1
|
Akhlaghipour I, Moghbeli M. Matrix metalloproteinases as the critical regulators of cisplatin response and tumor cell invasion. Eur J Pharmacol 2024; 982:176966. [PMID: 39216742 DOI: 10.1016/j.ejphar.2024.176966] [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: 06/30/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Cisplatin (CDDP) as one of the most common first-line chemotherapy drugs plays a vital role in the treatment of a wide range of malignant tumors. Nevertheless, CDDP resistance is observed as a therapeutic challenge in a large number of cancer patients. Considering the CDDP side effects in normal tissues, predicting the CDDP response of cancer patients can significantly help to choose the appropriate therapeutic strategy. In this regard, investigating the molecular mechanisms involved in CDDP resistance can lead to the introduction of prognostic markers in cancer patients. Matrix metalloproteinases (MMPs) have critical roles in tissue remodeling and cell migration through extracellular matrix degradation. Therefore, defects in MMPs functions can be associated with tumor metastasis and chemo resistance. In the present review, we discussed the role of MMPs in CDDP response and tumor cell invasion. PubMed, Scopus, Google Scholar, and Web of Science were searched using "MMP", "cisplatin", and "cancer" keywords for data retrieval that was limited to Apr 20, 2024. It has been reported that MMPs can increase CDDP resistance in tumor cells as the effectors of PI3K/AKT, MAPK, and NF-κB signaling pathways or independently through the regulation of structural proteins, autophagy, and epithelial-to-mesenchymal transition (EMT) process. This review has an effective role in introducing MMPs as the prognostic markers and therapeutic targets in CDDP-resistant cancer patients.
Collapse
Affiliation(s)
- Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Chen H, Gong Z, Zhou H, Han Y. Deciphering chemoresistance in osteosarcoma: Unveiling regulatory mechanisms and function through the lens of noncoding RNA. Drug Dev Res 2024; 85:e22167. [PMID: 38444106 DOI: 10.1002/ddr.22167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/24/2024] [Accepted: 02/18/2024] [Indexed: 03/07/2024]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor and is prevalent in children, adolescents, and elderly individuals. It has the characteristics of high invasion and metastasis. Neoadjuvant chemotherapy combined with surgical resection is the most commonly used treatment for OS. However, the efficacy of OS is considerably diminished by chemotherapy resistance. In recent years, noncoding RNAs (ncRNAs), including microRNAs, long noncoding RNAs, and circular RNAs, are hot topics in the field of chemotherapy resistance research. Several studies have demonstrated that ncRNAs are substantially associated with chemoresistance in OS. Thus, the present study overviews the abnormally expressed ncRNAs in OS and the molecular mechanisms involved in chemoresistance, with an emphasis on their function in promoting or inhibiting chemoresistance. ncRNAs are expected to become potential therapeutic targets for overcoming drug resistance and predictive biomarkers in OS, which are of great significance for enhancing the therapeutic effect and improving the prognosis.
Collapse
Affiliation(s)
- Hefen Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhujun Gong
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Hong Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Han
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Yin JY, Zhou Y, Ding XM, Gong RZ, Zhou Y, Hu HY, Liu Y, Lv XB, Zhang B. UCA1 Inhibits NKG2D-mediated Cytotoxicity of NK Cells to Breast Cancer. Curr Cancer Drug Targets 2024; 24:204-219. [PMID: 37076962 DOI: 10.2174/1568009623666230418134253] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/05/2023] [Accepted: 02/20/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND Natural killer cells play important roles in tumor immune surveillance, and cancer cells must resist this surveillance in order to progress and metastasise. INTRODUCTION The study aimed to explore the mechanism of how breast cancer cells become resistant to the cytotoxicity of NK cells. METHODS We established NK-resistant breast cancer cells by exposing MDA-MB-231 cells and MCF-7 cells to NK92 cells. Profiles of lncRNA were compared between the NK-resistant and parental cell lines. Primary NK cells were isolated by MACS, and the NK attacking effect was tested by non-radioactive cytotoxicity. The change in lncRNAs was analyzed by Gene-chip. The interaction between lncRNA and miRNA was displayed by Luciferase assay. The regulation of the gene was verified by QRT-PCR and WB. The clinical indicators were detected by ISH, IH, and ELISA, respectively. RESULTS UCA1 was found to be significantly up-regulated in both NK-resistant cell lines, and we confirmed such up-regulation on its own to be sufficient to render parental cell lines resistant to NK92 cells. We found that UCA1 up-regulated ULBP2 via the transcription factor CREB1, while it up-regulated ADAM17 by "sponging" the miR-26b-5p. ADAM17 facilitated the shedding of soluble ULBP2 from the surface of breast cancer cells, rendering them resistant to killing by NK cells. UCA1, ADAM17, and ULBP2 were found to be expressed at higher levels in bone metastases of breast cancer than in primary tumors. CONCLUSION Our data strongly suggest that UCA1 up-regulates ULBP2 expression and shedding, rendering breast cancer cells resistant to killing by NK cells.
Collapse
Affiliation(s)
- Jun-Yi Yin
- Orthopaedic Department of the Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, 445 Bayi Road, Donghu District, Nanchang, Jiangxi, 330006, China
- Oncology Department of Tongji Hospital of Tongji University, No. 389 Xincun Road, Shanghai, 200065, China
| | - Yao Zhou
- Department of Breast Surgery, the Third hospital of Nanchang, No. 2, Xiangshan Road, Xihu District, Nanchang, Jiangxi, 330009, China
| | - Xiao-Ming Ding
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Run-Ze Gong
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Yan Zhou
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Hai-Yan Hu
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Yuan Liu
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Xiao-Bin Lv
- Central Laboratory of the Third Affiliated Hospital of Nanchang University, No. 128 Xiangshan N Road, Donghu District, Nanchang, Jiangxi, 330008, China
| | - Bing Zhang
- Orthopaedic Department of the Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, 445 Bayi Road, Donghu District, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
4
|
Poddar NK, Khan A, Fatima F, Saxena A, Ghaley G, Khan S. Association of mTOR Pathway and Conformational Alterations in C-Reactive Protein in Neurodegenerative Diseases and Infections. Cell Mol Neurobiol 2023; 43:3815-3832. [PMID: 37665407 DOI: 10.1007/s10571-023-01402-z] [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: 06/02/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023]
Abstract
Inflammatory biomarkers have been very useful in detecting and monitoring inflammatory processes along with providing helpful information to select appropriate therapeutic strategies. C-reactive protein (CRP) is a nonspecific, but quite useful medical acute inflammatory biomarker and is associated with persistent chronic inflammatory processes. Several studies suggest that different levels of CRP are correlated with neurological disorders such as Alzheimer's disease (AD). However, dynamics of CRP levels have also been observed in virus/bacterial-related infections leading to inflammatory responses and this triggers mTOR-mediated pathways for neurodegeneration diseases. The biophysical structural transition from CRP to monomeric CRP (mCRP) and the significance of the ratio of CRP levels on the onset of symptoms associated with inflammatory response have been discussed. In addition, mTOR inhibitors act as immunomodulators by downregulating the expression of viral infection and can be explored as a potential therapy for neurological diseases.
Collapse
Affiliation(s)
- Nitesh Kumar Poddar
- Department of Biosciences, Manipal University Jaipur, Jaipur-Ajmer Express Highway, Dehmi Kalan, Near GVK Toll Plaza, Jaipur, Rajasthan, India, 303007.
| | - Arshma Khan
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India, 243123
| | - Falak Fatima
- Amity Institute of Biotechnology, Amity University, Uttar Pradesh, Noida, India, 201301
| | - Anshulika Saxena
- Department of Biosciences, Manipal University Jaipur, Jaipur-Ajmer Express Highway, Dehmi Kalan, Near GVK Toll Plaza, Jaipur, Rajasthan, India, 303007
| | - Garima Ghaley
- Department of Biosciences, Manipal University Jaipur, Jaipur-Ajmer Express Highway, Dehmi Kalan, Near GVK Toll Plaza, Jaipur, Rajasthan, India, 303007
| | - Shahanavaj Khan
- Department of Medical Lab Technology, Indian Institute of Health and Technology (IIHT), Deoband, Saharanpur, Uttar Pradesh, India, 247554.
| |
Collapse
|
5
|
Panwar V, Singh A, Bhatt M, Tonk RK, Azizov S, Raza AS, Sengupta S, Kumar D, Garg M. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct Target Ther 2023; 8:375. [PMID: 37779156 PMCID: PMC10543444 DOI: 10.1038/s41392-023-01608-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 10/03/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a protein kinase that controls cellular metabolism, catabolism, immune responses, autophagy, survival, proliferation, and migration, to maintain cellular homeostasis. The mTOR signaling cascade consists of two distinct multi-subunit complexes named mTOR complex 1/2 (mTORC1/2). mTOR catalyzes the phosphorylation of several critical proteins like AKT, protein kinase C, insulin growth factor receptor (IGF-1R), 4E binding protein 1 (4E-BP1), ribosomal protein S6 kinase (S6K), transcription factor EB (TFEB), sterol-responsive element-binding proteins (SREBPs), Lipin-1, and Unc-51-like autophagy-activating kinases. mTOR signaling plays a central role in regulating translation, lipid synthesis, nucleotide synthesis, biogenesis of lysosomes, nutrient sensing, and growth factor signaling. The emerging pieces of evidence have revealed that the constitutive activation of the mTOR pathway due to mutations/amplification/deletion in either mTOR and its complexes (mTORC1 and mTORC2) or upstream targets is responsible for aging, neurological diseases, and human malignancies. Here, we provide the detailed structure of mTOR, its complexes, and the comprehensive role of upstream regulators, as well as downstream effectors of mTOR signaling cascades in the metabolism, biogenesis of biomolecules, immune responses, and autophagy. Additionally, we summarize the potential of long noncoding RNAs (lncRNAs) as an important modulator of mTOR signaling. Importantly, we have highlighted the potential of mTOR signaling in aging, neurological disorders, human cancers, cancer stem cells, and drug resistance. Here, we discuss the developments for the therapeutic targeting of mTOR signaling with improved anticancer efficacy for the benefit of cancer patients in clinics.
Collapse
Affiliation(s)
- Vivek Panwar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Aishwarya Singh
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, 201313, India
| | - Manini Bhatt
- Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab, 140001, India
| | - Rajiv K Tonk
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Shavkatjon Azizov
- Laboratory of Biological Active Macromolecular Systems, Institute of Bioorganic Chemistry, Academy of Sciences Uzbekistan, Tashkent, 100125, Uzbekistan
- Faculty of Life Sciences, Pharmaceutical Technical University, 100084, Tashkent, Uzbekistan
| | - Agha Saquib Raza
- Rajive Gandhi Super Speciality Hospital, Tahirpur, New Delhi, 110093, India
| | - Shinjinee Sengupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, 201313, India.
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
6
|
Yu K, Basu A, Yau C, Wolf DM, Goodarzi H, Bandyopadhyay S, Korkola JE, Hirst GL, Asare S, DeMichele A, Hylton N, Yee D, Esserman L, van ‘t Veer L, Sirota M. Computational drug repositioning for the identification of new agents to sensitize drug-resistant breast tumors across treatments and receptor subtypes. Front Oncol 2023; 13:1192208. [PMID: 37384294 PMCID: PMC10294228 DOI: 10.3389/fonc.2023.1192208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Drug resistance is a major obstacle in cancer treatment and can involve a variety of different factors. Identifying effective therapies for drug resistant tumors is integral for improving patient outcomes. Methods In this study, we applied a computational drug repositioning approach to identify potential agents to sensitize primary drug resistant breast cancers. We extracted drug resistance profiles from the I-SPY 2 TRIAL, a neoadjuvant trial for early stage breast cancer, by comparing gene expression profiles of responder and non-responder patients stratified into treatments within HR/HER2 receptor subtypes, yielding 17 treatment-subtype pairs. We then used a rank-based pattern-matching strategy to identify compounds in the Connectivity Map, a database of cell line derived drug perturbation profiles, that can reverse these signatures in a breast cancer cell line. We hypothesize that reversing these drug resistance signatures will sensitize tumors to treatment and prolong survival. Results We found that few individual genes are shared among the drug resistance profiles of different agents. At the pathway level, however, we found enrichment of immune pathways in the responders in 8 treatments within the HR+HER2+, HR+HER2-, and HR-HER2- receptor subtypes. We also found enrichment of estrogen response pathways in the non-responders in 10 treatments primarily within the hormone receptor positive subtypes. Although most of our drug predictions are unique to treatment arms and receptor subtypes, our drug repositioning pipeline identified the estrogen receptor antagonist fulvestrant as a compound that can potentially reverse resistance across 13/17 of the treatments and receptor subtypes including HR+ and triple negative. While fulvestrant showed limited efficacy when tested in a panel of 5 paclitaxel resistant breast cancer cell lines, it did increase drug response in combination with paclitaxel in HCC-1937, a triple negative breast cancer cell line. Conclusion We applied a computational drug repurposing approach to identify potential agents to sensitize drug resistant breast cancers in the I-SPY 2 TRIAL. We identified fulvestrant as a potential drug hit and showed that it increased response in a paclitaxel-resistant triple negative breast cancer cell line, HCC-1937, when treated in combination with paclitaxel.
Collapse
Affiliation(s)
- Katharine Yu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Amrita Basu
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Denise M. Wolf
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hani Goodarzi
- University of California, San Francisco, San Francisco, CA, United States
| | | | - James E. Korkola
- Oregon Health and Science University, Portland, OR, United States
| | - Gillian L. Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Smita Asare
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- QuantumLeap Healthcare Collaborative, San Francisco, CA, United States
| | | | - Nola Hylton
- University of California, San Francisco, San Francisco, CA, United States
| | - Douglas Yee
- University of Minnesota, Minneapolis, MN, United States
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Laura van ‘t Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
7
|
Mafi A, Keshavarzmotamed A, Hedayati N, Boroujeni ZY, Reiter RJ, Dehmordi RM, Aarabi MH, Rezaee M, Asemi Z. Melatonin targeting non-coding RNAs in cancer: Focus on mechanisms and potential therapeutic targets. Eur J Pharmacol 2023; 950:175755. [PMID: 37119959 DOI: 10.1016/j.ejphar.2023.175755] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/15/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023]
Abstract
Despite, melatonin is mainly known as a regulatory factor for circadian rhythm, its notable role in other fundamental biological processes, such as redox homeostasis and programmed cell death, has been found. In this line, a growing body of evidence indicated that melatonin could apply an inhibitory effect on the tumorigenic processes. Hence, melatonin might be considered an efficient adjuvant agent for cancer treatment. Besides, the physiological and pathological functions of non-coding RNAs (ncRNAs) in various disease, particularly cancers, have been expanded over the past two decades. It is well-established that ncRNAs can modulate the gene expression at various levels, thereby, ncRNAs. can regulate the numerous biological processes, including cell proliferation, cell metabolism, apoptosis, and cell cycle. Recently, targeting the ncRNAs expression provides a novel insight in the therapeutic approaches for cancer treatment. Moreover, accumulating investigations have revealed that melatonin could impact the expression of different ncRNAs in a multiple disorders, including cancer. Therefore, in the precent study, we discuss the potential roles of melatonin in modulating the expression of ncRNAs and the related molecular pathways in different types of cancer. Also, we highlighted its importance in therapeutic application and translational medicine in cancer treatment.
Collapse
Affiliation(s)
- Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran.
| | - Zahra Yeganeh Boroujeni
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA.
| | - Rohollah Mousavi Dehmordi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mohammad-Hossein Aarabi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Malihe Rezaee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
8
|
Tian W, Niu X, Feng F, Wang X, Wang J, Yao W, Zhang P. The promising roles of exosomal microRNAs in osteosarcoma: A new insight into the clinical therapy. Biomed Pharmacother 2023; 163:114771. [PMID: 37119740 DOI: 10.1016/j.biopha.2023.114771] [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: 03/02/2023] [Revised: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023] Open
Abstract
Osteosarcoma is the most common malignant bone sarcoma in children. Chemotherapy drugs resistance significantly hinders the overall survival of patients. Due to high biocompatibility and immunocompatibility, exosomes have been explored extensively. Multiple parent cells can actively secrete numerous exosomes, and the membrane structure of exosomes can protect miRNAs from degradation. Based on these characteristics, exosomal miRNAs play an important role in the occurrence, development, drug resistance. Therefore, in-depth exploration of exosome biogenesis and role of exosomal miRNAs will provide new strategies and targets for understanding the pathogenesis of osteosarcoma and overcoming chemotherapy drug resistance. Moreover, advancing evidences have showed that engineering modification could attribute stronger targeting to exosomes to deliver cargos to recipient cells more effectively. In this review, we focus on the mechanisms of exosomal miRNAs on the occurrence and development of osteosarcoma and the potential to function as tumor biomarkers for diagnosis and prognosis prediction. In addition, we also summarize recent advances in the clinical application values of engineering exosomes to provide novel ideas and directions for overcoming the chemotherapy resistance in osteosarcoma.
Collapse
Affiliation(s)
- Wen Tian
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xiaoying Niu
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, Henan 450001, China
| | - Xin Wang
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Jiaqiang Wang
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Weitao Yao
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Peng Zhang
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| |
Collapse
|
9
|
Fu Y, Liu H, Long M, Song L, Meng Z, Lin S, Zhang Y, Qin J. Icariin attenuates the tumor growth by targeting miR-1-3p/TNKS2/Wnt/β-catenin signaling axis in ovarian cancer. Front Oncol 2022; 12:940926. [PMID: 36185280 PMCID: PMC9516086 DOI: 10.3389/fonc.2022.940926] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose Despite various therapy advances, ovarian cancer remains an incurable disease for which survival rates have only modestly improved. Natural products are important sources of anti-cancer lead compounds. Icariin exhibited broad anti-cancer efficacy. However, the mechanism of icariin against ovarian cancer is poorly elucidated. Methods Cell viability was detected to evaluate the effect of icariin on SKOV-3 cells. The cell cycle and apoptosis were analyzed. The transcript of SKOV-3 cells was profiled by RNA-seq. GSEA and DEGs analyses were performed to interpret gene expression data. Western blot and TOP/FOP flash assay were applied to detect Wnt/β-catenin signaling. MiRDB database and dual-luciferase reporter assay was applied to study the regulation of miR-1-3p on TNKS2. Anti-tumor efficacy of icariin was evaluated by xenograft mouse model. Immunohistochemistry was performed with antibodies against Ki67. Results Icariin significantly suppressed the proliferation of SKOV-3 cells. Furthermore, icariin stalled cell cycle and induced apoptosis by blocking TNKS2/Wnt/β-catenin pathway through upregulating the level of miR-1-3p. Finally, icariin dramatically suppressed tumor growth in vivo. Conclusions In this study, we demonstrated for the first time that icariin significantly attenuated the growth of ovarian tumor in xenograft mouse model. Furthermore, we systematically revealed that icariin attenuates the tumor progression by suppressing TNKS2/Wnt/β-catenin signaling via upregulating the level of miR-1-3p in ovarian cancer with transcriptome analysis.
Collapse
Affiliation(s)
- Yanjin Fu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Haiquan Liu
- Huizhou Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine, Huizhou, Guangdong, China
| | - Mengsha Long
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Linliang Song
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zuyu Meng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shaozi Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yiyao Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - JiaJia Qin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- *Correspondence: JiaJia Qin,
| |
Collapse
|
10
|
Gaulee P, Yang Z, Sura L, Xu H, Rossignol C, Weiss MD, Bliznyuk N. Concentration of Serum Biomarkers of Brain Injury in Neonates With a Low Cord pH With or Without Mild Hypoxic-Ischemic Encephalopathy. Front Neurol 2022; 13:934755. [PMID: 35873777 PMCID: PMC9301366 DOI: 10.3389/fneur.2022.934755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
Objective To determine the concentrations of four neuroprotein biomarkers and 68 miRNAs in neonates with low cord pH and/or mild hypoxic-ischemic encephalopathy (HIE). Study Design A prospective cohort study enrolled neonates with low cord pH (n = 18), moderate-severe HIE (n = 40), and healthy controls (n = 38). Groups provided serum samples at 0–6 h of life. The concentrations of biomarkers and miRNAs were compared between cohorts. Result The low cord pH and moderate-severe HIE groups had increased concentrations of GFAP, NFL and Tau compared to controls (P < 0.05, P < 0.001, respectively). NFL concentrations in mild HIE was higher than controls (P < 0.05) but less than moderate-severe HIE (P < 0.001). Of 68 miRNAs, 36 in low cord pH group and 40 in moderate-severe HIE were upregulated compared to controls (P < 0.05). Five miRNAs in low cord pH group (P < 0.05) and 3 in moderate-severe HIE were downregulated compared to controls (P < 0.05). Conclusion A biomarker panel in neonates with low cord pH may help clinicians make real-time decisions.
Collapse
Affiliation(s)
- Pratima Gaulee
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
- *Correspondence: Pratima Gaulee
| | - Zhihui Yang
- Department of Emergency Medicine, University of Florida, Gainesville, FL, United States
| | - Livia Sura
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Haiyan Xu
- Department of Emergency Medicine, University of Florida, Gainesville, FL, United States
| | - Candace Rossignol
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Michael D. Weiss
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Nikolay Bliznyuk
- Department of Agricultural and Biological Engineering, Biostatistics and Statistics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
11
|
Sritharan S, Guha S, Hazarika S, Sivalingam N. Meta analysis of bioactive compounds, miRNA, siRNA and cell death regulators as sensitizers to doxorubicin induced chemoresistance. Apoptosis 2022; 27:622-646. [PMID: 35716277 DOI: 10.1007/s10495-022-01742-z] [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] [Accepted: 05/30/2022] [Indexed: 11/02/2022]
Abstract
Cancer has presented to be the most challenging disease, contributing to one in six mortalities worldwide. The current treatment regimen involves multiple rounds of chemotherapy administration, alone or in combination. The treatment has adverse effects including cardiomyopathy, hepatotoxicity, and nephrotoxicity. In addition, the development of resistance to chemo has been attributed to cancer relapse and low patient overall survivability. Multiple drug resistance development may be through numerous factors such as up-regulation of drug transporters, drug inactivation, alteration of drug targets and drug degradation. Doxorubicin is a widely used first line chemotherapeutic drug for a myriad of cancers. It has multiple intracellular targets, DNA intercalation, adduct formation, topoisomerase inhibition, iron chelation, reactive oxygen species generation and promotes immune mediated clearance of the tumor. Agents that can sensitize the resistant cancer cells to the chemotherapeutic drug are currently the focus to improve the clinical efficiency of cancer therapy. This review summarizes the recent 10-year research on the use of natural phytochemicals, inhibitors of apoptosis and autophagy, miRNAs, siRNAs and nanoformulations being investigated for doxorubicin chemosensitization.
Collapse
Affiliation(s)
- Sruthi Sritharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Sampurna Guha
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Snoopy Hazarika
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
12
|
Wang C, Jia Q, Guo X, Li K, Chen W, Shen Q, Xu C, Fu Y. microRNA-34 Family: From Mechanism to Potential Applications. Int J Biochem Cell Biol 2022; 144:106168. [DOI: 10.1016/j.biocel.2022.106168] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/04/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
|
13
|
Serra M, Hattinger CM, Pasello M, Casotti C, Fantoni L, Riganti C, Manara MC. Impact of ABC Transporters in Osteosarcoma and Ewing's Sarcoma: Which Are Involved in Chemoresistance and Which Are Not? Cells 2021; 10:cells10092461. [PMID: 34572110 PMCID: PMC8467338 DOI: 10.3390/cells10092461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
The ATP-binding cassette (ABC) transporter superfamily consists of several proteins with a wide repertoire of functions. Under physiological conditions, ABC transporters are involved in cellular trafficking of hormones, lipids, ions, xenobiotics, and several other molecules, including a broad spectrum of chemical substrates and chemotherapeutic drugs. In cancers, ABC transporters have been intensely studied over the past decades, mostly for their involvement in the multidrug resistance (MDR) phenotype. This review provides an overview of ABC transporters, both related and unrelated to MDR, which have been studied in osteosarcoma and Ewing's sarcoma. Since different backbone drugs used in first-line or rescue chemotherapy for these two rare bone sarcomas are substrates of ABC transporters, this review particularly focused on studies that have provided findings that have been either translated to clinical practice or have indicated new candidate therapeutic targets; however, findings obtained from ABC transporters that were not directly involved in drug resistance were also discussed, in order to provide a more complete overview of the biological impacts of these molecules in osteosarcoma and Ewing's sarcoma. Finally, therapeutic strategies and agents aimed to circumvent ABC-mediated chemoresistance were discussed to provide future perspectives about possible treatment improvements of these neoplasms.
Collapse
Affiliation(s)
- Massimo Serra
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.M.H.); (M.P.); (C.C.); (L.F.); (M.C.M.)
- Correspondence: ; Tel.: +39-051-6366762
| | - Claudia Maria Hattinger
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.M.H.); (M.P.); (C.C.); (L.F.); (M.C.M.)
| | - Michela Pasello
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.M.H.); (M.P.); (C.C.); (L.F.); (M.C.M.)
| | - Chiara Casotti
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.M.H.); (M.P.); (C.C.); (L.F.); (M.C.M.)
| | - Leonardo Fantoni
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.M.H.); (M.P.); (C.C.); (L.F.); (M.C.M.)
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy;
| | - Maria Cristina Manara
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (C.M.H.); (M.P.); (C.C.); (L.F.); (M.C.M.)
| |
Collapse
|
14
|
Huang P, Li F, Mo Z, Geng C, Wen F, Zhang C, Guo J, Wu S, Li L, Brünner N, Stenvang J. A Comprehensive RNA Study to Identify circRNA and miRNA Biomarkers for Docetaxel Resistance in Breast Cancer. Front Oncol 2021; 11:669270. [PMID: 34055636 PMCID: PMC8162208 DOI: 10.3389/fonc.2021.669270] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
To investigate the relationship between non-coding RNAs [especially circular RNAs (circRNAs)] and docetaxel resistance in breast cancer, and to find potential predictive biomarkers for taxane-containing therapies, we have performed transcriptome and microRNA (miRNA) sequencing for two established docetaxel-resistant breast cancer (DRBC) cell lines and their docetaxel-sensitive parental cell lines. Our analyses revealed differences between circRNA signatures in the docetaxel-resistant and -sensitive breast cancer cells, and discovered circRNAs generated by multidrug-resistance genes in taxane-resistant cancer cells. In DRBC cells, circABCB1 was identified and validated as a circRNA that is strongly up-regulated, whereas circEPHA3.1 and circEPHA3.2 are strongly down-regulated. Furthermore, we investigated the potential functions of these circRNAs by bioinformatics analysis, and miRNA analysis was performed to uncover potential interactions between circRNAs and miRNAs. Our data showed that circABCB1, circEPHA3.1 and circEPHA3.2 may sponge up eight significantly differentially expressed miRNAs that are associated with chemotherapy and contribute to docetaxel resistance via the PI3K-Akt and AGE-RAGE signaling pathways. We also integrated differential expression data of mRNA, long non-coding RNA, circRNA, and miRNA to gain a global profile of multi-level RNA changes in DRBC cells, and compared them with changes in DNA copy numbers in the same cell lines. We found that Chromosome 7 q21.12-q21.2 was a common region dominated by multi-level RNA overexpression and DNA amplification, indicating that overexpression of the RNA molecules transcribed from this region may result from DNA amplification during stepwise exposure to docetaxel. These findings may help to further our understanding of the mechanisms underlying docetaxel resistance in breast cancer.
Collapse
Affiliation(s)
| | - Fengyu Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | | | | | - Fang Wen
- MGI, BGI-Shenzhen, Shenzhen, China
| | | | - Jia Guo
- BGI, BGI-Shenzhen, Shenzhen, China
| | - Song Wu
- Shenzhen Luohu Hospital Group, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen, China
| | - Lin Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,National Research Center for Translational Medicine, National Key Scientific Infrastructure for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nils Brünner
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Stenvang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Role of MicroRNAs in Human Osteosarcoma: Future Perspectives. Biomedicines 2021; 9:biomedicines9050463. [PMID: 33922820 PMCID: PMC8146779 DOI: 10.3390/biomedicines9050463] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma (OS) is a rare form of cancer with high death rate but is one of the most frequent forms of bone cancer in children and adolescents. MiRNAs are small endogenous RNAs that regulate gene expression post-transcriptionally. The discovery of miRNAs could allow us to obtain an earlier diagnosis, predict prognosis and chemoresistance, and lead to the discovery of new treatments in different types of tumors, including OS. Despite the fact that there is currently only one clinical trial being carried out on a single miRNA for solid tumors, it is very probable that the number of clinical trials including miRNAs as prognostic and diagnostic biomarkers, as well as potential therapeutic targets, will increase in the near future. This review summarizes the different miRNAs related to OS and their possible therapeutic application.
Collapse
|
16
|
Li M, Ma W. miR-26a Reverses Multidrug Resistance in Osteosarcoma by Targeting MCL1. Front Cell Dev Biol 2021; 9:645381. [PMID: 33816494 PMCID: PMC8012539 DOI: 10.3389/fcell.2021.645381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
The multidrug resistance (MDR) acquired in human osteosarcoma is a huge obstacle for effective chemotherapy. Recently, microRNA-26a (miR-26a) has been associated with the pathogenesis and progression of osteosarcoma. However, whether it regulates MDR in osteosarcoma is unknown. We show here that miR-26a expression declines in chemoresistant osteosarcoma after neoadjuvant chemotherapy, and its expression correlates with clinical outcome. In addition, compared with sensitive parental cells, miR-26a expression also declines in osteosarcoma MDR cells, together suggesting a negative correlation between miR-26a expression and MDR development in osteosarcoma. We also show that the enforced expression of miR-26a reverses MDR in osteosarcoma cells, and conversely, miR-26a knockdown confers MDR in chemosensitive osteosarcoma cells treated with doxorubicin, methotrexate, or cisplatin. Mechanistically, miR-26a directly targets the pro-survival protein myeloid cell leukemia 1 (MCL1), and in turn, the enforced expression of MCL1 markedly antagonizes miR-26a-decreased MDR in osteosarcoma MDR cells, therefore demonstrating that miR-26a reverses MDR in osteosarcoma by targeting MCL1. Lastly, miR-26a reverses resistance to doxorubicin in osteosarcoma MDR cells xenografted in nude mice. Collectively, these results reveal a negative role and the underlying mechanism of miR-26a in the regulation of MDR in human osteosarcoma, implying a potential tactic of manipulating miR-26a for overcoming MDR in osteosarcoma chemotherapy.
Collapse
Affiliation(s)
- Ming Li
- Department of Orthopaedic, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Ma
- Department of Orthopaedic, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
17
|
Lin Z, Xie X, Lu S, Liu T. Noncoding RNAs in osteosarcoma: Implications for drug resistance. Cancer Lett 2021; 504:91-103. [PMID: 33587978 DOI: 10.1016/j.canlet.2021.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 02/09/2023]
Abstract
Osteosarcoma is the most frequent bone malignancy in children and adolescents. Despite advances of surgery and chemotherapy in osteosarcoma over the past decades, overall survival rates of osteosarcoma have reached a plateau. The development of multi-drug resistance (MDR) has become the main obstacle in improving chemotherapeutic effects in osteosarcoma treatment. Therefore, understanding detailed mechanisms of chemoresistance and developing novel therapeutic targets to overcome chemoresistance are crucial to improve the prognosis of osteosarcoma patients. Accumulating evidence has proved that multiple noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) play pivotal roles in osteosarcoma progression. Notably, a great number of ncRNAs are abnormally expressed and can regulate chemosensitivity through various mechanisms in osteosarcoma. In this review, we systematically summarize the roles of ncRNAs as well as the molecular mechanisms in modulating drug resistance of osteosarcoma and discuss the potential roles of ncRNAs as biomarkers and novel therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Zhengjun Lin
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, 139 Renmin Road, Changsha, 410011, Hunan, People's Republic of China; Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Xubin Xie
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Shiyao Lu
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, 139 Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
18
|
Darici S, Alkhaldi H, Horne G, Jørgensen HG, Marmiroli S, Huang X. Targeting PI3K/Akt/mTOR in AML: Rationale and Clinical Evidence. J Clin Med 2020; 9:E2934. [PMID: 32932888 PMCID: PMC7563273 DOI: 10.3390/jcm9092934] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematopoietic malignancy characterized by excessive proliferation and accumulation of immature myeloid blasts in the bone marrow. AML has a very poor 5-year survival rate of just 16% in the UK; hence, more efficacious, tolerable, and targeted therapy is required. Persistent leukemia stem cell (LSC) populations underlie patient relapse and development of resistance to therapy. Identification of critical oncogenic signaling pathways in AML LSC may provide new avenues for novel therapeutic strategies. The phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathway, is often hyperactivated in AML, required to sustain the oncogenic potential of LSCs. Growing evidence suggests that targeting key components of this pathway may represent an effective treatment to kill AML LSCs. Despite this, accruing significant body of scientific knowledge, PI3K/Akt/mTOR inhibitors have not translated into clinical practice. In this article, we review the laboratory-based evidence of the critical role of PI3K/Akt/mTOR pathway in AML, and outcomes from current clinical studies using PI3K/Akt/mTOR inhibitors. Based on these results, we discuss the putative mechanisms of resistance to PI3K/Akt/mTOR inhibition, offering rationale for potential candidate combination therapies incorporating PI3K/Akt/mTOR inhibitors for precision medicine in AML.
Collapse
Affiliation(s)
- Salihanur Darici
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Hazem Alkhaldi
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Gillian Horne
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Heather G. Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| |
Collapse
|
19
|
Yao D, Li C, Rajoka MSR, He Z, Huang J, Wang J, Zhang J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Am J Cancer Res 2020; 10:9741-9766. [PMID: 32863957 PMCID: PMC7449905 DOI: 10.7150/thno.46913] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The p21-Activated kinase 1 (PAK1), a member of serine-threonine kinases family, was initially identified as an interactor of the Rho GTPases RAC1 and CDC42, which affect a wide range of processes associated with cell motility, survival, metabolism, cell cycle, proliferation, transformation, stress, inflammation, and gene expression. Recently, the PAK1 has emerged as a potential therapeutic target in cancer due to its role in many oncogenic signaling pathways. Many PAK1 inhibitors have been developed as potential preclinical agents for cancer therapy. Here, we provide an overview of essential roles that PAK1 plays in cancer, including its structure and autoactivation mechanism, its crucial function from onset to progression to metastasis, metabolism, immune escape and even drug resistance in cancer; endogenous regulators; and cancer-related pathways. We also summarize the reported PAK1 small-molecule inhibitors based on their structure types and their potential application in cancer. In addition, we provide overviews on current progress and future challenges of PAK1 in cancer, hoping to provide new ideas for the diagnosis and treatment of cancer.
Collapse
|
20
|
Gray A, Dang BN, Moore TB, Clemens R, Pressman P. A review of nutrition and dietary interventions in oncology. SAGE Open Med 2020; 8:2050312120926877. [PMID: 32537159 PMCID: PMC7268120 DOI: 10.1177/2050312120926877] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022] Open
Abstract
The complex cellular mechanisms and inter-related pathways of cancer proliferation, evasion, and metastasis remain an emerging field of research. Over the last several decades, nutritional research has prominent role in identifying emerging adjuvant therapies in our fight against cancer. Nutritional and dietary interventions are being explored to improve the morbidity and mortality for cancer patients worldwide. In this review, we examine several dietary interventions and their proposed mechanisms against cancer as well as identifying limitations in the currently available literature. This review provides a comprehensive review of the cancer metabolism, dietary interventions used during cancer treatment, anti metabolic drugs, and their impact on nutritional deficiencies along with a critical review of the following diets: caloric restriction, intermittent fasting, ketogenic diet, Mediterranean diet, Japanese diet, and vegan diet.
Collapse
Affiliation(s)
- Ashley Gray
- Division of Pediatric Hematology/Oncology, Mattel Children's Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Brian N Dang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Theodore B Moore
- Division of Pediatric Hematology/Oncology, Mattel Children's Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Roger Clemens
- Pharmacology & Pharmaceutical Sciences, USC School of Pharmacy, International Center for Regulatory Science, Los Angeles, CA, USA
| | - Peter Pressman
- Polyscience Consulting & Director of Nutrition and Public Health, The Daedalus Foundation, San Clemente, CA, USA
| |
Collapse
|
21
|
Jiang W, Cai X, Xu T, Liu K, Yang D, Fan L, Li G, Yu X. Tripartite Motif-Containing 46 Promotes Viability and Inhibits Apoptosis of Osteosarcoma Cells by Activating NF-B Signaling Through Ubiquitination of PPAR. Oncol Res 2020; 28:409-421. [PMID: 32295675 PMCID: PMC7851538 DOI: 10.3727/096504020x15868639303417] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS), the most common bone cancer, causes high morbidity in children and young adults. TRIM46 is a member of the family of tripartite motif (TRIM)-containing proteins that serve as important regulators of tumorigenesis. Here we investigate the possible role of TRIM46 in OS and the underlying molecular mechanism. We report an increase in the expression of TRIM46 in OS and its association with tumor size, Enneking’s stage, and patient prognosis. TRIM46 knockdown inhibits OS cell viability and cell cycle progression and induces apoptosis, while TRIM46 overexpression exerts inverse effects, which are inhibited by peroxisome proliferator-activated receptor alpha (PPARα) overexpression and the nuclear factor kappa B (NF-κB) inhibitor, pyrrolidine dithiocarbamate (PDTC). Furthermore, TRIM46 negatively regulates PPARα expression via ubiquitination-mediated protein degradation and modification. PPARα overexpression also inactivates NF-κB signaling and NF-κB promoter activity in OS cells overexpressing TRIM46. Moreover, TRIM46 knockdown inhibits tumor growth and induces apoptosis of OS cells in vivo. TRIM46 acts as an oncogene in OS by interacting with and ubiquitinating PPARα, resulting in the activation of NF-κB signaling pathway. Thus, TRIM46 may be a potential biomarker of carcinogenesis.
Collapse
Affiliation(s)
- Wenwei Jiang
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Xinyu Cai
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Tianyang Xu
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Kaiyuan Liu
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Dong Yang
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Lin Fan
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Guodong Li
- Department of Orthopedics, Shanghai Tenth Peoples Hospital, Tong Ji University School of MedicineShanghaiP.R. China
| | - Xiao Yu
- Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal HospitalSuzhouP.R. China
| |
Collapse
|
22
|
Hsieh MJ, Lin CW, Su SC, Reiter RJ, Chen AWG, Chen MK, Yang SF. Effects of miR-34b/miR-892a Upregulation and Inhibition of ABCB1/ABCB4 on Melatonin-Induced Apoptosis in VCR-Resistant Oral Cancer Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:877-889. [PMID: 31982774 PMCID: PMC6994412 DOI: 10.1016/j.omtn.2019.12.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 11/27/2019] [Accepted: 12/19/2019] [Indexed: 12/19/2022]
Abstract
Multidrug resistance (MDR) is the resistance of cells toward various drugs commonly used in tumor treatment. The mechanism of drug resistance in oral cancer is not completely understood. Melatonin is an endogenously produced molecule involved in active biological mechanisms including antiproliferation, oncogene expression modulation, antitumor invasion and migration, and anti-inflammatory, antioxidant, and antiangiogenic effects. Despite these functions, the effects of melatonin on vincristine (VCR)-resistant human oral cancer cells remain largely unknown. This study analyzed the role of melatonin in VCR-resistant human oral cancer cells along with the underlying mechanism. We determined that melatonin induced the apoptosis and autophagy of VCR-resistant oral cancer cells; these actions were mediated by AKT, p38, and c-Jun N-terminal kinase (JNK). Melatonin inhibited ATP-binding cassette B1 (ABCB1) and ABCB4 expression in vitro and in vivo. Melatonin reduced the drug resistance and promoted the apoptosis of VCR-resistant oral cancer cells through the upregulation of microRNA-892a (miR-892a) and miR-34b-5p expressions. The expression of miR-892a and miR-34b-5p was related to melatonin-induced apoptosis, but not autophagy. Therefore, melatonin is a potential novel chemotherapeutic agent for VCR-resistant human oral cancer cell lines.
Collapse
Affiliation(s)
- Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 402, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital 402, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan; Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Linkou, and Keelung 204, Taiwan
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Andy Wei-Ge Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Mu-Kuan Chen
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
23
|
Naghizadeh S, Mohammadi A, Duijf PHG, Baradaran B, Safarzadeh E, Cho WCS, Mansoori B. The role of miR-34 in cancer drug resistance. J Cell Physiol 2020; 235:6424-6440. [PMID: 32064620 DOI: 10.1002/jcp.29640] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/31/2020] [Indexed: 12/25/2022]
Abstract
Resistance to conventional chemotherapy remains a major cause of cancer relapse and cancer-related deaths. Therefore, there is an urgent need to overcome resistance barriers. To improve cancer treatment approaches, it is critical to elucidate the basic mechanisms underlying drug resistance. Increasingly, the mechanisms involving micro-RNAs (miRNAs) are studied because miRNAs are also considered practical therapeutic options due to high degrees of specificity, efficacy, and accuracy, as well as their ability to target multiple genes at the same time. Years of research have firmly established miR-34 as a key tumor suppressor miRNA whose target genes are involved in drug resistance mechanisms. Indeed, numerous articles show that low levels of circulating miR-34 or tumor-specific miR-34 expression are associated with poor response to chemotherapy. In addition, elevation of inherently low miR-34 levels in resistant cancer cells effectively restores sensitivity to chemotherapeutic agents. Here, we review this literature, also highlighting some contradictory observations. In addition, we discuss the potential utility of miR-34 expression as a predictive biomarker for chemotherapeutic drug response. Although caution needs to be exercised, miR-34 is emerging as a biomarker that could improve cancer precision medicine.
Collapse
Affiliation(s)
- Sanaz Naghizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia.,Institute of Health and Biomedical Innovation, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Safarzadeh
- Department of Microbiology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Williams M, Cheng YY, Phimmachanh M, Winata P, van Zandwijk N, Reid G. Tumour suppressor microRNAs contribute to drug resistance in malignant pleural mesothelioma by targeting anti-apoptotic pathways. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:1193-1206. [PMID: 35582270 PMCID: PMC9019216 DOI: 10.20517/cdr.2019.41] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/17/2019] [Accepted: 09/28/2019] [Indexed: 11/28/2022]
Abstract
Aim: Aberrant microRNA expression is a common event in cancer drug resistance, however its involvement in malignant pleural mesothelioma (MPM) drug resistance is largely unexplored. We aimed to investigate the contribution of microRNAs to the resistance to drugs commonly used in the treatment of MPM. Methods: Drug resistant MPM cell lines were generated by treatment with cisplatin, gemcitabine or vinorelbine. Expression of microRNAs was quantified using RT-qPCR. Apoptosis and drug sensitivity assays were carried out following transfection with microRNA mimics or BCL2 siRNAs combined with drugs. Results: Expression of miR-15a, miR-16 and miR-34a was downregulated in MPM cells with acquired drug resistance. Transfection with miR-15a or miR-16 mimics reversed the resistance to cisplatin, gemcitabine or vinorelbine, whereas miR-34a reversed cisplatin and vinorelbine resistance only. Similarly, in parental cell lines, miR-15a or miR-16 mimics sensitised cells to all drugs, whereas miR-34a increased response to cisplatin and vinorelbine. Increased microRNA expression increased drug-induced apoptosis and caused BCL2 mRNA and protein reduction. RNAi-mediated knockdown of BCL2 partly recapitulated the increase in drug sensitivity in cisplatin and vinorelbine treated cells. Conclusion: Drug-resistant MPM cell lines exhibited reduced expression of tumour suppressor microRNAs. Increasing tumour suppressor of microRNA expression sensitised both drug resistant and parental cell lines to chemotherapeutic agents, in part through targeting of BCL2. Taken together, these data suggest that miR-15a, miR-16 and miR-34a are involved in the acquired and intrinsic drug resistance phenotype of MPM cells.
Collapse
Affiliation(s)
- Marissa Williams
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
| | - Yuen Yee Cheng
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
| | - Monica Phimmachanh
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
- Current address: Garvan Institute of Medical Research, Darlinghurst, Sydney NSW2010, Australia
| | - Patrick Winata
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
| | - Nico van Zandwijk
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
- Current address: Sydney Local Health District, Concord, Sydney NSW2194, Australia
| | - Glen Reid
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
- Current address: Department of Pathology, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
25
|
Viera GM, Salomao KB, de Sousa GR, Baroni M, Delsin LEA, Pezuk JA, Brassesco MS. miRNA signatures in childhood sarcomas and their clinical implications. Clin Transl Oncol 2019; 21:1583-1623. [PMID: 30949930 DOI: 10.1007/s12094-019-02104-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
Abstract
Progresses in multimodal treatments have significantly improved the outcomes for childhood cancer. Nonetheless, for about one-third of patients with Ewing sarcoma, rhabdomyosarcoma, or osteosarcoma steady remission has remained intangible. Thus, new biomarkers to improve early diagnosis and the development of precision-targeted medicine remain imperative. Over the last decade, remarkable progress has been made in the basic understanding of miRNAs function and in interpreting the contribution of their dysregulation to cancer development and progression. On this basis, this review focuses on what has been learned about the pivotal roles of miRNAs in the regulation of key genes implicated in childhood sarcomas.
Collapse
Affiliation(s)
- G M Viera
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - K B Salomao
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - G R de Sousa
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - M Baroni
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - L E A Delsin
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - J A Pezuk
- Anhanguera University of Sao Paulo, UNIAN/SP, Sao Paulo, Brasil
| | - M S Brassesco
- Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brasil.
- Departamento de Biologia, FFCLRP-USP, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirao Preto, SP, CEP 14040-901, Brazil.
| |
Collapse
|
26
|
Chen R, Wang G, Zheng Y, Hua Y, Cai Z. Drug resistance-related microRNAs in osteosarcoma: Translating basic evidence into therapeutic strategies. J Cell Mol Med 2019; 23:2280-2292. [PMID: 30724027 PMCID: PMC6433687 DOI: 10.1111/jcmm.14064] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/14/2018] [Accepted: 11/08/2018] [Indexed: 12/19/2022] Open
Abstract
Although the application of multiple chemotherapy brought revolutionary changes to improve overall survival of osteosarcoma patients, the existence of multidrug resistance (MDR) has become a great challenge for successful osteosarcoma treatment in recent decades. Substantial studies have revealed various underlying mechanisms of MDR in cancers. As for osteosarcoma, evidence has highlighted that microRNAs (miRNAs) can mediate in the processes of DNA damage response, apoptosis avoidance, autophagy induction, activation of cancer stem cells, and signal transduction. Besides, these drug resistance‐related miRNAs showed much promise for serving as candidates for predictive biomarkers of poor outcomes and shorter survival time, and therapeutic targets to reverse drug resistance and overcome treatment refractoriness. This review aims to demonstrate the potential molecular mechanisms of miRNAs‐regulated drug resistance in osteosarcoma, and provide insight in translating basic evidence into therapeutic strategies.
Collapse
Affiliation(s)
- Ruiling Chen
- Department of Orthopedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gangyang Wang
- Department of Orthopedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Zheng
- Department of Orthopedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengdong Cai
- Department of Orthopedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Comparison of Cardiac miRNA Transcriptomes Induced by Diabetes and Rapamycin Treatment and Identification of a Rapamycin-Associated Cardiac MicroRNA Signature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8364608. [PMID: 30647817 PMCID: PMC6311877 DOI: 10.1155/2018/8364608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/16/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
Abstract
Rapamycin (Rap), an inhibitor of mTORC1, reduces obesity and improves lifespan in mice. However, hyperglycemia and lipid disorders are adverse side effects in patients receiving Rap treatment. We previously reported that diabetes induces pansuppression of cardiac cytokines in Zucker obese rats (ZO-C). Rap treatment (750 μg/kg/day for 12 weeks) reduced their obesity and cardiac fibrosis significantly; however, it increased their hyperglycemia and did not improve their cardiac diastolic parameters. Moreover, Rap treatment of healthy Zucker lean rats (ZL-C) induced cardiac fibrosis. Rap-induced changes in ZL-C's cardiac cytokine profile shared similarities with that of diabetes-induced ZO-C. Therefore, we hypothesized that the cardiac microRNA transcriptome induced by diabetes and Rap treatment could share similarities. Here, we compared the cardiac miRNA transcriptome of ZL-C to ZO-C, Rap-treated ZL (ZL-Rap), and ZO (ZO-Rap). We report that 80% of diabetes-induced miRNA transcriptome (40 differentially expressed miRNAs by minimum 1.5-fold in ZO-C versus ZL-C; p ≤ 0.05) is similar to 47% of Rap-induced miRNA transcriptome in ZL (68 differentially expressed miRNAs by minimum 1.5-fold in ZL-Rap versus ZL-C; p ≤ 0.05). This remarkable similarity between diabetes-induced and Rap-induced cardiac microRNA transcriptome underscores the role of miRNAs in Rap-induced insulin resistance. We also show that Rap treatment altered the expression of the same 17 miRNAs in ZL and ZO hearts indicating that these 17 miRNAs comprise a unique Rap-induced cardiac miRNA signature. Interestingly, only four miRNAs were significantly differentially expressed between ZO-C and ZO-Rap, indicating that, unlike the nondiabetic heart, Rap did not substantially change the miRNA transcriptome in the diabetic heart. In silico analyses showed that (a) mRNA-miRNA interactions exist between differentially expressed cardiac cytokines and miRNAs, (b) human orthologs of rat miRNAs that are strongly correlated with cardiac fibrosis may modulate profibrotic TGF-β signaling, and (c) changes in miRNA transcriptome caused by diabetes or Rap treatment include cardioprotective miRNAs indicating a concurrent activation of an adaptive mechanism to protect the heart in conditions that exacerbate diabetes.
Collapse
|
28
|
Biersack B. Interplay of non-coding RNAs and approved antimetabolites such as gemcitabine and pemetrexed in mesothelioma. Noncoding RNA Res 2018; 3:213-225. [PMID: 30809600 PMCID: PMC6257890 DOI: 10.1016/j.ncrna.2018.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 12/13/2022] Open
Abstract
Gemcitabine and pemetrexed are clinically approved antimetabolites for the therapy of mesothelioma diseases. These drugs are often applied in combination with platinum complexes and other drugs. The activity of antimetabolites depended on the expression levels of certain non-coding RNAs, in particular, of small microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). The development of tumor resistance towards antimetabolites was regulated by non-coding RNAs. An overview of the interplay between gemcitabine/pemetrexed antimetabolites and non-coding RNAs in mesothelioma is provided. Further to this, various non-coding RNA-modulating agents are discussed which displayed positive effects on gemcitabine or pemetrexed treatment of mesothelioma diseases. A detailed knowledge of the connections of non-coding RNAs with antimetabolites will be constructive for the design of improved therapies in future.
Collapse
Key Words
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- Bcl-2, B-cell lymphoma 2
- DADS, diallyl sulfide
- DHA, docosahexaenoic acid
- DIM, 3,3‘-diindolylmethane
- DMPM, diffuse malignant peritoneal mesothelioma
- EGCG, epigallocatechin-3-gallate
- EMT, epithelial-mesenchymal transition
- Gemcitabine
- HOTAIR, HOX transcript antisense RNA
- I3C, indole-3-carbinol
- Long non-coding RNA
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MPM, malignant pleural mesothelioma
- Mesothelioma
- MicroRNA
- NSCLC, non-small cell lung cancer
- NaB, sodium butyrate
- PDCD4, programmed cell death 4
- PEG, polyethylene glycole
- PEITC, phenethylisothiocyanate
- PTEN, phosphatase and tensin homolog
- Pemetrexed
- RA, retinoic acid
- SAHA, suberoylanilide hydroxamic acid
- SFN, sulforaphane
- TSA, trichostatin A
Collapse
|
29
|
Li Y, Ju K, Wang W, Liu Z, Xie H, Jiang Y, Jiang G, Lu J, Dong Z, Tang F. Dinitrosopiperazine-decreased PKP3 through upregulating miR-149 participates in nasopharyngeal carcinoma metastasis. Mol Carcinog 2018; 57:1763-1779. [PMID: 30144176 PMCID: PMC6282612 DOI: 10.1002/mc.22895] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/18/2022]
Abstract
Nasopharyngeal carcinoma (NPC) has a high metastatic clinicopathological feature. As a carcinogen factor, N,N'-dinitrosopiperazine (DNP) is involved in NPC metastasis, but its precise mechanism has not been fully elucidated. Herein, we showed that DNP promotes NPC metastasis through upregulating miR-149. DNP was found to decrease Plakophilin3 (PKP3) expression, further DNP-decreased PKP3 was verified to be through upregulating miR-149. We also found that DNP induced proliferation, adhesion, migration and invasion of NPC cell, which was inhibited by miR-149-inhibitor. DNP may promote NPC metastasis through miR-149-decreased PKP3 expression. Therefore, DNP-increased miR-149 expression may be an important factor of NPC high metastasis, and miR-149 may serve as a molecular target for anti-metastasis therapy of NPC.
Collapse
Affiliation(s)
- Yuejin Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Zhuhai Hospital of Jinan University, Zhuhai, China
| | - Kunyu Ju
- Metallurgical Science and Engineering, Central South University, Changsha, China
| | - Weiwei Wang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zheliang Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Haitao Xie
- The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yuan Jiang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Guanmin Jiang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jinping Lu
- Zhuhai Hospital of Jinan University, Zhuhai, China
| | - Zigang Dong
- Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Faqing Tang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Zhuhai Hospital of Jinan University, Zhuhai, China
| |
Collapse
|
30
|
Melatonin Inhibits the Progression of Hepatocellular Carcinoma through MicroRNA Let7i-3p Mediated RAF1 Reduction. Int J Mol Sci 2018; 19:ijms19092687. [PMID: 30201903 PMCID: PMC6163650 DOI: 10.3390/ijms19092687] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/06/2018] [Accepted: 09/08/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin is the main pineal hormone that relays light/dark-cycle information to the circadian system. Recent studies have examined the intrinsic antitumor activity of melatonin in various cancers, including hepatocellular carcinoma (HCC), the primary life-threatening malignancy in both sexes in Taiwan. However, the detailed regulatory mechanisms underlying melatonin’s anti-HCC activity remain incompletely understood. Here, we investigated the mechanisms by which the anti-HCC activity of melatonin is regulated. Human hepatoma cell lines were treated with 1 and 2 mM melatonin, and functional assays were used to dissect melatonin’s antitumor effect in HCC; small-RNA sequencing was performed to identify the microRNAs (miRNAs) involved in the anti-HCC activity of melatonin; and quantitative RT-PCR and Western blotting were used to elucidate how miRNAs regulate melatonin-mediated HCC suppression. Melatonin treatment at both doses strongly inhibited the proliferation, migration and invasion capacities of Huh7 and HepG2 cell lines, and melatonin treatment markedly induced the expression of the miRNA let7i-3p in cells. Notably, transfection of cells with a let7i-3p mimic drastically reduced RAF1 expression and activation of mitogen-activated protein kinase signaling downstream from RAF1, and rescue-assay results demonstrated that melatonin inhibited HCC progression by modulating let7i-3p-mediated RAF1 suppression. Our findings support the view that melatonin treatment holds considerable promise as a therapy for HCC.
Collapse
|
31
|
Emerging Role of mTOR Signaling-Related miRNAs in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6141902. [PMID: 30305865 PMCID: PMC6165581 DOI: 10.1155/2018/6141902] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
Mechanistic/mammalian target of rapamycin (mTOR), an atypical serine/threonine kinase of the phosphoinositide 3-kinase- (PI3K-) related kinase family, elicits a vital role in diverse cellular processes, including cellular growth, proliferation, survival, protein synthesis, autophagy, and metabolism. In the cardiovascular system, the mTOR signaling pathway integrates both intracellular and extracellular signals and serves as a central regulator of both physiological and pathological processes. MicroRNAs (miRs), a class of short noncoding RNA, are an emerging intricate posttranscriptional modulator of critical gene expression for the development and maintenance of homeostasis across a wide array of tissues, including the cardiovascular system. Over the last decade, numerous studies have revealed an interplay between miRNAs and the mTOR signaling circuit in the different cardiovascular pathophysiology, like myocardial infarction, hypertrophy, fibrosis, heart failure, arrhythmia, inflammation, and atherosclerosis. In this review, we provide a comprehensive state of the current knowledge regarding the mechanisms of interactions between the mTOR signaling pathway and miRs. We have also highlighted the latest advances on mTOR-targeted therapy in clinical trials and the new perspective therapeutic strategies with mTOR-targeting miRs in cardiovascular diseases.
Collapse
|
32
|
Zhang Y, Yang F. Analyzing the disease module associated with osteosarcoma via a network- and pathway-based approach. Exp Ther Med 2018; 16:2584-2592. [PMID: 30210606 PMCID: PMC6122582 DOI: 10.3892/etm.2018.6506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor observed in children and adolescents. The aim of the present study was to identify an osteosarcoma-related gene module (OSM) by looking for a dense module following the integration of signals from genome-wide association studies (GWAS) into the human protein-protein interaction (PPI) network. A dataset of somatic mutations in osteosarcoma was obtained from the dbGaP database and their testing P-values were incorporated into the PPI network from a recent study using the dmGWAS bioconductor package. An OSM containing 201 genes (OS genes) and 268 interactions, which were closely associated with immune response, intracellular signal transduction and cell activity was identified. Topological analysis of the OSM identified 11 genes, including APP, APPBP2, ATXN1, HSP90B1, IKZF1, KRTAP10-1, PAK1, PDPK1, SMAD4, SUZ12 and TP53 as potential diagnostic biomarkers for osteosarcoma. The overall survival analysis of osteosarcoma for those 11 genes based on a dataset from the Cancer Genome Atlas, identified APP, HSP90B1, SUZ12 and IKZF1 as osteosarcoma survival-related genes. The results of the present study should be helpful in understanding the diagnosis and treatment of osteosarcoma and its underlying mechanisms. In addition, the methodology used in the present study may be suitable for the analysis of other types of disease.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopaedic Microsurgery, Central Hospital of Zibo, Zibo, Shandong 255000, P.R. China
| | - Fei Yang
- Department of Orthopedic Joint Surgery, Central Hospital of Zibo, Zibo, Shandong 255000, P.R. China
| |
Collapse
|
33
|
Yu B, Jiang K, Zhang J. MicroRNA-124 suppresses growth and aggressiveness of osteosarcoma and inhibits TGF-β-mediated AKT/GSK-3β/SNAIL-1 signaling. Mol Med Rep 2018; 17:6736-6744. [PMID: 29488603 DOI: 10.3892/mmr.2018.8637] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 09/13/2017] [Indexed: 11/05/2022] Open
Abstract
Osteosarcoma is one of the most common malignant tumors in adolescent populations and the prognosis remains incompletely understand. Previous reports have demonstrated that microRNA‑124 (miR‑124) has inhibitory effects on various human malignancies and is associated with tumor progression. However, the clinical significance and potential mechanisms of miR‑124 in the progression of osteosarcoma is not clearly understood. In this study, the potential molecular mechanism of miR‑124 in osteosarcoma tumorigenesis, growth and aggressiveness was investigated. The growth, proliferation, apoptosis, migration and invasion of osteosarcoma cells were investigated following miR‑124 transfection were determined by colony formation assay, western blotting, immunofluorescence, migration/invasion assays and reverse transcription‑quantitative polymerase chain reaction. In vivo anti‑cancer effects of miR‑124 were analyzed by a tumor growth assay, immunohistochemistry and survival rate observations. The results demonstrated that miR‑124 transfection significantly decreased integrin expression in osteosarcoma cells, and further inhibited growth, proliferation, migration and invasion of osteosarcoma cells. Flow cytometry assays indicated that miR‑124 transfection attenuated apoptosis resistance of osteosarcoma to tunicamycin, potentially via the downregulation of P53 and Bcl‑2 apoptosis regulator expression. Mechanistic assays demonstrated that miR‑124 transfection suppressed TGF‑β expression in osteosarcoma. An animal study revealed that tumor growth was reduced in tumor cells transfected with miR‑124 compared with control cells, and the survival rate was prolonged in mice with miR‑124 transfected xenografts compared with control tumors. In conclusion, these results indicate that miR‑124 transection inhibits the growth and aggressive of osteosarcoma, potentially via suppression of TGF‑β‑mediated AKT/GSK‑3β/snail family transcriptional repressor 1 (SNAIL‑1) signaling, suggesting miR‑124 may be a potential anti‑cancer agent/target for osteosarcoma therapy.
Collapse
Affiliation(s)
- Bo Yu
- Department of Orthopedics, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Kaibiao Jiang
- Department of Orthopedics, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Jidong Zhang
- Department of Orthopedics, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| |
Collapse
|
34
|
Kun-Peng Z, Xiao-Long M, Chun-Lin Z. Overexpressed circPVT1, a potential new circular RNA biomarker, contributes to doxorubicin and cisplatin resistance of osteosarcoma cells by regulating ABCB1. Int J Biol Sci 2018; 14:321-330. [PMID: 29559849 PMCID: PMC5859477 DOI: 10.7150/ijbs.24360] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs (circRNAs) represent a widespread class of non-coding RNAs generated from back-splicing, with a circular loop structure. Many circRNAs have been reported to play essential roles in cancer development and have the potential to serve as a novel class of biomarkers for clinical diagnosis. However, the role of circRNA in osteosarcoma (OS) remains largely unknown. In the current study, we examined the expression level of circular RNA PVT1 (circPVT1), previously screened and identified the oncogenic role in gastric cancer, in OS and found that circPVT1 was significantly up-regulated in the OS tissues, serums and chemoresistant cell lines, correlated with poor prognosis of OS patients. Besides, ROC curve demonstrated that circPVT1 may be a better diagnostic biomarker than alkaline phosphatase (ALP) in OS with more sensitivity and specificity. In addition, functional assays revealed that circPVT1 knockdown by siRNA could weaken the resistance to doxorubicin and cisplatin of OS cells through decreasing the expression of classical drug resistance-related gene ABCB1. These findings may provide a new insight into the role of circPVT1 as a biomarker for the diagnosis and treatment target of OS.
Collapse
Affiliation(s)
- Zhu Kun-Peng
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, PR China.,Institute of Bone Tumor Affiliated to Tongji University, School of Medicine, Shanghai 200072, PR China
| | - Ma Xiao-Long
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, PR China.,Institute of Bone Tumor Affiliated to Tongji University, School of Medicine, Shanghai 200072, PR China
| | - Zhang Chun-Lin
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, PR China.,Institute of Bone Tumor Affiliated to Tongji University, School of Medicine, Shanghai 200072, PR China
| |
Collapse
|
35
|
Li YJ, Zhang GP, Zhao F, Li RQ, Liu SJ, Zhao ZR, Wang X. Target therapy of TRIM-14 inhibits osteosarcoma aggressiveness through the nuclear factor-κB signaling pathway. Exp Ther Med 2017; 15:2365-2373. [PMID: 29467844 PMCID: PMC5792772 DOI: 10.3892/etm.2017.5679] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 05/05/2017] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma is the most common cause of cancer-associated mortality and the prognosis is yet to be fully elucidated due to the paucity of effective therapeutic targets that significantly influence the quality of life and mean survival rates of patients with osteosarcoma. Studies have showed that tripartite motif-containing (TRIM)-14 is a member of the TRIM protein family that has a vital role in tumor progression and metastasis and promotes angiogenesis, invasion and apoptotic resistance of bone cancer. In this study, a chimeric antibody targeting TRIM-14 (Chanti-TRIM) was constructed and the molecular mechanism of target therapy for TRIM-14 was investigated in osteosarcoma cells and xenograft mice. The growth, migration and invasion properties of U-2OS cells were analyzed following incubation with 10–160 mg/ml Chanti-TRIM. Apoptosis of U-2OS cells was detected after Chanti-TRIM treatment. Matrix metalloproteinase (MMP)-9-mediated nuclear factor-κB (NF-κB) signal pathway was analyzed in U-2OS cells treated with Chanti-TRIM. The inhibitory efficacy of Chanti-TRIM was studied in U-2OS-bearing xenograft mice. Our results demonstrated that neutralizing TRIM-14 expression markedly inhibited the growth, migration and invasion of osteosarcoma cells, in vitro and in vivo. We found that TRIM-14 depletion decreased cell viability and induced cells apoptosis in vitro. In addition, we identified Chanti-TRIM inhibited growth and promoted apoptosis induced by cisplatin through MMP-9-mediated NF-κB signal pathway. Furthermore, we observed that Chanti-TRIM treatment inhibited osteosarcoma growth in vivo. Histological analysis indicated that apoptotic bodies were increased and NF-κB nuclear translocation factors, including Ikkβ, p65 and IkBα, were decreased in tumors treated by Chanti-TRIM. In conclusion, these results showed that Chanti-TRIM markedly inhibited the progression of osteosarcoma, suggesting Chanti-TRIM may be a potential anti-cancer agent that functions via the activation of the NF-κB pathway for osteosarcoma.
Collapse
Affiliation(s)
- Yi-Jiong Li
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Guo-Ping Zhang
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Feng Zhao
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Rui-Qi Li
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Shao-Jun Liu
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Zeng-Ren Zhao
- Department of General Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Xin Wang
- Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| |
Collapse
|
36
|
Liu J, Luo B, Zhao M. Bmi‑1‑targeting suppresses osteosarcoma aggressiveness through the NF‑κB signaling pathway. Mol Med Rep 2017; 16:7949-7958. [PMID: 28983587 PMCID: PMC5779877 DOI: 10.3892/mmr.2017.7660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 07/05/2017] [Indexed: 12/25/2022] Open
Abstract
Bone cancer is one of the most lethal malignancies and the specific causes of tumor initiation are not well understood. B‑cell‑specific Moloney murine leukemia virus integration site 1 protein (Bmi‑1) has been reported to be associated with the initiation and progression of osteosarcoma, and as a prognostic indicator in the clinic. In the current study, a full‑length antibody targeting Bmi‑1 (AbBmi‑1) was produced and the preclinical value of Bmi‑1‑targeted therapy was evaluated in bone carcinoma cells and tumor xenograft mice. The results indicated that the Bmi‑1 expression level was markedly upregulated in bone cancer cell lines, and inhibition of Bmi‑1 by AbBmi‑1 reduced the invasiveness and migration of osteosarcoma cells. Overexpression of Bmi‑1 promoted proliferation and angiogenesis, and increased apoptosis resistance induced by cisplatin via the nuclear factor‑κB (NF‑κB) signal pathway. In addition, AbBmi‑1 treatment inhibited the tumorigenicity of osteosarcoma cells in vivo. Furthermore, AbBmi‑1 blocked NF‑κB signaling and reduced MMP‑9 expression. Furthermore, Bmi‑1 promoted osteosarcoma tumor growth, whereas AbBmi‑1 significantly inhibited osteosarcoma tumor growth in vitro and in vivo. Notably, AbBmi‑1 decreased the percentages of Ki67‑positive cells and terminal deoxynucleotidyl transferase dUTP nick end labeling‑positive cells in tumors compared with Bmi‑1‑treated and PBS controls. Notably, MMP‑9 and NF‑κB expression were downregulated by treatment with AbBmi‑1 in MG‑63 osteosarcoma cells. In conclusion, the data provides evidence that AbBmi‑1 inhibited the progression of osteosarcoma, suggesting that AbBmi‑1 may be a novel anti‑cancer agent through the inhibition of Bmi‑1 via activating the NF‑κB pathway in osteosarcoma.
Collapse
Affiliation(s)
- Jiaguo Liu
- Department of Orthopedics, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Luo
- Department of Orthopedics, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Meng Zhao
- Department of Orthopedics, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
37
|
Begicevic RR, Falasca M. ABC Transporters in Cancer Stem Cells: Beyond Chemoresistance. Int J Mol Sci 2017; 18:E2362. [PMID: 29117122 PMCID: PMC5713331 DOI: 10.3390/ijms18112362] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/02/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022] Open
Abstract
The efficacy of chemotherapy is one of the main challenges in cancer treatment and one of the major obstacles to overcome in achieving lasting remission and a definitive cure in patients with cancer is the emergence of cancer resistance. Indeed, drug resistance is ultimately accountable for poor treatment outcomes and tumour relapse. There are various molecular mechanisms involved in multidrug resistance, such as the change in the activity of membrane transporters primarily belonging to the ATP binding cassette (ABC) transporter family. In addition, it has been proposed that this common feature could be attributed to a subpopulation of slow-cycling cancer stem cells (CSCs), endowed with enhanced tumorigenic potential and multidrug resistance. CSCs are characterized by the overexpression of specific surface markers that vary in different cancer cell types. Overexpression of ABC transporters has been reported in several cancers and more predominantly in CSCs. While the major focus on the role played by ABC transporters in cancer is polarized by their involvement in chemoresistance, emerging evidence supports a more active role of these proteins, in which they release specific bioactive molecules in the extracellular milieu. This review will outline our current understanding of the role played by ABC transporters in CSCs, how their expression is regulated and how they support the malignant metabolic phenotype. To summarize, we suggest that the increased expression of ABC transporters in CSCs may have precise functional roles and provide the opportunity to target, particularly these cells, by using specific ABC transporter inhibitors.
Collapse
Affiliation(s)
- Romana-Rea Begicevic
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth WA 6102, Australia.
| | - Marco Falasca
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth WA 6102, Australia.
| |
Collapse
|
38
|
Maroof H, Islam F, Ariana A, Gopalan V, Lam AK. The roles of microRNA-34b-5p in angiogenesis of thyroid carcinoma. Endocrine 2017; 58:153-166. [PMID: 28840508 DOI: 10.1007/s12020-017-1393-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/09/2017] [Indexed: 11/27/2022]
Abstract
PURPOSE This study aims to determine the expression of miR-34b-5p in thyroid carcinomas and to investigate the role of miR34b-5p in the modulation of proteins involved in angiogenesis of thyroid carcinoma cells. METHODS The expressions of miR-34b-5p levels in five cell lines and 65 tissue samples from thyroid carcinomas were examined by real-time polymerase chain reaction. An exogenous miR-34b-5p (mimic) transiently overexpress miR-34b-5p in theses thyroid carcinoma cells. The effects of miR-34b-5p overexpression on the proteins involved in angiogenesis and cell cycle regulations (VEGF-A, Bcl-2 and Notch1) were investigated by Western blot, immunofluorescence, enzyme-linked immunosorbent assay followed by cell cycle analysis and apoptosis assays. RESULTS miR-34b-5p is markedly downregulated in all thyroid carcinoma cell lines and tissues samples when compared with non-neoplastic immortalised thyroid cell line and non-neoplastic thyroid tissues, respectively. The expression levels of miR-34b were significantly associated with T-stages of thyroid carcinomas (p = 0.042). Downregulation of VEGF-A, Bcl-2 and Notch1 proteins in thyroid carcinoma cells were noted in cells that transiently transfected with miR-34b-5p mimic. In addition, enzyme-linked immunosorbent assay confirmed the decreased expression of VEGF in thyroid carcinoma cells after transfection with miR-34b-5p mimic. Furthermore, miR-34b-5p mimic transfection induces significant accumulation of cells in G0-G1 of the cell cycle by blocking of their entry into the S transitional phase as well as increasing the total apoptosis. CONCLUSIONS miR-34b-5p functions as a potent regulator of angiogenesis, apoptosis and cell proliferation via modulation of VEGF-A, Bcl-2 and Notch1 proteins. It could be a target for developing treatment strategies of thyroid carcinoma with aggressive clinical behaviour.
Collapse
Affiliation(s)
- Hamidreza Maroof
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Farhadul Islam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Armin Ariana
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4222, Australia.
| |
Collapse
|
39
|
Zhou Y, Wang X, Zhang J, He A, Wang YL, Han K, Su Y, Yin J, Lv X, Hu H. Artesunate suppresses the viability and mobility of prostate cancer cells through UCA1, the sponge of miR-184. Oncotarget 2017; 8:18260-18270. [PMID: 28209917 PMCID: PMC5392325 DOI: 10.18632/oncotarget.15353] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/09/2017] [Indexed: 12/03/2022] Open
Abstract
Artesunate (ART) is a sesquiterpene lactone isolated from the leafy portions of the Chinese herb Artemisia annua. Here, we evaluated the effect of ART on the prostate cancer (PCa) cell lines DU145 and LNCaP and explored its potential mechanisms. ART inhibited the viability and mobility of DU145 and LNCaP cells. Mechanistically, we found that UCA1, one of the most important lncRNAs in malignancies of the urinary system, may be a potential mediator contributing to the tumor suppressor function of ART. First, the UCA1 level was reduced significantly after being exposed to ART. In addition, UCA1 was up-regulated in prostate cancer tissues compared to hyperplastic prostatic tissues, and a higher UCA1 level predicted poor prognosis in PCa patients. Furthermore, reintroduction of UCA1 into PCa cells reversed the effect of ART on apoptosis and metastatic ability. Then we determined that the miR-184/Bcl-2 axis might be the downstream signaling pathway of UCA1 upon ART treatment. UCA1 binds to miR-184 through its seed sequences and may function as a sponge for miR-184.
Collapse
Affiliation(s)
- Yan Zhou
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Xiuju Wang
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Yuexiu District of Guangzhou City, Guangdong Province, China, 510282
| | - Jianjun Zhang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Aina He
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Ya Ling Wang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Kun Han
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Yang Su
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Junyi Yin
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| | - Xiaobin Lv
- Central Laboratory of the Third Affiliated Hospital, Nanchang University, Donghu District, Nanchang City, Jiangxi Province, China, 330008
| | - Haiyan Hu
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai, China, 200233
| |
Collapse
|
40
|
Yu GH, Li AM, Li X, Yang Z, Peng H. Bispecific antibody suppresses osteosarcoma aggressiveness through regulation of NF-κB signaling pathway. Tumour Biol 2017. [PMID: 28631557 DOI: 10.1177/1010428317705572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma is one of the most lethal malignancies, and the prognosis remains dismal due to the paucity of effective therapeutic targets. Bmi-1 and TRIM-14 are associated with the initiation and progression of osteosarcoma, which could promote angiogenesis, invasion, and apoptotic resistance in bone cancer tissue. In this study, we constructed a bispecific antibody of BsAbBmi/TRIM targeting Bmi-1 and TRIM-14 and investigated the therapeutic value in bone carcinoma cells and xenograft mice. Our results showed that Bmi-1 and TRIM-14 expression levels were markedly upregulated correlated with nuclear factor-κB nuclear translocation in bone cancer cells and clinical carcinoma tissues. Results have demonstrated that overexpression of Bmi-1 and TRIM-14 promoted growth, proliferation, aggressiveness, and apoptosis resistance of osteosarcoma cells. BsAbBmi/TRIM administration significantly inhibited nuclear factor-κB expression derived by matrix metalloproteinase-9 promoter. BsAbBmi/TRIM administration inhibited growth of osteosarcoma cells and downregulated Bmi-1 and TRIM-14 expression levels. Data also demonstrated that migration and invasion of osteosarcoma cells were also inhibited by BsAbBmi/TRIM. In addition, results illustrated that BsAbBmi/TRIM inhibited tumor growth and tumorigenicity by blockaded sensor expression in nuclear factor-κB signal pathway. Furthermore, in vivo study showed that BsAbBmi/TRIM treatment markedly inhibited the tumorigenicity and growth of osteosarcoma cells compared to either AbBmi-1 or AbTRIM-14 treatment. Notably, survival of xenograft mice was prolonged by BsAbBmi/TRIM treatment compared to either AbBmi-1 or AbTRIM-14 treatment. In conclusion, these results provided new evidence that BsAbBmi/TRIM inhibited the progression of osteosarcoma, which suggest that BsAbBmi/TRIM may be a novel anti-cancer agent for osteosarcoma therapy.
Collapse
Affiliation(s)
- Gui-Hua Yu
- 1 Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ai-Min Li
- 2 Basic Medical College, Wuhan University, Wuhan, Hubei, China
| | - Xiang Li
- 2 Basic Medical College, Wuhan University, Wuhan, Hubei, China
| | - Zhong Yang
- 2 Basic Medical College, Wuhan University, Wuhan, Hubei, China
| | - Hao Peng
- 1 Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
41
|
O'Flanagan CH, Smith LA, McDonell SB, Hursting SD. When less may be more: calorie restriction and response to cancer therapy. BMC Med 2017; 15:106. [PMID: 28539118 PMCID: PMC5442682 DOI: 10.1186/s12916-017-0873-x] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/15/2017] [Indexed: 12/18/2022] Open
Abstract
Calorie restriction (CR) extends lifespan and has been shown to reduce age-related diseases including cancer, diabetes, and cardiovascular and neurodegenerative diseases in experimental models. Recent translational studies have tested the potential of CR or CR mimetics as adjuvant therapies to enhance the efficacy of chemotherapy, radiation therapy, and novel immunotherapies. Chronic CR is challenging to employ in cancer patients, and therefore intermittent fasting, CR mimetic drugs, or alternative diets (such as a ketogenic diet), may be more suitable. Intermittent fasting has been shown to enhance treatment with both chemotherapy and radiation therapy. CR and fasting elicit different responses in normal and cancer cells, and reduce certain side effects of cytotoxic therapy. Findings from preclinical studies of CR mimetic drugs and other dietary interventions, such as the ketogenic diet, are promising for improving the efficacy of anticancer therapies and reducing the side effects of cytotoxic treatments. Current and future clinical studies will inform on which cancers, and at which stage of the cancer process, CR, fasting, or CR mimetic regimens will prove most effective.
Collapse
Affiliation(s)
- Ciara H O'Flanagan
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, 27517, USA
| | - Laura A Smith
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, 27517, USA
| | - Shannon B McDonell
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, 27517, USA
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, 27517, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27517, USA. .,Nutrition Research Institute, University of North Carolina, Kannapolis, NC, 28081, USA. .,Department of Nutrition, University of North Carolina at Chapel Hill, 2100 Michael Hooker Research Center, Campus Box 7461, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
42
|
Lu M, Chen WH, Wang CY, Mao CQ, Wang J. RETRACTED: Reciprocal regulation of miR-1254 and c-Myc in oral squamous cell carcinoma suppresses EMT-mediated metastasis and tumor-initiating properties through MAPK signaling. Biochem Biophys Res Commun 2017; 484:801-807. [DOI: 10.1016/j.bbrc.2017.01.170] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 01/28/2017] [Indexed: 12/16/2022]
|
43
|
Wang DX, Zou YJ, Zhuang XB, Chen SX, Lin Y, Li WL, Lin JJ, Lin ZQ. Sulforaphane suppresses EMT and metastasis in human lung cancer through miR-616-5p-mediated GSK3β/β-catenin signaling pathways. Acta Pharmacol Sin 2017; 38:241-251. [PMID: 27890917 DOI: 10.1038/aps.2016.122] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022] Open
Abstract
Sulforaphane is a common antioxidant selectively abundant in cruciferous plants, which exhibits effective anti-cancer actions in control of tumorigenesis or progression of various cancers. A recent study has shown that sulforaphane attenuates the EGFR signaling pathway in non-small cell lung cancer (NSCLC), suggesting its potential anti-metastatic effects. In this study we assessed the involvement of sulforaphane and miR-616-5p in epithelial-mesenchymal transition (EMT) and NSCLC metastasis. Sulforaphane suppressed the cell proliferation in human NSCLC cell lines H1299, 95C and 95D with IC50 values of 9.52±1.23, 9.04±1.90 and 17.35±2.03 μmol/L, respectively. At low concentrations (1-5 μmol/L), sulforaphane dose-dependently inhibited the migration and invasion of 95D and H1299 cells with relatively high metastatic potential. The anti-metastatic action of sulforaphane was confirmed in 95D and H1299 cell xenografts in vivo. In fresh NSCLC tissue samples from 179 patients, miR-616-5p levels were upregulated in late-stage NSCLCs, and strongly correlated with risk of NSCLC recurrence and metastasis. Consistent with the clinic observation, miR-616-5p levels in the 3 NSCLC cell lines were correlated with their metastatic ability, and were decreased by sulforaphane treatment. Silencing miR-616-5p markedly suppressed the migration and invasion of 95D cells in vitro and NSCLC metastasis in vivo. Further studies revealed that miR-616-5p directly targeted GSK3β and decreased its expression, whereas sulforaphane decreased miR-616-5p levels by histone modification, and followed by inactivation of the GSK3β/β-catenin signaling pathway and inhibition of EMT, which was characterized by loss of epithelial markers and acquisition of a mesenchymal phenotype in NSCLC cells. Our findings suggest that sulforaphane is a potential adjuvant chemotherapeutic agent for the prevention of NSCLC recurrence and metastasis, and miR-616-5p can be clinically utilized as a biomarker or therapeutic target to inhibit metastasis.
Collapse
|
44
|
Zhou Y, Lin S, Tseng KF, Han K, Wang Y, Gan ZH, Min DL, Hu HY. Selumetinib suppresses cell proliferation, migration and trigger apoptosis, G1 arrest in triple-negative breast cancer cells. BMC Cancer 2016; 16:818. [PMID: 27769200 PMCID: PMC5073736 DOI: 10.1186/s12885-016-2773-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 09/08/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) has aggressive progression with poor prognosis and ineffective treatments. Selumetinib is an allosteric, ATP-noncompetitive inhibitor of MEK1/2, which has benn known as effective antineoplastic drugs for several malignant tumors. We hypothesized that Selumetinib might be potential drug for TNBC and explore the mechanism. METHODS After treated with Selumetinib, the viability and mobility of HCC1937 and MDA-MB-231 were detected by MTT, tunnel, wound-healing assay, transwell assay and FCM methods. MiR array was used to analysis the change of miRs. We predicted and verified CUL1 is the target of miR-302a using Luciferase reporter assay. We also silenced the CUL1 by siRNA, to clarify whether CUL1 take part in the cell proliferation, migration and regulated its substrate TIMP1 and TRAF2. Moreover, after transfection, the antagomir of miR-302a and CUL1 over-expressed plasmid into HCC1937 and MDA-MB-231 cell accompanied with the Selumetinib treatment, we detected the proliferation and migration again. RESULTS Selumetinib reduce the proliferation, migration, triggered apoptosis and G1 arrest in TNBC cell lines. In this process, the miR-302a was up-regulated and inhibited the CUL1 expression. The later negatively regulated the TIMP1 and TRAF2. As soon as we knockdown miR-302a and over-expression CUL1 in TNBC cells, the cytotoxicity of Selumetinib was reversed. CONCLUSIONS MiR-302a targeted regulated the CUL1 expression and mediated the Selumetinib-induced cytotoxicity of triple-negative breast cancer.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Shuchen Lin
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Kuo-Fu Tseng
- Biophysics Department of Oregan State University, ALS-2139, Corvallis, OR 97330 USA
| | - Kun Han
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Yaling Wang
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Zhi-hua Gan
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Da-liu Min
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
- Biophysics Department of Oregan State University, ALS-2139, Corvallis, OR 97330 USA
| | - Hai-yan Hu
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| |
Collapse
|
45
|
Targeting the Mammalian Target of Rapamycin in Lung Cancer. Am J Med Sci 2016; 352:507-516. [PMID: 27865299 DOI: 10.1016/j.amjms.2016.08.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/09/2016] [Accepted: 08/18/2016] [Indexed: 12/19/2022]
Abstract
Lung cancer is the leading cause of cancer death worldwide. Despite advances in its prevention and management, the prognosis of patients with lung cancer remains poor. Therefore, much attention is being given to factors that contribute to the development of this disease, the mechanisms that drive oncogenesis and tumor progression and the search for novel targets that could lead to the development of more effective treatments. One cellular pathway implicated in lung cancer development and progression is that of the mammalian target of rapamycin. Studies involving human tissues have linked lung cancer with abnormalities in this pathway. Furthermore, studies in vitro and in vivo using animal models of lung cancer reveal that targeting this pathway might represent an effective means of treating this disease. As a result, there is significant effort invested in the development of drugs targeting mammalian target of rapamycin and related pathways in the clinical setting.
Collapse
|
46
|
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1:12-34. [PMID: 30159408 PMCID: PMC6096431 DOI: 10.1016/j.ncrna.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The epigenetic regulation of cancer cells by small non-coding RNA molecules, the microRNAs (miRNAs), has raised particular interest in the field of oncology. These miRNAs play crucial roles concerning pathogenic properties of cancer cells and the sensitivity of cancer cells towards anticancer drugs. Certain miRNAs are responsible for an enhanced activity of drugs, while others lead to the formation of tumor resistance. In addition, miRNAs regulate survival and proliferation of cancer cells, in particular of cancer stem-like cells (CSCs), that are especially drug-resistant and, thus, cause tumor relapse in many cases. Various small molecule compounds were discovered that target miRNAs that are known to modulate tumor aggressiveness and drug resistance. This review comprises the effects of naturally occurring small molecules (phenolic compounds and terpenoids) on miRNAs involved in cancer diseases.
Collapse
Key Words
- 1,25-D, 1,25-dihydroxyvitamin D3
- 18-AGA, 18α-glycyrrhetinic acid
- 3,6-DHF, 3,6-dihydroxyflavone
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- CAPE, caffeic acid phenethyl ester
- CDODA-Me, methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate
- Dox, doxorubicin
- EGCG, (−)-epigallocatechin-3-O-gallate
- MicroRNA
- PEG, polyethylene glycol
- PPAP, polycyclic polyprenylated acylphloroglucinol
- Polyphenols
- RA, retinoic acid
- ROS, reactive oxygen species
- TQ, thymoquinone
- Terpenes
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|