1
|
Zhang Z, Li M, Zhang X, Zhou F. Novel Strategies for Tumor Treatment: Harnessing ROS-Inducing Active Ingredients from Traditional Chinese Medicine Through Multifunctional Nanoformulations. Int J Nanomedicine 2024; 19:9659-9688. [PMID: 39309188 PMCID: PMC11416109 DOI: 10.2147/ijn.s479212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Reactive oxygen species (ROS) encompass a diverse group of chemically reactive molecules or ions distinguished by their substantial oxidative potential. Empirical studies have shown that the targeted administration of high toxic concentrations of ROS can effectively induce tumor cell death in various types. Numerous bioactive ingredients derived from traditional Chinese medicine (TCM), recognized for their ROS-inducing properties, have demonstrated significant anti-tumor activity. Nonetheless, their clinical application has been hindered by challenges such as low solubility, limited bioavailability, and poor selectivity. Multifunctional nanoformulations possess the potential to overcome these challenges and enhance the anticancer efficacy of ROS-inducing active compounds. Through extensive searches of various academic databases and a thorough review and screening of relevant literature, this study aims to systematically summarize and generalize multiple active ingredients in TCM that induce ROS generation, along with their multifunctional nanoformulations, from various perspectives. The objective is to provide new insights and references for fundamental cancer research and clinical treatments. Furthermore, we acknowledge that although numerous active ingredients and their nanoformulations in TCM have demonstrated ROS-inducing and anti-tumor potentials, potentially offering novel strategies for tumor therapy, the underlying mechanisms require further comprehensive investigation.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Xiaolong Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
Varlamova EG. Molecular Mechanisms of the Therapeutic Effect of Selenium Nanoparticles in Hepatocellular Carcinoma. Cells 2024; 13:1102. [PMID: 38994955 PMCID: PMC11240755 DOI: 10.3390/cells13131102] [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: 05/13/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
This review describes and summarizes, for the first time, the molecular mechanisms of the cytotoxic effect of selenium nanoparticles of various origins on hepatocellular carcinoma cells. The text provides information from recent years indicating the regulation of various signaling pathways and endoplasmic reticulum stress by selenium nanoparticles; the pathways of cell death of liver cancer cells as a result of exposure to selenium nanoparticles are considered. Particular attention is paid to the participation of selenoproteins and selenium-containing thioredoxin reductases and glutathione peroxidases in these processes. Previously, there were no reviews that fully reflected the cytotoxic effects of selenium nanoparticles specifically in hepatocellular carcinoma, despite the fact that many reviews and experimental articles have been devoted to the causes of this disease and the molecular mechanisms of regulation of cytotoxic effects by other agents. The relevance of this review is primarily explained by the fact that despite the development of various drugs and approaches for the treatment and prevention of hepatocellular carcinoma, this disease is still the fourth leading cause of death in the world. For this reason, a complete understanding of the latest trends in the treatment of oncology of various etiologies, especially hepatocellular carcinoma, is extremely important.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290 Pushchino, Russia
| |
Collapse
|
3
|
Khaledizade E, Tafvizi F, Jafari P. Anti-breast cancer activity of biosynthesized selenium nanoparticles using Bacillus coagulans supernatant. J Trace Elem Med Biol 2024; 82:127357. [PMID: 38103517 DOI: 10.1016/j.jtemb.2023.127357] [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: 06/21/2023] [Revised: 10/07/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND In the present study, Selenium Nanoparticles (SeNPs) were prepared using Bacillus coagulans, which is a type of Lactic Acid Bacteria (LAB), and then they were applied to treat breast cancer cells. METHODS The chemicophysical properties of the bioengineered SeNPs were investigated by Transmission Electron Microscopy (TEM), Field Emission Scanning Electron Microscopy (FE-SEM), zeta potential, dynamic light scattering, Fourier Transform Infrared Spectroscopy (FT-IR), energy dispersive X-ray spectroscopy (EDX) and X-ray diffraction analysis (XRD). The cytotoxic potential of SeNPs was evaluated by MTT assay against MCF-7 breast cancer cell line. The expression levels of apoptotic genes including BAX, BCL2, VEGF, ERBB2, CASP3, CASP9, CCNE1, CCND1, MMP2 and MMP9 were determined by real-time PCR. The rate of apoptosis and necrosis of the cancer cells as well as the results of the cell cycle were evaluated by flow cytometry method. RESULTS The synthesized SeNPs had an average particle size of about 24-40 nm and a zeta potential of -16.1 mV, indicating the high stability of SeNPs. EDX results showed presence of SeNPs because amount of selenium in SeNPs was 86.6 % by weight. The cytotoxicity results showed a concentration-dependent effect against MCF-7 cells. The half-maximal inhibitory concentration (IC50) values of B. coagulans supernatant and SeNPs against breast cancer cells were 389.7 µg/mL and 17.56 µg/mL, respectively. In addition, SeNPs synthesized by the green process exhibited enhanced apoptotic potential in MCF-7 cancer cells compared with bacterial supernatants. Cancer cells treated with IC50 concentration of SeNPs induced 32 % apoptosis compared to untreated cells (3 % apoptosis). The gene expression levels of BAX, CASP3, and CASP9 were upregulated, while the expression levels of BCL2, CCNE1, CCND1, MMP2, MMP9, VEGF, and ERBB2 were downregulated after SeNPs treatment of cells. The potential of SeNPs to induce cell apoptosis was demonstrated by the increase in the expression level of BAX gene and the decrease in the expression level of BCL2 after treatment of cancer cells with SeNPs. CONCLUSION The obtained results indicated that SeNPs had strong potential to induce significant cell apoptosis and are cytotoxic against the MCF-7 cancer cell line.
Collapse
Affiliation(s)
- Elaheh Khaledizade
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Farzaneh Tafvizi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran.
| | - Parvaneh Jafari
- Microbiology Department, Faculty of Science, Arak Branch, Islamic Azad University, Arak, Iran
| |
Collapse
|
4
|
Lu S, Huang J, Zhang J, Wu C, Huang Z, Tao X, You L, Stalin A, Chen M, Li J, Tan Y, Wu Z, Geng L, Li Z, Fan Q, Liu P, Lin Y, Zhao C, Wu J. The anti-hepatocellular carcinoma effect of Aidi injection was related to the synergistic action of cantharidin, formononetin, and isofraxidin through BIRC5, FEN1, and EGFR. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117209. [PMID: 37757991 DOI: 10.1016/j.jep.2023.117209] [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: 08/04/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aidi injection (ADI) is a popular anti-tumor Chinese patent medicine, widely used in clinics for the treatment of hepatocellular carcinoma (HCC) with remarkable therapeutic effects through multiple targets and pathways. However, the scientific evidence of the synergistic role of the complex chemical component system and the potential mechanism for treating diseases are ignored and remain to be elucidated. AIM OF THE STUDY This study aimed to elucidate and verify the cooperative association between the potential active ingredient of ADI, which is of significance to enlarge our understanding of its anti-HCC molecular mechanisms. MATERIALS AND METHODS Firstly, the anti-HCC effect of ADI was evaluated in various HCC cells and the zebrafish xenograft model. Subsequently, a variety of bioinformatic technologies, including network pharmacology, weighted gene co-expression network analysis (WGCNA), meta-analysis of gene expression profiles, and pathway enrichment analysis were performed to construct the competitive endogenous RNA (ceRNA) network of ADI intervention in HCC and to establish the relationship between the critical targets/pathways and the key corresponding components, which were involved in ADI against HCC in a synergistic way and were validated by molecular biology experiments. RESULTS ADI exerted remarkable anti-HCC in vitro cells and in vivo zebrafish model, especially that the Hep 3B2.1-7 cell showed substantial sensibility to ADI. The ceRNA network revealed that the EGFR/PI3K/AKT signaling pathway was identified as the promising pathway. Furthermore, the meta-analysis also demonstrated the critical role of BIRC5 and FEN1 as key targets. Finally, the synergistic effect of ADI was revealed by discovering the inhibitory effect of cantharidin on BIRC5, formononetin on FEN1 and EGFR, as well as isofraxidin on EGFR. CONCLUSION Our study unveiled that the incredible protective effect of ADI on HCC resulted from the synergistic inhibition effect of cantharidin, formononetin, and isofraxidin on multiple targets/pathways, including BIRC5, FEN1, and EGFR/PI3K/AKT, respectively, providing a scientific interpretation of ADI against HCC and a typical example of pharmacodynamic evaluation of other proprietary Chinese patent medicine.
Collapse
Affiliation(s)
- Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiaqi Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Xiaoyu Tao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Meilin Chen
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiaqi Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Libo Geng
- Guizhou Yibai Pharmaceutical Co. Ltd, Guiyang, 550008, Guizhou, China.
| | - Zhiqi Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Qiqi Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Pengyun Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yifan Lin
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chongjun Zhao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
5
|
Hu X, Huang S, Ye S, Jiang J. The Natural Product Oridonin as an Anticancer Agent: Current Achievements and Problems. Curr Pharm Biotechnol 2024; 25:655-664. [PMID: 37605407 DOI: 10.2174/1389201024666230821110116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 08/23/2023]
Abstract
Oridonin, an active diterpenoid isolated from traditional Chinese herbal medicine, has received a rising attention for its remarkable roles in cancer therapy. In recent years, increasing evidences have revealed that oridonin inhibits the occurrence and development of tumor cells through multiple mechanisms, including induction of apoptosis and autophagy, cell cycle arrest, and inhibition of angiogenesis as well as migration and invasion. In addition, several molecular signal targets have been identified, including ROS, EGFR, NF-κB, PI3K/Akt, and MAPK. In this paper, we review considerable knowledge about the molecular mechanisms and signal targets of oridonin, which has been studied in recent years. It is expected that oridonin may be developed as a novel anti-tumor herbal medicine in human cancer treatment.
Collapse
Affiliation(s)
- Xiangyan Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
| | - Sisi Huang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Pharmacy School of Fudan University, Shanghai, 200032, P.R. China
| | - Shiying Ye
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
| | - Jinhuan Jiang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
| |
Collapse
|
6
|
Tang B, Ma W, Lin Y. Emerging applications of anti-angiogenic nanomaterials in oncotherapy. J Control Release 2023; 364:61-78. [PMID: 37871753 DOI: 10.1016/j.jconrel.2023.10.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Angiogenesis is the process of generating new blood vessels from pre-existing vasculature. Under normal conditions, this process is delicately controlled by pro-angiogenic and anti-angiogenic factors. Tumor cells can produce plentiful pro-angiogenic molecules promoting pathological angiogenesis for uncontrollable growth. Therefore, anti-angiogenic therapy, which aims to inhibit tumor angiogenesis, has become an attractive approach for oncotherapy. However, classic anti-angiogenic agents have several limitations in clinical use, such as lack of specific targeting, low bioavailability, and poor therapeutic outcomes. Hence, alternative angiogenic inhibitors are highly desired. With the emergence of nanotechnology, various nanomaterials have been designed for anti-angiogenesis purposes, offering promising features like excellent targeting capabilities, reduced side effects, and enhanced therapeutic efficacy. In this review, we describe tumor vascular features, discuss current dilemma of traditional anti-angiogenic medicines in oncotherapy, and underline the potential of nanomaterials in tumor anti-angiogenic therapy. Moreover, we discuss the current challenges of anti-angiogenic cancer treatment. We expect that this summary of anti-angiogenic nanomaterials in oncotherapy will offer valuable insights, facilitating their extensive applications in the future.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
7
|
Xiao Q, Zhang Y, Zhao A, Duan Z, Yao J. Application and development of nanomaterials in the diagnosis and treatment of esophageal cancer. Front Bioeng Biotechnol 2023; 11:1268454. [PMID: 38026877 PMCID: PMC10657196 DOI: 10.3389/fbioe.2023.1268454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Esophageal cancer is a malignant tumor with a high incidence worldwide. Currently, there are a lack of effective early diagnosis and treatment methods for esophageal cancer. However, delivery systems based on nanoparticles (NPs) have shown ideal efficacy in real-time imaging and chemotherapy, radiotherapy, gene therapy, and phototherapy for tumors, which has led to their recent widespread design as novel treatment strategies. Compared to traditional drugs, nanomedicine has unique advantages, including strong targeting ability, high bioavailability, and minimal side effects. This article provides an overview of the application of NPs in the diagnosis and treatment of esophageal cancer and provides a reference for future research.
Collapse
Affiliation(s)
| | | | | | | | - Jun Yao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
8
|
Verimli N, Goralı Sİ, Abisoglu B, Altan CL, Sucu BO, Karatas E, Tulek A, Bayraktaroglu C, Beker MC, Erdem SS. Development of light and pH-dual responsive self-quenching theranostic SPION to make EGFR overexpressing micro tumors glow and destroy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 248:112797. [PMID: 37862898 DOI: 10.1016/j.jphotobiol.2023.112797] [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: 06/01/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023]
Abstract
Drug resistant and undetectable tumors easily escape treatment leading metastases and/or recurrence of the lethal disease. Therefore, it is vital to diagnose and destroy micro tumors using simple yet novel approaches. Here, we present fluorescence-based detection and light-based destruction of cancer cells that are known to be resistant to standard therapies. We developed a superparamagnetic iron oxide nanoparticle (SPION)-based theranostic agent that is composed of self-quenching light activated photosensitizer (BPD) and EGFR targeting ligand (Anti-EGFR ScFv or GE11 peptide). Photosensitizer (BPD) was immobilized to PEG-PEI modified SPION with acid-labile linker. Prior to stimulation of the theranostic system by light its accumulation within cancer cells is vital since BPD phototoxicity and fluorescence is activated by lysosomal proteolysis. As BPD is cleaved, the system switches from off to on position which triggers imaging and therapy. Targeting, therapeutic and diagnostic features of the theranostic system were evaluated in high and moderate level EGFR expressing pancreatic cancer cell lines. Our results indicate that the system distinguishes high and moderate EGFR expression levels and yields up to 4.3-fold increase in intracellular fluorescence intensity. Amplification of fluorescence signal was as low as 1.3-fold in the moderate or no EGFR expressing cell lines. Anti-EGFR ScFv targeted SPION caused nearly 2-fold higher cell death via apoptosis in high EGFR expressing Panc-1 cell line. The developed system, possessing advanced targeting, enhanced imaging and effective therapeutic features, is a promising candidate for multi-mode detection and destruction of residual drug-resistant cancer cells.
Collapse
Affiliation(s)
- Nihan Verimli
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey; International School of Medicine, Medical Biochemistry, Istanbul Medipol University, 34810 Istanbul, Turkey
| | - S İrem Goralı
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey; International School of Medicine, Medical Biochemistry, Istanbul Medipol University, 34810 Istanbul, Turkey
| | - Beyza Abisoglu
- Department of Chemical Engineering, Yeditepe University, Atasehir, Istanbul 34755, Turkey
| | - Cem Levent Altan
- Department of Chemical Engineering, Yeditepe University, Atasehir, Istanbul 34755, Turkey
| | - Bilgesu Onur Sucu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul Medipol University, Istanbul, Turkey; Center of Drug Discovery and Development, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Ersin Karatas
- Ağrı İbrahim Çeçen University, Patnos Vocational School, Department of Medical Services and Techniques, Ağrı, Turkey
| | - Ahmet Tulek
- Iğdır University, Vocational School of Health Services, Department of Care Services, Iğdır, Turkey
| | - Cigdem Bayraktaroglu
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey
| | - Mustafa Caglar Beker
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey
| | - S Sibel Erdem
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey; International School of Medicine, Medical Biochemistry, Istanbul Medipol University, 34810 Istanbul, Turkey.
| |
Collapse
|
9
|
Shen L, Liao K, Yang E, Yang F, Lin W, Wang J, Fan S, Huang X, Chen L, Shen H, Jin H, Ruan Y, Liu X, Zeng G, Xu JF, Pi J. Macrophage targeted iron oxide nanodecoys augment innate immunological and drug killings for more effective Mycobacterium Tuberculosis clearance. J Nanobiotechnology 2023; 21:369. [PMID: 37817142 PMCID: PMC10563239 DOI: 10.1186/s12951-023-02103-x] [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: 08/01/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, is still one of the top killers worldwide among infectious diseases. The escape of Mtb from immunological clearance and the low targeting effects of anti-TB drugs remain the substantial challenges for TB control. Iron is particularly required for Mtb growth but also toxic for Mtb in high dosages, which makes iron an ideal toxic decoy for the 'iron-tropic' Mtb. Here, a macrophage-targeted iron oxide nanoparticles (IONPs)-derived IONPs-PAA-PEG-MAN nanodecoy is designed to augment innate immunological and drug killings against intracellular Mtb. IONPs-PAA-PEG-MAN nanodecoy exhibits preferential uptake in macrophages to significantly increase drug uptake with sustained high drug contents in host cells. Moreover, it can serve as a specific nanodecoy for the 'iron-tropic' Mtb to realize the localization of Mtb contained phagosomes surrounding the drug encapsulated nanodecoys and co-localization of Mtb with the drug encapsulated nanodecoys in lysosomes, where the incorporated rifampicin (Rif) can be readily released under acidic lysosomal condition for enhanced Mtb killing. This drug encapsulated nanodecoy can also polarize Mtb infected macrophages into anti-mycobacterial M1 phenotype and enhance M1 macrophage associated pro-inflammatory cytokine (TNF-α) production to trigger innate immunological responses against Mtb. Collectively, Rif@IONPs-PAA-PEG-MAN nanodecoy can synergistically enhance the killing efficiency of intracellular Mtb in in vitro macrophages and ex vivo monocyte-derived macrophages, and also significantly reduce the mycobacterial burdens in the lung of infected mice with alleviated pathology. These results indicate that Rif@IONPs-PAA-PEG-MAN nanodecoy may have a potential for the development of more effective therapeutic strategy against TB by manipulating augmented innate immunity and drug killings.
Collapse
Affiliation(s)
- Ling Shen
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Kangsheng Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Enzhuo Yang
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fen Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Wensen Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiajun Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xueqin Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Lingming Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xing Liu
- Key Laboratory of Animal Disease Diagnostics and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gucheng Zeng
- Department of Microbiology, Zhongshan School of Medicine, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China.
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
10
|
Rathnayake K, Patel U, Hunt EC, Singh N. Fabrication of a Dual-Targeted Liposome-Coated Mesoporous Silica Core-Shell Nanoassembly for Targeted Cancer Therapy. ACS OMEGA 2023; 8:34481-34498. [PMID: 37779923 PMCID: PMC10536893 DOI: 10.1021/acsomega.3c02901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Nanoparticles have been suggested as drug-delivery systems for chemotherapeutic drugs to allow for controlled drug release profiles and selectivity to target cancer cells. In addition, nanoparticles can be used for the in situ generation and amplification of reactive oxygen species (ROS), which have been shown to be a promising strategy for cancer treatment. Thus, a targeted nanoscale drug-delivery platform could be used to synergistically improve cancer treatment by the action of chemotherapeutic drugs and ROS generation. Herein, we propose a promising chemotherapy strategy where the drug-loaded nanoparticles generate high doses of ROS together with the loaded ROS-generating chemotherapeutic drugs, which can damage the mitochondria and activate cell death, potentiating the therapeutic outcome in cancer therapy. In the present study, we have developed a dual-targeted drug-delivery nanoassembly consisting of a mesoporous silica core loaded with the chemotherapeutic, ROS-generating drug, paclitaxel (Px), and coated with a liposome layer for controlled drug release. Two different lung cancer-targeting ligands, folic acid and peptide GE11, were used to target the overexpressed nonsmall lung cancer receptors to create the final nanoassembly (MSN@Px) L-GF. Upon endocytosis by the cancer cells, the liposome layer was degraded by the intracellular lipases, and the drug was rapidly released at a rate of 65% within the first 20 h. In vitro studies confirmed that this nanoassembly was 8-fold more effective in cancer therapy compared to the free drug Px.
Collapse
Affiliation(s)
- Kavini Rathnayake
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Unnati Patel
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Emily C. Hunt
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Nirupama Singh
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| |
Collapse
|
11
|
Zhang L, Li X, Yue G, Guo L, Hu Y, Cui Q, Wang J, Tang J, Liu H. Nanodrugs systems for therapy and diagnosis of esophageal cancer. Front Bioeng Biotechnol 2023; 11:1233476. [PMID: 37520291 PMCID: PMC10373894 DOI: 10.3389/fbioe.2023.1233476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023] Open
Abstract
With the increasing incidence of esophageal cancer, its diagnosis and treatment have become one of the key issues in medical research today. However, the current diagnostic and treatment methods face many unresolved issues, such as low accuracy of early diagnosis, painful treatment process for patients, and high recurrence rate after recovery. Therefore, new methods for the diagnosis and treatment of esophageal cancer need to be further explored, and the rapid development of nanomaterials has brought new ideas for solving this problem. Nanomaterials used as drugs or drug delivery systems possess several advantages, such as high drug capacity, adjustably specific targeting capability, and stable structure, which endow nanomaterials great application potential in cancer therapy. However, even though the nanomaterials have been widely used in cancer therapy, there are still few reviews on their application in esophageal cancer, and systematical overview and analysis are deficient. Herein, we overviewed the application of nanodrug systems in therapy and diagnosis of esophageal cancer and summarized some representative case of their application in diagnosis, chemotherapy, targeted drug, radiotherapy, immunity, surgery and new therapeutic method of esophageal cancer. In addition, the nanomaterials used for therapy of esophageal cancer complications, esophageal stenosis or obstruction and oesophagitis, are also listed here. Finally, the challenge and the future of nanomaterials used in cancer therapy were discussed.
Collapse
Affiliation(s)
- Lihan Zhang
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xing Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Guangxing Yue
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lihao Guo
- Interdisciplinary Research Center of Smart Sensors, School of Advanced Materials and Nanotechnology, Xidian University, Xi’an, China
| | - Yanhui Hu
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Qingli Cui
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jia Wang
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jingwen Tang
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Huaimin Liu
- Department of Integrated Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
12
|
Toubhans B, Alkafri N, Quintela M, James DW, Bissardon C, Gazze S, Knodel F, Proux O, Gourlan AT, Rathert P, Bohic S, Gonzalez D, Francis LW, Charlet L, Conlan RS. Selenium nanoparticles modulate histone methylation via lysine methyltransferase activity and S-adenosylhomocysteine depletion. Redox Biol 2023; 61:102641. [PMID: 36842241 PMCID: PMC9988660 DOI: 10.1016/j.redox.2023.102641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
At physiological levels, the trace element selenium plays a key role in redox reactions through the incorporation of selenocysteine in antioxidant enzymes. Selenium has also been evaluated as a potential anti-cancer agent, where selenium nanoparticles have proven effective, and are well tolerated in vivo at doses that are toxic as soluble Se. The use of such nanoparticles, coated with either serum albumin or the naturally occurring alkaline polysaccharide chitosan, also serves to enhance biocompatibility and bioavailability. Here we demonstrate a novel role for selenium in regulating histone methylation in ovarian cancer cell models treated with inorganic selenium nanoparticles coated with serum albumin or chitosan. As well as inducing thioredoxin reductase expression, ROS activity and cancer cell cytotoxicity, coated nanoparticles caused significant increases in histone methylation. Specifically, selenium nanoparticles triggered an increase in the methylation of histone 3 at lysines K9 and K27, histone marks involved in both the activation and repression of gene expression, thus suggesting a fundamental role for selenium in these epigenetic processes. This direct function was confirmed using chemical inhibitors of the histone lysine methyltransferases EZH2 (H3K27) and G9a/EHMT2 (H3K9), both of which blocked the effect of selenium on histone methylation. This novel role for selenium supports a distinct function in histone methylation that occurs due to a decrease in S-adenosylhomocysteine, an endogenous inhibitor of lysine methyltransferases, the metabolic product of methyl-group transfer from S-adenosylmethionine in the one-carbon metabolism pathway. These observations provide important new insights into the action of selenium nanoparticles. It is now important to consider both the classic antioxidant and novel histone methylation effects of this key redox element in its development in cancer therapy and other applications.
Collapse
Affiliation(s)
- Benoit Toubhans
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK; Université Grenoble Alpes, ISTerre, 38000, Grenoble, France
| | - Nour Alkafri
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Marcos Quintela
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - David W James
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Caroline Bissardon
- Université Grenoble Alpes, INSERM, UA7 STROBE, Synchrotron Radiation for Biomedicine, Grenoble, France
| | - Salvatore Gazze
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Franziska Knodel
- Department of Biochemistry, Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, D-70550, Stuttgart, Germany
| | - Olivier Proux
- OSUG, UAR 832 CNRS, Université Grenoble Alpes, 38041, Grenoble, France
| | | | - Philipp Rathert
- Department of Biochemistry, Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, D-70550, Stuttgart, Germany
| | - Sylvain Bohic
- Université Grenoble Alpes, INSERM, UA7 STROBE, Synchrotron Radiation for Biomedicine, Grenoble, France; ESRF, European Synchrotron Radiation Facility, CS, 40220, 38043, Grenoble, Cedex 9, France
| | - Deyarina Gonzalez
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Lewis W Francis
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | | | - R Steven Conlan
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK.
| |
Collapse
|
13
|
Toprakcioglu Z, Wiita EG, Jayaram AK, Gregory RC, Knowles TPJ. Selenium Silk Nanostructured Films with Antifungal and Antibacterial Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10452-10463. [PMID: 36802477 PMCID: PMC9982822 DOI: 10.1021/acsami.2c21013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
The rapid emergence of drug-resistant bacteria and fungi poses a threat for healthcare worldwide. The development of novel effective small molecule therapeutic strategies in this space has remained challenging. Therefore, one orthogonal approach is to explore biomaterials with physical modes of action that have the potential to generate antimicrobial activity and, in some cases, even prevent antimicrobial resistance. Here, to this effect, we describe an approach for forming silk-based films that contain embedded selenium nanoparticles. We show that these materials exhibit both antibacterial and antifungal properties while crucially also remaining highly biocompatible and noncytotoxic toward mammalian cells. By incorporating the nanoparticles into silk films, the protein scaffold acts in a 2-fold manner; it protects the mammalian cells from the cytotoxic effects of the bare nanoparticles, while also providing a template for bacterial and fungal eradication. A range of hybrid inorganic/organic films were produced and an optimum concentration was found, which allowed for both high bacterial and fungal death while also exhibiting low mammalian cell cytotoxicity. Such films can thus pave the way for next-generation antimicrobial materials for applications such as wound healing and as agents against topical infections, with the added benefit that bacteria and fungi are unlikely to develop antimicrobial resistance to these hybrid materials.
Collapse
Affiliation(s)
- Zenon Toprakcioglu
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Elizabeth G. Wiita
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Akhila K. Jayaram
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- Cavendish
Laboratory, Department of Physics, University
of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Rebecca C. Gregory
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Tuomas P. J. Knowles
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- Cavendish
Laboratory, Department of Physics, University
of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| |
Collapse
|
14
|
Ogunleye GE, Adebayo-Tayo BC, Oyinlola KA. Biological evaluation of extracellular mycosynthesized silver nanoparticles by Trichoderma asperellum. Biometals 2023; 36:97-109. [PMID: 36414828 DOI: 10.1007/s10534-022-00463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
Mycosynthesis of silver nano-scale particles by fungi is an important aspect in nanotechnology due to its eco-friendly, safe, and cost-effective nature. It also provides protein-capped nanoparticles, which are very stable and have good dispersion. The Mycosynthesis of silver nano-scale particles (SNPs) by Trichoderma asperellum Filtrate (TAF) was evaluated. The characterizations of TAFSNPs were done by UV-visible spectroscopy, FTIR, SEM, and EDX. Biological evaluations such as antibacterial activity against some pathogens, in-vivo immuno-stimulatory, and antitumor potential and hematological parameters of SNPs of T. asperellum were investigated. Changes in color from yellow to brown indicate the formation of nanoparticles. TAFSNPs biosynthesis had a Surface Plasmon Resonance peak at 450 nm. The nanoparticles were spherical, with a size ranging from 0.7 to 10.0 nm. EDX analysis revealed a strong signal for the silver element. TAFSNPs had profound antagonistic activity against the test microorganism. B. subtilis, S. aureus, and Salmonella typhi were highly susceptible with inhibition zone of 24.3 mm, 21.8 mm, and 21.3 mm. TAFSNPs had significant immune-stimulation in the treated mice with an increase in IgA and IgM production, which ranged from 169 0.83d-269 0.56a mg/dl and 26 0.16d-69 0.27a mg/dl. TAFSNPs had CEA level reduction capability (3.62-4.05 ng/L). TAFSNPs had the highest amounts of PCV, Red blood cells, heamoglobin, and neutrophils (38%, 5.7 × 1012%, 14.09 g/dl, and 71%). Significant improvement was recorded on the lifespan of the TAFSNPs treated tumor-induced mice. Conclusively, TAFSNPs have good antimicrobial, immuno-stimulatory, and antitumor potential.
Collapse
|
15
|
Zambonino MC, Quizhpe EM, Mouheb L, Rahman A, Agathos SN, Dahoumane SA. Biogenic Selenium Nanoparticles in Biomedical Sciences: Properties, Current Trends, Novel Opportunities and Emerging Challenges in Theranostic Nanomedicine. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:424. [PMID: 36770385 PMCID: PMC9921003 DOI: 10.3390/nano13030424] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Selenium is an important dietary supplement and an essential trace element incorporated into selenoproteins with growth-modulating properties and cytotoxic mechanisms of action. However, different compounds of selenium usually possess a narrow nutritional or therapeutic window with a low degree of absorption and delicate safety margins, depending on the dose and the chemical form in which they are provided to the organism. Hence, selenium nanoparticles (SeNPs) are emerging as a novel therapeutic and diagnostic platform with decreased toxicity and the capacity to enhance the biological properties of Se-based compounds. Consistent with the exciting possibilities offered by nanotechnology in the diagnosis, treatment, and prevention of diseases, SeNPs are useful tools in current biomedical research with exceptional benefits as potential therapeutics, with enhanced bioavailability, improved targeting, and effectiveness against oxidative stress and inflammation-mediated disorders. In view of the need for developing eco-friendly, inexpensive, simple, and high-throughput biomedical agents that can also ally with theranostic purposes and exhibit negligible side effects, biogenic SeNPs are receiving special attention. The present manuscript aims to be a reference in its kind by providing the readership with a thorough and comprehensive review that emphasizes the current, yet expanding, possibilities offered by biogenic SeNPs in the biomedical field and the promise they hold among selenium-derived products to, eventually, elicit future developments. First, the present review recalls the physiological importance of selenium as an oligo-element and introduces the unique biological, physicochemical, optoelectronic, and catalytic properties of Se nanomaterials. Then, it addresses the significance of nanosizing on pharmacological activity (pharmacokinetics and pharmacodynamics) and cellular interactions of SeNPs. Importantly, it discusses in detail the role of biosynthesized SeNPs as innovative theranostic agents for personalized nanomedicine-based therapies. Finally, this review explores the role of biogenic SeNPs in the ongoing context of the SARS-CoV-2 pandemic and presents key prospects in translational nanomedicine.
Collapse
Affiliation(s)
- Marjorie C. Zambonino
- School of Biological Sciences and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| | - Ernesto Mateo Quizhpe
- School of Biological Sciences and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| | - Lynda Mouheb
- Laboratoire de Recherche de Chimie Appliquée et de Génie Chimique, Hasnaoua I, Université Mouloud Mammeri, BP 17 RP, Tizi-Ouzou 15000, Algeria
| | - Ashiqur Rahman
- Center for Midstream Management and Science, Lamar University, 211 Redbird Ln., Beaumont, TX 77710, USA
| | - Spiros N. Agathos
- Earth and Life Institute, Catholic University of Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Si Amar Dahoumane
- Department of Chemical Engineering, Polytechnique Montréal, C.P. 6079, Succ. Centre-Ville, Montréal, QC H3C 3A7, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, 18, Ave Antonine-Maillet, Moncton, NB E1A 3E9, Canada
| |
Collapse
|
16
|
An engineered three-in-one hybrid nanosystem from elastin-like polypeptides for enhanced cancer suppression. Colloids Surf B Biointerfaces 2022. [DOI: 10.1016/j.colsurfb.2022.112976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
Progress in the Surface Functionalization of Selenium Nanoparticles and Their Potential Application in Cancer Therapy. Antioxidants (Basel) 2022; 11:antiox11101965. [PMID: 36290687 PMCID: PMC9598587 DOI: 10.3390/antiox11101965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
As an essential micronutrient, selenium participates in numerous life processes and plays a key role in human health. In the past decade, selenium nanoparticles (SeNPs) have attracted great attention due to their excellent functionality for potential applications in pharmaceuticals. However, the utilization of SeNPs has been restricted by their instability and low targeting ability. Since the existing reviews mainly focused on the applications of SeNPs, this review highlights the synthesis of SeNPs and the strategies to improve their stability and targeting ability through surface functionalization. In addition, the utilization of functionalized SeNPs for the single and co-delivery of drugs or genes to achieve the combination of therapy are also presented, with the emphasis on the potential mechanism. The current challenges and prospects of functionalized SeNPs are also summarized. This review may provide valuable information for the design of novel functionalized SeNPs and promote their future application in cancer therapy.
Collapse
|
18
|
Fakhrolmobasheri M, Mazaheri-Tehrani S, Kieliszek M, Zeinalian M, Abbasi M, Karimi F, Mozafari AM. COVID-19 and Selenium Deficiency: a Systematic Review. Biol Trace Elem Res 2022; 200:3945-3956. [PMID: 34739678 PMCID: PMC8569840 DOI: 10.1007/s12011-021-02997-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/24/2021] [Indexed: 12/16/2022]
Abstract
Several studies have indicated that selenium deficiency may be detrimental in the context of various viral disorders, and in the case of COVID-19, several studies have reported heterogeneous results concerning the association of selenium deficiency with the severity of disease. To summarize the available data surrounding the association of body selenium levels with the outcomes of COVID-19, a systematic search was performed in the Medline database (PubMed), Scopus, Cochrane Library, Embase, and Web of Science using keywords including "SARS-CoV-2," "COVID-19," and "selenium," Studies evaluating the association of COVID-19 with body selenium levels were included. Among 1,862 articles viewed in the database search, 10 articles were included after title, abstract, and full-text review. One study was further included after searching the literature again for any newly published articles. Out of 11 included studies, 10 studies measured serum selenium level, and one study investigated urinary selenium level. Three of 10 studies measured serum SELENOP level as well as selenium level. Glutathione peroxidase-3 level in serum was also assessed in one study. The reported outcomes were severity, mortality, and risk of COVID-19. Nine studies indicated that a lower serum selenium level is associated with worse outcomes. Two studies reported no significant association between serum selenium level and COVID-19. In one study, urinary selenium level was reported to be higher in severe and fatal cases compared to non-severe and recovered patients, respectively. In most cases, selenium deficiency was associated with worse outcomes, and selenium levels in COVID-19 patients were lower than in healthy individuals. Thus, it could be concluded that cautious selenium supplementation in COVID-19 patients may be helpful to prevent disease progression. However, randomized clinical trials are needed to confirm this.
Collapse
Affiliation(s)
| | - Sadegh Mazaheri-Tehrani
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, 81746-73461, Isfahan, Iran
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159C, 02-776, Warsaw, Poland.
| | - Mehrdad Zeinalian
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, 81746-73461, Isfahan, Iran.
| | - Mehdi Abbasi
- School of Medicine, Isfahan University of Medical Sciences, 81746-73461, Esfahan, Iran
| | - Fateme Karimi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, 81746-73461, Isfahan, Iran
| | - Amir Mohamad Mozafari
- Medical Library and Information Sciences Department, Health Information Technology Research Center, School of Management and Medical Information Sciences, Isfahan University of Medical Sciences, 81746-73461, Isfahan, Iran
| |
Collapse
|
19
|
Cai M, Liang W, Wang K, Yin D, Fu T, Zhu R, Qu C, Dong X, Ni J, Yin X. Aperture Modulation of Isoreticular Metal Organic Frameworks for Targeted Antitumor Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36366-36378. [PMID: 35897121 DOI: 10.1021/acsami.2c07450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The introduction of different pore diameters in metal organic frameworks (MOFs) could adjust their drug delivery performance. MOFs with customized structures have potential application value in targeted drug delivery. However, no research on this topic has been found so far. In this report, isoreticular metal organic frameworks (IRMOFs) have been taken as a typical case of tailor-made MOFs, the pore size of which is enlarged (average BJH pore sizes of about 2.43, 3.06, 5.47, and 6.50 nm were determined for IRMOF-1, IRMOF-8, IRMOF-10, and IRMOF-16, respectively), emphasizing the relationship between pore size and model drugs (Oridonin, ORI) and clarifying its potential working mechanism. IRMOF-1, whose pore size matches the size of ORI, has an outstanding drug loading capacity (57.93% by wt) and release profile (about 90% in 24 h at pH 7.4). IRMOF-1 was further coated with polyethylene glycol (PEG) modified with a cell penetrating peptide (CPP44) bound to M160 (CD163L1) protein for targeting of hepatic tumor lines. This nanoplatform (CPP44-PEG@ORI@IRMOF-1) exhibited acid-responsive drug release behavior (37.86% in 10 h at pH 7.4 and 66.66% in 10 h at pH 5.5) and significantly enhanced antitumor effects. The results of cell targeting and in vivo animal imaging indicated that CPP44-PEG@ORI@IRMOF-1 may serve as a tumor-selective drug delivery nanoplatform. Toxicity assessment confirmed that PEGylated IRMOF-1 did not cause organ or systemic toxicity. Furthermore, it is encouraging that the IRMOF-based targeted drug delivery system with pore size modulation showed rapid clearance (most administered NPs are metabolized from urine and feces within 1 week) and avoided accumulation in the body, indicating their promise for biomedical applications. This MOF-based aperture modulation combined with a targeted modification strategy might find broad applications in cancer theranostics. Thus, it is convenient to customize personalized MOFs according to the size of drug molecules in future research.
Collapse
Affiliation(s)
- Mengru Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wulin Liang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Kaixin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongge Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tingting Fu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rongyue Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhai Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoxv Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
20
|
Ali ES, Akter S, Ramproshad S, Mondal B, Riaz TA, Islam MT, Khan IN, Docea AO, Calina D, Sharifi-Rad J, Cho WC. Targeting Ras-ERK cascade by bioactive natural products for potential treatment of cancer: an updated overview. Cancer Cell Int 2022; 22:246. [PMID: 35941592 PMCID: PMC9358858 DOI: 10.1186/s12935-022-02666-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/27/2022] [Indexed: 12/11/2022] Open
Abstract
MAPK (mitogen-activated protein kinase) or ERK (extracellular-signal-regulated kinase) pathway is an important link in the transition from extracellular signals to intracellular responses. Because of genetic and epigenetic changes, signaling cascades are altered in a variety of diseases, including cancer. Extant studies on the homeostatic and pathologic behavior of MAPK signaling have been conducted; however, much remains to be explored in preclinical and clinical research in terms of regulation and action models. MAPK has implications for cancer therapy response, more specifically in response to experimental MAPK suppression, compensatory mechanisms are activated. The current study investigates MAPK as a very complex cell signaling pathway that plays roles in cancer treatment response, cellular normal conduit maintenance, and compensatory pathway activation. Most MAPK inhibitors, unfortunately, cause resistance by activating compensatory feedback loops in tumor cells and tumor microenvironment components. As a result, innovative combinatorial treatments for cancer management must be applied to limit the likelihood of alternate pathway initiation as a possibility for generating novel therapeutics based on incorporation in translational research. We summarize current knowledge about the implications of ERK (MAPK) in cancer, as well as bioactive products from plants, microbial organisms or marine organisms, as well as the correlation with their chemical structures, which modulate this pathway for the treatment of different types of cancer.
Collapse
Affiliation(s)
- Eunus S Ali
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042, Australia
| | - Shamima Akter
- Department of Bioinformatics and Computational Biology, George Mason University, Fairfax, VA, 22030, USA
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400, Bangladesh
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400, Bangladesh
| | - Thoufiqul Alam Riaz
- Department of Pharmacology and Institute of New Drug Development, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea
| | - Muhammad Torequl Islam
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Ishaq N Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, 25100, Pakistan
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | | | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong.
| |
Collapse
|
21
|
Chen G, Yang F, Fan S, Jin H, Liao K, Li X, Liu GB, Liang J, Zhang J, Xu JF, Pi J. Immunomodulatory roles of selenium nanoparticles: Novel arts for potential immunotherapy strategy development. Front Immunol 2022; 13:956181. [PMID: 35958612 PMCID: PMC9361286 DOI: 10.3389/fimmu.2022.956181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022] Open
Abstract
Current chemotherapy strategies used in clinic appear with lots of disadvantages due to the low targeting effects of drugs and strong side effects, which significantly restricts the drug potency, causes multiple dysfunctions in the body, and even drives the emergence of diseases. Immunotherapy has been proved to boost the body’s innate and adaptive defenses for more effective disease control and treatment. As a trace element, selenium plays vital roles in human health by regulating the antioxidant defense, enzyme activity, and immune response through various specific pathways. Profiting from novel nanotechnology, selenium nanoparticles have been widely developed to reveal great potential in anticancer, antibacterial, and anti-inflammation treatments. More interestingly, increasing evidence has also shown that functional selenium nanoparticles can be applied for potential immunotherapy, which would achieve more effective treatment efficiency as adjunctive therapy strategies for the current chemotherapy. By directly interacting with innate immune cells, such as macrophages, dendritic cells, and natural killer cells, selenium nanoparticles can regulate innate immunity to intervene disease developments, which were reported to boost the anticancer, anti-infection, and anti-inflammation treatments. Moreover, selenium nanoparticles can also activate and recover different T cells for adaptive immunity regulations to enhance their cytotoxic to combat cancer cells, indicating the potential of selenium nanoparticles for potential immunotherapy strategy development. Here, aiming to enhance our understanding of the potential immunotherapy strategy development based on Se NPs, this review will summarize the immunological regulation effects of selenium nanoparticles and the application of selenium nanoparticle-based immunotherapy strategies. Furthermore, we will discuss the advancing perspective of selenium nanoparticle-based potential immunotherapy as a kind of novel adjunctive therapy to enhance the efficiency of current chemotherapies and also introduce the current obstacles for the development of selenium nanoparticles for potential immunotherapy strategy development. This work is expected to promote the future research on selenium nanoparticle-assisted immunotherapy and finally benefit the more effective disease treatments against the threatening cancer and infectious and chronic diseases.
Collapse
Affiliation(s)
- Gengshi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Fen Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Kangsheng Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xuemeng Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Pathogenic Biology and Immunology, School of Basic Medicine, Guangdong Medical University, Dongguan, China
| | - Gan-Bin Liu
- Department of Respiration, Dongguan 6th Hospital, Dongguan, China
| | - Jing Liang
- Department of Respiration, Dongguan 6th Hospital, Dongguan, China
| | - Junai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Junai Zhang, ; Jun-Fa Xu, ; Jiang Pi,
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Junai Zhang, ; Jun-Fa Xu, ; Jiang Pi,
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Junai Zhang, ; Jun-Fa Xu, ; Jiang Pi,
| |
Collapse
|
22
|
Sarkar J, Das S, Aich S, Bhattacharyya P, Acharya K. Antiviral potential of nanoparticles for the treatment of Coronavirus infections. J Trace Elem Med Biol 2022; 72:126977. [PMID: 35397331 PMCID: PMC8957383 DOI: 10.1016/j.jtemb.2022.126977] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND On 31st December 2019 in Wuhan, China, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), was acknowledged. This virus spread quickly throughout the world causing a global pandemic. The World Health Organization declared COVID-19 a pandemic disease on 11th March 2020. Since then, the whole world has come together and have developed several vaccines against this deadly virus. Similarly, several alternative searches for pandemic disease therapeutics are still ongoing. One of them has been identified as nanotechnology. It has demonstrated significant promise for detecting and inhibiting a variety of viruses, including coronaviruses. Several nanoparticles, including gold nanoparticles, silver nanoparticles, quantum dots, carbon dots, graphene oxide nanoparticles, and zinc oxide nanoparticles, have previously demonstrated remarkable antiviral activity against a diverse array of viruses. OBJECTIVE This review aims to provide a basic and comprehensive overview of COVID-19's initial global outbreak and its mechanism of infiltration into human host cells, as well as the detailed mechanism and inhibitory effects of various nanoparticles against this virus. In addition to nanoparticles, this review focuses on the role of several antiviral drugs used against COVID-19 to date. CONCLUSION COVID-19 has severely disrupted the social and economic lives of people all over the world. Due to a lack of adequate medical facilities, countries have struggled to maintain control of the situation. Neither a drug nor a vaccine has a 100% efficacy rate. As a result, nanotechnology may be a better therapeutic alternative for this pandemic disease.
Collapse
Affiliation(s)
- Joy Sarkar
- Department of Botany, Dinabandhu Andrews College, Garia, Kolkata, West Bengal 700084, India
| | - Sunandana Das
- Department of Botany, Dinabandhu Andrews College, Garia, Kolkata, West Bengal 700084, India
| | - Sahasrabdi Aich
- Department of Botany, Vivekananda College, Thakurpukur, Kolkata, West Bengal 700063, India
| | - Prithu Bhattacharyya
- Department of Botany, Dinabandhu Andrews College, Garia, Kolkata, West Bengal 700084, India
| | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Centre of Advanced Study, Department of Botany, University of Calcutta, Kolkata, West Bengal 700019, India; Center for Research in Nanoscience & Nanotechnology, Technology Campus, University of Calcutta, Kolkata, West Bengal 700098, India.
| |
Collapse
|
23
|
Ren Y, Hong Y, He W, Liu Y, Chen W, Wen S, Sun M. EGF/EGFR Promotes Salivary Adenoid Cystic Carcinoma Cell Malignant Neural Invasion via Activation of PI3K/AKT and MEK/ERK Signaling. Curr Cancer Drug Targets 2022; 22:603-616. [PMID: 35410600 DOI: 10.2174/1568009622666220411112312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Salivary adenoid cystic carcinoma (SACC) is one of the most common malignant cancers of the salivary gland, and 32.4-72.0% of SACC cases exhibit neural invasion (NI), however, the molecular mechanism underlying the high invasion potential of SACC remains unclear. METHODS The present study investigated the role of epidermal growth factor receptor (EGFR) in the AKT inhibition- or mitogen-activated protein kinase kinase (MEK)-induced NI and epithelial-mesenchymal transition (EMT) in SACC cells using EGFR, PI3K and MEK inhibitors. SACC 83 cell viability was assessed using an MTT assay, and a wound healing assay was performed to evaluate cell migration. Immunohistochemical staining with streptavidin peroxidase was used to detect the positive expression rate of EMT, AKT, phosphorylated (p)-AKT, ERK and p-ERK proteins. The impact of EGFR, PI3K and MEK inhibitors on tumor growth and NI was examined in a xenograft model in nude mice. RESULTS EGF and EGFR are effective in increasing cell viability, migration and invasion. SACC metastasis is affected by the PI3K/AKT and MEK/ERK pathways, both of which are initiated by EGF/EGFR. The EMT and NI are regulated by the EGF/EGFR, PI3K/AKT and MEK/ERK pathways. The present findings demonstrate the importance of suppressed EGFR/AKT/MEK signaling in NI in SACC by neural-tumor co-culture in vitro. Furthermore, our preclinical experiment provides solid evidence that injection of EGFR, PI3K and MEK inhibitors obviously suppressed the tumor growth and NI of SACC cells in nude mice. CONCLUSION It was identified that inhibitors of EGFR, PI3K/AKT or MEK/ERK suppressed the proliferation, migration and NI of SACC-83 cells via downregulation of the PI3K/AKT or MEK/ERK pathways. It was also demonstrated that inhibition of EGFR abolishes EMT in SACC by inhibiting the signaling of PI3K/AKT and MEK/ERK. The present results suggest the potential effectiveness of targeting multiple oncogenes associated with downstream pathways of EGF/EGFR, as well as potential therapeutic targets to limit NI in SACC by PI3K/AKT or MEK/ERK inhibition.
Collapse
Affiliation(s)
- Yixiong Ren
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi, China.,Department of Oral and Maxillofacial Surgery, Shanxi Province People's Hospital, Taiyuan, Shanxi, China
| | - Yonglong Hong
- Department of Oral and Maxillofacial Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Wenting He
- Nursing College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yakun Liu
- Department of Obstetrics, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenge Chen
- Department of Oral and Maxillofacial Surgery, Shanxi Province People's Hospital, Taiyuan, Shanxi, China
| | - Sui Wen
- Department of Oral and Maxillofacial Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Moyi Sun
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Air Force Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
24
|
The development and progress of nanomedicine for esophageal cancer diagnosis and treatment. Semin Cancer Biol 2022; 86:873-885. [DOI: 10.1016/j.semcancer.2022.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
|
25
|
Huang X, Chen L, Lin Y, Tou KIP, Cai H, Jin H, Lin W, Zhang J, Cai J, Zhou H, Pi J. Tumor targeting and penetrating biomimetic mesoporous polydopamine nanoparticles facilitate photothermal killing and autophagy blocking for synergistic tumor ablation. Acta Biomater 2021; 136:456-472. [PMID: 34562660 DOI: 10.1016/j.actbio.2021.09.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/23/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022]
Abstract
The synergistic manipulation of autophagy blocking with tumor targeting and penetration effects to enhance cancer cell killing during photothermal therapy (PTT) remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous prostate cancer cell membranes (CMs) onto the surface of mesoporous polydopamine nanoparticles (mPDA NPs) encapsulating the autophagy inhibitor chloroquine (CQ) for synergistically manipulating PTT and autophagy for anticancer treatment. The resulting biomimetic mPDA@CMs NPs-CQ system could escape macrophage phagocytosis, overcome the vascular barrier, and home in the homologous prostate tumor xenograft with high tumor targeting and penetrating efficiency. The mPDA NPs core endowed the mPDA@CMs NPs-CQ with good photothermal capability to mediate PTT killing of prostate cancer cells, while NIR-triggered CQ release from the nanosystem further arrested PTT-induced protective autophagy of cancer cells, thus weakening the resistance of prostate cancer cells to PTT. This combined PTT killing and autophagy blocking anticancer strategy could induce significant autophagosome accumulation, ROS generation, mitochondrial damage, endoplasmic reticulum stress, and apoptotic signal transduction, which finally results in synergistic prostate tumor ablation in vivo. This prostate cancer biomimetic nanosystem with synergistically enhanced anticancer efficiency achieved by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy against prostate cancer. STATEMENT OF SIGNIFICANCE: Autophagy is considered as one of the most efficient rescuer and reinforcement mechanisms of cancer cells against photothermal therapy (PTT)-induced cancer cell eradication. How to synergistically manipulate autophagy blocking with significant tumor targeting and penetration to enhance PTT-mediated cancer cell killing remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous cancer cell membranes onto the surface of mesoporous polydopamine nanoparticles with encapsulation of the autophagy inhibitor chloroquine for synergistic antitumor treatment with high tumor targeting and penetrating efficiency both in vitro and in vivo. This biomimetic nanosystem with synergistically enhanced anticancer efficiency by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy.
Collapse
|
26
|
Yang L, Fang H, Jiang J, Sha Y, Zhong Z, Meng F. EGFR-targeted pemetrexed therapy of malignant pleural mesothelioma. Drug Deliv Transl Res 2021; 12:2527-2536. [PMID: 34802094 DOI: 10.1007/s13346-021-01094-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2021] [Indexed: 12/17/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy with poor prognosis, for which chemotherapy with pemetrexed (PEM) is among the few clinical treatments. PEM suffers, however, fast clearance, moderate drug exposure, and dose-limiting toxicities. Here, we report on epidermal growth factor receptor (EGFR)-targeted disulfide-crosslinked biodegradable chimaeric polymersomes (EGFR-CPs) to firmly load PEM and boost chemotherapy of MPM. EGFR-CPs encapsulating 8.7-16.4 wt.% PEM (EGFR-CPs-PEM) showed diameters of 62-65 nm and reduction-responsive drug release property. EGFR-CPs-PEM was more efficiently taken up by EGFR-overexpressed MSTO-211H cells, inducing about 4.7-fold enhanced anticancer activity compared with non-targeted CPs-PEM control. Intriguingly, the in vivo experiments in MSTO-211H xenograft mouse model revealed that EGFR-CPs-PEM brought about superior tumor deposition and penetration to CPs-PEM, and significantly more potent tumor repression than CPs-PEM and free PEM. This polymersome-enabled EGFR-targeted delivery of PEM offers an appealing therapeutic strategy for MPM.
Collapse
Affiliation(s)
- Liang Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Hanghang Fang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Jingjing Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Yongjie Sha
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
27
|
Nguyen PV, Hervé-Aubert K, Chourpa I, Allard-Vannier E. Active targeting strategy in nanomedicines using anti-EGFR ligands - A promising approach for cancer therapy and diagnosis. Int J Pharm 2021; 609:121134. [PMID: 34571073 DOI: 10.1016/j.ijpharm.2021.121134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022]
Abstract
As active targeting using nanomedicines establishes itself as a strategy of choice in cancer therapy, several target receptors or ligands overexpressed in cancer cells have been identified and exploited. Among them, the epidermal growth factor receptor (EGFR) has emerged as one of the most promising oncomarkers for active targeting nanomedicines due to its overexpression and its active involvement in a wide range of cancer types. Henceforth, many novel EGFR-targeted nanomedicines for cancer therapy have been developed, giving encouraging results both in vitro and in vivo. This review focuses on different applications of such medicines in oncotherapy. On an important note, the contribution of EGFR-targeting ligands to final therapy efficacy along with current challenges and possible solutions or alternatives are emphasized.
Collapse
Affiliation(s)
- Phuoc Vinh Nguyen
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Katel Hervé-Aubert
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Igor Chourpa
- EA6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | | |
Collapse
|
28
|
Li S, Pang X, Zhao J, Zhang Q, Shan Y. Evaluating the single-molecule interactions between targeted peptides and the receptors on living cell membrane. NANOSCALE 2021; 13:17318-17324. [PMID: 34642724 DOI: 10.1039/d1nr05547j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
As potential ligands, targeted peptides have become an important part in the construction of intelligent drug delivery systems (DDSs). The targeting interaction of peptides with receptors is a key point affecting the efficacy of targeted nano-drugs. Herein, three common peptides (HAIYPRH (T7), YHWYGYTPQNVI (GE11), and RGD) that have been widely used in cancer targeted therapy and tumor diagnostics, targeting the corresponding receptors (transferrin receptor (TfR), epidermal growth factor receptor (EGFR), and ανβ3 integrin receptor), were selected as examples to study the targeting interacton on living cell surface at the single-molecule level by using single-molecule force spectroscopy (SMFS) based on atomic force microscopy (AFM). The dissociation activation energy in the absence of an external force (ΔGβ,0) of T7-TfR, GE11-EGFR, and RGD-ανβ3 integrin is evaluated at single-molecule level. Among these three peptide-receptor pairs, the T7-TfR bond is the most stable with a smaller dissociation kinetic rate constant at zero force (Koff), larger kinetic on-rate constant (Kon), and shorter interaction time (τ). Furthermore, T7 can target TfR even more effectively on A549 cell membrane after treatment with drugs. Our methodology can also be applicable to the study of other ligand targeted DDSs.
Collapse
Affiliation(s)
- Siying Li
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
| | - Xuelei Pang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Jing Zhao
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Qingrong Zhang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Yuping Shan
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| |
Collapse
|
29
|
He S, Tian S, He X, Le X, Ning Y, Chen J, Chen H, Mu J, Xu K, Xiang Q, Wu Y, Chen J, Xiang T. Multiple targeted self-emulsifying compound RGO reveals obvious anti-tumor potential in hepatocellular carcinoma. Mol Ther Oncolytics 2021; 22:604-616. [PMID: 34589579 PMCID: PMC8449031 DOI: 10.1016/j.omto.2021.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly vascularized, inflammatory, and abnormally proliferating tumor. Monotherapy is often unable to effectively and comprehensively inhibit the progress of HCC. In present study, we selected ginsenoside Rg3, ganoderma lucidum polysaccharide (GLP), and oridonin as the combined therapy. These three plant monomers play important roles in anti-angiogenesis, immunological activation, and apoptosis promotion, respectively. However, the low solubility and poor bioavailability seriously hinder their clinical application. To resolve these problems, we constructed a new drug, Rg3, GLP, and oridonin self-microemulsifying drug delivery system (RGO-SMEDDS). We found that this drug effectively inhibits the progression of HCC by simultaneously targeting multiple signaling pathways. RGO-SMEDDS restored immune function by suppressing the production of immunosuppressive cytokine and M2-polarized macrophages, reduced angiogenesis by downregulation of vascular endothelial growth factor and its receptor, and retarded proliferation by inhibiting the epidermal growth factor receptor EGFR/AKT/epidermal growth factor receptor/protein kinase B/glycogen synthase kinase-3 (GSK3) signaling pathway. In addition, RGO-SMEDDS showed considerable safety in acute toxicity tests. Results from this study show that RGO-SMEDDS is a promising therapy for the treatment of HCC.
Collapse
Affiliation(s)
- Sanxiu He
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shaorong Tian
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqian He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Le
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yijiao Ning
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jialin Chen
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Hongyi Chen
- Chongqing College of Humanities, Science & Technology, Chongqing, China
| | - Junhao Mu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Xu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiong Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Tingxiu Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
30
|
Gaurav I, Wang X, Thakur A, Iyaswamy A, Thakur S, Chen X, Kumar G, Li M, Yang Z. Peptide-Conjugated Nano Delivery Systems for Therapy and Diagnosis of Cancer. Pharmaceutics 2021; 13:1433. [PMID: 34575511 PMCID: PMC8471603 DOI: 10.3390/pharmaceutics13091433] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 11/16/2022] Open
Abstract
Peptides are strings of approximately 2-50 amino acids, which have gained huge attention for theranostic applications in cancer research due to their various advantages including better biosafety, customizability, convenient process of synthesis, targeting ability via recognizing biological receptors on cancer cells, and better ability to penetrate cell membranes. The conjugation of peptides to the various nano delivery systems (NDS) has been found to provide an added benefit toward targeted delivery for cancer therapy. Moreover, the simultaneous delivery of peptide-conjugated NDS and nano probes has shown potential for the diagnosis of the malignant progression of cancer. In this review, various barriers hindering the targeting capacity of NDS are addressed, and various approaches for conjugating peptides and NDS have been discussed. Moreover, major peptide-based functionalized NDS targeting cancer-specific receptors have been considered, including the conjugation of peptides with extracellular vesicles, which are biological nanovesicles with promising ability for therapy and the diagnosis of cancer.
Collapse
Affiliation(s)
- Isha Gaurav
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (X.W.); (A.I.); (X.C.); (M.L.)
| | - Xuehan Wang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (X.W.); (A.I.); (X.C.); (M.L.)
| | - Abhimanyu Thakur
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation-CAS Limited, Hong Kong, China;
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (X.W.); (A.I.); (X.C.); (M.L.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Sudha Thakur
- National Institute for Locomotor Disabilities (Divyangjan), Kolkata 700090, India;
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (X.W.); (A.I.); (X.C.); (M.L.)
| | - Gaurav Kumar
- School of Basic and Applied Science, Galgotias University, Greater Noida 203201, India;
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (X.W.); (A.I.); (X.C.); (M.L.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (I.G.); (X.W.); (A.I.); (X.C.); (M.L.)
- Changshu Research Institute, Hong Kong Baptist University, Changshu Economic and Technological Development (CETD) Zone, Changshu 215500, China
| |
Collapse
|
31
|
Ferro C, Florindo HF, Santos HA. Selenium Nanoparticles for Biomedical Applications: From Development and Characterization to Therapeutics. Adv Healthc Mater 2021; 10:e2100598. [PMID: 34121366 DOI: 10.1002/adhm.202100598] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/16/2021] [Indexed: 12/11/2022]
Abstract
Selenium (Se) is an essential element to human health that can be obtained in nature through several sources. In the human body, it is incorporated into selenocysteine, an amino acid used to synthesize several selenoproteins, which have an active center usually dependent on the presence of Se. Although Se shows several beneficial properties in human health, it has also a narrow therapeutic window, and therefore the excessive intake of inorganic and organic Se-based compounds often leads to toxicity. Nanoparticles based on Se (SeNPs) are less toxic than inorganic and organic Se. They are both biocompatible and capable of effectively delivering combinations of payloads to specific cells following their functionalization with active targeting ligands. Herein, the main origin of Se intake, its role on the human body, and its primary biomedical applications are revised. Particular focus will be given to the main therapeutic targets that are explored for SeNPs in cancer therapies, discussing the different functionalization methodologies used to improve SeNPs stability, while enabling the extensive delivery of drug-loaded SeNP to tumor sites, thus avoiding off-target effects.
Collapse
Affiliation(s)
- Cláudio Ferro
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki FI‐00014 Finland
- Research Institute for Medicines iMed.ULisboa Faculty of Pharmacy Universidade de Lisboa Lisbon 1649‐003 Portugal
| | - Helena F. Florindo
- Research Institute for Medicines iMed.ULisboa Faculty of Pharmacy Universidade de Lisboa Lisbon 1649‐003 Portugal
| | - Hélder A. Santos
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki FI‐00014 Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki Helsinki FI‐00014 Finland
| |
Collapse
|
32
|
Lin W, Zhang J, Xu JF, Pi J. The Advancing of Selenium Nanoparticles Against Infectious Diseases. Front Pharmacol 2021; 12:682284. [PMID: 34393776 PMCID: PMC8361478 DOI: 10.3389/fphar.2021.682284] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Infectious diseases, caused by the direct exposure of cellular or acellular pathogens, are found to be closely associated with multiple inflammation and immune responses, keeping one of the top threats to human health. As an indispensable trace element, Selenium (Se) plays important roles in antioxidant defence and redox state regulation along with a variety of specific metabolic pathways. In recent decades, with the development of novel nanotechnology, Selenium nanoparticles (Se NPs) emerged as a promising agent for biomedical uses due to their low toxicity, degradability and high bioavailability. Taking the advantages of the strong ability to trigger apoptosis or autophagy by regulating reactive oxygen species (ROS), Se NPs have been widely used for direct anticancer treatments and pathogen killing/clearance in host cells. With excellent stability and drug encapsulation capacity, Se NPs are now serving as a kind of powerful nano-carriers for anti-cancer, anti-inflammation and anti-infection treatments. Notably, Se NPs are also found to play critical roles in immunity regulations, such as macrophage and T effector cell activation, which thus provides new possibilities to achieve novel nano-immune synergetic strategy for anti-cancer and anti-infection therapies. In this review, we summarized the progress of preparation methods for Se NPs, followed by the advances of their biological functions and mechanisms for biomedical uses, especially in the field of anti-infection treatments. Moreover, we further provide some prospects of Se NPs in anti-infectious diseases, which would be helpful for facilitating their future research progress for anti-infection therapy.
Collapse
Affiliation(s)
- Wensen Lin
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Junai Zhang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jun-Fa Xu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
33
|
Recent developments in mitogen activated protein kinase inhibitors as potential anticancer agents. Bioorg Chem 2021; 114:105161. [PMID: 34328852 DOI: 10.1016/j.bioorg.2021.105161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 01/06/2023]
Abstract
The mitogen activated protein kinase (MAPK) belongs to group of kinase that links the extracellular stimuli to intracellular response. The MAPK signalling pathway (RAS-RAF-MEK-ERK) involved in different pathological conditions like cancer, caused due to genetic or any other factor such as physical or environmental. Many studies have been conducted on the pathological view of MAPK cascade and its associated element like RAS, RAF, MEK, ERK or its isoforms, and still the research is going on particularly with respect to its activation, regulation and inhibition. The MAPK signalling pathway has become the area of research to identify new target for the management of cancer. A number of heterocyclics are key to fight with the cancer associated with these enzymes thus give some hope in the management of cancer by inhibiting MAPK cascade. In the present article, we have focussed on MAPK signalling pathway and role of different heterocyclic scaffolds bearing nitrogen, sulphur and oxygen and about their potential to block MAPK signalling pathway. The heterocyclics are gaining importance due to high potency and selectivity with less off-target effects against different targets involved in the MAPK signalling pathway. We have tried to cover recent advancements in the MAPK signalling pathway inhibitors with an aim to get better understanding of the mechanism of action of the compounds. Several compounds in the preclinical and clinical studies have been thoroughly dealt with. In addition to the synthetic compounds, a significant number of natural products containing heterocyclic moieties as MAPK signalling pathway inhibitors have been put together. The structure activity relationship along with docking studies have been discussed to apprehend the mechanistic studies of various compounds that will ultimately help to design and develop more MAPK signalling pathway inhibitors.
Collapse
|
34
|
Sun M, Wang T, Li L, Li X, Zhai Y, Zhang J, Li W. The Application of Inorganic Nanoparticles in Molecular Targeted Cancer Therapy: EGFR Targeting. Front Pharmacol 2021; 12:702445. [PMID: 34322025 PMCID: PMC8311435 DOI: 10.3389/fphar.2021.702445] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is an anticancer drug target for a number of cancers, such as non-small cell lung cancer. However, unsatisfying treatment effects, terrible side-effects, and development of drug resistance are current insurmountable challenges of EGFR targeting treatments for cancers. With the advancement of nanotechnology, an increasing number of inorganic nanomaterials are applied in EGFR-mediated therapy to improve those limitations and further potentiate the efficacy of molecular targeted cancer therapy. Given their facile preparation, easy modification, and biosecurity, inorganic nanoparticles (iNPs) have been extensively explored in cancer treatments to date. This review presents an overview of the application of some typical metal nanoparticles and nonmetallic nanoparticles in EGFR-targeted therapy, then discusses and summarizes the relevant advantages. Moreover, we also highlight future perspectives regarding their remaining issues. We hope these discussions inspire future research on EGFR-targeted iNPs.
Collapse
Affiliation(s)
- Meng Sun
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, China
| | - Ting Wang
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Leijiao Li
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, China
| | - Xiangyang Li
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, China
| | - Yutong Zhai
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, China
| | - Jiantao Zhang
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wenliang Li
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| |
Collapse
|
35
|
Nanomedicines functionalized with anti-EGFR ligands for active targeting in cancer therapy: Biological strategy, design and quality control. Int J Pharm 2021; 605:120795. [PMID: 34119579 DOI: 10.1016/j.ijpharm.2021.120795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.
Collapse
|
36
|
Cai G, Wang Y, Houda T, Yang C, Wang L, Gu M, Mueck A, Croteau S, Ruan X, Hardy P. MicroRNA-181a suppresses norethisterone-promoted tumorigenesis of breast epithelial MCF10A cells through the PGRMC1/EGFR-PI3K/Akt/mTOR signaling pathway. Transl Oncol 2021; 14:101068. [PMID: 33730679 PMCID: PMC7974027 DOI: 10.1016/j.tranon.2021.101068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Research suggests that hormone replacement therapy may increase the risk of breast cancer, and progestins such as norethisterone (NET) play a key role in this phenomenon. We have demonstrated that microRNA-181a (miR-181a) suppresses NET-promoted breast cancer cell survival. Nonetheless, the effects of NET and miR-181a on the tumorigenesis of human breast epithelial cells have not yet been elaborated. METHODS Assays of cell viability, proliferation, migration, apoptosis, and colony formation were performed to investigate the pro-tumorigenesis effect of NET and the effects of miR-181a on human breast epithelial MCF10A cells. The expressions of cell-proliferation-related genes and apoptotic factors were analyzed by quantitative RT-PCR and Western blot in MCF10A cells treated with NET and miR-181a. RESULTS NET significantly increased MCF10A cell viability, proliferation, migration, and colony formation, but reduced cellular apoptosis. In addition, NET increased the expression of progesterone receptor membrane component 1 (PGRMC1), EGFR, B-cell lymphoma 2, cyclin D1, and proliferating cell nuclear antigen, but decreased the expression of pro-apoptosis factors, such as Bax, caspase-7, and caspase-9. Overexpression of miR-181a strongly inhibited the effects of NET on MCF10A cells and abrogated NET-stimulated PGRMC1, EGFR, and mTOR expression. CONCLUSIONS Activation of the PGRMC1/EGFR-PI3K/Akt/mTOR signaling pathway is the primary mechanism underlying the pro-tumorigenesis effects of NET on human breast epithelial MCF10A cells. Additionally, miR-181a can suppress the effects of NET on these cells. These data suggest a therapeutic potential for miR-181a in reducing or preventing the risk of breast cancer in hormone replacement therapy using NET.
Collapse
Affiliation(s)
- Guiju Cai
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada; Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yuejiao Wang
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada; Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Tahiri Houda
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada
| | - Chun Yang
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada
| | - Lijuan Wang
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada; Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Muqing Gu
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada; Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Alfred Mueck
- University Women's Hospital and Research Centre for Women's Health, Department of Women's Health, University of Tübingen, D-72076 Tübingen, Germany
| | - Stephane Croteau
- Departments of Medicine, Pediatrics, Pharmacology, and Physiology, University of Montréal, Canada
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China.
| | - Pierre Hardy
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec, Canada; Departments of Medicine, Pediatrics, Pharmacology, and Physiology, University of Montréal, Canada.
| |
Collapse
|
37
|
Hu W, Zhao C, Hu H, Yin S. Food Sources of Selenium and Its Relationship with Chronic Diseases. Nutrients 2021; 13:nu13051739. [PMID: 34065478 PMCID: PMC8160805 DOI: 10.3390/nu13051739] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022] Open
Abstract
Selenium (Se) is an essential micronutrient for mammals, and its deficiency seriously threatens human health. A series of biofortification strategies have been developed to produce Se-enriched foods for combating Se deficiency. Although there have been some inconsistent results, extensive evidence has suggested that Se supplementation is beneficial for preventing and treating several chronic diseases. Understanding the association between Se and chronic diseases is essential for guiding clinical practice, developing effective public health policies, and ultimately counteracting health issues associated with Se deficiency. The current review will discuss the food sources of Se, biofortification strategies, metabolism and biological activities, clinical disorders and dietary reference intakes, as well as the relationship between Se and health outcomes, especially cardiovascular disease, diabetes, chronic inflammation, cancer, and fertility. Additionally, some concepts were proposed, there is a non-linear U-shaped dose-responsive relationship between Se status and health effects: subjects with a low baseline Se status can benefit from Se supplementation, while Se supplementation in populations with an adequate or high status may potentially increase the risk of some diseases. In addition, at supra-nutritional levels, methylated Se compounds exerted more promising cancer chemo-preventive efficacy in preclinical trials.
Collapse
|
38
|
Tan RZ, Yan Y, Yu Y, Diao H, Zhong X, Lin X, Liao YY, Wang L. Renoprotective Effect of Oridonin in a Mouse Model of Acute Kidney Injury via Suppression of Macrophage Involved Inflammation. Biol Pharm Bull 2021; 44:714-723. [PMID: 33952827 DOI: 10.1248/bpb.b21-00071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemia-reperfusion injury (IRI) is the major cause of acute kidney injury (AKI). The previous studies demonstrated that Oridonin can protect kidney against IRI-induced AKI, but the underlying molecular mechanism is unclear. In this study, it showed that Oridonin significantly improved kidney damage, and inhibited the expression of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and MCP-1, as well as macrophage marker F4/80 in kidney and the secretion of inflammatory cytokins in serum of AKI mice in vivo. In addition, Oridonin also effectively reduced the expression and secretion of lipopolysaccharide (LPS)-induced inflammatory factors in macrophage cell line RAW264.7 in vitro. Notably, Oridonin strongly downregulated Mincle and AKT/nuclear factor-kappaB (NF-κB) signaling both in vivo and in vitro, and the results of cellular recovery experiments of overexpression of Mincle in macrophage suggested that Oridonin suppressed inflammatory response of macrophage through inhibiting Mincle, which may be the underlying mechanism of Oridonin improving injury in kidney of AKI mice. In summary, the above results indicated that Oridonin can protect kidney from IRI-induced inflammation and injury by inhibiting the expression of Mincle in macrophage.
Collapse
Affiliation(s)
- Rui-Zhi Tan
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University
| | - Ying Yan
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University.,Clinical Laboratory, Affiliated Traditional Medicine Hospital, Southwest Medical University
| | - Yan Yu
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University.,College of Integrated Chinese and Western Medicine, Southwest Medical University
| | - Hui Diao
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University
| | - Xia Zhong
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University
| | - Xiao Lin
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University
| | - Yi-Yi Liao
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University.,College of Integrated Chinese and Western Medicine, Southwest Medical University
| | - Li Wang
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University
| |
Collapse
|
39
|
GE11 Peptide Conjugated Liposomes for EGFR-Targeted and Chemophotothermal Combined Anticancer Therapy. Bioinorg Chem Appl 2021; 2021:5534870. [PMID: 33868396 PMCID: PMC8035035 DOI: 10.1155/2021/5534870] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/13/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023] Open
Abstract
How to actively target tumor sites manipulating the controllable release of the encapsulated anticancer drugs and photosensitizers for synergistic anticancer therapy remains a big challenge. In this study, a cancer cell-targeted, near-infrared (NIR) light-triggered and anticancer drug loaded liposome system (LPs) was developed for synergistic cancer therapy. Photosensitizer indocyanine green (ICG) and chemotherapy drug Curcumin (CUR) were coencapsulated into the liposomes, followed by the surface conjugation of GE11 peptide for epidermal growth factor receptor (EGFR) targeting on the cancer cell surface. Strictly controlled by NIR light, GE11 peptide modified and CUR/ICG-loaded LPs (GE11-CUR/ICG-LPs) could introduce hyperthermia in EGFR overexpressed A549 cancer cells for photothermal therapy, which could also trigger the increased release of CUR for enhanced cancer cell inhibition. GE11-CUR/ICG-LPs synergized photochemotherapy could induce reactive oxygen species (ROS) generation and cytoskeleton disruption to activate stronger apoptotic signaling events than the photothermal therapy or chemotherapy alone by regulating Bax/Bcl-2 and PI3K/AKT pathways. This EGFR-targeted drug-delivery nanosystem with NIR sensitivity may potentially serve in more effective anticancer therapeutics with reduced off-target effects.
Collapse
|
40
|
Huang X, Cai H, Zhou H, Li T, Jin H, Evans CE, Cai J, Pi J. Cobalt oxide nanoparticle-synergized protein degradation and phototherapy for enhanced anticancer therapeutics. Acta Biomater 2021; 121:605-620. [PMID: 33259958 DOI: 10.1016/j.actbio.2020.11.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022]
Abstract
How to enable protein degradation pathways including the autophagy-lysosome pathway (ALP) and the ubiquitin-proteasome system (UPS) to enhance the efficacy of anticancer treatments remains a substantial challenge. Cobalt oxide nanoparticles (Co3O4 NPs) have attracted interest in recent years for their potential use as a synergistic anticancer treatment, although their therapeutic mechanisms of action are still poorly understood. Here, we describe the synergistic use of Co3O4 NPs as an autophagy inhibitor, chemosensitizer and photosensitizer, which manipulate protein degradation pathways (ALP and UPS) and photothermal therapy for enhanced anticancer treatments both in vitro and in vivo. We show that Co3O4 NPs can induce autolysosome accumulation and lysosomal functions damage by inhibiting lysosomal proteolytic activity and reducing intracellular ATP levels. Notably, Co3O4 NPs can be combined with the proteasome inhibitor, Carfilzomib (Cfz), to promote the accumulation of autophagic substrates, protein ubiquitination, and endoplasmic reticulum stress, and in doing so, inhibit cancer progression. By taking advantage of their photothermal conversion efficiency, Co3O4 NPs can also serve as photothermal sensitizer, which synergistically enhances the anticancer efficacy of Cfz both in vitro and in vivo. In summary, we provide evidence of a nanomaterial-synergized, photothermal anticancer strategy that synergistically targets cancer cell survival pathways and may eventually serve to enhance the anticancer efficacy of established cancer therapeutics.
Collapse
Affiliation(s)
- Xueqin Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 000583, China
| | - Huaihong Cai
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Haibo Zhou
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 000583, China
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, the Scientific Research Center of Dongguan, College of Pharmacy, Institute of Clinical Laboratory Medicine, Guangdong Medical University, Dongguan 523808, China
| | - Colin E Evans
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jiye Cai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 000583, China; Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Jiang Pi
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
41
|
He L, Zhao J, Wang L, Liu Q, Fan Y, Li B, Yu YL, Chen C, Li YF. Using nano-selenium to combat Coronavirus Disease 2019 (COVID-19)? NANO TODAY 2021; 36:101037. [PMID: 33250930 PMCID: PMC7683300 DOI: 10.1016/j.nantod.2020.101037] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 05/20/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic represents a severe global health threat. Selenium (Se), as one of the essential trace elements in human body, is well known for its antioxidant and immunity-boosting capabilities that induce a strong antiviral effect. In response to the global pandemic, we highlight here the current status of Se in combating different viruses, as well as the potential application of nano-selenium (nanoSe) in combating COVID-19.
Collapse
Affiliation(s)
- Lina He
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, & Beijing Metallomics Facility, & National Consortium for Excellence in Metallomics, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Jiating Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, & Beijing Metallomics Facility, & National Consortium for Excellence in Metallomics, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, & Beijing Metallomics Facility, & National Consortium for Excellence in Metallomics, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Quancheng Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Yuqin Fan
- Shandong Provincial Maternal & Child Health Care Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, Shandong, China
| | - Bai Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, & Beijing Metallomics Facility, & National Consortium for Excellence in Metallomics, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Yong-Liang Yu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, & CAS Center for Excellence in Nanoscience, & Beijing Metallomics Facility, National Centre for Nanoscience and Technology, Beijing 100191, China
| | - Yu-Feng Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, & CAS-HKU Joint Laboratory of Metallomics on Health and Environment, & Beijing Metallomics Facility, & National Consortium for Excellence in Metallomics, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
42
|
Gill MSA, Saleem H, Ahemad N. Plant Extracts and their Secondary Metabolites as Modulators of Kinases. Curr Top Med Chem 2021; 20:1093-1104. [PMID: 32091334 DOI: 10.2174/1568026620666200224100219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 12/20/2022]
Abstract
Natural Products (NP), specifically from medicinal plants or herbs, have been extensively utilized to analyze the fundamental mechanisms of ultimate natural sciences as well as therapeutics. Isolation of secondary metabolites from these sources and their respective biological properties, along with their lower toxicities and cost-effectiveness, make them a significant research focus for drug discovery. In recent times, there has been a considerable focus on isolating new chemical entities from natural flora to meet the immense demand for kinase modulators, and also to overcome major unmet medical challenges in relation to signal transduction pathways. The signal transduction systems are amongst the foremost pathways involved in the maintenance of life and protein kinases play an imperative part in these signaling pathways. It is important to find a kinase inhibitor, as it can be used not only to study cell biology but can also be used as a drug candidate for cancer and metabolic disorders. A number of plant extracts and their isolated secondary metabolites such as flavonoids, phenolics, terpenoids, and alkaloids have exhibited activities against various kinases. In the current review, we have presented a brief overview of some important classes of plant secondary metabolites as kinase modulators. Moreover, a number of phytocompounds with kinase inhibition potential, isolated from different plant species, are also discussed.
Collapse
Affiliation(s)
- Muhammad Shoaib Ali Gill
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore 54000, Pakistan
| | - Hammad Saleem
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Institute of Pharmaceutical Sciences (IPS), University of Veterinary and Animal Sciences (UVAS), Lahore 54000, Pakistan
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia.,Global Asia in 21st Century Platform, Monash University Malaysia, Jalan Lagoon Selatan, Petaling Jaya 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
43
|
Selenium nanoparticles reduce glucose metabolism and promote apoptosis of glioma cells through reactive oxygen species-dependent manner. Neuroreport 2021; 31:226-234. [PMID: 31876687 DOI: 10.1097/wnr.0000000000001386] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gliomas are the most common, malignant, and lethal tumors in adults. Furthermore, gliomas are highly resistant to current chemotherapeutic drugs. Thus, new effective anticancer drugs for glioma are urgently needed. Selenium nanoparticles have been reported to have potent anti-tumor activity, although the specific mechanism is not fully understood. This study aimed to test the anti-tumor effect of selenium nanoparticles and its mechanism. We used selenium nanoparticles to treat commercial glioma cell lines, and patient-derived glioma cells, and then used the MTT assay to determine selenium nanoparticles effect against these. Apoptotic cell death was determined by annexin V-Fluos staining kit. Glucose uptake, lactate, and adenosine triphosphate production, together with hexokinase 2 and pyruvate kinase activities were measured to determine the glucose metabolism level. Reactive oxygen species production was tested using 2',7'-dichlorodihydrofluorescein diacetate. Our results showed that selenium nanoparticles had a potent cytotoxic effect in glioma cells, regardless of whether they were drug-resistant or not, whereas it showed less toxic effect in normal healthy cells. Further tests showed that selenium nanoparticles treatment leads to apoptotic cell death enhancement and glucose metabolism reduction, and this process was in a reactive oxygen species pathway-dependent manner. These results may provide a novel direction for glioma therapy in the future.
Collapse
|
44
|
Selenium nanostructure: Progress towards green synthesis and functionalization for biomedicine. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-020-00510-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
45
|
Sheng H, Zhang Y, Nai J, Wang S, Dai M, Lin G, Zhu L, Zhang Q. Preparation of oridonin nanocrystals and study of their endocytosis and transcytosis behaviours on MDCK polarized epithelial cells. PHARMACEUTICAL BIOLOGY 2020; 58:518-527. [PMID: 32501184 PMCID: PMC8641689 DOI: 10.1080/13880209.2020.1767160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 06/06/2023]
Abstract
Context: Oridonin (ORI) has obvious anticancer effects, but its solubility is poor. Nanocrystal (NC) is a novel nano-drug delivery system for increasing bioavailability for ORI. However, the endocytosis and transcytosis behaviours of oridonin nanocrystals (ORI-NCs) through epithelial membrane are still unclear.Objectives: ORI-NCs were prepared and characterized. The in vitro cytotoxicity and endocytosis and transcytosis process on Madin-Darby canine kidney (MDCK) monolayer were investigated.Materials and methods: Anti-solvent precipitation method was adopted in preparation of ORI-NCs. Differential scanning calorimetry (DSC) and X-ray diffraction (XRD) were adopted to explore crystallography of ORI-NCs. Sulforhodamine B (SRB) method was used to test the inhibition effect on proliferation of MDCK cells. Quantitative analysis by HPLC was performed to study the endocytosis and transcytosis of ORI-NCs and ORI bulk drug, and the process was observed by confocal laser spectrum microscopy (CLSM) and flow cytometry.Results: The particle size of ORI-NCs was about 274 nm. The crystallography form of ORI was not changed after prepared into NCs. The dissolution rate of ORI-NCs was higher than pure ORI in 120 min. At higher concentrations (34, 84 and 135 μg/mL), ORI-NCs significantly reduced the cell viability compared with free ORI (p < 0.05, p < 0.01). ORI-NCs demonstrated higher endocytosis in MDCK cells than free ORI (p < 0.01). In the transport process, ORI-NC was taken up into cells in an intact form, and excreted out from basolateral membrane of polarized epithelial cells in an intact form. The internalization and transmembrane amount increased as a function of time.Conclusions: ORI-NCs transported through the MDCK monolayers in an intact form.
Collapse
Affiliation(s)
- Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanyuan Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jijuan Nai
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shaohua Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengmeng Dai
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guitao Lin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
46
|
Targeted nanomedicine with anti-EGFR scFv for siRNA delivery into triple negative breast cancer cells. Eur J Pharm Biopharm 2020; 157:74-84. [DOI: 10.1016/j.ejpb.2020.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/04/2020] [Accepted: 10/08/2020] [Indexed: 01/01/2023]
|
47
|
Forest CR, Silva CAC, Thordarson P. Dual‐peptide functionalized nanoparticles for therapeutic use. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Chelsea R. Forest
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of New South Wales Sydney New South Wales Australia
| | - Caitlin A. C. Silva
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of New South Wales Sydney New South Wales Australia
| | - Pall Thordarson
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of New South Wales Sydney New South Wales Australia
| |
Collapse
|
48
|
Zhou C, Xia Y, Wei Y, Cheng L, Wei J, Guo B, Meng F, Cao S, van Hest JCM, Zhong Z. GE11 peptide-installed chimaeric polymersomes tailor-made for high-efficiency EGFR-targeted protein therapy of orthotopic hepatocellular carcinoma. Acta Biomater 2020; 113:512-521. [PMID: 32562803 DOI: 10.1016/j.actbio.2020.06.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) remains a leading malignancy with a high mortality and little improvement in treatments. Protein drugs though known for their extraordinary potency and specificity have rarely been investigated for HCC therapy owing to lack of appropriate delivery systems. Here, we designed GE11 peptide-installed chimaeric polymersomes (GE11-CPs) for high-efficiency EGFR-targeted protein therapy of orthotopic SMMC-7721 HCC-bearing nude mice. GE11-CPs were assembled from poly(ethylene glycol)-b-poly(trimethylene carbonate-co-dithiolane trimethylene carbonate)-b-poly(aspartic acid) (PEG-P(TMC-DTC)-PAsp) and GE11-functionalized PEG-P(TMC-DTC), which allowed efficient loading and protection of proteins in the watery interior and fine-tuning of GE11 densities at the surface. CPs with short PAsp segments (degree of polymerization (DP) = 5, 10 and 15) exhibited a protein loading efficiency of 60%-72% and glutathione-responsive protein release. Saporin-loaded GE11-CPs had a size of 36 - 62 nm depending on GE11 densities and DP of PAsp. Notably, GE11-CPs with 10% GE11 revealed greatly enhanced uptake in SMMC-7721 cells, boosting the anticancer potency of saporin for over 3-folds compared with non-targeted control (half-maximal inhibitory concentration (IC50) = 11.0 versus 36.3 nM). The biodistribution studies using Cy5-labeled cytochrome C as a model protein demonstrated about 3-fold higher accumulation of GE11-CPs formulation than CPs counterpart in both subcutaneous and orthotopic SMMC-7721 tumor models. Notably, saporin-loaded GE11-CPs revealed low toxicity, effective tumor inhibition and significant improvement of survival rate compared with PBS and non-targeted groups (median survival time: 99 versus 37 and 42 days). EGFR-targeted chimaeric polymersomes carrying proteins appear an interesting HCC treatment modality.
Collapse
Affiliation(s)
- Cheng Zhou
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China
| | - Yifeng Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China
| | - Yaohua Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China
| | - Liang Cheng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China; Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China.
| | - Jingjing Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China.
| | - Shoupeng Cao
- Eindhoven University of Technology, P.O. Box 513 (STO 3.31), 5600MB Eindhoven, the Netherlands
| | - Jan C M van Hest
- Eindhoven University of Technology, P.O. Box 513 (STO 3.31), 5600MB Eindhoven, the Netherlands
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou, 215123, PR China.
| |
Collapse
|
49
|
Yan Y, Tan RZ, Liu P, Li JC, Zhong X, Liao Y, Lin X, Wei C, Wang L. Oridonin Alleviates IRI-Induced Kidney Injury by Inhibiting Inflammatory Response of Macrophages via AKT-Related Pathways. Med Sci Monit 2020; 26:e921114. [PMID: 32362652 PMCID: PMC7219002 DOI: 10.12659/msm.921114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is one of the most common complications in clinic, but there is still no effective treatment. Oridonin, extracted from Rabdosia rubescens, has been identified to promote inhibitory effects on tumor, inflammatory and fibrosis by previous study. This study aimed to assess the kidney-protective role of Oridonin in AKI and the underlying mechanism by which Oridonin improves AKI in vivo and inhibits inflammation in LPS-induced bone marrow-derived macrophages (BMDM) in vitro. MATERIAL AND METHODS SPF C57BL/6J male mice (8 - 10 weeks old, body weight 20 - 25 g) were divided into 3 groups - sham group, AKI group, and Oridonin-treated AKI group - with 6 mice in each group. In the in vitro study, LPS-induced inflammatory BMDM cells were treated with Oridonin and agonist of AKT. The expression and secretion levels of inflammation-related indicators and AKT-related signaling molecules were detected by real-time PCR, ELISA, Western blot, and immunofluorescence. Also, various methods are used to assess renal function and pathological changes. RESULTS The results showed that Oridonin treatment significantly improved the serum creatinine and BUN levels in AKI mice. Interestingly, treatment with Oridonin also resulted in decreased the infiltration of macrophages in renal tissues of AKI mice, which was associated with decreased expression and activation of AKT and its related signaling pathways, such as NF-kappaB and STAT3, suggesting that Oridonin attenuates AKI kidney injury via a mechanism associated with reducing the inflammatory response of macrophages in the AKI kidney. This was investigated in vitro in macrophages, and the results showed that Oridonin reduced the LPS-stimulated inflammatory response in macrophages. Mechanistically, the addition of Oridonin reversed LPS-induced downregulation of AKT, NF-kappaB, and STAT3 expression and inflammatory response in macrophages, suggesting that Oridonin has a protective role, via the AKT-related signaling pathways, in reducing the inflammatory response of macrophages in AKI mice. This was further confirmed by adding agonist of AKT of IGF-1 to block the inhibitory effect of Oridonin on inflammatory response in vitro. CONCLUSIONS Oridonin ameliorates AKI kidney injuries by suppressing AKT-mediated inflammatory response of macrophages.
Collapse
Affiliation(s)
- Ying Yan
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Rui-Zhi Tan
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Peng Liu
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Beijing, China (mainland)
| | - Jian-Chun Li
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Xia Zhong
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Yuan Liao
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Xiao Lin
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Cong Wei
- Clinical Laboratory, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Li Wang
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| |
Collapse
|
50
|
Li S, Lin L, Wang W, Yan X, Chen B, Jiang S, Liu S, Ma X, Tian H, Yu X. Aza-crown ether locked on polyethyleneimine: solving the contradiction between transfection efficiency and safety during in vivo gene delivery. Chem Commun (Camb) 2020; 56:5552-5555. [PMID: 32297607 DOI: 10.1039/c9cc10041e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We proposed a method using an aza-crown ether derivative to lock a hyperbranched polyethyleneimine, which endows the PEI25k with tumor targeting ability, anti-serum ability and extended circulation in the blood meanwhile retaining the high gene complexation and high transfection efficiency. The method we proposed here simultaneously endows cationic materials with high transfection efficiency and high safety, which greatly pushed the cationic materials to be applied in in vivo gene delivery.
Collapse
Affiliation(s)
- Shengran Li
- Laboratory of Polymer Composites Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|