1
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Sun Q, Lei X, Yang X. The crosstalk between non-coding RNAs and oxidative stress in cancer progression. Genes Dis 2025; 12:101286. [PMID: 40028033 PMCID: PMC11870203 DOI: 10.1016/j.gendis.2024.101286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2025] Open
Abstract
As living standards elevate, cancers are appearing in growing numbers among younger individuals globally and these risks escalate with advancing years. One of the reasons is that instability in the cancer genome reduces the effectiveness of conventional drug treatments and chemotherapy, compared with more targeted therapies. Previous research has discovered non-coding RNAs' crucial role in shaping genetic networks involved in cancer cell growth and invasion through their influence on messenger RNA production or protein binding. Additionally, the interaction between non-coding RNAs and oxidative stress, a crucial process in cancer advancement, cannot be overlooked. Essentially, oxidative stress results from the negative effects of radicals within the body and ties directly to cancer gene expression and signaling. Therefore, this review focuses on the mechanism between non-coding RNAs and oxidative stress in cancer progression, which is conducive to finding new cancer treatment strategies.
Collapse
Affiliation(s)
- Qiqi Sun
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
3
|
Liu R, Guo L, Shi D, Sun X, Shang M, Zhao Y, Wang X, Yang Y, Xiao S, Li J. Multilayer cascade-response nanoplatforms as metabolic symbiotic disruptors to reprogram the immunosuppressive microenvironment. J Control Release 2025; 383:113797. [PMID: 40318807 DOI: 10.1016/j.jconrel.2025.113797] [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: 02/05/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Nanomedicine is extensively utilized in tumor treatment, however, the restricted permeability of nanomaterials within tumor tissues, along with the inherent metabolic complexity of these tissues, have hindered effective control of tumor progression. Hypoxic and normoxic tumor cells utilize monocarboxylic acid transporters (MCTs) for the rapid reutilization of lactate, facilitating accelerated tumor growth. Here, cascade-response nanoplatforms (NPs) with contrast-enhanced ultrasound imaging (CEUI) capability had been established, incorporating basigin siRNA internally and featuring hyaluronidase (HAase) and γ-glutamyltranspeptidase (GGT)-responsive lipid coatings externally (GHB NPs). The GHB NPs took advantage of GGT-responsive HAase release to facilitate deep tumor penetration. Furthermore, ultrasound (US) irradiation decreased the expression of glycolysis-related proteins through the modulation of the β-catenin/c-Myc pathway, and US irradiation induced mitochondrial damage, leading to a low-energy state in tumor cells. On this basis, GHB NPs was paired with US stimulation to provide a combination therapy that disturbed tumor cell metabolic symbiosis and remodeled the immunosuppressive tumor microenvironment. This study formulates an effective therapeutic approach for metabolic-immunotherapy, potentially offering a viable candidate for tumor treatment.
Collapse
Affiliation(s)
- Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yuanyuan Yang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Department of Ultrasound, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong 266035, China.
| |
Collapse
|
4
|
Fabiano AR, Newman MW, Dombroski JA, Rowland SJ, Knoblauch SV, Kusunose J, Gibson‐Corley KN, Kaufman BG, Ren L, Caskey CF, King MR. Applying Ultrasound to Mechanically and Noninvasively Sensitize Prostate Tumors to TRAIL-Mediated Apoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412995. [PMID: 39976192 PMCID: PMC12005757 DOI: 10.1002/advs.202412995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Non-surgical and safe prostate cancer (PCa) therapies are in demand. Soluble tumor necrosis factor (TNF-α) related apoptosis inducing ligand (TRAIL), a cancer-specific drug, shows preclinical efficacy but has a short circulation half-life. This research has shown that physiological fluid shear stress activates mechanosensitive ion channels (MSCs), such as Piezo1, enhancing TRAIL-mediated apoptosis in cancer cells. Herein, noninvasive, focal ultrasound (FUS) is implemented to augment the pro-apoptotic effects of TRAIL. Using thermally safe FUS parameters, it is observed that TRAIL sensitivity increases with higher FUS pressure in PCa cells, mediated by Piezo1. This is confirmed by examining the effects of calcium chelation, MSC inhibitors, and PIEZO knockdown. In vivo, a multi-dose study with 10 min FUS exposure shows that 0 and 4-h intervals between TRAIL and FUS significantly reduce tumor burden, with an increase in apoptosis evident by enhanced cleaved-caspase 3 expression. This mechanotherapy offers a clinically translatable approach by utilizing widely available FUS technology, applicable to treat additional cancer types.
Collapse
Affiliation(s)
- Abigail R. Fabiano
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Malachy W. Newman
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Jenna A. Dombroski
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Schyler J. Rowland
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | | | - Jiro Kusunose
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTN37235USA
| | - Katherine N. Gibson‐Corley
- Department of PathologyMicrobiology and ImmunologyDivision of Comparative MedicineVanderbilt University Medical CenterNashvilleTN37235USA
| | | | - Liqin Ren
- Department of BioengineeringRice UniversityHoustonTX77005USA
| | - Charles F. Caskey
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTN37235USA
- Department of Radiology and Radiological SciencesVanderbilt UniversityNashvilleTN37235USA
| | - Michael R. King
- Department of BioengineeringRice UniversityHoustonTX77005USA
| |
Collapse
|
5
|
Doodmani SM, Safari MH, Akbari M, Farahani N, Alimohammadi M, Aref AR, Tajik F, Maghsoodlou A, Daneshi S, Tabari T, Taheriazam A, Entezari M, Nabavi N, Hashemi M. Metastasis and chemoresistance in breast cancer: Crucial function of ZEB1/2 proteins. Pathol Res Pract 2025; 267:155838. [PMID: 39954369 DOI: 10.1016/j.prp.2025.155838] [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: 10/14/2024] [Revised: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Breast cancer remains one of the leading causes of mortality worldwide. While advancements in chemotherapy, immunotherapy, radiotherapy, and targeted therapies have significantly improved breast cancer treatment, many patients are diagnosed at advanced stages, where tumor cells exhibit aggressive behavior and therapy resistance. Understanding the mechanisms driving breast cancer progression is therefore critical. Metastasis is a major factor that drastically reduces patient prognosis and survival, accounting for most breast cancer-related deaths. ZEB proteins have emerged as key regulators of cancer metastasis. Beyond their role in metastasis, ZEB proteins also influence drug resistance. This review focuses on the role of ZEB1 and ZEB2 in regulating breast cancer metastasis. These proteins interact with components of the tumor microenvironment (TME) to drive cancer progression and metastasis. Additionally, ZEB proteins regulate angiogenesis through interactions with VEGF. Targeting ZEB proteins offers potential therapeutic benefits, particularly for aggressive breast cancer subtypes such as triple-negative breast cancer (TNBC), which often show poor therapeutic response. ZEB proteins also influence the sensitivity of breast cancer cells to chemotherapy, making them promising targets for enhancing treatment efficacy. Given their upregulation in breast cancer, ZEB proteins can serve as valuable diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Seyed Mohammad Doodmani
- Department of Pathobiology, Faculty of Specialized Veterinary Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences,Tehran, Iran
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Fatemeh Tajik
- Department of Surgery, University of California, Irvine Medical Center, Orange, CA, USA
| | - Amin Maghsoodlou
- Young Researchers and Elite Club, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Teimour Tabari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
6
|
Wu Y, Liu Y, Wu H, Tong M, Du L, Ren S, Che Y. Advances in Ultrasound-Targeted Microbubble Destruction (UTMD) for Breast Cancer Therapy. Int J Nanomedicine 2025; 20:1425-1442. [PMID: 39925678 PMCID: PMC11804227 DOI: 10.2147/ijn.s504363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
Breast cancer is one of the most common types of cancer in women worldwide and is a leading cause of cancer deaths among women. As a result, various treatments have been developed to combat this disease. Breast cancer treatment varies based on its stage and type of pathology. Among the therapeutic options, ultrasound has been employed to assist in the treatment of breast cancer, including radiation therapy, chemotherapy, targeted immunotherapy, hormonal therapy, and, more recently, radiofrequency ablation for early-stage and inoperable patients. One notable advancement is ultrasound-targeted microbubble destruction (UTMD), which is gradually becoming a highly effective and non-invasive anti-tumor modality. This technique can enhance chemical, genetic, immune, and anti-vascular therapies through its physical and biological effects. Specifically, UTMD improves drug transfer efficiency and destroys tumor neovascularization while reducing toxic side effects on the body during tumor treatment. Given these developments, the application of ultrasound-assisted therapy to breast cancer has gained significant attention from research scholars. In this review, we will discuss the development of various therapeutic modalities for breast cancer and, importantly, highlight the application of ultrasound microbubble-targeted disruption techniques in breast cancer treatment.
Collapse
Affiliation(s)
- Yunfeng Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Yuxi Liu
- Department of Ultrasound, Shandong Second Medical University Affiliated Hospital, Shan Dong, Weifang, People’s Republic of China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Mengying Tong
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Linyao Du
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| |
Collapse
|
7
|
Huang B, Li X. Mechanisms of GPM6A in Malignant Tumors. Cancer Rep (Hoboken) 2025; 8:e70137. [PMID: 39957375 PMCID: PMC11831008 DOI: 10.1002/cnr2.70137] [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: 06/28/2024] [Revised: 09/20/2024] [Accepted: 01/25/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Glycoprotein M6A (GPM6A) encodes a transmembrane protein, expressing in large quantities on the cell surface of central nervous system (CNS) neurons. GPM6A acts importantly in neurodevelopment by modulating neuronal differentiation, migration, axon growth, synaptogenesis, and spine formation, but its role in malignancy remains controversial and requires further research. This article reviewed the mechanisms of GPM6A in colorectal cancer, liver cancer, lung cancer, glioblastoma, and other malignant tumors, and made a "one-stop" summary of the relevant mechanisms. RECENT FINDINGS Researches have indicated that GPM6A is related to malignant tumors. It affects epithelial-mesenchymal transition and induces the formation of filopodia, participating in the adhesion, migration, and metastasis of cancer cells. Its role in malignant tumors remains controversial, however. On the one hand, GPM6A may have carcinogenic properties and is related to poor prognosis of malignant tumors. It is highly expressed in lymphoblastic leukemia and is a potential oncogene. It also shows carcinogenic properties in colorectal cancer, glioblastoma, gonadotroph adenomas and so on. On the other hand, the expression of GPM6A decreases in lung adenocarcinoma, liver cancer, thyroid cancer, and so forth as the tumor progresses, and it can inhibit the progression of malignant tumors by inhibiting some signaling pathways, suggesting that it may be a tumor suppressor gene. CONCLUSION Carcinogenic or tumor suppressive? Although the biological function of GPM6A in the development of malignant tumors is still unclear, according to the current research progress, it is still expected to become an effective molecular marker for predicting tumor occurrence, metastasis and prognosis, as well as a new target for diagnosis and treatment.
Collapse
Affiliation(s)
- Bei Huang
- Operation Management and Evaluation Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
- Emergency Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
8
|
Xing P, Chen Z, Zhu W, Lin B, Quan M. NRF3 suppresses the malignant progression of TNBC by promoting M1 polarization of macrophages via ROS/HMGB1 axis. Cancer Biol Ther 2024; 25:2416221. [PMID: 39443820 PMCID: PMC11509002 DOI: 10.1080/15384047.2024.2416221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer. Due to its lack of targeted therapy options, TNBC remains a significant clinical challenge. In this study, we investigated the role of nuclear respiratory factor 3 (NRF3) and high-mobility group box 1 (HMGB1) in the progression of TNBC. METHODS The study analyzed NRF3's clinical expression, differentially expressed genes (DEGs), and immune infiltration in TNBC using the TCGA database and bioinformatics tools. Cellular functions of MDA-MB-468 and Hs578t cells were evaluated through MTT, colony formation, transwell, flow cytometry, and western blotting. The regulatory function of NRF3 in TNBC cell lines was assessed using Immunofluorescence, Immunohistochemistry, qRT-PCR, CHIP, luciferase assay, and ELISA. Moreover, a xenograft model was established to investigate the role of NRF3 in TNBC in vivo. RESULTS Low expression of NRF3 in TNBC tumors was associated with unfavorable prognosis and transcripts from tumors with higher NRF3 levels were enriched in oxidative stress and immune-related pathways. The subsequent gain- and loss-functional experiments indicated that NRF3 overexpression significantly suppressed malignant phenotypes, MAPK/ERK signaling pathways, and epithelial-mesenchymal transition (EMT), whereas it promoted reactive oxygen species (ROS) levels in TNBC. Further mechanistic exploration showed that NRF3 inhibited TNBC cell function by regulating oxidative stress-related genes to inhibit the MAPK/ERK signaling pathway by promoting the release of HMGB1 via ROS, thereby promoting M1 macrophage polarization. CONCLUSION NRF3 promotes M1 macrophage polarization through the ROS/HMGB1 axis, thereby inhibiting the malignant progression of TNBC. It is expected to become a therapeutic biomarker for TNBC.
Collapse
Affiliation(s)
- Ping Xing
- Department of Surgical Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- Department of Surgical Oncology, Enze Hospital, Taizhou Enze Medical Center, Taizhou, Zhejiang, China
| | - Zhenzhen Chen
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Wenbo Zhu
- Department of Surgical Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
- Department of Surgical Oncology, Enze Hospital, Taizhou Enze Medical Center, Taizhou, Zhejiang, China
| | - Bangyi Lin
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Mingming Quan
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
9
|
Du C, Guo X, Qiu X, Jiang W, Wang X, An H, Wang J, Luo Y, Du Q, Wang R, Cheng C, Guo Y, Teng H, Ran H, Wang Z, Li P, Zhou Z, Ren J. Self-Reinforced Bimetallic Mito-Jammer for Ca 2+ Overload-Mediated Cascade Mitochondrial Damage for Cancer Cuproptosis Sensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306031. [PMID: 38342617 PMCID: PMC11022715 DOI: 10.1002/advs.202306031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/21/2024] [Indexed: 02/13/2024]
Abstract
Overproduction of reactive oxygen species (ROS), metal ion accumulation, and tricarboxylic acid cycle collapse are crucial factors in mitochondria-mediated cell death. However, the highly adaptive nature and damage-repair capabilities of malignant tumors strongly limit the efficacy of treatments based on a single treatment mode. To address this challenge, a self-reinforced bimetallic Mito-Jammer is developed by incorporating doxorubicin (DOX) and calcium peroxide (CaO2) into hyaluronic acid (HA) -modified metal-organic frameworks (MOF). After cellular, Mito-Jammer dissociates into CaO2 and Cu2+ in the tumor microenvironment. The exposed CaO2 further yields hydrogen peroxide (H2O2) and Ca2+ in a weakly acidic environment to strengthen the Cu2+-based Fenton-like reaction. Furthermore, the combination of chemodynamic therapy and Ca2+ overload exacerbates ROS storms and mitochondrial damage, resulting in the downregulation of intracellular adenosine triphosphate (ATP) levels and blocking of Cu-ATPase to sensitize cuproptosis. This multilevel interaction strategy also activates robust immunogenic cell death and suppresses tumor metastasis simultaneously. This study presents a multivariate model for revolutionizing mitochondria damage, relying on the continuous retention of bimetallic ions to boost cuproptosis/immunotherapy in cancer.
Collapse
Affiliation(s)
- Chier Du
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xun Guo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaoling Qiu
- Department of Intensive Care Unitthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Weixi Jiang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaoting Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hongjin An
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Jingxue Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Yuanli Luo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Qianying Du
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Ruoyao Wang
- Department of Breast and Thyroid SurgerySecond Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Chen Cheng
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Yuan Guo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hua Teng
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Haitao Ran
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Zhigang Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Pan Li
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Zhiyi Zhou
- Department of General PracticeChongqing General HospitalChongqing400010P. R. China
| | - Jianli Ren
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| |
Collapse
|
10
|
Liu Y, Niu R, Deng R, Wang Y, Song S, Zhang H. Multi-Enzyme Co-Expressed Nanomedicine for Anti-Metastasis Tumor Therapy by Up-Regulating Cellular Oxidative Stress and Depleting Cholesterol. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307752. [PMID: 37734072 DOI: 10.1002/adma.202307752] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Tumor cells movement and migration are inseparable from the integrity of lipid rafts and the formation of lamellipodia, and lipid rafts are also a prerequisite for the formation of lamellipodia. Therefore, destroying the lipid rafts is an effective strategy to inhibit tumor metastasis. Herein, a multi-enzyme co-expressed nanomedicine: cholesterol oxidase (CHO) loaded Co─PN3 single-atom nanozyme (Co─PN3 SA/CHO) that can up-regulate cellular oxidative stress, disrupt the integrity of lipid rafts, and inhibit lamellipodia formation to induce anti-metastasis tumor therapy, is developed. In this process, Co─PN3 SA can catalyze oxygen (O2 ) and hydrogen peroxide (H2 O2 ) to generate reactive oxygen species (ROS) via oxidase-like and Fenton-like properties. The doping of P atoms optimizes the adsorption process of the intermediate at the active site and enhances the ROS generation properties of nanomedicine. Meantime, O2 produced by catalase-like catalysis can combine with excess cholesterol to generate more H2 O2 under CHO catalysis, achieving enhanced oxidative damage to tumor cells. Most importantly, cholesterol depletion in tumor cells also disrupts the integrity of lipid rafts and inhibits the formation of lamellipodia, greatly inhibiting the proliferation and metastasis of tumor cells. This strategy by up-regulating cellular oxidative stress and depleting cellular cholesterol constructs a new idea for anti-metastasis-oriented cancer therapy strategies.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Rui Niu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Ruiping Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Department of Chemistry, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
11
|
Hashemi M, Paskeh MDA, Orouei S, Abbasi P, Khorrami R, Dehghanpour A, Esmaeili N, Ghahremanzade A, Zandieh MA, Peymani M, Salimimoghadam S, Rashidi M, Taheriazam A, Entezari M, Hushmandi K. Towards dual function of autophagy in breast cancer: A potent regulator of tumor progression and therapy response. Biomed Pharmacother 2023; 161:114546. [PMID: 36958191 DOI: 10.1016/j.biopha.2023.114546] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023] Open
Abstract
As a devastating disease, breast cancer has been responsible for decrease in life expectancy of females and its morbidity and mortality are high. Breast cancer is the most common tumor in females and its treatment has been based on employment of surgical resection, chemotherapy and radiotherapy. The changes in biological behavior of breast tumor relies on genomic and epigenetic mutations and depletions as well as dysregulation of molecular mechanisms that autophagy is among them. Autophagy function can be oncogenic in increasing tumorigenesis, and when it has pro-death function, it causes reduction in viability of tumor cells. The carcinogenic function of autophagy in breast tumor is an impediment towards effective therapy of patients, as it can cause drug resistance and radio-resistance. The important hallmarks of breast tumor such as glucose metabolism, proliferation, apoptosis and metastasis can be regulated by autophagy. Oncogenic autophagy can inhibit apoptosis, while it promotes stemness of breast tumor. Moreover, autophagy demonstrates interaction with tumor microenvironment components such as macrophages and its level can be regulated by anti-tumor compounds in breast tumor therapy. The reasons of considering autophagy in breast cancer therapy is its pleiotropic function, dual role (pro-survival and pro-death) and crosstalk with important molecular mechanisms such as apoptosis. Moreover, current review provides a pre-clinical and clinical evaluation of autophagy in breast tumor.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sima Orouei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pegah Abbasi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esmaeili
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azin Ghahremanzade
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari 4815733971, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
12
|
Li W, Jin Q, Zhang L, He S, Song Y, Xu L, Deng C, Wang L, Qin X, Xie M. Ultrasonic Microbubble Cavitation Deliver Gal-3 shRNA to Inhibit Myocardial Fibrosis after Myocardial Infarction. Pharmaceutics 2023; 15:pharmaceutics15030729. [PMID: 36986588 PMCID: PMC10051524 DOI: 10.3390/pharmaceutics15030729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Galectin-3 (Gal-3) participates in myocardial fibrosis (MF) in a variety of ways. Inhibiting the expression of Gal-3 can effectively interfere with MF. This study aimed to explore the value of Gal-3 short hairpin RNA (shRNA) transfection mediated by ultrasound-targeted microbubble destruction (UTMD) in anti-myocardial fibrosis and its mechanism. A rat model of myocardial infarction (MI) was established and randomly divided into control and Gal-3 shRNA/cationic microbubbles + ultrasound (Gal-3 shRNA/CMBs + US) groups. Echocardiography measured the left ventricular ejection fraction (LVEF) weekly, and the heart was harvested to analyze fibrosis, Gal-3, and collagen expression. LVEF in the Gal-3 shRNA/CMB + US group was improved compared with the control group. On day 21, the myocardial Gal-3 expression decreased in the Gal-3 shRNA/CMBs + US group. Furthermore, the proportion of the myocardial fibrosis area in the Gal-3 shRNA/CMBs + US group was 6.9 ± 0.41% lower than in the control group. After inhibition of Gal-3, there was a downregulation in collagen production (collagen I and III), and the ratio of Col I/Col III decreased. In conclusion, UTMD-mediated Gal-3 shRNA transfection can effectively silence the expression of Gal-3 in myocardial tissue, reduce myocardial fibrosis, and protect the cardiac ejection function.
Collapse
Affiliation(s)
- Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Shukun He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yishu Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lingling Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lufang Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Correspondence: (X.Q.); (M.X.); Tel.: +86-136-0710-8938 (M.X.); Fax: +86-27-85726386 (M.X.)
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Correspondence: (X.Q.); (M.X.); Tel.: +86-136-0710-8938 (M.X.); Fax: +86-27-85726386 (M.X.)
| |
Collapse
|
13
|
Xiao S, Guo L, Ai C, Shang M, Shi D, Meng D, Sun X, Wang X, Liu R, Zhao Y, Li J. pH-/Redox-Responsive Nanodroplet Combined with Ultrasound-Targeted Microbubble Destruction for the Targeted Treatment of Drug-Resistant Triple Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8958-8973. [PMID: 36757913 DOI: 10.1021/acsami.2c20478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Multiple drug resistance (MDR) exists in divergent cancers including triple negative breast cancer (TNBC) and partly results in the resistance to many first-line anti-cancer agents, bringing a big challenge to TNBC management. To develop novel TNBC therapeutics, in our study, a hyaluronic acid (HA)-carboxymethyl chitosan (CMC) conjugate linked via a disulfide-bond (HA-SS-CMC, HSC) was synthesized to fabricate nanodroplets (NDs). The NDs encapsulating doxorubicin (DOX) and perfluorohexane (DOX-HSC-NDs) were prepared via a homogenization/emulsification strategy and exhibited not only high biocompatibility but also noticeable tumor cell targeting ability and dual pH/redox responsiveness. Besides, DOX-HSC-NDs can be used as a contrast-enhanced ultrasound imaging agent for specific tumor imaging. DOX-HSC-NDs in combination with ultrasound targeted microbubble destruction could improve intracellular drug aggregation and retention of MDR cells and work against multiple mechanisms of drug resistance through synergistic strategies, including up-regulating the reactive oxygen species (ROS) level, promoting apoptosis, and scavenging glutathione, while reducing the expression levels of P-glycoprotein and inhibiting the epithelial-mesenchymal transition. This combination strategy showed protective effects against TNBC in both MDA-MB-231/ADR cells and tumor-bearing mice. Our study for the first time developed and reported the ultrasound-augmented HSC-NDs as the DOX nanocarrier and provided scientific evidence to support the future application of DOX-HSC-NDs as a potential TNBC therapy.
Collapse
Affiliation(s)
- Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chen Ai
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
14
|
Wu Y, Deng C, Xu J, Wang W, Chen Y, Qin X, Lv Q, Xie M. Enhanced Local Delivery of microRNA-145a-5P into Mouse Aorta via Ultrasound-Targeted Microbubble Destruction Inhibits Atherosclerotic Plaque Formation. Mol Pharm 2023; 20:1086-1095. [PMID: 36656656 DOI: 10.1021/acs.molpharmaceut.2c00799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) play a key role in the formation and rupture of atherosclerotic plaques. Previous studies have confirmed that microRNA-145 (miR-145) is involved in the phenotypic regulation of VSMCs and reduction of atherosclerosis. At present, seeking safe and effective gene delivery remains a key problem restricting the development of gene therapy. In recent years, ultrasound-targeted microbubble destruction (UTMD) has become a safe and effective transfection method that is widely used in the basic research of gene therapy for heart and tumor diseases. Here, we synthesized cationic microbubbles to encapsulate miR-145 and targeted their release into VSMCs in vitro and in vivo using ultrasound. The feasibility of this gene therapy was verified by fluorescence microscopy and an in vivo imaging system. The results showed that treatment with miR-145 delivered via UTMD considerably improved the gene transfection efficiency and promoted the contraction phenotype of VSMCs in vitro. In vivo, this treatment reduced the atherosclerotic plaque area by 48.04% compared with treatment with free miR-145. Therefore, UTMD-mediated miRNA therapy may provide a new targeted therapeutic approach for atherosclerotic plaques.
Collapse
Affiliation(s)
- Yu Wu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jia Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wei Wang
- Department of Ultrasound, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
15
|
Liu R, Shi D, Guo L, Xiao S, Shang M, Sun X, Meng D, Zhao Y, Wang X, Li J. Ultrasound-Targeted Microbubble Disruption with Key Nanodroplets for Effective Ferroptosis in Triple-Negative Breast Cancer Using Animal Model. Int J Nanomedicine 2023; 18:2037-2052. [PMID: 37155504 PMCID: PMC10122866 DOI: 10.2147/ijn.s400495] [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: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is known to be the most aggressive form of breast cancer. Due to its high recurrence and mortality rates, the treatment of TNBC is a significant challenge for the medical community. Besides, ferroptosis is an emerging regulatory cell death that may provide new insights into the treatment of TNBC. As a central inhibitor of the ferroptosis process, the selenoenzyme glutathione peroxidase 4 (GPX4) is its classical therapeutic target. However, inhibition of GPX4 expression is quite detrimental to normal tissues. Ultrasound contrast agents, as an emerging visualization precision treatment, may provide a solution to the existing problem. Methods In this study, nanodroplets (NDs) carrying simvastatin (SIM) were constructed using the homogeneous/emulsification method. Then, the characterization of SIM-NDs was systematically evaluated. Meanwhile, in this study, the ability of SIM-NDs combined with ultrasound-targeted microbubble disruption (UTMD) to initiate ferroptosis and its respective mechanisms of ferroptosis induction were verified. Finally, the antitumor activity of SIM-NDs was investigated in vitro and in vivo using MDA-MB-231 cells and TNBC animal models. Results SIM-NDs exhibited excellent pH- and ultrasound-responsive drug release and noticeable ultrasonographic imaging ability, also showing good biocompatibility and biosafety. UTMD could promote increased intracellular reactive oxygen species and consume intracellular glutathione. However, SIM-NDs were efficiently internalized into cells under ultrasound irradiation, followed by the rapid release of SIM, which inhibited intracellular mevalonate production, and synergistically downregulated GPX4 expression, thereby promoting ferroptosis. Moreover, this combined treatment demonstrated strong antitumor ability in vitro and in vivo. Conclusion The combination of UTMD and SIM-NDs presents a promising avenue for harnessing ferroptosis in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Correspondence: Jie Li, Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China, Email
| |
Collapse
|
16
|
circ-ZEB1 regulates epithelial-mesenchymal transition and chemotherapy resistance of colorectal cancer through acting on miR-200c-5p. Transl Oncol 2022; 28:101604. [PMID: 36542990 PMCID: PMC9792398 DOI: 10.1016/j.tranon.2022.101604] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/27/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Circular RNAs (circRNAs) have been demonstrated to be important regulators in human malignant tumors, including colorectal cancer (CRC). While the role circ-ZEB1 played in CRC remains unclear. In this study, we aim to explore the biological function and the underlying mechanism of circ-ZEB1 in CRC. RNAscope was used to analyze the expression and localization of circ-ZEB1 in CRC tissues. Loss of function experiments were conducted, including CCK-8, transwell assays, flow cytometry analysis, and murine xenograft models, so as to detect the effect of circ-ZEB1 on CRC cells. IC50 assay was used to evaluate the influence of circ-ZEB1 on the chemoresistance of CRC cells. Epithelial-mesenchymal transition (EMT) related markers were detected. The relationship between circ-ZEB1 and miR-200c-5p was investigated by FISH, dual-luciferase reporter assay, and RIP assay. We found in our study that circ-ZEB1 was significantly upregulated in CRC tissues. Downregulation of circ-ZEB1 inhibited cell proliferation, colony formation, as well as cell migration and invasion abilities of CRC cell lines. In vivo experiments indicated that knockdown of circ-ZEB1 suppressed tumorigenesis and distant metastasis of CRC cells in nude mice. What's more, EMT and chemoresistance of CRC cells were also attenuated following circ-ZEB1 knockdown. Mechanistically, we proved that circ-ZEB1 could directly bind with miR-200c and functioned as miR-200c sponge to exert its biological functions in CRC cells. In conclusion, circ-ZEB1 could promote CRC cells progression, EMT, and chemoresistance via acting on miR-200c, elucidating a potential therapeutic target to inhibit CRC progression.
Collapse
|
17
|
Dong L, Li N, Wei X, Wang Y, Chang L, Wu H, Song L, Guo K, Chang Y, Yin Y, Pan M, Shen Y, Wang F. A Gambogic Acid-Loaded Delivery System Mediated by Ultrasound-Targeted Microbubble Destruction: A Promising Therapy Method for Malignant Cerebral Glioma. Int J Nanomedicine 2022; 17:2001-2017. [PMID: 35535034 PMCID: PMC9078874 DOI: 10.2147/ijn.s344940] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Background Purpose Methods Results Conclusion
Collapse
Affiliation(s)
- Lei Dong
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Xixi Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Yongling Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Liansheng Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Hongwei Wu
- Department of Chemistry, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Liujiang Song
- Department of Ophthalmology, Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Kang Guo
- Department of Oncology, The Third affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Yuqiao Chang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Min Pan
- Department of Ultrasound, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518034, People’s Republic of China
- Min Pan, Department of Ultrasound, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, No. 6001 Beihuan Avenue, Shenzhen, 518034, People’s Republic of China, Email
| | - Yuanyuan Shen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, People’s Republic of China
| | - Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
- Correspondence: Feng Wang, Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453002, People’s Republic of China, Email
| |
Collapse
|
18
|
Chen X, Zhang X, Qian Y, Xia E, Wang Y, Zhou Q. Ultrasound-targeted microbubble destruction-mediated miR-144-5p overexpression enhances the anti-tumor effect of paclitaxel on thyroid carcinoma by targeting STON2. Cell Cycle 2022; 21:1058-1076. [PMID: 35184686 PMCID: PMC9037415 DOI: 10.1080/15384101.2022.2040778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The effects of miR-144-5p and paclitaxel (PTX) on thyroid carcinoma were less explored. Thus, we investigated the effects of miR-144-5p and PTX on thyroid carcinoma. The expression and target gene of miR-144-5p in thyroid carcinoma were analyzed by bioinformatics, y qRT-PCR and dual-luciferase reporter assay. After the transfection mediated by ultrasound-targeted microbubble destruction (UTMD) or liposome, or the treatment of PTX, the viability, proliferation, migration, and invasion of thyroid carcinoma cells were detected by MTT, colony formation, wound-healing, and transwell assays. The expressions of miR-144-5p, STON2, MMP-9, E-cadherin, and N-cadherin in cells were calculated via qRT-PCR or Western blotting. After a subcutaneous-xenotransplant tumor model was established using BALB/c nude mice and further treated with PTX and UTMD-mediated miR-144-5p, the volume, weight, and Ki67 level of tumor were recorded or evaluated by immunohistochemical assays. MiR-144-5p, which was low-expressed in thyroid carcinoma, directly down-regulated STON2 level. MiR-144-5p overexpression and PTX inhibited the viability, proliferation, migration, and invasion of thyroid carcinoma cells, while miR-144-5p silencing caused the opposite results. MiR-144-5p overexpression and PTX further up-regulated E-cadherin level and down-regulated those of MMP-9 and N-cadherin in thyroid carcinoma cells. STON2 overexpression reversed the effects of miR-144-5p overexpression.. MiR-144-5p overexpression enhanced the inhibiting effect of PTX on tumor volume, weight, and Ki67 level of xenotransplant tumor, and the effects of UTMD-mediated miR-144-5p overexpression were stronger than those mediated by liposome. Collectively, UTMD-mediated miR-144-5p overexpression enhanced the anti-tumor effect of PTX on thyroid carcinoma by targeting STON2.
Collapse
Affiliation(s)
- Xuefeng Chen
- Ultrasound Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an City, China,Ultrasound Department Yancheng No. 1 People’s Hospital, Yancheng City, China
| | - Xinyuan Zhang
- Ultrasound Department Yancheng No. 1 People’s Hospital, Yancheng City, China
| | - Yangyang Qian
- General Surgery Department, Yancheng No. 1 People’s Hospital, Yancheng City, China
| | - Enhui Xia
- Ultrasound Department Yancheng No. 1 People’s Hospital, Yancheng City, China
| | - Yu Wang
- Emergency Department, Yancheng No. 1 People’s Hospital, Yancheng City, China
| | - Qi Zhou
- Ultrasound Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an City, China,CONTACT Qi Zhou Ultrasound Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Siwu Road, Xincheng Distrcit, Xi’an City, Shaanxi Province710004, China
| |
Collapse
|
19
|
Fang F, Xu W, Zhang J, Gu J, Yang G. Ultrasound microbubble-mediated RNA interference targeting WNT1 inducible signaling pathway protein 1(WISP1) suppresses the proliferation and metastasis of breast cancer cells. Bioengineered 2022; 13:11050-11060. [PMID: 35481425 PMCID: PMC9208516 DOI: 10.1080/21655979.2022.2068738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
In the context of relatively sufficient research that annotated WNT1 inducible signaling pathway protein 1 (WISP1) as a promoting factor in tumor progression of breast cancer, and identified the effects of ultrasound microbubble technology on enhancing the transfection efficiency and achieving better gene interference, this study managed to investigate the effects of ultrasound microbubble-mediated siWISP1 transfection on proliferation and metastasis of breast cancer cells. To achieve our research objectives, the expression of WISP1 in breast cancer tissues was retrieved from GEPIA website, and the viability of breast cancer cells (SK-BR-3 and MCF7) was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for ultrasound intensity screening. After the transfection of siWISP1 by ultrasound microbubble or lipofectamine 6000, the content of WISP1 secreted by cells was detected through Enzyme-linked immunosorbent assay (ELISA), and WISP1 expression in cells was determined by quantitative reverse transcription polymerase-chain reaction (qRT-PCR). Besides, the cell invasion, migration, and proliferation were evaluated by wound healing, transwell, and EdU assays, respectively. In accordance with experimental results, WISP1 was highly expressed in breast cancer tissues, and the 1 W/cm2 intensity was the onset of a notable decrease in cell viability. Compared with lipofectamine 6000 transfection, the transfection of siWISP1 mediated by ultrasound microbubble further reduced the expression of WISP1, and meanwhile suppressed cell invasion, migration, and proliferation. Collectively, ultrasound microbubble-mediated transfection of siWISP1 worked rather effectively in improving transfection efficiency and inhibiting the progression of breast cancer.
Collapse
Affiliation(s)
- Faying Fang
- Department of Special Examination, Maternal and Child Health Hospital of Chun'an County, Hangzhou, Zhejiang, China
| | - Weizhi Xu
- Department of Ultrasound, Sanmen People's Hospital, Taizhou, Zhejiang, China
| | - Jian Zhang
- Department of Ultrasound, Pingyi County Hospital of Traditional Chinese Medicine, Linyi, Shandong, China
| | - Jin Gu
- Department of Ultrasound, Chongqing Public Health Medical Center, Chongqing, Shandong, China
| | - Gaoyi Yang
- Department of Ultrasound, Sanmen People's Hospital, Taizhou, Zhejiang, China
| |
Collapse
|
20
|
Entezari M, Sadrkhanloo M, Rashidi M, Asnaf SE, Taheriazam A, Hashemi M, Ashrafizadeh M, Zarrabi A, Rabiee N, Hushmandi K, Mirzaei S, Sethi G. Non-coding RNAs and macrophage interaction in tumor progression. Crit Rev Oncol Hematol 2022; 173:103680. [PMID: 35405273 DOI: 10.1016/j.critrevonc.2022.103680] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/25/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
The macrophages are abundantly found in TME and their M2 polarization is in favor of tumor malignancy. On the other hand, non-coding RNAs (ncRNAs) can modulate macrophage polarization in TME to affect cancer progression. The miRNAs can dually induce/suppress M2 polarization of macrophages and by affecting various molecular pathways, they modulate tumor progression and therapy response. The lncRNAs can affect miRNAs via sponging and other molecular pathways to modulate macrophage polarization. A few experiments have also examined role of circRNAs in targeting signaling networks and affecting macrophages. The therapeutic targeting of these ncRNAs can mediate TME remodeling and affect macrophage polarization. Furthermore, exosomal ncRNAs derived from tumor cells or macrophages can modulate polarization and TME remodeling. Suppressing biogenesis and secretion of exosomes can inhibit ncRNA-mediated M2 polarization of macrophages and prevent tumor progression. The ncRNAs, especially exosomal ncRNAs can be considered as non-invasive biomarkers for tumor diagnosis.
Collapse
Affiliation(s)
- Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sholeh Etehad Asnaf
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
21
|
Huang L, Ding L, Yu S, Huang X, Ren Q. Propofol postconditioning alleviates diabetic myocardial ischemia‑reperfusion injury via the miR‑200c‑3p/AdipoR2/STAT3 signaling pathway. Mol Med Rep 2022; 25:137. [PMID: 35211763 PMCID: PMC8908333 DOI: 10.3892/mmr.2022.12653] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 08/20/2021] [Indexed: 11/23/2022] Open
Abstract
Myocardial ischemia/reperfusion (MI/RI) syndrome is one of the leading causes of mortality and disability. Propofol postconditioning is known to improve myocardial ischemia/reperfusion injury (MI/RI). The present study aimed to explore the mechanism of propofol postconditioning in diabetic MI/RI. Diabetic MI/RI rat models were established and the rats were treated via propofol postconditioning. Staining with 2,3,5-triphenyl-2H-tetrazolium chloride, H&E staining, TUNEL staining and ELISA were applied to detect infarct size, pathological changes, apoptosis and oxidative stress-related factor and apoptotic factor levels, respectively. Subsequently, the effect of propofol on H9C2 cells was also assessed using the Cell Counting Kit-8 assay. High-glucose hypoxia/reperfusion (H/R) models of H9C2 cardiomyocytes were established. miR-200c-3p overexpression or AdipoR2 silencing combined with propofol postconditioning was performed in H/R-induced H9C2 cells and STAT3 protein expression levels were determined. Propofol postconditioning significantly reduced myocardial infarct size, oxidative stress and apoptosis in diabetic MI/RI models. Furthermore, propofol postconditioning significantly reduced the oxidative stress and apoptosis of H9C2 cells in high-glucose H/R models. Propofol postconditioning also significantly downregulated miR-200c-3p expression levels and promoted AdipoR2 expression levels. miR-200c-3p overexpression or AdipoR2 downregulation significantly reversed the effects of propofol postconditioning on its antioxidation and anti-apoptotic effects in H9C2 cells and on decreasing STAT3 phosphorylation levels. Together, the results of the present study demonstrated that propofol postconditioning inhibited miR-200c-3p, upregulated AdipoR2 and activated the STAT3 signaling pathway, thus alleviating diabetic MI/RI and therefore highlighting its potential as a treatment of diabetic MI/RI.
Collapse
Affiliation(s)
- Lijun Huang
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Li Ding
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Shenghui Yu
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xin Huang
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Qiusheng Ren
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
22
|
Tang X, Hao N, Zhou Y, Liu Y. Ultrasound targeted microbubble destruction-mediated SOCS3 attenuates biological characteristics and epithelial-mesenchymal transition (EMT) of breast cancer stem cells. Bioengineered 2022; 13:3896-3910. [PMID: 35109743 PMCID: PMC8973955 DOI: 10.1080/21655979.2022.2031384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
SOCS3 is low-expressed in breast cancer and may be a potential target. Ultrasound targeted microbubble destruction (UTMD) improved the efficiency of gene transfection. We explored the effects of UTMD-mediated transfection of SOCS3 on the biological characteristics and epithelial-mesenchymal transition (EMT) of breast cancer stem cells (BCSCs). The expression of SOCS3 in breast cancer (BC) and its association with prognosis were evaluated by GEPIA and The Cancer Genome Atlas (TCGA) websites. BCSCs were sorted by flow cytometry and immunomagnetic bead method, followed by sphere formation, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and xenograft assays to test their effects in vitro and in vivo. The levels of SOCS3, EMT- and STAT3 pathway-related genes were determined by RT-qPCR and Western blot, respectively. The effects of liposome and UTMD on BCSCs and mice were compared by the gain-of-function experiments. Low expression of SOCS3 was associated with poor prognosis of BC patients, and found in BC and BCSCs. BCSCs were successfully sorted, with high viability and tumorigenicity. UTMD increased the transfection rate of SOCS3. Moreover, UTMD- and liposome-mediated SOCS3 reduced cell viability, proliferation, migration and invasion, blocked cell cycle, inhibited sphere formation in BCSCs, and retarded tumor growth in mice. Mechanistically, overexpressed SOCS3 inhibited the expressions of EMT-related genes and the activation of STAT3 pathway in BCSCs and mice. The regulatory effects of UTMD-mediated SOCS3 on the above-mentioned biological characteristics were better than liposome-mediated SOCS3. UTMD-mediated SOCS3 has a better therapeutic effect in BC, providing new experimental evidence for the treatment of BC.
Collapse
Affiliation(s)
- Xiaojiang Tang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Na Hao
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuhui Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Liu
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Han X, Long Y, Duan X, Liu Z, Hu X, Zhou J, Li N, Wang Y, Qin J. ZEB1 induces ROS generation through directly promoting MCT4 transcription to facilitate breast cancer. Exp Cell Res 2022; 412:113044. [DOI: 10.1016/j.yexcr.2022.113044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/31/2021] [Accepted: 01/22/2022] [Indexed: 12/17/2022]
|
24
|
Williams MM, Christenson JL, O'Neill KI, Hafeez SA, Ihle CL, Spoelstra NS, Slansky JE, Richer JK. MicroRNA-200c restoration reveals a cytokine profile to enhance M1 macrophage polarization in breast cancer. NPJ Breast Cancer 2021; 7:64. [PMID: 34045467 PMCID: PMC8160264 DOI: 10.1038/s41523-021-00273-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/03/2021] [Indexed: 11/21/2022] Open
Abstract
Many immune suppressive mechanisms utilized by triple negative breast cancer (TNBC) are regulated by oncogenic epithelial-to-mesenchymal transition (EMT). How TNBC EMT impacts innate immune cells is not fully understood. To determine how TNBC suppresses antitumor macrophages, we used microRNA-200c (miR-200c), a powerful repressor of EMT, to drive mesenchymal-like mouse mammary carcinoma and human TNBC cells toward a more epithelial state. MiR-200c restoration significantly decreased growth of mouse mammary carcinoma Met-1 cells in culture and in vivo. Cytokine profiling of Met-1 and human BT549 cells revealed that miR-200c upregulated cytokines, such as granulocyte-macrophage colony-stimulating factor (GM-CSF), promoted M1 antitumor macrophage polarization. Cytokines upregulated by miR-200c correlated with an epithelial gene signature and M1 macrophage polarization in BC patients and predicted a more favorable overall survival for TNBC patients. Our findings demonstrate that immunogenic cytokines (e.g., GM-CSF) are suppressed in aggressive TNBC, warranting further investigation of cytokine-based therapies to limit disease recurrence.
Collapse
Affiliation(s)
- Michelle M Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sabrina A Hafeez
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire L Ihle
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill E Slansky
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
25
|
Cai J, Cui Y, Yang J, Wang S. Epithelial-mesenchymal transition: When tumor cells meet myeloid-derived suppressor cells. Biochim Biophys Acta Rev Cancer 2021; 1876:188564. [PMID: 33974950 DOI: 10.1016/j.bbcan.2021.188564] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous myeloid cell population characterized by protumoral functions in the tumor immune network. An increasing number of studies have focused on the biological functions of MDSCs in tumor immunity. Epithelial-mesenchymal transition (EMT) is a cellular plasticity process accompanied by a loss of epithelial phenotypes and an acquisition of mesenchymal phenotypes. In general, tumor cells that undergo EMT are more likely to invade and metastasize. Recently, extensive evidence suggests that EMT is closely related to a highly immunosuppressive environment. This review will summarize the immunosuppressive capacities of MDSC subsets and their distinct role in tumor EMT and further discuss immunotherapy for tumor EMT by targeting MDSCs.
Collapse
Affiliation(s)
- Jingshan Cai
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yudan Cui
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jun Yang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
26
|
Chong ZX, Yeap SK, Ho WY. Transfection types, methods and strategies: a technical review. PeerJ 2021; 9:e11165. [PMID: 33976969 PMCID: PMC8067914 DOI: 10.7717/peerj.11165] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
Transfection is a modern and powerful method used to insert foreign nucleic acids into eukaryotic cells. The ability to modify host cells' genetic content enables the broad application of this process in studying normal cellular processes, disease molecular mechanism and gene therapeutic effect. In this review, we summarized and compared the findings from various reported literature on the characteristics, strengths, and limitations of various transfection methods, type of transfected nucleic acids, transfection controls and approaches to assess transfection efficiency. With the vast choices of approaches available, we hope that this review will help researchers, especially those new to the field, in their decision making over the transfection protocol or strategy appropriate for their experimental aims.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Wan Yong Ho
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Selangor, Malaysia
| |
Collapse
|
27
|
Hussen BM, Shoorei H, Mohaqiq M, Dinger ME, Hidayat HJ, Taheri M, Ghafouri-Fard S. The Impact of Non-coding RNAs in the Epithelial to Mesenchymal Transition. Front Mol Biosci 2021; 8:665199. [PMID: 33842553 PMCID: PMC8033041 DOI: 10.3389/fmolb.2021.665199] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a course of action that enables a polarized epithelial cell to undertake numerous biochemical alterations that allow it to adopt features of mesenchymal cells such as high migratory ability, invasive properties, resistance to apoptosis, and importantly higher-order formation of extracellular matrix elements. EMT has important roles in implantation and gastrulation of the embryo, inflammatory reactions and fibrosis, and transformation of cancer cells, their invasiveness and metastatic ability. Regarding the importance of EMT in the invasive progression of cancer, this process has been well studies in in this context. Non-coding RNAs (ncRNAs) have been shown to exert critical function in the regulation of cellular processes that are involved in the EMT. These processes include regulation of some transcription factors namely SNAI1 and SNAI2, ZEB1 and ZEB2, Twist, and E12/E47, modulation of chromatin configuration, alternative splicing, and protein stability and subcellular location of proteins. In the present paper, we describe the influence of ncRNAs including microRNAs and long non-coding RNAs in the EMT process and their application as biomarkers for this process and cancer progression and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Pharmacognosy Department, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdi Mohaqiq
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Marcel E. Dinger
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Tian P, Wang Y, Du W. Ultrasound-targeted microbubble destruction enhances the anti-tumor action of miR-4284 inhibitor in non-small cell lung cancer cells. Exp Ther Med 2021; 21:551. [PMID: 33850523 PMCID: PMC8027739 DOI: 10.3892/etm.2021.9983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are known to be involved in various human cancer types. Ultrasound-targeted microbubble destruction (UTMD) may improve the transfection efficiency of exogenous genes into target tissues and organs, thereby improving cancer treatment. In the present study, the role of miR-4284 in non-small cell lung cancer (NSCLC) was investigated and the effect of UTMD-mediated inhibition of miR-4284 on tumor progression was further analyzed. The expression of miR-4284 in NSCLC cells and tissues was detected by reverse transcription-quantitative PCR. UTMD-mediated inhibition of miR-4284 was achieved by co-transfection of microvesicles and miR-4284 inhibitors into NSCLC cells. A Cell Counting Kit-8 assay was used to determine NSCLC cell proliferation, and the migration and invasion of NSCLC cells were examined by Transwell assays. Compared with that in the control group, the expression of miR-4284 was increased in NSCLC tissues and cells. Knockdown of miR-4284 in NSCLC cells inhibited cell proliferation, migration and invasion. UTMD improved the transfection efficiency of miR-4284 inhibitors in NSCLC cells, resulting in more significant inhibition of tumor cell proliferation, migration and invasion. In conclusion, the results indicated that the expression of miR-4284 was increased in clinical samples and cell lines of NSCLC and that knockdown of miR-4284 inhibited the proliferation, migration and invasion of tumor cells. UTMD-mediated miR-4284 inhibition further promoted this effect, indicating that this technique may represent a novel strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Peng Tian
- Department of Ultrasonics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Yanzhen Wang
- Department of Ultrasonics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Wenyan Du
- Department of Science and Education, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
29
|
Advances in Understanding Mitochondrial MicroRNAs (mitomiRs) on the Pathogenesis of Triple-Negative Breast Cancer (TNBC). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5517777. [PMID: 33824695 PMCID: PMC8007369 DOI: 10.1155/2021/5517777] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022]
Abstract
Triple-negative breast cancer (TNBC) is characterized by poor outcome and the most challenging breast cancer type to treat worldwide. TNBC manifests distinct profile of mitochondrial functions, which dictates reprogrammed metabolism, fosters tumor progression, and notably serves as therapeutic targets. Mitochondrial microRNAs (mitomiRs) are a group of microRNAs that critically modulate mitochondrial homeostasis. By a pathway-centric manner, mitomiRs tightly orchestrate metabolic reprogramming, redox status, cell apoptosis, mitochondrial dynamics, mitophagy, mitochondrial DNA (mtDNA) maintenance, and calcium balance, leading to an emerging field of study in various cancer types, including TNBC. We herein review the recent insights into the roles and mechanism of mitomiRs in TNBC and highlight its clinical value in diagnosis and prognosis as well as vital advances on therapeutics of preclinical and clinical studies.
Collapse
|
30
|
Ashrafizadeh M, Hushmandi K, Rahmani Moghadam E, Zarrin V, Hosseinzadeh Kashani S, Bokaie S, Najafi M, Tavakol S, Mohammadinejad R, Nabavi N, Hsieh CL, Zarepour A, Zare EN, Zarrabi A, Makvandi P. Progress in Delivery of siRNA-Based Therapeutics Employing Nano-Vehicles for Treatment of Prostate Cancer. Bioengineering (Basel) 2020; 7:E91. [PMID: 32784981 PMCID: PMC7552721 DOI: 10.3390/bioengineering7030091] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa) accounts for a high number of deaths in males with no available curative treatments. Patients with PCa are commonly diagnosed in advanced stages due to the lack of symptoms in the early stages. Recently, the research focus was directed toward gene editing in cancer therapy. Small interfering RNA (siRNA) intervention is considered as a powerful tool for gene silencing (knockdown), enabling the suppression of oncogene factors in cancer. This strategy is applied to the treatment of various cancers including PCa. The siRNA can inhibit proliferation and invasion of PCa cells and is able to promote the anti-tumor activity of chemotherapeutic agents. However, the off-target effects of siRNA therapy remarkably reduce its efficacy in PCa therapy. To date, various carriers were designed to improve the delivery of siRNA and, among them, nanoparticles are of importance. Nanoparticles enable the targeted delivery of siRNAs and enhance their potential in the downregulation of target genes of interest. Additionally, nanoparticles can provide a platform for the co-delivery of siRNAs and anti-tumor drugs, resulting in decreased growth and migration of PCa cells. The efficacy, specificity, and delivery of siRNAs are comprehensively discussed in this review to direct further studies toward using siRNAs and their nanoscale-delivery systems in PCa therapy and perhaps other cancer types.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran; (K.H.); (S.B.)
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
| | | | - Saied Bokaie
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran; (K.H.); (S.B.)
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran;
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614525, Iran;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kermaan 55425147, Iran;
| | - Noushin Nabavi
- Research Services, University of Victoria, Victoria, BC V8W 2Y2, Canada;
| | - Chia-Ling Hsieh
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei City 110, Taiwan;
| | - Atefeh Zarepour
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 8174673441, Iran;
| | | | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
- Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 61537-53843, Iran
| |
Collapse
|
31
|
Dissecting miRNA facilitated physiology and function in human breast cancer for therapeutic intervention. Semin Cancer Biol 2020; 72:46-64. [PMID: 32497683 DOI: 10.1016/j.semcancer.2020.05.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/17/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are key epigenomic regulators of biological processes in animals and plants. These small non coding RNAs form a complex networks that regulate cellular function and development. MiRNAs prevent translation by either inactivation or inducing degradation of mRNA, a major concern in post-transcriptional gene regulation. Aberrant regulation of gene expression by miRNAs is frequently observed in cancer. Overexpression of various 'oncomiRs' and silencing of tumor suppressor miRNAs are associated with various types of human cancers, although overall downregulation of miRNA expression is reported as a hallmark of cancer. Modulations of the total pool of cellular miRNA by alteration in genetic and epigenetic factors associated with the biogenesis of miRNA machinery. It also depends on the availability of cellular miRNAs from its store in the organelles which affect tumor development and cancer progression. Here, we have dissected the roles and pathways of various miRNAs during normal cellular and molecular functions as well as during breast cancer progression. Recent research works and prevailing views implicate that there are two major types of miRNAs; (i) intracellular miRNAs and (ii) extracellular miRNAs. Concept, that the functions of intracellular miRNAs are driven by cellular organelles in mammalian cells. Extracellular miRNAs function in cell-cell communication in extracellular spaces and distance cells through circulation. A detailed understanding of organelle driven miRNA function and the precise role of extracellular miRNAs, pre- and post-therapeutic implications of miRNAs in this scenario would open several avenues for further understanding of miRNA function and can be better exploited for the treatment of breast cancers.
Collapse
|