1
|
Shi Y, Shao Q, Ren Z, Shang G, Han J, Cheng J, Zheng Y, Cheng F, Li C, Wang Q, Wang X. Mechanisms of pulmonary fibrosis and lung cancer induced by chronic PM 2.5 exposure: Focus on the airway epithelial barrier and epithelial-mesenchymal transition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118253. [PMID: 40311473 DOI: 10.1016/j.ecoenv.2025.118253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/03/2025]
Abstract
This study aims to provide new insights into PM2.5-induced lung diseases through a focus on the pulmonary epithelial barrier and epithelial-mesenchymal transition (EMT). Firstly, we analyzed the mechanisms by which PM2.5 damages the airway epithelial barrier, including inflammatory responses, immune imbalance, oxidative stress, apoptosis, and autophagy. Subsequently, we investigated the mechanisms by which PM2.5 induces EMT, which involve the synergistic effect of oxidative stress and inflammation, the activation of key signaling pathways, and the regulatory role of non-coding RNAs. Furthermore, we explored the interaction between the airway epithelial barrier and EMT, especially the induction of EMT by epithelial barrier damage and the impact of EMT on epithelial barrier repair. Regarding lung injury diseases, we focused on the roles of the epithelial barrier and EMT in the development of pulmonary fibrosis and lung cancer, providing evidence from in vitro and in vivo studies. Emphasizing the translational prospects from basic research to clinical applications, and we proposed new ideas for treating PM2.5-related lung diseases from four aspects-anti-inflammatory and antioxidant drugs, signaling pathway inhibitors, non-coding RNA-targeted therapies, and gene editing and cell therapies-by focusing on the two key links of the airway epithelial barrier and EMT.
Collapse
Affiliation(s)
- Yuyu Shi
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Shao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zilin Ren
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guojiao Shang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinhua Han
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jialin Cheng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuxiao Zheng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fafeng Cheng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Changxiang Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Qingguo Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Xueqian Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, 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
|
Hosseini H, Heydari S, Hushmandi K, Daneshi S, Raesi R. Bone tumors: a systematic review of prevalence, risk determinants, and survival patterns. BMC Cancer 2025; 25:321. [PMID: 39984867 PMCID: PMC11846205 DOI: 10.1186/s12885-025-13720-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Though relatively rare, bone tumors significantly impact patient health and treatment outcomes. OBJECTIVE This systematic review analyzes the incidence, types, survival rates, and risk factors associated with bone tumors, including both benign and malignant forms. METHODS This systematic review was conducted using the keywords "bone tumors," "epidemiology," "benign bone tumors," "malignant bone tumors," "osteosarcoma," "Ewing sarcoma," "chondrosarcoma," "risk factors," and "survival" in electronic databases including PubMed, Scopus, Web of Science, and Google Scholar from 2000 to 2024. The search strategy was based on the PRISMA statement. Finally, 9 articles were selected for inclusion in the study. RESULTS The systematic review highlights that primary bone tumors can be classified into benign and malignant types, with osteosarcoma being the most prevalent malignant form, particularly among adolescents and young adults. The epidemiology of bone tumors is influenced by factors such as age, gender, geographic location, and genetic predispositions. Recent advancements in imaging techniques have improved the detection of these tumors, contributing to an increasing recognition of their prevalence. Data shows that the limited-duration prevalence of malignant bone tumors has increased significantly. This increase is from 0.00069% in 2000 to 0.00749% in 2018, indicating an increasing recognition and diagnosis of these rare tumors over time. Survival rates vary significantly by tumor type, with approximately 50-60% for osteosarcoma and around 70% for Ewing's sarcoma, though these rates decrease with metastasis. Key risk factors identified include genetic predispositions such as Li-Fraumeni syndrome and TP53 mutations, environmental exposures like radiation, and growth patterns related to height. CONCLUSION The review highlights the importance of early diagnosis and treatment intervention, as survival rates are significantly better for patients with localized disease compared to those with metastatic conditions. The observed variations in survival rates across different tumor types underscore the need for tailored treatment strategies. Key risk factors include genetic predispositions and environmental exposures, highlighting the need for targeted screening and ongoing research to enhance diagnostic accuracy and treatment strategies.
Collapse
Affiliation(s)
- Hasan Hosseini
- Department of Orthopedics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Heydari
- School of Medicine, Imam Khomeini Hospital, Jiroft University of Medical Science, Jiroft, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Rasoul Raesi
- Department of Public Health, School of Health, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran.
- Department of Health Services Management, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Jalali P, Yaghoobi A, Rezaee M, Zabihi MR, Piroozkhah M, Aliyari S, Salehi Z. Decoding common genetic alterations between Barrett's esophagus and esophageal adenocarcinoma: A bioinformatics analysis. Heliyon 2024; 10:e31194. [PMID: 38803922 PMCID: PMC11128929 DOI: 10.1016/j.heliyon.2024.e31194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Background Esophageal adenocarcinoma (EAC) is a common cancer with a poor prognosis in advanced stages. Therefore, early EAC diagnosis and treatment have gained attention in recent decades. It has been found that various pathological changes, particularly Barrett's Esophagus (BE), can occur in the esophageal tissue before the development of EAC. In this study, we aimed to identify the molecular contributor in BE to EAC progression by detecting the essential regulatory genes that are differentially expressed in both BE and EAC. Materials and methods We conducted a comprehensive bioinformatics analysis to detect BE and EAC-associated genes. The common differentially expressed genes (DEGs) and common single nucleotide polymorphisms (SNPs) were detected using the GEO and DisGeNET databases, respectively. Then, hub genes and the top modules within the protein-protein interaction network were identified. Moreover, the co-expression network of the top module by the HIPPIE database was constructed. Additionally, the gene regulatory network was constructed based on miRNAs and circRNAs. Lastly, we inspected the DGIdb database for possible interacted drugs. Results Our microarray dataset analysis identified 92 common DEGs between BE and EAC with significant enrichment in skin and epidermis development genes. The study also identified 22 common SNPs between BE and EAC. The top module of PPI network analysis included SCEL, KRT6A, SPRR1A, SPRR1B, SPRR3, PPL, SPRR2B, EVPL, and CSTA. We constructed a ceRNA network involving three specific mRNAs, 23 miRNAs, and 101 selected circRNAs. According to the results from the DGIdb database, TD101 was found to interact with the KRT6A gene. Conclusion The present study provides novel potential candidate genes that may be involved in the molecular association between Esophageal adenocarcinoma and Barrett's Esophagus, resulting in developing the diagnostic tools and therapeutic targets to prevent progression of BE to EAC.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Yaghoobi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zabihi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Aliyari
- Division of Applied Bioinformatics, German Cancer Research Center DKFZ Heidelberg, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Wang D, Chen S, Shao Y, Deng Y, Huang L. EIF4A3 modulated circ_000999 promotes epithelial-mesenchymal transition in cadmium-induced malignant transformation through the miR-205-5p/ZEB1 axis. ENVIRONMENT INTERNATIONAL 2024; 186:108656. [PMID: 38621321 DOI: 10.1016/j.envint.2024.108656] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/07/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
Cadmium (Cd) is an accumulative toxic metal which poses a serious threat to human health, even in trace amounts. One of the most important steps in the pathophysiology of lung cancer (LC) is the epithelial-mesenchymal transition (EMT). In this investigation, a cell malignant transformation model was established by exposing human bronchial epithelial cells (16HBE) to a low dose of Cd for 30 weeks, after which a highly expressed circular RNA (circ_000999) was identified. Cd-induced EMT was clearly observed in rat lungs and 16HBE cells, which was further enhanced following circ_000999-overexpression. Furthermore, upregulated EIF4A3 interacted with the parental gene AGTPBP1 to promote high expression of circ_000999. Subsequent experiments confirmed that circ_000999 could regulate the EMT process by competitively binding miR-205-5p and inhibiting its activity, consequently upregulating expression of zinc finger E-box binding protein 1 (ZEB1). Importantly, the circ_000999 expression level in LC tissues was significantly increased, exhibiting a strong correlation with EMT indicators. Overall, these findings provide a new objective and research direction for reversing lung EMT and subsequent treatment and prevention of LC.
Collapse
Affiliation(s)
- Donglei Wang
- School of Public Health, Baotou Medical College, Baotou 014030, Inner Mongolia, China
| | - Shijie Chen
- School of Public Health, Baotou Medical College, Baotou 014030, Inner Mongolia, China
| | - Yueqing Shao
- School of Public Health, Baotou Medical College, Baotou 014030, Inner Mongolia, China
| | - Yang Deng
- School of Public Health, Baotou Medical College, Baotou 014030, Inner Mongolia, China
| | - Lihua Huang
- School of Public Health, Baotou Medical College, Baotou 014030, Inner Mongolia, China.
| |
Collapse
|
6
|
Dehghanzad M, Mohammadi M, Nejati M, Pouremamali F, Maroufi NF, Akbarzadeh M, Samadi N, Nouri M. The potential therapeutic effect of melatonin in oxaliplatin combination therapy against chemoresistant colorectal cancer cells. Mol Biol Rep 2024; 51:348. [PMID: 38401018 DOI: 10.1007/s11033-024-09316-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 02/02/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Oxaliplatin is one of the main therapeutics in colorectal cancer (CRC) chemotherapy. However, in light of multidrug resistance (MDR) phenotype development, the efficacy of oxaliplatin has decreased. This study aimed to assess the potential therapeutic effect of melatonin in oxaliplatin combination therapy for drug-resistant colorectal cancer cells. METHODS AND RESULTS Initially, the oxaliplatin-resistant cell line was created of LS174T (LS174T/DR) by using the oxaliplatin IC50 concentration and resting cycles. MTT assays and flow cytometry were applied for assessing cell viability and apoptotic cells. The mRNA expression level of Bax, Bcl2, MT1, MT2, and ABCB1 as well as protein levels of ABCB1, Bcl2, BAX were measured by the qRT-PCR and western blot techniques respectively. P-gp activity was assessed by Rho123 staining. The IC50 concentration of oxaliplatin in resistant cells was increased from 500.7 ± 0.2 nM to 7119 ± 0.1 nM. Bcl2, MT1, MT2, and ABCB1 mRNA plus protein expression levels of Bcl2 and ABCB1 were significantly reduced in resistant cells, along with a marked increase in Bax mRNA and protein levels compared to parental cells. Rho 123 staining revealed a marked reduction in P-gp activities in the combination-treated group compared to the oxaliplatin-treated group. CONCLUSIONS The results of cytotoxicity assays, MTT, and flow cytometry revealed that the combination of melatonin and oxaliplatin exerts synergistic effects on induction of oxaliplatin's cytotoxicity in CRC. Our research suggests that combining the treatments of melatonin and oxaliplatin may be considered as a new approach to overcoming oxaliplatin resistance in CRC patients.
Collapse
Affiliation(s)
- Masoumeh Dehghanzad
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Gholgasht Ave, Tabriz, Iran
| | - Mohammad Mohammadi
- Department of Medical Laboratory Science, Faculty of Medicine, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Mohaddeseh Nejati
- Department of Biology, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Farhad Pouremamali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Gholgasht Ave, Tabriz, Iran
- Department of Human Genetics, McGill University, Montreal, Canada
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Canada
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Naser Samadi
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Gholgasht Ave, Tabriz, Iran
| | - Mohammad Nouri
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Gholgasht Ave, Tabriz, Iran.
| |
Collapse
|
7
|
Mirabedini S, Musavi H, Makhlough A, Hashemi-Sooteh MB, Zargari M. Association of S19W polymorphism in APOA5 gene and serum lipid levels in patients with type 2 diabetic nephropathy. Horm Mol Biol Clin Investig 2023; 44:243-249. [PMID: 36855913 DOI: 10.1515/hmbci-2022-0056] [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: 06/02/2022] [Accepted: 02/11/2023] [Indexed: 03/02/2023]
Abstract
OBJECTIVES Type 2 diabetic Mellitus (T2DM) is the most common systemic and endocrine disease in humans, and diabetic nephropathy is one of the most serious complications of this disorder. The polymorphisms in the apolipoprotein A5 (ApoA5) gene are strongly related to hypertriglyceridemia and are considered a predisposing factor for diabetic nephropathy. The current study proposed to examine the association of APOA5-S19W polymorphism with serum lipids levels in patients with type 2 diabetic nephropathy in Mazandaran province. METHODS This case-control study was designed to determine the association of APOA5-S19W polymorphism with plasma lipid profile in 161 T2DM patients with nephropathy (DN+), without nephropathy (DN-), and in 58 healthy individuals. Lipid profile values were measured using Pars Azmoun commercial kits. S19W variant, one of the polymorphisms of the APOA5 gene, was determined by PCR-restriction fragment length polymorphism (PCR-RFLP) and Taq1 restriction enzyme. RESULTS In comparison between the three groups, DN+ had a higher mean TG than DN- and the control group (p<0.001). The incidence of the G allele in DN+ was not significant compared to groups of DN-. Comparing the relationship between the mean of biochemical variables with CC and CG genotypes showed that the mean level of TG in people with CC genotype was increased compared to people with CG genotype in diabetic patients. However, this increase was not significant (p=0.19). CONCLUSIONS There was no association between SNP APOA5 S19W and serum lipids in diabetic patients with and without nephropathy.
Collapse
Affiliation(s)
- Shivasadat Mirabedini
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hadis Musavi
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Atieh Makhlough
- Department of Internal Medicine, Diabetes Research Center, Sari Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad-Bagher Hashemi-Sooteh
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
8
|
Hashemi M, Zandieh MA, Talebi Y, Rahmanian P, Shafiee SS, Nejad MM, Babaei R, Sadi FH, Rajabi R, Abkenar ZO, Rezaei S, Ren J, Nabavi N, Khorrami R, Rashidi M, Hushmandi K, Entezari M, Taheriazam A. Paclitaxel and docetaxel resistance in prostate cancer: Molecular mechanisms and possible therapeutic strategies. Biomed Pharmacother 2023; 160:114392. [PMID: 36804123 DOI: 10.1016/j.biopha.2023.114392] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Prostate cancer is among most malignant tumors around the world and this urological tumor can be developed as result of genomic mutations and their accumulation during progression towards advanced stage. Due to lack of specific symptoms in early stages of prostate cancer, most cancer patients are diagnosed in advanced stages that tumor cells display low response to chemotherapy. Furthermore, genomic mutations in prostate cancer enhance the aggressiveness of tumor cells. Docetaxel and paclitaxel are suggested as well-known compounds for chemotherapy of prostate tumor and they possess a similar function in cancer therapy that is based on inhibiting depolymerization of microtubules, impairing balance of microtubules and subsequent delay in cell cycle progression. The aim of current review is to highlight mechanisms of paclitaxel and docetaxel resistance in prostate cancer. When oncogenic factors such as CD133 display upregulation and PTEN as tumor-suppressor shows decrease in expression, malignancy of prostate tumor cells enhances and they can induce drug resistance. Furthermore, phytochemicals as anti-tumor compounds have been utilized in suppressing chemoresistance in prostate cancer. Naringenin and lovastatin are among the anti-tumor compounds that have been used for impairing progression of prostate tumor and enhancing drug sensitivity. Moreover, nanostructures such as polymeric micelles and nanobubbles have been utilized in delivery of anti-tumor compounds and decreasing risk of chemoresistance development. These subjects are highlighted in current review to provide new insight for reversing drug resistance in prostate cancer.
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
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Yasmin Talebi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sareh Sadat Shafiee
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Melina Maghsodlou Nejad
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Roghayeh Babaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Farzaneh Hasani Sadi
- General Practitioner, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Shamin Rezaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, 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.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, 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.
| | - 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.
| |
Collapse
|
9
|
Hashemi M, Rashidi M, Hushmandi K, Ten Hagen TLM, Salimimoghadam S, Taheriazam A, Entezari M, Falahati M. HMGA2 regulation by miRNAs in cancer: affecting cancer hallmarks and therapy response. Pharmacol Res 2023; 190:106732. [PMID: 36931542 DOI: 10.1016/j.phrs.2023.106732] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
High mobility group A 2 (HMGA2) is a protein that modulates the structure of chromatin in the nucleus. Importantly, aberrant expression of HMGA2 occurs during carcinogenesis, and this protein is an upstream mediator of cancer hallmarks including evasion of apoptosis, proliferation, invasion, metastasis, and therapy resistance. HMGA2 targets critical signaling pathways such as Wnt/β-catenin and mTOR in cancer cells. Therefore, suppression of HMGA2 function notably decreases cancer progression and improves outcome in patients. As HMGA2 is mainly oncogenic, targeting expression by non-coding RNAs (ncRNAs) is crucial to take into consideration since it affects HMGA2 function. MicroRNAs (miRNAs) belong to ncRNAs and are master regulators of vital cell processes, which affect all aspects of cancer hallmarks. Long ncRNAs (lncRNAs) and circular RNAs (circRNAs), other members of ncRNAs, are upstream mediators of miRNAs. The current review intends to discuss the importance of the miRNA/HMGA2 axis in modulation of various types of cancer, and mentions lncRNAs and circRNAs, which regulate this axis as upstream mediators. Finally, we discuss the effect of miRNAs and HMGA2 interactions on the response of cancer cells to therapy. Regarding the critical role of HMGA2 in regulation of critical signaling pathways in cancer cells, and considering the confirmed interaction between HMGA2 and one of the master regulators of cancer, miRNAs, targeting miRNA/HMGA2 axis in cancer therapy is promising and this could be the subject of future clinical trial experiments.
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.
| | - 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.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, 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.
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
10
|
Bakhshandeh N, Mohammadi M, Mohammadi P, Nazari E, Damchi M, Khodabandelu S, Mokhtari H. Increased expression of androgen receptor and PSA genes in LNCaP (prostate cancer) cell line due to high concentrations of EGCG, an active ingredient in green tea. Horm Mol Biol Clin Investig 2022:hmbci-2022-0054. [PMID: 36578191 DOI: 10.1515/hmbci-2022-0054] [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: 06/02/2022] [Accepted: 12/11/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Androgen receptor (AR) play a key role in the onset and progression of prostate cancer. Epigallocatechin-3-gallate (EGCG) is a polyphenolic compound and the active ingredient in green tea, which is involved in modulating gene expression through epigenetic alterations. Previous studies have shown that EGCG at low concentrations reduces the expression of AR and prostate-specific antigen (PSA) in the LNCaP cell line of prostate cancer. In this study, the effect of higher EGCG concentrations on AR and PSA expression in LNCaP prostate cancer cell line was investigated. METHODS In this study, LNCaP prostate cancer cell line was used and after MTT test, concentrations of 40, 60 and 80 μg/mL EGCG were used for treatment. Then, the expression of AR and PSA genes was evaluated by RT-PCR. AR protein expression was also assessed by Western blotting. RESULTS The present study showed that treatment of LNCaPs cells by EGCG reduces cell proliferation. The IC50 value was 42.7 μg/mL under experimental conditions. It was also observed that EGCG at concentrations of 40 and 80 μg/mL increased the expression of AR and PSA (p<0.05). CONCLUSIONS The present study showed that the effect of EGCG on AR expression was different at different concentrations, so that unlike previous studies, higher concentrations of EGCG (80 and 40 μg/mL) increased AR and PSA expression. It seems that due to the toxic effects of EGCG in high concentrations on cancer cells and the possibility of its effect on normal cells, more caution should be exercised in its use.
Collapse
Affiliation(s)
- Nadereh Bakhshandeh
- Department of Medical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Mohammadi
- Health System Research, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Parisa Mohammadi
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Elahe Nazari
- Department of Biology, Islamic Azad University, Gorgan Branch, Gorgan, Iran
| | - Mehdi Damchi
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sajad Khodabandelu
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hossein Mokhtari
- Amol Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
11
|
Zhang XP, Pei JP, Zhang CD, Yusupu M, Han MH, Dai DQ. Exosomal circRNAs: A key factor of tumor angiogenesis and therapeutic intervention. Biomed Pharmacother 2022; 156:113921. [DOI: 10.1016/j.biopha.2022.113921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 11/02/2022] Open
|