1
|
Zhang Y, Li J. Recent advancements in understanding of biological role of homeobox C9 in human cancers. World J Clin Oncol 2024; 15:1168-1176. [PMID: 39351453 PMCID: PMC11438841 DOI: 10.5306/wjco.v15.i9.1168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/14/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Homeobox (HOX) C9, a member of the HOX family, is an important transcription factor, and it plays a significant role in various biological processes. This family of genes is highly valued for their essential roles in establishing and maintaining the body axis during embryonic development and adult tissues. Further, HOXC9 plays a central role in neuronal differentiation, angiogenesis, and adipose distribution, which are essential for the development of the nervous system, maturation of tissues and organs, and maintenance of energy balance and metabolic health. Recent research has found that abnormal HOXC9 expression is closely associated with the development and progression of various tumor types. The HOXC9 expression level can be an indicator of tumor prognosis. Therefore, elucidating the association between HOXC9 expression and its regulatory mechanisms and tumorigenesis can provide novel insights on the diagnosis and treatment of patients with cancer.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Clinical Laboratory, The Affiliated Lianyungang Oriental Hospital of Kangda College of Nanjing Medical University, Lianyungang 222042, Jiangsu Province, China
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, The Affiliated Lianyungang Oriental Hospital of Kangda College of Nanjing Medical University, Lianyungang 222042, Jiangsu Province, China
| |
Collapse
|
2
|
Cocîrlea MD, Soare A, Petrovici AR, Silion M, Călin T, Oancea S. Phenolic Composition and Bioactivities of Invasive Ailanthus altissima (Mill.) Swingle Leaf Extracts Obtained by Two-Step Sequential Extraction. Antioxidants (Basel) 2024; 13:824. [PMID: 39061893 PMCID: PMC11273599 DOI: 10.3390/antiox13070824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Ailanthus altissima, a highly invasive species, contains valuable compounds in different plant parts, indicating great practical potential. This paper proposes the use of non-polar (n-hexane) and polar (ethanol) solvents for the extraction of antioxidant compounds from A. altissima (family Simaroubaceae) leaves in a sequential two-step process. Fresh and dried leaves were examined for their microstructure by scanning electron microscopy, and for color changes in the CIELAB color space co-ordinates. An investigation of the harvesting season, processing (freezing and drying), and solvent indicates ethanol can be used for the highly efficient extraction of phenolics, flavonoids, tannins, and carotenoids. Statistically significant differences were found between the autumn and summer samples for phenolic content, and between dried and frozen samples for tannin content. The HPLC phenolic profile indicates more phenolics (nine polyphenols) in dried leaves harvested in both seasons compared to those in frozen ones (five to six polyphenols). Frozen leaves showed a higher antioxidant activity in a ferric-reducing antioxidant power assay than that of the dried samples, which exhibited a higher antioxidant activity using the 1, 1-diphenyl-2-picryl-hydrazyl assay, but it was not statistically significant. The phenolic, flavonoid, and carotenoid contents significantly influenced the antioxidant activities. Among the ethanolic extracts, those from dried leaves showed better antibacterial activity, in particular, on Staphylococcus aureus and Enterococcus faecalis. The high bioactive content and activity of A. altissima leaves make them suitable natural raw materials for various applications.
Collapse
Affiliation(s)
- Maria Denisa Cocîrlea
- Department of Agricultural Sciences and Food Engineering, “Lucian Blaga” University of Sibiu, 7–9 Ion Ratiu Street, 550024 Sibiu, Romania;
| | - Amalia Soare
- National Research and Development Institute for Cryogenic and Isotopic Technologies ICSI, 4 Uzinei Street, 240050 Râmnicu-Vâlcea, Romania;
| | - Anca Roxana Petrovici
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 41A Aleea Grigore Ghica-Voda, 700487 Iasi, Romania;
| | - Mihaela Silion
- Physics of Polymers and Polymeric Materials Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania;
| | - Teodora Călin
- Laboratory of Diagnostic and Investigation, Directorate of Public Health, 3 George Barițiu Street, 550178 Sibiu, Romania;
| | - Simona Oancea
- Department of Agricultural Sciences and Food Engineering, “Lucian Blaga” University of Sibiu, 7–9 Ion Ratiu Street, 550024 Sibiu, Romania;
| |
Collapse
|
3
|
Hushmandi K, Saadat SH, Raei M, Daneshi S, Aref AR, Nabavi N, Taheriazam A, Hashemi M. Implications of c-Myc in the pathogenesis and treatment efficacy of urological cancers. Pathol Res Pract 2024; 259:155381. [PMID: 38833803 DOI: 10.1016/j.prp.2024.155381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Urological cancers, including prostate, bladder, and renal cancers, are significant causes of death and negatively impact the quality of life for patients. The development and progression of these cancers are linked to the dysregulation of molecular pathways. c-Myc, recognized as an oncogene, exhibits abnormal levels in various types of tumors, and current evidence supports the therapeutic targeting of c-Myc in cancer treatment. This review aims to elucidate the role of c-Myc in driving the progression of urological cancers. c-Myc functions to enhance tumorigenesis and has been documented to increase growth and metastasis in prostate, bladder, and renal cancers. Furthermore, the dysregulation of c-Myc can result in a diminished response to therapy in these cancers. Non-coding RNAs, β-catenin, and XIAP are among the regulators of c-Myc in urological cancers. Targeting and suppressing c-Myc therapeutically for the treatment of these cancers has been explored. Additionally, the expression level of c-Myc may serve as a prognostic factor in clinical settings.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Epidemiology and Biostatistics, School of Health, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health,School of Health,Jiroft University Of Medical Sciences, Jiroft, Iran
| | - Amir Reza Aref
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Wu L, Hu Z, Song XF, Liao YJ, Xiahou JH, Li Y, Zhang ZH. Targeting Nrf2 signaling pathways in the role of bladder cancer: From signal network to targeted therapy. Biomed Pharmacother 2024; 176:116829. [PMID: 38820972 DOI: 10.1016/j.biopha.2024.116829] [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: 10/03/2023] [Revised: 05/09/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Bladder cancer (BC) is the most common malignancy of the urinary system and often recurs after tumor removal and/or is resistant to chemotherapy. In cancer cells, the activity of the signaling pathway changes significantly, affecting a wide range of cell activities from growth and proliferation to apoptosis, invasion and metastasis. Nrf2 is a transcription factor that plays an important role in cellular defense responses to a variety of cellular stresses. There is increasing evidence that Nrf2 acts as a tumor driver and that it is involved in the maintenance of malignant cell phenotypes. Abnormal expression of Nrf2 has been found to be common in a variety of tumors, including bladder cancer. Over-activation of Nrf2 can lead to DNA damage and the development of bladder cancer, and is also associated with various pathological phenomena of bladder cancer, such as metastasis, angiogenesis, and reduced toxicity and efficacy of therapeutic anticancer drugs to provide cell protection for cancer cells. However, the above process can be effectively inhibited or reversed by inhibiting Nrf2. Therefore, Nrf2 signaling may be a potential targeting pathway for bladder cancer. In this review, we will characterize this signaling pathway and summarize the effects of Nrf2 and crosstalk with other signaling pathways on bladder cancer progression. The focus will be on the impact of Nrf2 activation on bladder cancer progression and current therapeutic strategies aimed at blocking the effects of Nrf2. To better determine how to promote new chemotherapy agents, develop new therapeutic agents, and potential therapeutic targets.
Collapse
Affiliation(s)
- Liang Wu
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China.
| | - Zhao Hu
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Xiao-Fen Song
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Yu-Jian Liao
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Jiang-Huan Xiahou
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Yuan Li
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China
| | - Zhong-Hua Zhang
- Department of Urinary Surgery, Xinyu People's Hospital, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China; Department of Urinary Surgery, The Affiliated Xinyu Hospital of Nanchang University, 369 Xinxin North Road, Xinyu, Jiangxi Province 338000, PR China.
| |
Collapse
|
5
|
Fang C, Wu W, Ni Z, Liu Y, Luo J, Zhou Y, Gong C, Hu D, Yao C, Chen X, Wang L, Zhu S. Ailanthone inhibits non-small cell lung cancer growth and metastasis through targeting UPF1/GAS5/ULK1 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155333. [PMID: 38518633 DOI: 10.1016/j.phymed.2023.155333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 03/24/2024]
Abstract
BACKGROUND Targeting long non-coding RNAs (LncRNAs) is a novel and promising approach in cancer therapy. In our previous study, we investigated the effects of ailanthone (aila), the main active compound derived from the stem barks of Ailanthus altissima (Mill.) Swingle, on the growth of non-small cell lung cancer (NSCLC) cells. Although we observed significant inhibition of NSCLC cell growth of aila, the underlying mechanisms involving LncRNAs, specifically LncRNA growth arrest specific 5 (GAS5), remain largely unknown. METHODS To further explore the impact of aila on NSCLC, we performed a series of experiments. Firstly, we confirmed the inhibitory effect of aila on NSCLC cell growth using multiple assays, including MTT, wound healing, transwell assay, as well as subcutaneous and metastasis tumor mice models in vivo. Next, we utilized cDNA microarray and RT-QPCR to identify GAS5 as the primary target of aila. To verify the importance of GAS5 in aila-induced tumor inhibition, we manipulated GAS5 expression levels by constructing GAS5 over-expression and knockdown NSCLC cell lines. Furthermore, we investigated the upstream and downstream signaling pathways of GAS5 through western blot and RT-QPCR analysis. RESULTS Our results showed that aila effectively increased GAS5 expression, as determined by microarray analysis. We also observed that aila significantly enhanced GAS5 expression in a dose- and time-dependent manner across various NSCLC cell lines. Notably, over-expression of GAS5 led to a significant suppression of NSCLC cell tumor growth; while aila had minimal inhibitory effect on GAS5-knockdown NSCLC cells. Additionally, we discovered that aila inhibited ULK1 and autophagy, and this inhibition was reversed by GAS5 knockdown. Moreover, we found that aila up-regulated GAS5 expression by suppressing UPF1-mediated nonsense-mediated mRNA decay (NMD). CONCLUSION In summary, our findings suggest that aila promotes GAS5 expression by inhibiting UPF1-mediated NMD, leading to the repression of ULK1-mediated autophagy and subsequent inhibitory effects on NSCLC cells. These results indicate that aila is a potent enhancer of GAS5 and holds promising potential for application in NSCLC therapy. However, our research is currently focused only on NSCLC. It remains to be determined whether aila can also inhibit the growth of other types of tumors through the UPF1/GAS5/ULK1 signaling pathway. In future studies, we can further investigate the mechanisms by which aila suppresses other types of tumors and potentially broaden the scope of its application in cancer therapy.
Collapse
Affiliation(s)
- Cheng Fang
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenbin Wu
- Experiment Animal Center, Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhongya Ni
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yangli Liu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaojiao Luo
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yufu Zhou
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyuan Gong
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- School of Acupuncture, Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Yao
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Chen
- Department of Nei Jing, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Lixin Wang
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Shiguo Zhu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
6
|
Kumar A, BharathwajChetty B, Manickasamy MK, Unnikrishnan J, Alqahtani MS, Abbas M, Almubarak HA, Sethi G, Kunnumakkara AB. Natural compounds targeting YAP/TAZ axis in cancer: Current state of art and challenges. Pharmacol Res 2024; 203:107167. [PMID: 38599470 DOI: 10.1016/j.phrs.2024.107167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
Cancer has become a burgeoning global healthcare concern marked by its exponential growth and significant economic ramifications. Though advancements in the treatment modalities have increased the overall survival and quality of life, there are no definite treatments for the advanced stages of this malady. Hence, understanding the diseases etiologies and the underlying molecular complexities, will usher in the development of innovative therapeutics. Recently, YAP/TAZ transcriptional regulation has been of immense interest due to their role in development, tissue homeostasis and oncogenic transformations. YAP/TAZ axis functions as coactivators within the Hippo signaling cascade, exerting pivotal influence on processes such as proliferation, regeneration, development, and tissue renewal. In cancer, YAP is overexpressed in multiple tumor types and is associated with cancer stem cell attributes, chemoresistance, and metastasis. Activation of YAP/TAZ mirrors the cellular "social" behavior, encompassing factors such as cell adhesion and the mechanical signals transmitted to the cell from tissue structure and the surrounding extracellular matrix. Therefore, it presents a significant vulnerability in the clogs of tumors that could provide a wide window of therapeutic effectiveness. Natural compounds have been utilized extensively as successful interventions in the management of diverse chronic illnesses, including cancer. Owing to their capacity to influence multiple genes and pathways, natural compounds exhibit significant potential either as adjuvant therapy or in combination with conventional treatment options. In this review, we delineate the signaling nexus of YAP/TAZ axis, and present natural compounds as an alternate strategy to target cancer.
Collapse
Affiliation(s)
- Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mukesh Kumar Manickasamy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Jyothsna Unnikrishnan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Hassan Ali Almubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, Abha 61421, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India.
| |
Collapse
|
7
|
Li D, Wang J, Tuo Z, Yoo KH, Yu Q, Miyamoto A, Zhang C, Ye X, Wei W, Wu R, Feng D. Natural products and derivatives in renal, urothelial and testicular cancers: Targeting signaling pathways and therapeutic potential. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155503. [PMID: 38490077 DOI: 10.1016/j.phymed.2024.155503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Natural products have demonstrated significant potential in cancer drug discovery, particularly in renal cancer (RCa), urothelial carcinoma (UC), and testicular cancer (TC). PURPOSE This review aims to examine the effects of natural products on RCa, UC and TC. STUDY DESIGN systematic review METHODS: PubMed and Web of Science databases were retrieved to search studies about the effects of natural products and derivatives on these cancers. Relevant publications in the reference list of enrolled studies were also checked. RESULTS This review highlighted their diverse impacts on key aspects such as cell growth, apoptosis, metastasis, therapy response, and the immune microenvironment. Natural products not only hold promise for novel drug development but also enhance the efficacy of existing chemotherapy and immunotherapy. Importantly, we exert their effects through modulation of critical pathways and target genes, including the PI3K/AKT pathway, NF-κB pathway, STAT pathway and MAPK pathway, among others in RCa, UC, and TC. CONCLUSION These mechanistic insights provide valuable guidance for researchers, facilitating the selection of promising natural products for cancer management and offering potential avenues for further gene regulation studies in the context of cancer treatment.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - Qingxin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, 315211, China
| | - Akira Miyamoto
- Department of Rehabilitation, West Kyushu University, Japan
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Xing Ye
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| |
Collapse
|
8
|
Ma S, Guo X, Han R, Meng Q, Zhang Y, Quan W, Miao S, Yang Z, Shi X, Wang S. Elucidation of the mechanism of action of ailanthone in the treatment of colorectal cancer: integration of network pharmacology, bioinformatics analysis and experimental validation. Front Pharmacol 2024; 15:1355644. [PMID: 38384287 PMCID: PMC10880095 DOI: 10.3389/fphar.2024.1355644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/29/2024] [Indexed: 02/23/2024] Open
Abstract
Background: Ailanthone, a small compound derived from the bark of Ailanthus altissima (Mill.) Swingle, has several anti-tumour properties. However, the activity and mechanism of ailanthone in colorectal cancer (CRC) remain to be investigated. This study aims to comprehensively investigate the mechanism of ailanthone in the treatment of CRC by employing a combination of network pharmacology, bioinformatics analysis, and molecular biological technique. Methods: The druggability of ailanthone was examined, and its targets were identified using relevant databases. The RNA sequencing data of individuals with CRC obtained from the Cancer Genome Atlas (TCGA) database were analyzed. Utilizing the R programming language, an in-depth investigation of differentially expressed genes was carried out, and the potential target of ailanthone for anti-CRC was found. Through the integration of protein-protein interaction (PPI) network analysis, GO and KEGG enrichment studies to search for the key pathway of the action of Ailanthone. Then, by employing molecular docking verification, flow cytometry, Transwell assays, and Immunofluorescence to corroborate these discoveries. Results: Data regarding pharmacokinetic parameters and 137 target genes for ailanthone were obtained. Leveraging The Cancer Genome Atlas database, information regarding 2,551 differentially expressed genes was extracted. Subsequent analyses, encompassing protein-protein interaction network analysis, survival analysis, functional enrichment analysis, and molecular docking verification, revealed the PI3K/AKT signaling pathway as pivotal mediators of ailanthone against CRC. Additionally, the in vitro experiments indicated that ailanthone substantially affects the cell cycle, induces apoptosis in CRC cells (HCT116 and SW620 cells), and impedes the migration and invasion capabilities of these cells. Immunofluorescence staining showed that ailanthone significantly inhibited the phosphorylation of AKT protein and suppressed the activation of the PI3K/AKT signaling pathway, thereby inhibiting the proliferation and metastasis of CRC cells. Conclusion: Therefore, our findings indicate that Ailanthone exerts anti-CRC effects primarily by inhibiting the activation of the PI3K/AKT pathway. Additionally, we propose that Ailanthone holds potential as a therapeutic agent for the treatment of human CRC.
Collapse
Affiliation(s)
- Shanbo Ma
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Xiaodi Guo
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Ruisi Han
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Qian Meng
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Yan Zhang
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Wei Quan
- Department of Pharmacy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Shan Miao
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Zhao Yang
- Department of Military Medical Psychology, Air Force Military Medical University, Xi’an, Shaanxi, China
| | - Xiaopeng Shi
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Siwang Wang
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
9
|
Wang C, Yi T, Li X, Cui J, Li B, Qin Y, Tang S, Zhang J. Ailanthone synergizes with PARP1 inhibitor in tumour growth inhibition through crosstalk of DNA repair pathways in gastric cancer. J Cell Mol Med 2024; 28:e18033. [PMID: 38009603 PMCID: PMC10826444 DOI: 10.1111/jcmm.18033] [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: 08/30/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/29/2023] Open
Abstract
In our previous research, we proved that ailanthone (AIL) inhibits the growth of gastric cancer (GC) cells and causes apoptosis by inhibiting P23. However, we still find some GC organoids are insensitive to AIL. We have done some sequencing analysis and found that the insensitive strains are highly expressed in PARP1. In this study, we investigated whether AIL can enhance the anti-tumour effect of PARPi in GC. CCK8 and spheroid colony formation assay were used to measure anti-tumour effects. SynergyFinder software was used to calculate the synergy score of the drug combination and flow cytometry was used to detect apoptosis. Western blot, IHC, IF tests were used to measure protein expression. Finally, nude mouse xenograft models were used to verify the in vitro mechanisms. High expression of PARP1 was found to be the cause of drug insensitivity. When AIL is paired with a PARP1 inhibitor, olaparib (OLP), drug sensitivity improves. We discovered that this combination functions by blocking off HSP90-BRCA1 interaction and inhibiting the activity of PARP1, thus in turn inhibiting the homologous recombination deficiency and base excision repair pathway to finally achieve synthetic lethality through increased sensitivity. Moreover, P23 can regulate BRCA1 in GC in vitro. This study proves that the inhibitory effect of AIL on BRCA1 allowed even cancer cells with normal BRCA1 function to be sensitive to PARP inhibitors when it is simultaneously administered with OLP. The results greatly expanded the scope of the application of PARPi.
Collapse
Affiliation(s)
- Chunming Wang
- Department of General SurgeryThe Second Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Tingzhuang Yi
- Department of OncologyAffiliated Hospital of YouJiang Medical University For NationalitiesBaiseChina
| | - Xiangde Li
- Department of RadiotherapyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jiarui Cui
- College of StomatologyShanghai Jiao Tong UniversityShanghaiChina
| | - Biqi Li
- Department of PathologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Yankai Qin
- Department of General SurgeryThe Second Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Shixiong Tang
- Department of General SurgeryThe Second Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jianfeng Zhang
- Department of EmergencyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
10
|
Lin L, Wu Q, Lu F, Lei J, Zhou Y, Liu Y, Zhu N, Yu Y, Ning Z, She T, Hu M. Nrf2 signaling pathway: current status and potential therapeutic targetable role in human cancers. Front Oncol 2023; 13:1184079. [PMID: 37810967 PMCID: PMC10559910 DOI: 10.3389/fonc.2023.1184079] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/18/2023] [Indexed: 10/10/2023] Open
Abstract
Cancer is a borderless global health challenge that continues to threaten human health. Studies have found that oxidative stress (OS) is often associated with the etiology of many diseases, especially the aging process and cancer. Involved in the OS reaction as a key transcription factor, Nrf2 is a pivotal regulator of cellular redox state and detoxification. Nrf2 can prevent oxidative damage by regulating gene expression with antioxidant response elements (ARE) to promote the antioxidant response process. OS is generated with an imbalance in the redox state and promotes the accumulation of mutations and genome instability, thus associated with the establishment and development of different cancers. Nrf2 activation regulates a plethora of processes inducing cellular proliferation, differentiation and death, and is strongly associated with OS-mediated cancer. What's more, Nrf2 activation is also involved in anti-inflammatory effects and metabolic disorders, neurodegenerative diseases, and multidrug resistance. Nrf2 is highly expressed in multiple human body parts of digestive system, respiratory system, reproductive system and nervous system. In oncology research, Nrf2 has emerged as a promising therapeutic target. Therefore, certain natural compounds and drugs can exert anti-cancer effects through the Nrf2 signaling pathway, and blocking the Nrf2 signaling pathway can reduce some types of tumor recurrence rates and increase sensitivity to chemotherapy. However, Nrf2's dual role and controversial impact in cancer are inevitable consideration factors when treating Nrf2 as a therapeutic target. In this review, we summarized the current state of biological characteristics of Nrf2 and its dual role and development mechanism in different tumor cells, discussed Keap1/Nrf2/ARE signaling pathway and its downstream genes, elaborated the expression of related signaling pathways such as AMPK/mTOR and NF-κB. Besides, the main mechanism of Nrf2 as a cancer therapeutic target and the therapeutic strategies using Nrf2 inhibitors or activators, as well as the possible positive and negative effects of Nrf2 activation were also reviewed. It can be concluded that Nrf2 is related to OS and serves as an important factor in cancer formation and development, thus provides a basis for targeted therapy in human cancers.
Collapse
Affiliation(s)
- Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qing Wu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Feifei Lu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanhong Zhou
- Department of Medical School of Facial Features, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yifei Liu
- School of Biomedical Engineering, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ni Zhu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - You Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhifeng Ning
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Tonghui She
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
11
|
Wang Y, Zhu H, Xu H, Qiu Y, Zhu Y, Wang X. Senescence-related gene c-Myc affects bladder cancer cell senescence by interacting with HSP90B1 to regulate cisplatin sensitivity. Aging (Albany NY) 2023; 15:7408-7423. [PMID: 37433010 PMCID: PMC10457043 DOI: 10.18632/aging.204863] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/19/2023] [Indexed: 07/13/2023]
Abstract
Patients with advanced bladder cancer gradually become less sensitive to chemotherapeutic agents, leading to tumor recurrence. Initiating the senescence program in solid tumors may be an important means of improving short-term drug sensitivity. The important role of c-Myc in bladder cancer cell senescence was determined using bioinformatics methods. The response to cisplatin chemotherapy in bladder cancer sample was analyzed according to the Genomics of Drug Sensitivity in Cancer database. Cell Counting Kit-8 assay, clone formation assay, and senescence-associated β-galactosidase staining were used to assess bladder cancer cell growth, senescence, and sensitivity to cisplatin, respectively. Western blot and immunoprecipitation were performed to understand the regulation of p21 by c-Myc/HSP90B1. Bioinformatic analysis showed that c-Myc, a cellular senescence gene, was significantly associated with bladder cancer prognosis and sensitivity to cisplatin chemotherapy. c-Myc and HSP90B1 expression were highly correlated in bladder cancer. Reducing the level of c-Myc significantly inhibited bladder cancer cell proliferation, promoted cellular senescence, and enhanced cisplatin chemosensitivity. Immunoprecipitation assays confirmed that HSP90B1 interacted with c-Myc. Western blot analysis showed that reducing the level of HSP90B1 could redeem the p21 overexpression caused by c-Myc overexpression. Further studies showed that reducing HSP90B1 expression could alleviate the rapid growth and accelerate cellular senescence of bladder cancer cells caused by c-Myc overexpression, and that reducing HSP90B1 levels could also improve cisplatin sensitivity in bladder cancer cells. HSP90B1/c-Myc interaction regulates the p21 signaling pathway, which affects cisplatin chemosensitivity by modulating bladder cancer cell senescence.
Collapse
Affiliation(s)
- Yaxuan Wang
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong 226361, China
| | - Haixia Zhu
- Department of Central Laboratory, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong 226361, China
| | - Haifei Xu
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong 226361, China
| | - Yifan Qiu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yonghong Zhu
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong 226361, China
| | - Xiaolin Wang
- Department of Urology, Affiliated Tumor Hospital of Nantong University and Nantong Tumor Hospital, Nantong 226361, China
| |
Collapse
|
12
|
Becker AL, Indra AK. Oxidative Stress in Melanoma: Beneficial Antioxidant and Pro-Oxidant Therapeutic Strategies. Cancers (Basel) 2023; 15:cancers15113038. [PMID: 37297001 DOI: 10.3390/cancers15113038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Cutaneous melanoma ranks as the fifth most common cancer in the United States and represents one of the deadliest forms of skin cancer. While recent advances in systemic targeted therapies and immunotherapies have positively impacted melanoma survival, the survival rate of stage IV melanoma remains at a meager 32%. Unfortunately, tumor resistance can impede the effectiveness of these treatments. Oxidative stress is a pivotal player in all stages of melanoma progression, with a somewhat paradoxical function that promotes tumor initiation but hinders vertical growth and metastasis in later disease. As melanoma progresses, it employs adaptive mechanisms to lessen oxidative stress in the tumor environment. Redox metabolic rewiring has been implicated in acquired resistance to BRAF/MEK inhibitors. A promising approach to enhance the response to therapy involves boosting intracellular ROS production using active biomolecules or targeting enzymes that regulate oxidative stress. The complex interplay between oxidative stress, redox homeostasis, and melanomagenesis can also be leveraged in a preventive context. The purpose of this review is to provide an overview of oxidative stress in melanoma, and how the antioxidant system may be manipulated in a therapeutic context for improved efficacy and survival.
Collapse
Affiliation(s)
- Alyssa L Becker
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR 97331, USA
- John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI 96813, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University (OSU), Corvallis, OR 97331, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- Department of Biochemistry and Biophysics, Oregon State University (OSU), Corvallis, OR 97331, USA
- Linus Pauling Science Center, Oregon State University (OSU), Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| |
Collapse
|
13
|
Zhang FL, Chen XW, Wang YF, Hu Z, Zhang WJ, Zhou BW, Ci PF, Liu KX. Microbiota-derived tryptophan metabolites indole-3-lactic acid is associated with intestinal ischemia/reperfusion injury via positive regulation of YAP and Nrf2. J Transl Med 2023; 21:264. [PMID: 37072757 PMCID: PMC10111656 DOI: 10.1186/s12967-023-04109-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Lactobacillus has been demonstrated to serve a protective role in intestinal injury. However, the relationship between Lactobacillus murinus (L. murinus)-derived tryptophan metabolites and intestinal ischemia/reperfusion (I/R) injury yet to be investigated. This study aimed to evaluate the role of L. murinus-derived tryptophan metabolites in intestinal I/R injury and the underlying molecular mechanism. METHODS Liquid chromatograph mass spectrometry analysis was used to measure the fecal content of tryptophan metabolites in mice undergoing intestinal I/R injury and in patients undergoing cardiopulmonary bypass (CPB) surgery. Immunofluorescence, quantitative RT-PCR, Western blot, and ELISA were performed to explore the inflammation protective mechanism of tryptophan metabolites in WT and Nrf2-deficient mice undergoing intestinal I/R, hypoxia-reoxygenation (H/R) induced intestinal organoids. RESULTS By comparing the fecal contents of three L. murinus-derived tryptophan metabolites in mice undergoing intestinal I/R injury and in patients undergoing cardiopulmonary bypass (CPB) surgery. We found that the high abundance of indole-3-lactic acid (ILA) in the preoperative feces was associated with better postoperative intestinal function, as evidenced by the correlation of fecal metabolites with postoperative gastrointestinal function, serum I-FABP and D-Lactate levels. Furthermore, ILA administration improved epithelial cell damage, accelerated the proliferation of intestinal stem cells, and alleviated the oxidative stress of epithelial cells. Mechanistically, ILA improved the expression of Yes Associated Protein (YAP) and Nuclear Factor erythroid 2-Related Factor 2 (Nrf2) after intestinal I/R. The YAP inhibitor verteporfin (VP) reversed the anti-inflammatory effect of ILA, both in vivo and in vitro. Additionally, we found that ILA failed to protect epithelial cells from oxidative stress in Nrf2 knockout mice under I/R injury. CONCLUSIONS The content of tryptophan metabolite ILA in the preoperative feces of patients is negatively correlated with intestinal function damage under CPB surgery. Administration of ILA alleviates intestinal I/R injury via the regulation of YAP and Nrf2. This study revealed a novel therapeutic metabolite and promising candidate targets for intestinal I/R injury treatment.
Collapse
Affiliation(s)
- Fang-Ling Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Xiao-Wei Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
- Department of Anaesthesiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, Guangdong, China
| | - Yi-Fan Wang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Zhen Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Wen-Juan Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Bo-Wei Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Peng-Fei Ci
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Ave N, Guangzhou, 510515, China.
| |
Collapse
|
14
|
The Roles of miRNAs in Predicting Bladder Cancer Recurrence and Resistance to Treatment. Int J Mol Sci 2023; 24:ijms24020964. [PMID: 36674480 PMCID: PMC9864802 DOI: 10.3390/ijms24020964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Bladder cancer (BCa) is associated with significant morbidity, with development linked to environmental, lifestyle, and genetic causes. Recurrence presents a significant issue and is managed in the clinical setting with intravesical chemotherapy or immunotherapy. In order to address challenges such as a limited supply of BCG and identifying cases likely to recur, it would be advantageous to use molecular biomarkers to determine likelihood of recurrence and treatment response. Here, we review microRNAs (miRNAs) that have shown promise as predictors of BCa recurrence. MiRNAs are also discussed in the context of predicting resistance or susceptibility to BCa treatment.
Collapse
|
15
|
Creation of Value Chains for the Sustainability of Control and Eradication Actions on Ailanthus altissima (Mill.) Swingle. ENVIRONMENTS 2022. [DOI: 10.3390/environments9050064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Actions to control and eradicate Ailanthus altissima (Mill.) Swingle are essential to the prevention of uncontrolled growth and expansion of this species and its competition with native species. This competition leads to biodiversity and productivity losses in forests. The present study evaluated the potential to create value chains to maintain the sustainability of control actions through the energy recovery of collected A. altissima biomass. Other possibilities were also discussed, such as the extraction of allelopathic compounds. For this purpose, and to assess the potential for energy recovery, samples of A. altissima were collected and analyzed in the laboratory to discuss the potential of using extracted compounds in nature-based applications, and a literature review was carried out. It was found that, although there is potential for the use of these biomasses for energy production, the high levels of chlorine and heavy metals pose some obstacles to their large-scale use, mainly due to their corrosive potential. On the other hand, the extraction of allelopathic compounds was shown to be potentially interesting for use in the control of other invasive species. Used in this application, it may be possible to create value chains to sustain, control, and eradicate the actions of this invasive species.
Collapse
|
16
|
The Double-Edged Sword of Oxidative Stress in Skin Damage and Melanoma: From Physiopathology to Therapeutical Approaches. Antioxidants (Basel) 2022; 11:antiox11040612. [PMID: 35453297 PMCID: PMC9027913 DOI: 10.3390/antiox11040612] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
The skin is constantly exposed to exogenous and endogenous sources of reactive oxygen species (ROS). An adequate balance between ROS levels and antioxidant defenses is necessary for the optimal cell and tissue functions, especially for the skin, since it must face additional ROS sources that do not affect other tissues, including UV radiation. Melanocytes are more exposed to oxidative stress than other cells, also due to the melanin production process, which itself contributes to generating ROS. There is an increasing amount of evidence that oxidative stress may play a role in many skin diseases, including melanoma, being the primary cause or being a cofactor that aggravates the primary condition. Indeed, oxidative stress is emerging as another major force involved in all the phases of melanoma development, not only in the arising of the malignancy but also in the progression toward the metastatic phenotype. Furthermore, oxidative stress seems to play a role also in chemoresistance and thus has become a target for therapy. In this review, we discuss the existing knowledge on oxidative stress in the skin, examining sources and defenses, giving particular consideration to melanocytes. Therefore, we focus on the significance of oxidative stress in melanoma, thus analyzing the possibility to exploit the induction of oxidative stress as a therapeutic strategy to improve the effectiveness of therapeutic management of melanoma.
Collapse
|
17
|
Robust Quantification of Regional Patterns of Migration in Three-Dimensional Cell Culture Models. J Med Biol Eng 2022. [DOI: 10.1007/s40846-022-00680-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Abstract
Purpose
Wound healing assays is a common two-dimensional migration model, with the spheroid assay three-dimensional migration model recently emerging as being more representative of in vivo migration behaviours. These models provide insight into the overall migration of cells in response to various factors such as biological, chemotactic and molecular agents. However, currently available analysis techniques for these assays fall short on providing quantifiable means to measure regional migration patterns, which is essential to allow a more robust assessment of drug treatments on cell migration in a chemotactic fashion. Therefore, this study aims to develop a finite element (FE) based pipeline that can objectively quantify regional migration patterns of cells.
Methods
We have developed a novel FE based approach that is able to accurately measure changes in overall migration areas of 3D Glioblastoma Multiforme (GBM) spheroids that we generated using the primary cell lines from patients undergoing tumour resection surgery. We live-imaged the migration patterns of GBM spheroids and analysed them, first with the standard ImageJ method. We then performed the same analysis with the proposed FE method.
Results
When compared to the standard ImageJ method, our proposed method was able to measure the changes in a more quantitative and accurate manner. Furthermore, our regional migration analysis provided means to analyse the migration pattern seen in the phantom data and our experimental results.
Conclusion
Our FE based method will be a a robust tool for analysing cell migration patterns of GBM and other migrating cells in various diseases and degenerations.
Collapse
|
18
|
Argenziano M, Bessone F, Dianzani C, Cucci MA, Grattarola M, Pizzimenti S, Cavalli R. Ultrasound-Responsive Nrf2-Targeting siRNA-Loaded Nanobubbles for Enhancing the Treatment of Melanoma. Pharmaceutics 2022; 14:341. [PMID: 35214073 PMCID: PMC8878772 DOI: 10.3390/pharmaceutics14020341] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
The siRNA-mediated inhibition of nuclear factor E2-related factor 2 (Nrf2) can be an attractive approach to overcome chemoresistance in various malignant tumors, including melanoma. This work aims at designing a new type of chitosan-shelled nanobubble for the delivery of siRNA against Nrf2 in combination with an ultrasound. A new preparation method based on a water-oil-water (W/O/W) double-emulsion was purposely developed for siRNA encapsulation in aqueous droplets within a nanobubble core. Stable, very small NB formulations were obtained, with sizes of about 100 nm and a positive surface charge. siRNA was efficiently loaded in NBs, reaching an encapsulation efficiency of about 90%. siNrf2-NBs downregulated the target gene in M14 cells, sensitizing the resistant melanoma cells to the cisplatin treatment. The combination with US favored NB cell uptake and transfection efficiency. Based on the results, nanobubbles have shown to be a promising US responsive tool for siRNA delivery, able to overcome chemoresistance in melanoma cancer cells.
Collapse
Affiliation(s)
- Monica Argenziano
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (M.A.); (F.B.); (C.D.)
| | - Federica Bessone
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (M.A.); (F.B.); (C.D.)
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (M.A.); (F.B.); (C.D.)
| | - Marie Angèle Cucci
- Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (M.A.C.); (M.G.); (S.P.)
| | - Margherita Grattarola
- Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (M.A.C.); (M.G.); (S.P.)
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (M.A.C.); (M.G.); (S.P.)
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (M.A.); (F.B.); (C.D.)
| |
Collapse
|
19
|
Ding H, Yu X, Yan Z. Ailanthone suppresses the activity of human colorectal cancer cells through the STAT3 signaling pathway. Int J Mol Med 2021; 49:21. [PMID: 34958109 PMCID: PMC8722763 DOI: 10.3892/ijmm.2021.5076] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022] Open
Abstract
Ailanthone (AIL) is a major quassinoid extracted from the Chinese medicinal herb, Ailanthus altissima, which has been reported to exert anti-proliferative effects on various cancer cells. The present study aimed to investigate the anti-tumor effects of AIL on HCT116 and SW620 colon cancer cells, and to analyze the underlying molecular mechanisms. CCK-8 assay was used to detect cell viability. Furthermore, colony formation and Transwell assays, and flow cytometry were used to examine the effects of AIL on cell proliferation, apoptosis and migration. Finally, the expression levels of cell cycle control proteins, and caspase and Bcl-2 family-related proteins involved in the regulation of apoptosis, as well as those of cell migration- and pathway-related proteins were examined using western blot analysis. Reverse transcription-quantitative PCR was used to quantitatively analyze the changes in the JAK and STAT3 gene levels in each group. The in vitro cell function tests revealed that AIL inhibited the proliferation and migration, and induced the apoptosis and cell cycle arrest of HCT116 and SW620 cells. It was further found exerted these effects via the JAK/STAT3 signaling pathway, as well as through caspase and Bcl-2 family proteins. On the whole, the present study demonstrates that AIL suppresses the activity of colon cancer cells via the STAT3 pathway.
Collapse
Affiliation(s)
- Haixiang Ding
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Xiuchong Yu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Zhilong Yan
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
20
|
Oxidative Stress-Related Mechanisms in Melanoma and in the Acquired Resistance to Targeted Therapies. Antioxidants (Basel) 2021; 10:antiox10121942. [PMID: 34943045 PMCID: PMC8750393 DOI: 10.3390/antiox10121942] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a highly aggressive cancer with the poorest prognosis, representing the deadliest form of skin cancer. Activating mutations in BRAF are the most frequent genetic alterations, present in approximately 50% of all melanoma cases. The use of specific inhibitors towards mutant BRAF variants and MEK, a downstream signaling target of BRAF in the MAPK pathway, has significantly improved progression-free and overall survival in advanced melanoma patients carrying BRAF mutations. Nevertheless, despite these improvements, resistance still develops within the first year of therapy in around 50% of patients, which is a significant problem in managing BRAF-mutated advanced melanoma. Understanding these mechanisms is one of the mainstreams of the research on BRAFi/MEKi acquired resistance. Both genetic and epigenetic mechanisms have been described. Moreover, in recent years, oxidative stress has emerged as another major force involved in all the phases of melanoma development, from initiation to progression until the onsets of the metastatic phenotype and chemoresistance, and has thus become a target for therapy. In the present review, we discuss the current knowledge on oxidative stress and its signaling in melanoma, as well as the oxidative stress-related mechanisms in the acquired resistance to targeted therapies.
Collapse
|
21
|
Paiboonrungruang C, Simpson E, Xiong Z, Huang C, Li J, Li Y, Chen X. Development of targeted therapy of NRF2 high esophageal squamous cell carcinoma. Cell Signal 2021; 86:110105. [PMID: 34358647 PMCID: PMC8403639 DOI: 10.1016/j.cellsig.2021.110105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly disease and one of the most aggressive cancers of the gastrointestinal tract. As a master transcription factor regulating the stress response, NRF2 is often mutated and becomes hyperactive, and thus causes chemo-radioresistance and poor survival in human ESCC. There is a great need to develop NRF2 inhibitors for targeted therapy of NRF2high ESCC. In this review, we mainly focus on three aspects, NRF2 inhibitors and their mechanisms of action, screening novel drug targets, and evaluation of NRF2 activity in the esophagus. A research strategy has been proposed to develop NRF2 inhibitors using human ESCC cells and mouse models.
Collapse
Affiliation(s)
- Chorlada Paiboonrungruang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Emily Simpson
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Zhaohui Xiong
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Caizhi Huang
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27607, USA
| | - Jianying Li
- Euclados Bioinformatics Solutions, Cary, NC 27519, USA
| | - Yahui Li
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Xiaoxin Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA; Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
22
|
Grattarola M, Cucci MA, Roetto A, Dianzani C, Barrera G, Pizzimenti S. Post-translational down-regulation of Nrf2 and YAP proteins, by targeting deubiquitinases, reduces growth and chemoresistance in pancreatic cancer cells. Free Radic Biol Med 2021; 174:202-210. [PMID: 34364982 DOI: 10.1016/j.freeradbiomed.2021.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
The intrinsic chemoresistance of pancreatic ductal adenocarcinoma (PDAC) represents the main obstacle in treating this aggressive malignancy. It has been observed that high antioxidant levels and upregulated Nrf2 and the YAP protein expression can be involved in PDAC chemoresistance. The mechanisms of Nrf2 and YAP increase need to be clarified. We chose a panel of PDAC cell lines with diverse sensitivity to cisplatin and gemcitabine. In PANC-1 chemoresistant cells, we found a low level of oxidative stress and high levels of Nrf2 and YAP protein expressions and their respective targets. On the contrary, in CFPAC-1 chemosensitive cells, we found high levels of oxidative stress and low level of these two proteins, as well as their respective targets. In MiaPaCa-2 cells with a middle chemoresistance, we observed intermediate features. When Nrf2 and YAP were inhibited in PANC-1 cells by Ailanthone, a plant extract, we observed a reduction of viability, thus sustaining the role of these two proteins in maintaining the PDAC chemoresistance. We then delved into the mechanisms of the Nrf2 and YAP protein upregulation in chemoresistance, discovering that it was at a post-translational level since the mRNA expressions did not match the protein levels. Treatments of PANC-1 cells with the proteasome inhibitor MG-132 and the protein synthesis inhibitor cycloheximide further confirmed this observation. The expression of DUB3 and OTUD1 deubiquitinases, involved in the control of Nrf2 and YAP protein level, respectively, was also investigated. Both protein expressions were higher in PANC-1 cells, intermediate in MiaPaCa-2 cells, and lower in CFPAC-1 cells. When DUB3 or OTUD1 were silenced, both Nrf2 and YAP expressions were downregulated. Importantly, in deubiquitinase-silenced cells, we observed a great reduction of proliferation and a higher sensitivity to gemcitabine treatment, suggesting that DUB3 and OTUD1 can represent a suitable target to overcome chemoresistance in PDAC cells.
Collapse
Affiliation(s)
- Margherita Grattarola
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Marie Angèle Cucci
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy.
| |
Collapse
|
23
|
Liao Y, Zou X, Wang K, Wang Y, Wang M, Guo T, Zhong B, Jiang N. Comprehensive analysis of Transcription Factors identified novel prognostic biomarker in human bladder cancer. J Cancer 2021; 12:5605-5621. [PMID: 34405021 PMCID: PMC8364643 DOI: 10.7150/jca.58484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Transcriptional factors (TFs) are responsible for regulating the transcription of pro-oncogenes and tumor suppressor genes in the process of tumor development. However, the role of these transcription factors in Bladder cancer (BCa) remains unclear. And the main purpose of this research is to explore the possibility of these TFs serving as biomarkers for BCa. Methods: We analyzed the differential expression of TFs in BCa from The Cancer Genome Atlas (TCGA) online database, identified 408 up-regulated TFs and 751down-regulated TFs. We obtained some hub genes via WGCNA model and detected the RNAs level in BCa cells and tissues. Then, the relationship between the expression and clinicopathological parameters was further investigated. Kaplan-Meier curves and the log-rank test were carried out to analyze the relationship between NFATC1, AKNA and five-TFs combination and overall survival (OS). And RT-PCR assay was conducted to further consolidate and verify these results. Results: There were significant differences in the expression of five TFs (CBX7, AKNA, HDAC4, EBF2 and NFATC1) between bladder cancer and normal bladder tissue. In BCa tissue and cell lines, the five TFs were frequently down-regulated, and closely related to poor prognosis. Moreover, the RT-PCR results of five TFs in bladder cancer and normal bladder tissue were consistent with the database results, and reduced TFs could significantly induce or restrain the transcription of many critical factors. The expression level of AKNA and NFATC1 could serve as independent biomarker to predict the overall survival (P<0.05). And the above five TFs combined detection of bladder cancer has higher sensitivity and specificity. Furthermore, differential neutrophils expression between high-risk and low-risk were found, which consolidated the role and function of the five TFs combination model in the progression of BCa. Conclusions: Our analysis effectively provides a newly TFs-associated prognostic model for bladder cancer. The combination of five identified-TFs is an independent prognostic biomarker, which could serve as a more effective therapeutic target for BCa patients.
Collapse
Affiliation(s)
- Yihao Liao
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xuanxuan Zou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Keke Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Youzhi Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Miaomiao Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tao Guo
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Boqiang Zhong
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ning Jiang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
24
|
Hussain Y, Islam L, Khan H, Filosa R, Aschner M, Javed S. Curcumin-cisplatin chemotherapy: A novel strategy in promoting chemotherapy efficacy and reducing side effects. Phytother Res 2021; 35:6514-6529. [PMID: 34347326 DOI: 10.1002/ptr.7225] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
The efficacy of chemotherapy in cancer therapy is limited due to resistance, treatment selectivity, and severe adverse effects. Immunotherapy, chemotherapy, targeted therapy, radiation, and surgery are the most common therapeutic strategies for treatment, with chemotherapy being the most successful. Nonetheless, these treatments exhibit poor effectiveness due to toxicity and resistance. Therefore, combination therapies of natural products may be used as an effective and novel strategy to overcome such barriers. Cisplatin is a platinum-based chemotherapy agent, and when administered alone, it can lead to severe adverse effects and resistance mechanism resulting in therapeutic failure. Curcumin is a polyphenolic compound extracted from turmeric (Curcuma longa) exhibiting anticancer potential with minimal adverse effects. The combination therapy of curcumin and cisplatin is a novel strategy to mitigate/attenuate cisplatin-related adverse effects and improve the barrier of resistance reducing unwanted effects. However, there are uncertainties on the efficacy of curcumin, and more in depth and high-quality studies are needed. This review aims to explain the adverse effects related to individual cisplatin delivery, the positive outcome of individual curcumin delivery, and the combination therapy of curcumin and cisplatin from nano platform as a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Controlled Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Lubna Islam
- Department of Pharmacy, University of Malakand, Dir Lower Chakdara, KPK, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Rosanna Filosa
- Department of Experimental Medicine, University of Campania, "L. Vanvitelli", Naples, Italy
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Saba Javed
- Department of Zoology, Hazara University, Mansehra, Pakistan
| |
Collapse
|
25
|
The Role of Non-Coding RNAs in the Regulation of the Proto-Oncogene MYC in Different Types of Cancer. Biomedicines 2021; 9:biomedicines9080921. [PMID: 34440124 PMCID: PMC8389562 DOI: 10.3390/biomedicines9080921] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/17/2023] Open
Abstract
Alterations in the expression level of the MYC gene are often found in the cells of various malignant tumors. Overexpressed MYC has been shown to stimulate the main processes of oncogenesis: uncontrolled growth, unlimited cell divisions, avoidance of apoptosis and immune response, changes in cellular metabolism, genomic instability, metastasis, and angiogenesis. Thus, controlling the expression of MYC is considered as an approach for targeted cancer treatment. Since c-Myc is also a crucial regulator of many cellular processes in healthy cells, it is necessary to find ways for selective regulation of MYC expression in tumor cells. Many recent studies have demonstrated that non-coding RNAs play an important role in the regulation of the transcription and translation of this gene and some RNAs directly interact with the c-Myc protein, affecting its stability. In this review, we summarize current data on the regulation of MYC by various non-coding RNAs that can potentially be targeted in specific tumor types.
Collapse
|
26
|
Li X, Li Y, Ma S, Zhao Q, Wu J, Duan L, Xie Y, Wang S. Traditional uses, phytochemistry, and pharmacology of Ailanthus altissima (Mill.) Swingle bark: A comprehensive review. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114121. [PMID: 33862103 DOI: 10.1016/j.jep.2021.114121] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The dried bark of Ailanthus altissima (Mill.) Swingle (BAA), commonly designated as "Chunpi" in Chinese, is extensively used as a common traditional medicine in China, Korea, and India. It has been used to treat multiple ailments, including asthma, epilepsy, spermatorrhea, bleeding, and ophthalmic diseases, for thousands of years. AIM OF THE REVIEW To present a comprehensive and constructive review on the phytochemistry, pharmacology, pharmacokinetics, traditional uses, quality control, and toxicology of BAA; to aid the assessment of the therapeutic potential of BAA; to guide researchers working on the development of novel therapeutic agents. MATERIALS AND METHODS Information related to BAA (from 1960 to 2020) was retrieved from a wide variety of electronic databases, such as PubMed, Web of Science, China Knowledge Resource Integrated Database, ScienceDirect, SciFinder, and Google Scholar. Additional information and materials were acquired from Chinese Medicine Monographs, the 2020 edition of the Chinese Pharmacopoeia, and several web sources, such as the official website of The Plant List and Flora of China. Additionally, perspectives for future investigations and applications of BAA were extensively explored. RESULTS Approximately 221 chemical compounds, including alkaloids, quassinoids, phenylpropanoids, triterpenoids, volatile oils, and other compounds, have been isolated and characterized from BAA; among these, the quassinoid ailanthone is the most typical. The crude extracts and active compounds of BAA have been reported to exert a wide range of pharmacological activities, such as antitumor, anti-inflammatory, antiviral, herbicidal, and insecticidal activities. Although BAA is safe when administered at a conventional dose, at higher doses, it exhibits toxicity due to the presence of quassinoids. Thus, more studies are required to evaluate the efficacy and safety of BAA. CONCLUSION Modern pharmacological studies have revealed that BAA, as a valuable medicinal resource, possesses the potential to treat a wide variety of ailments, especially, cancer and gastrointestinal inflammation. These studies present a wide range of perspectives for the development of new drugs related to BAA. However, only a few traditional uses are associated with the reported pharmacological activities of BAA and have been confirmed by preclinical and clinical studies. Moreover, the pharmacokinetics, toxicology, and quality control of BAA should be considered indispensable research topics.
Collapse
Affiliation(s)
- Xiang Li
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Yao Li
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Shanbo Ma
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Qianqian Zhao
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Junsheng Wu
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Linrui Duan
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Yanhua Xie
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Siwang Wang
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
27
|
Wang CM, Li HF, Wang XK, Li WG, Su Q, Xiao X, Hao TF, Chen W, Zhang YW, Zhang HY, Wu W, Hu ZR, Zhao GY, Huo MY, He YL, Zhang CH. Ailanthus Altissima-derived Ailanthone enhances Gastric Cancer Cell Apoptosis by Inducing the Repression of Base Excision Repair by Downregulating p23 Expression. Int J Biol Sci 2021; 17:2811-2825. [PMID: 34345209 PMCID: PMC8326126 DOI: 10.7150/ijbs.60674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy plays an irreplaceable role in the treatment of GC, but currently available chemotherapeutic drugs are not ideal. The application of medicinal plants is an important direction for new drug discovery. Through drug screening of GC organoids, we determined that ailanthone has an anticancer effect on GC cells in vitro and in vivo. We also found that AIL can induce DNA damage and apoptosis in GC cells. Further transcriptome sequencing of PDX tissue indicated that AIL inhibited the expression of XRCC1, which plays an important role in DNA damage repair, and the results were also confirmed by western blotting. In addition, we found that AIL inhibited the expression of P23 and that inhibition of P23 decreased the expression of XRCC1, indicating that AIL can regulate XRCC1 via P23. The results of coimmunoprecipitation showed that AIL can inhibit the binding of P23 and XRCC1 to HSP90. These findings indicate that AIL can induce DNA damage and apoptosis in GC cells. Meanwhile, AIL can decrease XRCC1 activity by downregulating P23 expression to inhibit DNA damage repair. The present study sheds light on the potential application of new drugs isolated from natural medicinal plants for GC therapy.
Collapse
Affiliation(s)
- Chun-Ming Wang
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China.,Department of Intervention, The People's Hospital of Guangxi Zhuang Autonomous Region,Nanning Guangxi 530021,P.R. China
| | - Hua-Fu Li
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Adult Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Xiao-Kun Wang
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wu-Guo Li
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Xing Xiao
- Scientific research center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R.China
| | - Teng-Fei Hao
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wei Chen
- Scientific research center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R.China
| | - Ya-Wei Zhang
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hai-Yong Zhang
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wang Wu
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhen-Ran Hu
- Scientific research center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R.China
| | - Guang-Yin Zhao
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Ming-Yu Huo
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Yu-Long He
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China.,Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chang-Hua Zhang
- Digestive Disease Center, The Seventh Affiliated Hospital of Sun Yat‑Sen University, Shenzhen, Guangdong 518107, P.R. China
| |
Collapse
|
28
|
Chen L, Wu C, Wang H, Chen S, Ma D, Tao Y, Wang X, Luan Y, Wang T, Shi Y, Song G, Zhao Y, Dong X, Wang B. Analysis of Long Noncoding RNAs in Aila-Induced Non-Small Cell Lung Cancer Inhibition. Front Oncol 2021; 11:652567. [PMID: 34235076 PMCID: PMC8255921 DOI: 10.3389/fonc.2021.652567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/21/2021] [Indexed: 01/24/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) has the highest morbidity and mortality among all carcinomas. However, it is difficult to diagnose in the early stage, and current therapeutic efficacy is not ideal. Although numerous studies have revealed that Ailanthone (Aila), a natural product, can inhibit multiple cancers by reducing cell proliferation and invasion and inducing apoptosis, the mechanism by which Aila represses NSCLC progression in a time-dependent manner remains unclear. In this study, we observed that most long noncoding RNAs (lncRNAs) were either notably up- or downregulated in NSCLC cells after treatment with Aila. Moreover, alterations in lncRNA expression induced by Aila were crucial for the initiation and metastasis of NSCLC. Furthermore, in our research, expression of DUXAP8 was significantly downregulated in NSCLC cells after treatment with Aila and regulated expression levels of EGR1. In conclusion, our findings demonstrate that Aila is a potent natural suppressor of NSCLC by modulating expression of DUXAP8 and EGR1.
Collapse
Affiliation(s)
- Lin Chen
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.,College of Animal Science, Jilin University, Changchun, China
| | - Cui Wu
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Heming Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Sinuo Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Danhui Ma
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Ye Tao
- Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xingye Wang
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yanhe Luan
- Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Tiedong Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Yan Shi
- School of Pharmacy, Jilin University, Changchun, China
| | - Guangqi Song
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yicheng Zhao
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xijun Dong
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.,Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Bingmei Wang
- College of Clinical Medicine, College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
29
|
Serwetnyk MA, Blagg BS. The disruption of protein-protein interactions with co-chaperones and client substrates as a strategy towards Hsp90 inhibition. Acta Pharm Sin B 2021; 11:1446-1468. [PMID: 34221862 PMCID: PMC8245820 DOI: 10.1016/j.apsb.2020.11.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/12/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
The 90-kiloDalton (kD) heat shock protein (Hsp90) is a ubiquitous, ATP-dependent molecular chaperone whose primary function is to ensure the proper folding of several hundred client protein substrates. Because many of these clients are overexpressed or become mutated during cancer progression, Hsp90 inhibition has been pursued as a potential strategy for cancer as one can target multiple oncoproteins and signaling pathways simultaneously. The first discovered Hsp90 inhibitors, geldanamycin and radicicol, function by competitively binding to Hsp90's N-terminal binding site and inhibiting its ATPase activity. However, most of these N-terminal inhibitors exhibited detrimental activities during clinical evaluation due to induction of the pro-survival heat shock response as well as poor selectivity amongst the four isoforms. Consequently, alternative approaches to Hsp90 inhibition have been pursued and include C-terminal inhibition, isoform-selective inhibition, and the disruption of Hsp90 protein-protein interactions. Since the Hsp90 protein folding cycle requires the assembly of Hsp90 into a large heteroprotein complex, along with various co-chaperones and immunophilins, the development of small molecules that prevent assembly of the complex offers an alternative method of Hsp90 inhibition.
Collapse
Key Words
- ADP, adenosine diphosphate
- ATP, adenosine triphosphate
- Aha1, activator of Hsp90 ATPase homologue 1
- CTD, C-terminal domain
- Cdc37, cell division cycle 37
- Disruptors
- Grp94, 94-kD glucose-regulated protein
- HIF-1α, hypoxia-inducing factor-1α
- HIP, Hsp70-interaction protein
- HOP, Hsp70‒Hsp90 organizing protein
- HSQC, heteronuclear single quantum coherence
- Her-2, human epidermal growth factor receptor-2
- Hsp90
- Hsp90, 90-kD heat shock protein
- MD, middle domain
- NTD, N-terminal domain
- Natural products
- PPI, protein−protein interaction
- Peptidomimetics
- Protein−protein interactions
- SAHA, suberoylanilide hydroxamic acid
- SAR, structure–activity relationship
- SUMO, small ubiquitin-like modifier
- Small molecules
- TPR2A, tetratricopeptide-containing repeat 2A
- TRAP1, Hsp75tumor necrosis factor receptor associated protein 1
- TROSY, transverse relaxation-optimized spectroscopy
- hERG, human ether-à-go-go-related gene
Collapse
|
30
|
Tannous M, Caldera F, Hoti G, Dianzani U, Cavalli R, Trotta F. Drug-Encapsulated Cyclodextrin Nanosponges. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2207:247-283. [PMID: 33113141 DOI: 10.1007/978-1-0716-0920-0_19] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To date, a number of nanocarriers, either inorganic or organic, have been developed to improve the delivery and therapeutic efficacy of various drugs. Drug delivery systems have attempted to overcome the undesirable pharmacokinetic problems encountered. Among the various nanomaterials that have been designed as potential nanocarriers, cyclodextrin-based polymers are of particular interest in this review.Cyclodextrins (CD) are a class of cyclic glucopyranose oligomers, obtained from starch by enzymatic action, with a characteristic toroidal shape that forms a truncated cone-shaped lipophilic cavity. The main common native cyclodextrins are named α, β, and γ which comprise six, seven, and eight glucopyranose units, respectively. Cyclodextrins have the capability to include compounds whose size and polarity are compatible with those of their cavity.Cyclodextrin-based cross-linked polymers, often referred to as "cyclodextrin nanosponges" (CDNSs), attract great attention from researchers for solving major bioavailability problems such as inadequate solubility, poor dissolution rate, and limited stability of some agents, as well as increasing their effectiveness and decreasing unwanted side effects.Registered patents about this novel system in various fields, different pharmaceutical applications, and classes of drugs encapsulated by CDNSs are detailed. The features outlined make CDNSs a promising platform for the development of innovative and advanced delivery systems.
Collapse
Affiliation(s)
- Maria Tannous
- Dipartimento di Chimica, Università di Torino, Torino, Italy.,Department of Chemistry, University of Balamand, Tripoli, Lebanon
| | | | - Gjylije Hoti
- Dipartimento di Chimica, Università di Torino, Torino, Italy
| | - Umberto Dianzani
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Torino, Italy
| | - Roberta Cavalli
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Torino, Italy
| | | |
Collapse
|
31
|
Barrera G, Cucci MA, Grattarola M, Dianzani C, Muzio G, Pizzimenti S. Control of Oxidative Stress in Cancer Chemoresistance: Spotlight on Nrf2 Role. Antioxidants (Basel) 2021; 10:antiox10040510. [PMID: 33805928 PMCID: PMC8064392 DOI: 10.3390/antiox10040510] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Chemoresistance represents the main obstacle to cancer treatment with both conventional and targeted therapy. Beyond specific molecular alterations, which can lead to targeted therapy, metabolic remodeling, including the control of redox status, plays an important role in cancer cell survival following therapy. Although cancer cells generally have a high basal reactive oxygen species (ROS) level, which makes them more susceptible than normal cells to a further increase of ROS, chemoresistant cancer cells become highly adapted to intrinsic or drug-induced oxidative stress by upregulating their antioxidant systems. The antioxidant response is principally mediated by the transcription factor Nrf2, which has been considered the master regulator of antioxidant and cytoprotective genes. Nrf2 expression is often increased in several types of chemoresistant cancer cells, and its expression is mediated by diverse mechanisms. In addition to Nrf2, other transcription factors and transcriptional coactivators can participate to maintain the high antioxidant levels in chemo and radio-resistant cancer cells. The control of expression and function of these molecules has been recently deepened to identify which of these could be used as a new therapeutic target in the treatment of tumors resistant to conventional therapy. In this review, we report the more recent advances in the study of Nrf2 regulation in chemoresistant cancers and the role played by other transcription factors and transcriptional coactivators in the control of antioxidant responses in chemoresistant cancer cells.
Collapse
Affiliation(s)
- Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
- Correspondence:
| | - Marie Angele Cucci
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| | - Margherita Grattarola
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 11, 10125 Turin, Italy;
| | - Giuliana Muzio
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| |
Collapse
|
32
|
Mirzaei S, Mohammadi AT, Gholami MH, Hashemi F, Zarrabi A, Zabolian A, Hushmandi K, Makvandi P, Samec M, Liskova A, Kubatka P, Nabavi N, Aref AR, Ashrafizadeh M, Khan H, Najafi M. Nrf2 signaling pathway in cisplatin chemotherapy: Potential involvement in organ protection and chemoresistance. Pharmacol Res 2021; 167:105575. [PMID: 33771701 DOI: 10.1016/j.phrs.2021.105575] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 12/14/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a vital transcription factor and its induction is of significant importance for protecting against oxidative damage. Increased levels of Reactive Oxygen Species (ROS) stimulate Nrf2 signaling, enhancing the activity of antioxidant enzymes such as catalase, superoxide dismutase and glutathione peroxidase. These enzymes are associated with retarding oxidative stress. On the other hand, Nrf2 activation in cancer cells is responsible for the development of chemoresistance due to disrupting oxidative mediated-cell death by reducing ROS levels. Cisplatin (CP), cis-diamminedichloroplatinum(II), is a potent anti-tumor agent extensively used in cancer therapy, but its frequent application leads to the development of chemoresistance as well. In the present study, association of Nrf2 signaling with chemoresistance to CP and protection against its deleterious effects is discussed. Anti-tumor compounds, mainly phytochemicals, retard chemoresistance by suppressing Nrf2 signaling. Upstream mediators such as microRNAs can regulate Nrf2 expression during CP chemotherapy regimens. Protection against side effects of CP is mediated via activating Nrf2 signaling and its downstream targets activating antioxidant defense system. Protective agents that activate Nrf2 signaling, can ameliorate CP-mediated ototoxicity, nephrotoxicity and neurotoxicity. Reducing ROS levels and preventing cell death are the most important factors involved in alleviating CP toxicity upon Nrf2 activation. As pre-clinical experiments advocate the role of Nrf2 in chemoprotection and CP resistance, translating these findings to the clinic can provide a significant progress in treatment of cancer patients.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Aliasghar Tabatabaei Mohammadi
- Asu Vanda Gene Research Company, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Science Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interface, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, 56025 Pisa, Pontedera, Italy
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6 Canada
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanashah University of Medical Sciences, Kermanshah 6715847141, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
33
|
Hu L, Lv X, Li D, Zhang W, Ran G, Li Q, Hu J. The anti-angiogenesis role of FBXW7 in diabetic retinopathy by facilitating the ubiquitination degradation of c-Myc to orchestrate the HDAC2. J Cell Mol Med 2021; 25:2190-2202. [PMID: 33369138 PMCID: PMC7882985 DOI: 10.1111/jcmm.16204] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/08/2020] [Accepted: 12/04/2020] [Indexed: 01/10/2023] Open
Abstract
Diabetic retinopathy (DR) is the most prevalently occurring microvascular complication in diabetic patients that triggers severe visual impairments. The anti-angiogenesis role of FBXW7 has been identified in breast cancer. Therefore, this study intends to decipher the mechanism of FBXW7 in angiogenesis of DR. DR model was induced on mice using high-glucose (HG) and high-fat diet, and retinal microvascular endothelial cells (RMECs) isolated from normal mice were induced with HG, followed by evaluation of FBXW7, Ki67, HIF-1α and VEGF expression by immunofluorescence, immunohistochemistry or Western blot analysis. After gain- and loss-of-function assays in normal and DR mice, angiogenesis was assessed by CD31 fluorescence staining and Western blot analysis. After ectopic expression and silencing experiments in HG-induced RMECs, RMEC proliferation, migration and angiogenesis were, respectively, determined by EdU, Transwell and in vitro angiogenesis assays. The impact of FBXW7 on the ubiquitination of c-Myc was studied by cycloheximide chase assay and proteasome inhibition, and the binding of c-Myc to HDAC2 promoter by dual-luciferase reporter gene experiment. DR mice and HG-induced RMECs possessed down-regulated FBXW7 and up-regulated Ki67, HIF-1α and VEGF. Silencing FBXW7 enhanced angiogenesis in normal mouse retinal tissue, but overexpressing FBXW7 or silencing c-Myc diminished angiogenesis in DR mouse retinal tissue. Overexpressing FBXW7 or silencing c-Myc depressed proliferation, migration and angiogenesis in HG-induced RMECs. FBXW7 induced c-Myc ubiquitination degradation, and c-Myc augmented HDAC2 expression by binding to HDAC2 promoter. Conclusively, our data provided a novel sight of anti-angiogenesis role of FBXW7 in DR by modulating the c-Myc/HDAC2 axis.
Collapse
Affiliation(s)
- Lihua Hu
- Aier Eye Hospital of Wuhan UniversityWuhanChina
| | - Xiangyun Lv
- Aier Eye Hospital of Wuhan UniversityWuhanChina
| | - Dai Li
- School of OptometryHubei University of Science and TechnologyXianningChina
| | | | | | - Qingchun Li
- School of OptometryHubei University of Science and TechnologyXianningChina
| | - Jun Hu
- Aier Eye Hospital of Wuhan UniversityWuhanChina
- School of OptometryHubei University of Science and TechnologyXianningChina
| |
Collapse
|
34
|
Dong Y, Hao L, Fang K, Han XX, Yu H, Zhang JJ, Cai LJ, Fan T, Zhang WD, Pang K, Ma WM, Wang XT, Han CH. A network pharmacology perspective for deciphering potential mechanisms of action of Solanum nigrum L. in bladder cancer. BMC Complement Med Ther 2021; 21:45. [PMID: 33494738 PMCID: PMC7836472 DOI: 10.1186/s12906-021-03215-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Solanum nigrum L. decoction has been used as a folklore medicine in China to prevent the postoperative recurrence of bladder cancer (BC). However, there are no previous pharmacological studies on the protective mechanisms of this activity of the plant. Thus, this study aimed to perform a systematic analysis and to predict the potential action mechanisms underlying S. nigrum activity in BC based on network pharmacology. METHODS Based on network pharmacology, the active ingredients of S. nigrum and the corresponding targets were identified using the Traditional Chinese Medicines for Systems Pharmacology Database and Analysis Platform database, and BC-related genes were screened using GeneCards and the Online Mendelian Inheritance in Man database. In addition, ingredient-target (I-T) and protein-protein interaction (PPI) networks were constructed using STRING and Cytoscape, Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted, and then the pathways directly related to BC were integrated manually to reveal the pharmacological mechanism underlying S. nigrum-medicated therapeutic effects in BC. RESULTS Seven active herbal ingredients from 39 components of S. nigrum were identified, which shared 77 common target genes related to BC. I-T network analysis revealed that quercetin was associated with all targets and that NCOA2 was targeted by four ingredients. Besides, interleukin 6 had the highest degree value in the PPI network, indicating a hub role. A subsequent gene enrichment analysis yielded 86 significant GO terms and 89 significant pathways, implying that S. nigrum had therapeutic benefits in BC through multi-pathway effects, including the HIF-1, TNF, P53, MAPK, PI3K/Akt, apoptosis and bladder cancer pathway. CONCLUSIONS S. nigrum may mediate pharmacological effects in BC through multi-target and various signaling pathways. Further validation is required experimentally. Network pharmacology approach provides a predicative novel strategy to reveal the holistic mechanism of action of herbs.
Collapse
Affiliation(s)
- Yang Dong
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China.,Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Lin Hao
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China.,Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Kun Fang
- Xuzhou Clinical Medical College of Integrated Traditional Chinese and Western Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Xuzhou, China
| | - Xiao-Xiao Han
- Center of Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Yu
- Yantai Hospital of Traditional Chinese Medicine, Yantai, China
| | - Jian-Jun Zhang
- Department of Urology, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Long-Jun Cai
- Department of Urology, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Tao Fan
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Wen-da Zhang
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Kun Pang
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Wei-Ming Ma
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Xi-Tao Wang
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Cong-Hui Han
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China. .,Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, China. .,Department of Biotechnology, College of Life Sciences, Jiangsu Normal University, Xuzhou, China.
| |
Collapse
|
35
|
|
36
|
Smolková K, Mikó E, Kovács T, Leguina-Ruzzi A, Sipos A, Bai P. Nuclear Factor Erythroid 2-Related Factor 2 in Regulating Cancer Metabolism. Antioxid Redox Signal 2020; 33:966-997. [PMID: 31989830 PMCID: PMC7533893 DOI: 10.1089/ars.2020.8024] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Nuclear factor erythroid 2 (NFE2)-related factor 2 (NFE2L2, or NRF2) is a transcription factor predominantly affecting the expression of antioxidant genes. NRF2 plays a significant role in the control of redox balance, which is crucial in cancer cells. NRF2 activation regulates numerous cancer hallmarks, including metabolism, cancer stem cell characteristics, tumor aggressiveness, invasion, and metastasis formation. We review the molecular characteristics of the NRF2 pathway and discuss its interactions with the cancer hallmarks previously listed. Recent Advances: The noncanonical activation of NRF2 was recently discovered, and members of this pathway are involved in carcinogenesis. Further, cancer-related changes (e.g., metabolic flexibility) that support cancer progression were found to be redox- and NRF2 dependent. Critical Issues: NRF2 undergoes Janus-faced behavior in cancers. The pro- or antineoplastic effects of NRF2 are context dependent and essentially based on the specific molecular characteristics of the cancer in question. Therefore, systematic investigation of NRF2 signaling is necessary to clarify its role in cancer etiology. The biggest challenge in the NRF2 field is to determine which cancers can be targeted for better clinical outcomes. Further, large-scale genomic and transcriptomic studies are missing to correlate the clinical outcome with the activity of the NRF2 system. Future Directions: To exploit NRF2 in a clinical setting in the future, the druggable members of the NRF2 pathway should be identified. In addition, it will be important to study how the modulation of the NRF2 system interferes with cytostatic drugs and their combinations.
Collapse
Affiliation(s)
- Katarína Smolková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alberto Leguina-Ruzzi
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary.,Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
37
|
Xue D, Zhou X, Qiu J. Emerging role of NRF2 in ROS-mediated tumor chemoresistance. Biomed Pharmacother 2020; 131:110676. [PMID: 32858502 DOI: 10.1016/j.biopha.2020.110676] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/16/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
Chemoresistance is a central cause for the tumor management failure. Cancer cells disrupt the redox homeostasis through reactive oxygen species (ROS) regulatory mechanisms, leading to tumor progression and chemoresistance. The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of neutralizing cellular ROS and restoring redox balance. Understanding the role of NRF2 in ROS-mediated chemoresistance can be helpful in the development of chemotherapy strategies with better efficiency. In this review, we sum up the roles of ROS in the development of chemoresistance to classical chemotherapy agents including cisplatin, 5-fluorouracil, gemcitabine, oxaliplatin, paclitaxel, and doxorubicin, and how to overcome ROS-mediated tumor chemoresistance by targeting NRF2. Finally, we propose that targeting NRF2 might be a promising strategy to resist ROS-driven chemoresistance and acquire better efficacy in cancer treatment.
Collapse
Affiliation(s)
- Danfeng Xue
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiongming Zhou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jiaxuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
38
|
Carbosilane Dendrimers Loaded with siRNA Targeting Nrf2 as a Tool to Overcome Cisplatin Chemoresistance in Bladder Cancer Cells. Antioxidants (Basel) 2020; 9:antiox9100993. [PMID: 33066634 PMCID: PMC7602517 DOI: 10.3390/antiox9100993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is considered as the master regulator of antioxidant and cytoprotective gene expressions. Moreover, it plays a pivotal role in cancer progression. Nrf2 mediates the adaptive response which contributes to the resistance to chemotherapeutic pro-oxidant drugs, such as cisplatin (CDDP), in various tumors, including bladder cancers. For this reason, Nrf2 could be a promising target to overcome chemoresistance. There are several known Nrf2 pharmacological inhibitors; however, most of them are not specific. The use of a specific small interfering RNA (siRNA) targeting the Nrf2 gene (siNrf2) loaded into nanovehicles is an attractive alternative, since it can increase specificity. This study aimed to evaluate the biological activity of siNrf2 loaded on guanidine-terminated carbosilane dendrimers (GCDs) in overcoming CDDP resistance in bladder cancer cells with a high level of Nrf2. Parameters such as viability, proliferation, apoptosis, migration, and oxidative stress level were taken into account. Results demonstrated that siNrf2-GCD treatment sensitized CDDP-resistant cells to CDDP treatment. Moreover, data obtained by treating the non-cancerous human kidney HK-2 cell line strongly suggest a good safety profile of the carbosilane dendrimers loaded with siNrf2. In conclusion, we suggest that siNrf2-GCD is a promising drug delivery system for gene therapy to be used in vivo; and it may represent an important tool in the therapy of CDDP-resistant cancer.
Collapse
|
39
|
Salimian J, Baradaran B, Azimzadeh Jamalkandi S, Moridikia A, Ahmadi A. MiR-486-5p enhances cisplatin sensitivity of human muscle-invasive bladder cancer cells by induction of apoptosis and down-regulation of metastatic genes. Urol Oncol 2020; 38:738.e9-738.e21. [PMID: 32527702 DOI: 10.1016/j.urolonc.2020.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/28/2020] [Accepted: 05/09/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Cisplatin is one of the common chemotherapy drugs for bladder cancer, and resistance to this drug is one of the major obstacles to effective chemotherapy. MicroRNAs (miRNAs) are a category of small noncoding RNAs that can regulate the expression of numerous genes. Recent studies showed that miRNAs can act as a powerful regulator of chemo-sensitivity in cancer cells. Hence, this study aimed to investigate the effects of miRNA-486-5p on cisplatin-sensitivity of different bladder cancer cells. MATERIAL AND METHODS The 5637 and EJ138 cancer cells were treated with miRNA-486-5p and cisplatin, individually or in combination. RESULTS Afterward, the cytotoxicity effects of these treatments were determined by MTT assay and the increased cisplatin-sensitivity observed in both cell lines, especially, 5637 cells. Moreover, subG1 phase cell cycle arrest, changes in the expression of caspase-9, caspase-3, P53, SIRT1, OLFM4, SMAD2, and Bcl-2 genes and nuclear fragmentation also revealed the induction of apoptosis in all treatments, which increased in combination groups. Also, the combination of miRNA-486-5p with cisplatin significantly down-regulated the expression of migration associated genes including ROCK, CD44, and MMP-9 as compared with cisplatin alone. CONCLUSION Altogether, these results indicated that the miRNA-486-5p could induce apoptosis and inhibit cell migration ability of the cells. It seems that pre-electroporation of cells with miRNA-486-5p has useful results in the enhancement of cisplatin sensitivity of 5637 and EJ138 cancer cells and this combination may be a promising treatment strategy for bladder cancer therapy.
Collapse
Affiliation(s)
- Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abdollah Moridikia
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Chou ST, Ho BY, Tai YT, Huang CJ, Chao WW. Bidirect effects from cisplatin combine with rosmarinic acid (RA) or hot water extracts of Glechoma hederacea (HWG) on renal cancer cells. Chin Med 2020; 15:77. [PMID: 32760434 PMCID: PMC7391823 DOI: 10.1186/s13020-020-00358-2] [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: 03/27/2020] [Accepted: 07/20/2020] [Indexed: 01/21/2023] Open
Abstract
Background Cisplatin (CDDP) is a chemotherapeutic drug which also causes adverse side effects. Glechoma hederacea is a traditional Chinese herb belonging to the Labiatae family and has many biological activities. Our previous study indicated that rosmarinic acid (RA) was the most abundant phytochemical in G. hederacea. However, the antioxidant or anti-inflammatory effects of the combined treatment of G. hederacea, RA and CDDP on human renal cell carcinoma (RCC) 786-O cells have not been clearly demonstrated. We aimed to investigate the bioefficacy of hot water extracts of G. hederacea (HWG) and RA in inhibiting RCC 786-O cell activity and its synergism with CDDP against metastatic renal cancer cell. Methods Bioactivities of the combination treatment of HWG, RA, HWG/CDDP and RA/CDDP were assessed using the MTT assay and transwell migration, and the crude extract/compound efficacy was evaluated using wound healing migration assays, flow cytometry and western blotting. Results Our study indicates that CDDP inhibits 786-O cell proliferation and migration and HWG and RA protect against these effects. On the other hand, HWG and RA demonstrate a low cytotoxic effect in human renal proximal tubular epithelial cell line -2 (HK-2 cells). Cell cycle analysis found that HWG/CDDP and RA/CDDP combined treatment exerted cytotoxicity by inducing G2/M arrest and apoptosis. RA in combined with CDDP significantly inhibiting the expression of p-FAK (Tyr 925) in RCC 786-O cells in vitro. Conclusion We propose that the inhibition of RA on RCC 786-O cell invasion and migration may partly occur through the downregulation of FAK phosphorylation. The HWG/CDDP and RA/CDDP combined treatments may be effective strategies for intervention of RCC 786-O cell activity.
Collapse
Affiliation(s)
- Su-Tze Chou
- Department of Food and Nutrition, Providence University, Taichung, 433 Taiwan
| | - Bing-Ying Ho
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, 116 Taiwan.,Department of General Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, 116 Taiwan
| | - Yu-Ting Tai
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, 116 Taiwan
| | - Chun-Jen Huang
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, 116 Taiwan
| | - Wen-Wan Chao
- Department of Nutrition and Health Sciences, Kainan University, No.1 Kainan Road, Luzhu Dist, Taoyuan City, 33857 Taiwan, ROC
| |
Collapse
|
41
|
Ding H, Yu X, Hang C, Gao K, Lao X, Jia Y, Yan Z. Ailanthone: A novel potential drug for treating human cancer. Oncol Lett 2020; 20:1489-1503. [PMID: 32724391 PMCID: PMC7377054 DOI: 10.3892/ol.2020.11710] [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: 09/12/2019] [Accepted: 05/05/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is the second leading cause of death after cardiovascular disease. In 2015, >8.7 million people died worldwide due to cancer, and by 2030 this figure is expected to increase to ~13.1 million. Tumor chemotherapy drugs have specific toxicity and side effects, and patients can also develop secondary drug resistance. To prevent and treat cancer, scientists have developed novel drugs with improved antitumor effects and decreased toxicity. Ailanthone (AIL) is a quassinoid extract from the traditional Chinese medicine plant Ailanthus altissima, which is known to have anti-inflammatory and antimalarial effects. An increasing number of studies have focused on AIL due to its antitumor activity. AIL can inhibit cell proliferation and induce apoptosis by up- or downregulating cancer-associated molecules, which ultimately leads to cancer cell death. Antitumor effects of AIL have been observed in melanoma, acute myeloid leukemia, bladder, lung, breast, gastric and prostate cancer and vestibular neurilemmoma. To the best of our knowledge, the present study is the first review to describe the antitumor mechanisms of AIL.
Collapse
Affiliation(s)
- Haixiang Ding
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiuchong Yu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of The Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Chen Hang
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Kaijun Gao
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xifeng Lao
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yangtao Jia
- Medical School of Ningbo University, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Zhilong Yan
- Department of Gastrointestinal Surgery, The Affiliated Hospital of The Medical School of Ningbo University and Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
42
|
Bailly C. Anticancer properties and mechanism of action of the quassinoid ailanthone. Phytother Res 2020; 34:2203-2213. [PMID: 32239572 DOI: 10.1002/ptr.6681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/03/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
Ailanthone (AIT) is a quassinoid natural product isolated from the worldwide-distributed plant Ailanthus altissima. The drug displays multiple pharmacological properties, in particular significant antitumor effects against a variety of cancer cell lines in vitro. Potent in vivo activities have been evidenced in mice bearing hepatocellular carcinoma, nonsmall cell lung cancer and castration-resistant prostate cancer. This review focusses on the mechanism of action of AIT, notably to highlight the capacity of the drug to activate DNA damage responses, to inhibit the Hsp90 co-chaperone p23 and to modulate the expression of several microRNA. The interconnexion between these effects is discussed. The unique capacity of AIT to downregulate oncogenic miR-21 and to upregulate the tumor suppressor miRNAs miR-126, miR-148a, miR-195, and miR-449a is presented. AIT exploits several microRNAs to exert its anticancer effects in distinct tumor types. AIT is one of the rare antitumor natural products that binds to and strongly inhibits cochaperone p23, opening interesting perspectives to treat cancers. However, the toxicity profile of the molecule may limit its development as an anticancer drug, unless it can be properly formulated to prevent AIT-induced gastro-intestinal damages in particular. The antitumor properties of AIT and analogs are underlined, with the aim to encourage further pharmacological studies with this underexplored natural product and related quassinoids. HIGHLIGHTS: Ailanthone (AIT) is an anticancer quassinoid isolated from Ailanthus altissima It inhibits proliferation and induces cell death of many cancer cell types The drug activates DNA damage response and targets p23 cochaperone Up or downregulation of several microRNA by AIT contributes to the anticancer activity Analogs or specific formulations must be developed to prevent the toxicity of AIT.
Collapse
|
43
|
Cucci MA, Grattarola M, Dianzani C, Damia G, Ricci F, Roetto A, Trotta F, Barrera G, Pizzimenti S. Ailanthone increases oxidative stress in CDDP-resistant ovarian and bladder cancer cells by inhibiting of Nrf2 and YAP expression through a post-translational mechanism. Free Radic Biol Med 2020; 150:125-135. [PMID: 32101771 DOI: 10.1016/j.freeradbiomed.2020.02.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
Chemoresistance represents one of the main obstacles in treating several types of cancer, including bladder and ovarian cancers, and it is characterized by an increase of cellular antioxidant potential. Nrf2 and YAP proteins play an important role in increasing chemoresistance and in inducing antioxidant enzymes. It has been reported that Ailanthone (Aila), a compound extracted from the Ailanthus Altissima, has an anticancer activity toward several cancer cell lines, including chemoresistant cell lines. We have examined the effect of Aila on proliferation, migration and expression of Nrf2 and YAP proteins in A2780 (CDDP-sensitive) and A2780/CP70 (CDDP-resistant) ovarian cancer cells. Furthermore, to clarify the mechanism of Aila action we extended our studies to sensitive and CDDP-resistant 253J-BV bladder cancer cells, which have been used in a previous study on the effect of Aila. Results demonstrated that Aila exerted an inhibitory effect on growth and colony formation of sensitive and CDDP-resistant ovarian cancer cells and reduced oriented cell migration with higher effectiveness in CDDP resistant cells. Moreover, Aila strongly reduced Nrf2 and YAP protein expression and reduced the expression of the Nrf2 target GSTA4, and the YAP/TEAD target survivin. In CDDP-resistant ovarian and bladder cancer cells the intracellular oxidative stress level was lower with respect to the sensitive cells. Moreover, Aila treatment further reduced the superoxide anion content of CDDP-resistant cells in correlation with the reduction of Nrf2 and YAP proteins. However, Aila treatment increased Nrf2 and YAP mRNA expression in all cancer cell lines. The inhibition of proteolysis by MG132, a proteasoma inhibitor, restored Nrf2 and YAP protein expressions, suggesting that the Aila effect was at post-translational level. In accordance with this observation, we found an increase of the Nrf2 inhibitor Keap1, a reduction of p62/SQSTM1, a Nrf2 target which leads Keap1 protein to the autophagic degradation, and a reduction of P-YAP. Moreover, UCHL1 deubiquitinase expression, which was increased in bladder and ovarian resistant cells, was down-regulated by Aila treatment. In conclusion we demonstrated that Aila can reduce proliferation and migration of cancer cells through a mechanism involving a post translational reduction of Nrf2 and YAP proteins which, in turn, entailed an increase of oxidative stress particularly in the chemoresistant lines.
Collapse
Affiliation(s)
- Marie Angèle Cucci
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Margherita Grattarola
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, Università di Torino, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Giovanna Damia
- Istituto di Ricerche Farmacologiche "Mario Negri-IRCCS", Via Mario Negri 2, 20156, Milan, Italy
| | - Francesca Ricci
- Istituto di Ricerche Farmacologiche "Mario Negri-IRCCS", Via Mario Negri 2, 20156, Milan, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Francesco Trotta
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125, Torino, Italy.
| |
Collapse
|
44
|
Girek M, Kłosiński K, Grobelski B, Pizzimenti S, Cucci MA, Daga M, Barrera G, Pasieka Z, Czarnecka K, Szymański P. Novel tetrahydroacridine derivatives with iodobenzoic moieties induce G0/G1 cell cycle arrest and apoptosis in A549 non-small lung cancer and HT-29 colorectal cancer cells. Mol Cell Biochem 2019; 460:123-150. [PMID: 31313023 PMCID: PMC6745035 DOI: 10.1007/s11010-019-03576-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022]
Abstract
A series of nine tetrahydroacridine derivatives with iodobenzoic moiety were synthesized and evaluated for their cytotoxic activity against cancer cell lines—A549 (human lung adenocarcinoma), HT-29 (human colorectal adenocarcinoma) and somatic cell line—EA.hy926 (human umbilical vein cell line). All compounds displayed high cytotoxicity activity against A549 (IC50 59.12–14.87 µM) and HT-29 (IC50 17.32–5.90 µM) cell lines, higher than control agents—etoposide and 5-fluorouracil. Structure–activity relationship showed that the position of iodine in the substituent in the para position and longer linker most strongly enhanced the cytotoxic effect. Among derivatives, 1i turned out to be the most cytotoxic and displayed IC50 values of 14.87 µM against A549 and 5.90 µM against HT-29 cell lines. In hyaluronidase inhibition assay, all compounds presented anti-inflammatory activity, however, slightly lower than reference compound. ADMET prediction showed that almost all compounds had good pharmacokinetic profiles. 1b, 1c and 1f compounds turned out to act against chemoresistance in cisplatin-resistant 253J B-V cells. Compounds intercalated into DNA and inhibited cell cycle in G0/G1 phase—the strongest inhibition was observed for 1i in A549 and 1c in HT-29. Among compounds, the highest apoptotic effect in both cell lines was observed after treatment with 1i. Compounds caused DNA damage and H2AX phosphorylation, which was detected in A549 and HT-29 cells. All research confirmed anticancer properties of novel tetrahydroacridine derivatives and explained a few pathways of their mechanism of cytotoxic action.
Collapse
Affiliation(s)
- Małgorzata Girek
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Karol Kłosiński
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Pabianicka 62, 93-513, Lodz, Poland
| | - Bartłomiej Grobelski
- Animal House, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Marie Angele Cucci
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Martina Daga
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Corso Raffaello 30, 10125, Turin, Italy
| | - Zbigniew Pasieka
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Pabianicka 62, 93-513, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland.
| |
Collapse
|
45
|
Pingyu Z, Binglei J, Qilong J, Tao W, Wei T. Cyclic Stretch Promotes Proliferation and Contraction of Human Bladder Smooth Muscle Cells by Cajal-Mediated c-kit Expression in Interstitial Cells. Med Sci Monit 2019; 25:4784-4792. [PMID: 31249285 PMCID: PMC6612242 DOI: 10.12659/msm.917549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background The present study was performed to assess the effect of mechanical stretch on the proliferation and contractile function of hBSMCs. Material/Methods hBSMCs and ICCs were seeded at 8×104 cells/well in 6-well silicone elastomer-bottomed culture plates coated with type I collagen, and grown to 80% confluence in DMEM/10% FBS and a 5% CO2 humidified atmosphere at 37°C. Cells of hBSMCs and hBSMCs/ICCs of co-culture were then subjected to continuous cycles of stretch-relaxation using a computer-driven, stretch-inducing device. The treated concentration of imatinib was 10 μM. Mechanisms underlying observed hBSMCs contraction were examined using Western blotting and RT-PCR. The 0.1 μM carbachol was separately added to the experimental groups, and 300 s was recorded by laser scanning confocal microscope. Results We found that mechanical stretch increased contraction and proliferation of hBSMCs. Calcium ion activity increased significantly after mechanical stretch. The number of hBSMCs was significantly increased after the combination mechanical stretch with ICCs treatment. After combination mechanical stretch with hBSMCs/ICCs treatment, the mRNA and protein level of M2, M3, and c-kit were significantly increased. After combination of mechanical stretch with no imatinib treatment, the proliferation of hBSMCs was higher than others, and the mRNA and protein level of M2 and M3 were significantly increased. Conclusions We revealed that ICCs could promote hBSMC proliferation and contraction, and cyclic stretch could promote acetylcholine receptor M2 and M3 caused by c-kit in the ICCs, which promoted the contraction of hBSMCs.
Collapse
Affiliation(s)
- Zhu Pingyu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland).,Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Jiang Binglei
- Department of Ultrasound, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Jiang Qilong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Wu Tao
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Tang Wei
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|