which was positively correlated with thyroid gland carcinoma risk. PANDAR could promote thyroid gland carcinoma cell proliferation and metastasis. PANDAR negatively correlated with miR-637, and miR-637 overexpression suppressed thyroid gland carcinoma progression, which could be reversed by PANDAR. MiR-637 could target Kallikrein-related peptidases 4 (KLK4) to inhibit its expression, which was high in thyroid gland carcinoma. KLK4 inhibited cell progression in thyroid gland carcinoma cells. Knockdown of PANDAR expression inhibited cancer progression in nude mice. Overall, PANDAR can suppress miR-637 and induce KLK4 to regulate invasion and migration in thyroid gland carcinoma. Additionally, we identified miR-637 as a target of PANDAR in thyroid gland carcinoma, and PANDAR can be used as a novel therapeutic target for the treatment of thyroid gland carcinoma.
Collapse
Affiliation(s)
- Yi Qing
- Department of Oncology, Affiliated Hospital of Chengdu University, Chengdu 610081, People's Republic of China
| | - Qian Li
- Department of Oncology, Daping hospital, Army Medical University, Chongqing, 400042, China
| | - Ling-Yan Zhao
- Department of respiratory and critical care medicine, Guangyuan Central Hospital, Guangyuan City, Sichuan Province, 628000, China
| | - Ping Shi
- Department of respiratory and critical care medicine, Guangyuan Central Hospital, Guangyuan City, Sichuan Province, 628000, China
| | - Jin-Lu Shan
- Department of Oncology, Daping hospital, Army Medical University, Chongqing, 400042, China
| | - Wei Zhang
- Department of medical oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Centre, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, China
| |
Collapse
|
16
|
Sun R, Chen C, Deng X, Wang F, Song S, Cai Q, Wang J, Zhang T, Shi M, Ke Q, Luo Z. IL-11 mediates the Radioresistance of Cervical Cancer Cells via the PI3K/Akt Signaling Pathway. J Cancer 2021; 12:4638-4647. [PMID: 34149927 PMCID: PMC8210555 DOI: 10.7150/jca.56185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/10/2021] [Indexed: 12/13/2022] Open
Abstract
Cervical cancer is one of the most common malignant tumors in the female reproductive system. Radioresistance remains a significant factor that limits the efficacy of radiotherapy for cervical cancer. Interleukin-11 (IL-11) has been reported to be upregulated in various types of human cancer and correlate with clinical stage and poor survival. However, the exact effects and mechanisms of IL-11 in the radioresistance of cervical cancer have not yet been defined. In this research, TCGA databases revealed that IL-11 expression was upregulated in cervical cancer tissues and was associated with clinical stages and poor prognosis in cervical cancer patients. We discovered that IL-11 concentration was significantly upregulated in radioresistant cervical cancer cells. Knocking down IL-11 in Hela cells could reduce clonogenic survival rate, decrease cell viability, induce G2/M phase block, and facilitate cell apoptosis. In contrast, Exogeneous IL-11 in C33A cells could upregulate clonogenic survival rate, increase cell viability, curb G2/M phase block, and cell apoptosis. Mechanistic investigations showed that radioresistance conferred by IL-11 was attributed to the activation of the PI3K/Akt signaling pathway. Altogether, our results demonstrate that IL-11 might be involved in radioresistance, and IL-11 may be a potent radiosensitization target for cervical cancer therapy.
Collapse
Affiliation(s)
- Ruige Sun
- Postgraduate Training Basement of Jinzhou Medical University, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province 442000, China.,Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| | - Chunli Chen
- Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| | - Xinzhou Deng
- Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| | - Fengqin Wang
- Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| | - Shimao Song
- Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| | - Qiang Cai
- Graduate School of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jincheng Wang
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan City, Hubei Province 442000, China
| | - Te Zhang
- Biomedical Research Institute, Hubei University of Medicine, Shiyan City, Hubei Province 442000, China
| | - Mingliang Shi
- Department of Gastroenterology, Zhushan People's Hospital, Shiyan City, Hubei Province 442000, China
| | - Qing Ke
- Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| | - Zhiguo Luo
- Department of Clinical Oncology, Taihe Hospital, Shiyan City, Hubei Province 442000, China
| |
Collapse
|
17
|
Zhou P, Liu W, Cheng Y, Qian D. Nanoparticle-based applications for cervical cancer treatment in drug delivery, gene editing, and therapeutic cancer vaccines. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1718. [PMID: 33942532 PMCID: PMC8459285 DOI: 10.1002/wnan.1718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/25/2022]
Abstract
Cervical cancer is a leading cause of gynecological tumor related deaths worldwide. The applications of conventional approaches such as chemoradiotherapy and surgery are restricted due to their side effects and drug resistances. Although immune checkpoint inhibitors (ICIs) have emerged as novel choices, their clinical response rates are rather limited. To date there is a lack of effective treatment regimens for patients with metastatic or recurrent cervical cancer. Recently nanomaterials like liposomes, dendrimers, and polymers are considered as promising delivery carriers with advantages of tumor‐specific administration, reduced toxicity, and improved biocompatibility. Here, we review the applications of nanoparticles in the fields of drug delivery, CRISPR based genome‐editing and therapeutic vaccines in cervical cancer treatment. This article is categorized under:Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease
Collapse
Affiliation(s)
- Peijie Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yong Cheng
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
18
|
Zou D, Li Z, Lv F, Yang Y, Yang C, Song J, Chen Y, Jin Z, Zhou J, Jiang Y, Ma Y, Jing Z, Tang Y, Zhang Y. Pan-Cancer Analysis of NOS3 Identifies Its Expression and Clinical Relevance in Gastric Cancer. Front Oncol 2021; 11:592761. [PMID: 33747912 PMCID: PMC7969995 DOI: 10.3389/fonc.2021.592761] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background:NOS3 (endothelial NOS, eNOS) is a member of the nitric oxide synthase (NOS) enzyme family, mainly participating in nitric oxide (NO) generation. NOS3 has been reported to inhibit apoptosis and promote angiogenesis, proliferation, and invasiveness. However, the expression pattern of NOS3 and its diagnostic and prognostic potential has not been investigated in a pan-cancer perspective. Methods: Data from the Genotype-Tissue Expression (GTEx), the Cancer Genome Atlas (TCGA), the Cancer Cell Line Encyclopedia (CCLE), and the Cancer Therapeutics Response Portal (CTRP) were employed and NOS3 expression was comprehensively analyzed in normal tissues, cancer tissues, and cell lines. Immunohistochemical staining of tissue sections were used to validate the prognostic role of NOS3 in gastric cancer patients. GSVA and GSEA analyses were performed to investigate signaling pathways related to NOS3 expression. Results: In normal tissues, NOS3 was expressed highest in the spleen and lowest in the blood. NOS3 expression was increased in stomach adenocarcinoma (STAD) and significantly associated with poor prognosis of patients. Immunohistochemical staining validated that NOS3 was an independent prognostic factor of gastric cancer. Several canonical cancer-related pathways were found to be correlated with NOS3 expression in STAD. The expression of NOS3 was related to the response to QS-11 and brivinib in STAD. Conclusions:NOS3 was an independent prognostic factor for patients with STAD. Increased expression of NOS3 influenced occurrence and development of STAD through several canonical cancer-related pathways. Drug response analysis reported drugs to suppress NOS3. NOS3 might be a novel target for gastric cancer treatment.
Collapse
Affiliation(s)
- Dan Zou
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Fei Lv
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yi Yang
- Laboratory Animal Center, China Medical University, Shenyang, China
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Jincheng Song
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China.,Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Chen
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Zi Jin
- The First Department of Oncology, Shenyang Fifth People's Hospital, Shenyang, China
| | - Jinpeng Zhou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yang Jiang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China.,Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanju Ma
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, China
| | - Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yu Tang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
Caston RA, Gampala S, Armstrong L, Messmann RA, Fishel ML, Kelley MR. The multifunctional APE1 DNA repair-redox signaling protein as a drug target in human disease. Drug Discov Today 2021; 26:218-228. [PMID: 33148489 PMCID: PMC7855940 DOI: 10.1016/j.drudis.2020.10.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/27/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Apurinic/apyrimidinic (AP) endonuclease-reduction/oxidation factor 1 (APE1/Ref-1, also called APE1) is a multifunctional enzyme with crucial roles in DNA repair and reduction/oxidation (redox) signaling. APE1 was originally described as an endonuclease in the Base Excision Repair (BER) pathway. Further study revealed it to be a redox signaling hub regulating critical transcription factors (TFs). Although a significant amount of focus has been on the role of APE1 in cancer, recent findings support APE1 as a target in other indications, including ocular diseases [diabetic retinopathy (DR), diabetic macular edema (DME), and age-related macular degeneration (AMD)], inflammatory bowel disease (IBD) and others, where APE1 regulation of crucial TFs impacts important pathways in these diseases. The central responsibilities of APE1 in DNA repair and redox signaling make it an attractive therapeutic target for cancer and other diseases.
Collapse
Affiliation(s)
- Rachel A Caston
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | - Silpa Gampala
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | - Lee Armstrong
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | | | - Melissa L Fishel
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA
| | - Mark R Kelley
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, 1044 W. Walnut, Indianapolis, IN 46202, USA.
| |
Collapse
|
20
|
Li KK, Mao CY, Ma Q, Bao T, Wang YJ, Guo W, Zhao XL. U three protein 14a (UTP14A) promotes tumour proliferation and metastasis via the PERK/eIF2a/GRP78 signalling pathway in oesophageal squamous cell carcinoma. J Cancer 2021; 12:134-140. [PMID: 33391409 PMCID: PMC7738832 DOI: 10.7150/jca.44649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 08/29/2020] [Indexed: 12/22/2022] Open
Abstract
Metastasis and malignant proliferation are major obstacles to the treatment of oesophageal squamous cell carcinoma (ESCC), and UTP14A is associated with poor prognosis in ESCC. However, its mechanisms have not been fully elucidated. The TCGA and GEO databases were used to identify candidate target genes and possible downstream targets. Then, the effects were determined in vitro and in vivo through knockdown and overexpression techniques, and the mechanism was explored. UTP14A was significantly higher in the tumour tissue of ESCC patients than in normal tissue. Knockdown of UTP14A significantly suppressed the migration and proliferation of ESCC cells. The PERK/eIF2a signalling pathway was positively regulated by UTP14A, and its tumour-promoting effect was further activated by overexpression of UTP14A. In conclusion, UTP14A might promote the proliferation and metastasis of ESCC cells by inducing PERK/eIF2a signalling pathway expression.
Collapse
Affiliation(s)
- Kun-Kun Li
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, PR China
| | - Cheng-Yi Mao
- Department of Pathology, Daping Hospital, Army Medical University, PR China
| | - Qiang Ma
- Department of Pathology, Daping Hospital, Army Medical University, PR China
| | - Tao Bao
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, PR China
| | - Ying-Jian Wang
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, PR China
| | - Wei Guo
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, PR China
| | - Xiao-Long Zhao
- Department of Pathology, Daping Hospital, Army Medical University, PR China
| |
Collapse
|
21
|
Yang F, Liu WW, Chen H, Zhu J, Huang AH, Zhou F, Gan Y, Zhang YH, Ma L. Carfilzomib inhibits the growth of lung adenocarcinoma via upregulation of Gadd45a expression. J Zhejiang Univ Sci B 2020; 21:64-76. [PMID: 31898443 DOI: 10.1631/jzus.b1900551] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.
Collapse
Affiliation(s)
- Fang Yang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Wang-Wang Liu
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Hui Chen
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Jia Zhu
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Ai-Hua Huang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Fei Zhou
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yi Gan
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yan-Hua Zhang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Li Ma
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
22
|
Park M, Kwon J, Shin HJ, Moon SM, Kim SB, Shin US, Han YH, Kim Y. Butyrate enhances the efficacy of radiotherapy via FOXO3A in colorectal cancer patient‑derived organoids. Int J Oncol 2020; 57:1307-1318. [PMID: 33173975 PMCID: PMC7646587 DOI: 10.3892/ijo.2020.5132] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022] Open
Abstract
Enhancing the radioresponsiveness of colorectal cancer (CRC) is essential for local control and prognosis. However, consequent damage to surrounding healthy cells can lead to treatment failure. We hypothesized that short‑chain fatty acids (SCFAs) could act as radiosensitizers for cancer cells, allowing the administration of a lower and safer dose of radiation. To test this hypothesis, the responses of three‑dimensional‑cultured organoids, derived from CRC patients, to radiotherapy, as well as the effects of combined radiotherapy with the SCFAs butyrate, propionate and acetate as candidate radiosensitizers, were evaluated via reverse transcription‑quantitative polymerase chain reaction, immunohistochemistry and organoid viability assay. Of the three SCFAs tested, only butyrate suppressed the proliferation of the organoids. Moreover, butyrate significantly enhanced radiation‑induced cell death and enhanced treatment effects compared with administration of radiation alone. The radiation‑butyrate combination reduced the proportion of Ki‑67 (proliferation marker)‑positive cells and decreased the number of S phase cells via FOXO3A. Meanwhile, 3/8 CRC organoids were found to be non‑responsive to butyrate with lower expression levels of FOXO3A compared with the responsive cases. Notably, butyrate did not increase radiation‑induced cell death and improved regeneration capacity after irradiation in normal organoids. These results suggest that butyrate could enhance the efficacy of radiotherapy while protecting the normal mucosa, thus highlighting a potential strategy for minimizing the associated toxicity of radiotherapy.
Collapse
Affiliation(s)
- Misun Park
- Department of Radiological and Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Junhye Kwon
- Department of Radiological and Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Hye-Jin Shin
- Department of Radiological and Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Sun Mi Moon
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Sang Bum Kim
- Department of Radiological and Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Ui Sup Shin
- Department of Radiological and Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Young-Hoon Han
- Department of Radiological and Medico‑Oncological Sciences, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Younjoo Kim
- Department of Radiological and Clinical Research, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| |
Collapse
|
23
|
CB13, a novel PPARγ ligand, overcomes radio-resistance via ROS generation and ER stress in human non-small cell lung cancer. Cell Death Dis 2020; 11:848. [PMID: 33051435 PMCID: PMC7555888 DOI: 10.1038/s41419-020-03065-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a well-known therapeutic target for type 2 diabetes as well as is a potential target for effective anti-cancer drug, since PPARγ ligands such as ciglitazone (Cig) frequently cause cell death in many types of cancer cells and suppress tumor growth. However, many cancer patients acquire chemo-resistance or radio-resistance after chemo or radiotherapy, and it is still unclear. In the difficulty of well-known anti-cancer drugs, we developed a novel PPARγ agonist CB13 (1-benzyl-5-(4-methylphenyl) pyrido [2,3-d]pyrimidine-2,4(1H,3H)-dione) and investigated the anti-cancer effect and cell death mechanism on human non-small cell lung cancer (NSCLC) cells. With anti-cancer effect of Cig, CB13 also causes inhibition of cell growth by decreasing cell viability, increasing the release of LDH, and increasing caspase-3, and caspase-9 activities. CB13 generates reactive oxygen species (ROS) and causes cell death via ER stress in NSCLC and radio-resistant NSCLC cells (A549R and H460R), and a combination of CB13 and radiation induces greater ER stress and cell death when compared to CB13 alone. Taken together, our results suggest that a combination of CB13 and radiation may overcome radio-resistance caused by radiotherapy.
Collapse
|
24
|
Wang LA, Yang B, Tang T, Yang Y, Zhang D, Xiao H, Xu J, Wang L, Lin L, Jiang J. Correlation of APE1 with VEGFA and CD163 + macrophage infiltration in bladder cancer and their prognostic significance. Oncol Lett 2020; 20:2881-2887. [PMID: 32782604 PMCID: PMC7401005 DOI: 10.3892/ol.2020.11814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
The present study sought to estimate the applicability of apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1), vascular endothelial growth factor A (VEGFA) expression and CD163+ tumor-associated macrophage (TAM) ratio as prognostic factors in bladder cancer (BCa). A total of 127 patients with bladder urothelial cancer who underwent radical cystectomy at Daping Hospital were recruited between January 2013 and January 2017, including 45 cases of non-muscle invasive BCa (NMIBC) and 82 of MIBC. Immunohistochemical detection of APE1, VEGFA and CD163, as well as multiple immunofluorescence staining for APE1, VEGFA, CD163 and CD34, were performed on tissue samples. For APE1 and VEGFA, the staining was graded based on intensity (0–3), while CD163 was graded (0–3) based on the percentage of positively stained cells. The prognostic value of APE1, VEGF and CD163 was assessed using Kaplan-Meier and Cox regression analysis. The results suggested that in BCa, high APE1 expression was associated with high VEGFA expression and more infiltration of CD163+ TAM. Furthermore, high expression of APE1 was associated with lymphovascular invasion of BCa, as well as reduced survival time. This indicates that APE1 may be associated with CD163+ TAM infiltration in BCa, with VEGFA as a possible influencing factor.
Collapse
Affiliation(s)
- Lin-Ang Wang
- Department of Urology, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Bo Yang
- Cancer Center, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Tang Tang
- Department of Urology, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Yuxin Yang
- Cancer Center, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA
| | - Hualiang Xiao
- Department of Pathology, Daping Hospital/Army Medical Center of PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Jing Xu
- Department of Urology, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Luofu Wang
- Department of Urology, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Li Lin
- Department of Pathology, Daping Hospital/Army Medical Center of PLA, Army Medical University, Chongqing 400042, P.R. China
| | - Jun Jiang
- Department of Urology, Daping Hospital/Army Medical Center of the PLA, Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
25
|
Yao Y, Li N. MIR600HG suppresses metastasis and enhances oxaliplatin chemosensitivity by targeting ALDH1A3 in colorectal cancer. Biosci Rep 2020; 40:BSR20200390. [PMID: 32270866 PMCID: PMC7189477 DOI: 10.1042/bsr20200390] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metastasis and chemoresistance indicate a poor prognosis in colorectal cancer (CRC) patients. However, the mechanisms that lead to the development of chemoresistance and metastasis in CRC remain unclear. MATERIALS AND METHODS We combined clinical and experimental studies to determine the role of MIR600HG in CRC metastasis and chemoresistance. The statistical analysis was performed using GraphPad Prism software, version 8.0. RESULTS We detected down-regulated expression of long non-coding RNA (lncRNA) MIR600HG in CRC specimens and cell lines compared with normal controls, and the expression level of MIR600HG was inversely correlated with the overall survival of CRC patients. The inhibition of MIR600HG stimulated CRC cell metastasis and chemoresistance. In addition, our data showed that the inhibition of MIR600HG stimulated CRC stemness, while the overexpression of MIR600HG suppressed stemness. Importantly, our animal experiments showed that MIR600HG inhibited tumour formation and that the combination of MIR600HG inhibition and oxaliplatin (Oxa) treatment significantly inhibited tumour growth compared with that with either intervention alone. Furthermore, we demonstrated that MIR600HG exerts its anticancer role by targeting ALDH1A3 in CRC. CONCLUSIONS Our data suggest that MIR600HG functions as a tumour suppressor and that the overexpression of MIR600HG inhibits tumour invasion and enhances chemosensitivity, providing a new strategy for CRC treatment.
Collapse
Affiliation(s)
- Yi Yao
- Department of Gastroenterology, Eighth Medical Center of PLA General Hospital, Beijing 100853, China
| | - Nan Li
- Medical School of Chinese PLA, Department of Gastroenterology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
26
|
Liu Y, Zhang Z, Zhang L, Zhong Z. Cytoplasmic APE1 promotes resistance response in osteosarcoma patients with cisplatin treatment. Cell Biochem Funct 2020; 38:195-203. [PMID: 31930546 DOI: 10.1002/cbf.3461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/02/2019] [Accepted: 10/28/2019] [Indexed: 11/09/2022]
Abstract
Chemotherapy resistance has become a hold back and major clinical challenge in osteosarcoma cancer. The alteration and subcellular distribution of apurinic/apyrimidinic endonuclease 1 (APE1) has been reported to be involved in chemotherapy resistance in many cancers. Here, we report that the cytoplasmic distribution of APE1 plays a key role in the sensitivity of combination platinum chemotherapy in osteosarcoma. Interestingly, the prevalence of cisplatin-induced DNA damage and apoptosis in low cytoplasmic APE1 osteosarcoma cell lines was higher than in high expression of cytoplasmic APE1 cell lines. Overexpression of cytoplasmic APE1 protected the osteosarcoma cells from CDDP-induced apoptosis. In addition, clinical data also show that the level of cytoplasmic APE1 was negatively associated with sensitivity to combination chemotherapy of cisplatin in osteosarcoma patients. Our findings suggest that cytoplasmic APE1 plays a significant role in chemotherapy resistance. This role is a supplement to the extranuclear function of APE1, and cytoplasmic APE1 expression level could be a promising predictor of platinum treatment prognosis for osteosarcoma patients.
Collapse
Affiliation(s)
- Yufeng Liu
- Cancer Center, The Third Affiliated Hospital and Research Institute of Surgery of Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Zhimin Zhang
- Cancer Center, The Third Affiliated Hospital and Research Institute of Surgery of Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Liang Zhang
- Cancer Center, The Third Affiliated Hospital and Research Institute of Surgery of Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Zhaoyang Zhong
- Cancer Center, The Third Affiliated Hospital and Research Institute of Surgery of Army Medical University (Third Military Medical University), Chongqing, PR China
| |
Collapse
|
27
|
Gong Y, Dai HS, Shu JJ, Liu W, Bie P, Zhang LD. LNC00673 suppresses proliferation and metastasis of pancreatic cancer via target miR-504/ HNF1A. J Cancer 2020; 11:940-948. [PMID: 31949497 PMCID: PMC6959011 DOI: 10.7150/jca.32855] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 09/22/2019] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer is a highly invasive malignant tumor of the digestive system. To explore the mechanism of pancreatic cancer development, development, invasion and metastasis, in this study we focused on long non-coding RNA (LncRNA), which has been reported to be involved in tumorigenesis. We identified a LINC00673, which is highly correlated with the pancreatic cancer risk. LINC00673 Overexpression is associated with good survival in pancreatic cancer patients, Effects of LINC00673 on pancreatic cancer cell apoptosis, viability, migration. LINC00673 negatively correlated with miR-504 and MiR-504 overexpression promoted cancer progression in Pancreatic cancer. MiR-504 negatively correlated with HNF1A, which was highly expressed Pancreatic cancer. HNF1A inhibited cell progression in pancreatic cancer cells. LINC00673 overexpression inhibited caner progression in nude mice. Taken together, LINC00673 can through suppress miR-504/ HNF1A regulating invasion and migration in pancreatic cancer. Also, we identified miR-504 as a target of LINC00673 in pancreatic cancer and LINC00673 can be used as a novel therapeutic target for the pancreatic cancer.
Collapse
Affiliation(s)
- Yi Gong
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hai-Su Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jun-Jie Shu
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wei Liu
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Ping Bie
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Lei-da Zhang
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
28
|
Sun Z, Zhu Y, Aminbuhe, Fan Q, Peng J, Zhang N. Differential expression of APE1 in hepatocellular carcinoma and the effects on proliferation and apoptosis of cancer cells. Biosci Trends 2019; 12:456-462. [PMID: 30473552 DOI: 10.5582/bst.2018.01239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This research aimed to investigate the differential expression of apurinic-apyrimidinic endonuclease 1 (APE1) in hepatocellular carcinoma (HCC) tissues and cells and the effects on proliferation and apoptosis of cancer cells. Immunohistochemical techniques were used to detect the expression of APE1 in 80 cases of HCC and the corresponding paracancerous tissue microarrays; meanwhile, Western blots were used to detect the expression of APE1 in both human HCC BEL-7402, BEL-7405, HCC-9204, Hep3B, HepG2, SMMC-7721 and Huh-7 cells, and normal hepatocyte L-02 cells. The relationship between APE1 expression and clinical pathological characteristics of HCC was statistically analyzed. APE1 shRNA vector was constructed in Hep 3B cells to establish a stably transfected cell line, using Western blots to determine the interference efficiency. Cell proliferation activity was detected with MTT assays, while apoptosis was detected with the Annexin V-FITC/PI double-labeling technique. The expression of APE1 in HCC tissues and cells was significantly up-regulated, and its expression was significantly different from TNM staging and histopathological grading. Down-regulation of APE1 expression significantly reduced the proliferative activity and increased the apoptosis rate of Hep 3B cells. In conclusion, APE1 demonstrates cancer progression potential at the clinical, tissue and cell level. It provides a new idea and theoretical basis for APE1-based clinical diagnosis, prognosis determination and molecular targeted therapy in treatment of HCC.
Collapse
Affiliation(s)
- Zhipeng Sun
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine)
| | - Yubing Zhu
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine)
| | - Aminbuhe
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine)
| | - Qing Fan
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine)
| | - Jirun Peng
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine)
| | - Nengwei Zhang
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University (Peking University Ninth School of Clinical Medicine)
| |
Collapse
|
29
|
Jiang L, Hong L, Yang W, Zhao Y, Tan A, Li Y. Co-expression network analysis of the lncRNAs and mRNAs associated with cervical cancer progression. Arch Med Sci 2019; 15:754-764. [PMID: 31110543 PMCID: PMC6524193 DOI: 10.5114/aoms.2019.84740] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/07/2018] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Cervical cancer is the second most common type of cancer and the third leading cause of cancer deaths in females in developing countries. Recent studies showed that long non-coding RNAs play a key role in human cancers. However, the molecular mechanisms underlying the initiation and progression of cervical cancer remained to be further explored. MATERIAL AND METHODS In this study, we explored the differential expression of lncRNAs and mRNAs in cervical cancer progression by analyzing the public dataset GSE63514. Next, PPI and co-expression networks were constructed to reveal the potential roles of cervical cancer related mRNAs and lncRNAs. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed to explore functions of differentially expressed genes (DEGs) in cervical cancer. RESULTS In the present study, we observed that 3021 mRNAs were up-regulated and 1605 mRNAs were down-regulated in cervical cancer progression. Meanwhile, we for the first time found that 172 lncRNAs were up-regulated and 106 lncRNAs were down-regulated in cervical cancer progression. Co-expression network analysis showed that lncRNAs were widely co-expressed with cell cycle related genes in cervical cancer, implicating the important roles of these lncRNAs in cell proliferation regulation. Of note, we identified two hub lncRNA-mRNA networks involved in regulating various biological processes in cervical cancer progression. CONCLUSIONS Our results identified key mRNAs and lncRNAs in cervical cancer progression. This study will provide novel insights to explore the potential mechanisms underlying cervical cancer progression.
Collapse
Affiliation(s)
- Li Jiang
- Department of Gynaecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hong
- Department of Gynaecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenwu Yang
- Department of Gynaecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuzi Zhao
- Department of Gynaecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Aili Tan
- Department of Gynaecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Li
- Department of Gynaecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|