1
|
Srimathi R, Sondak T, Kim KS. Cell-free supernatant-assisted biogenic silver nanoparticles enhance the antibacterial efficacy of communicating bacterial pathogens. BIOTECHNOL BIOPROC E 2024; 29:902-914. [DOI: 10.1007/s12257-024-00122-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 05/14/2025]
|
2
|
Wang K, Xia Z, Yu R, Zhang W, Wang Z, Zhu M, Li R, Hu Z, Chen Z, Xu K, Mu C. Novel Hydrogel Adjuvant of Chinese Medicine External Preparations for Accelerated Healing of Deep Soft Tissue Injuries. ACS Biomater Sci Eng 2024; 10:4425-4436. [PMID: 38597148 DOI: 10.1021/acsbiomaterials.4c00165] [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] [Indexed: 04/11/2024]
Abstract
Traditional Chinese medicine external prescriptions have displayed excellent clinical effects for treating deep soft tissue injuries. However, the effects cannot be fully utilized due to the limitations of their dosage forms and usage methods. It is still a challenge to develop a satisfactory adjuvant of traditional Chinese medicine external prescriptions. Herein, a hydrogel adjuvant was prepared based on gallic acid coupled ε-poly-l-lysine and partially oxidized hyaluronic acid. The resulting adjuvant shows great physicochemical properties, low hemolysis rate (still much less than 5% at 5 mg/mL), excellent antibacterial ability (about 95% at 2 mg/mL), strong antioxidant ability (1.687 ± 0.085 mmol FeSO4/(g hydrogel) at 1 mg/mL), as well as outstanding biocompatibility. A clinically used Chinese medicine external preparation was selected as an example to investigate the effectiveness of the adjuvant in treating deep soft tissue injuries. The results show that the prescription can be evenly dispersed in the adjuvant. Moreover, the introduction of the prescription has not significantly changed these advanced properties of the adjuvant. Importantly, the hydrogel adjuvant significantly improves the effectiveness of the prescription in treating deep soft tissue injuries. This work offers an alternative approach to the development of a new-type adjuvant of Chinese medicine external preparations and also provides a new strategy for the combination of traditional Chinese medicine and hydrogel to treat clinical diseases.
Collapse
Affiliation(s)
- Kai Wang
- The First Clinical Medical School of Anhui University of Chinese Medicine, Hefei 230038, Anhui, P. R. China
| | - Zuyan Xia
- The First Clinical Medical School of Anhui University of Chinese Medicine, Hefei 230038, Anhui, P. R. China
| | - Rui Yu
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei 230012, Anhui, P. R. China
| | - Wendi Zhang
- College of Acumox and Tuina, Anhui University of Chinese Medicine, Hefei 230012, Anhui, P. R. China
| | - Zijian Wang
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei 230012, Anhui, P. R. China
| | - Minshun Zhu
- The First Clinical Medical School of Anhui University of Chinese Medicine, Hefei 230038, Anhui, P. R. China
| | - Renzhong Li
- The First Clinical Medical School of Anhui University of Chinese Medicine, Hefei 230038, Anhui, P. R. China
| | - Zhimu Hu
- The First Clinical Medical School of Anhui University of Chinese Medicine, Hefei 230038, Anhui, P. R. China
| | - Zhaohui Chen
- College of Acumox and Tuina, Anhui University of Chinese Medicine, Hefei 230012, Anhui, P. R. China
| | - Kui Xu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei 230038, Anhui, P. R. China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, P. R. China
| | - Caiyun Mu
- College of Acumox and Tuina, Anhui University of Chinese Medicine, Hefei 230012, Anhui, P. R. China
| |
Collapse
|
3
|
Tran THM, Wang R, Kim H, Kim YJ. The anti-inflammation and skin-moisturizing effects of Boehmeria tricuspis-mediated biosynthesized gold nanoparticles in human keratinocytes. Front Pharmacol 2023; 14:1258057. [PMID: 37869754 PMCID: PMC10588637 DOI: 10.3389/fphar.2023.1258057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction: Recently, nanotechnology has emerged as a potential technique for skin generation, which has several treatment advantages, such as decreased drug cytotoxicity and enhanced skin penetration. Boehmeria tricuspis (BT) belongs to the Urticaceae family and is rich in phenolic and flavonoid compounds. In this study, we biosynthesized gold nanoparticles (BT-AuNPs) using BT extract to explore their anti-inflammatory and skin-moisturizing properties in keratinocytes. Methods: Field-emission transmission electron microscopy, energydispersive X-ray spectrometry, dynamic light scattering, and Fourier-transforminfrared spectroscopy were used to examine the synthesized BT-AuNPs. qRT-PCR, western blot, and ELISA were applied for investigating the effect of BT-AuNPs on anti-inflammation and moisturizing activity in HaCaT cells. Results: At concentrations below 200 μg/mL, BT-AuNPs had no cytotoxic effect on keratinocytes. BT-AuNPs dramatically alleviated the expression and secretion of inflammatory chemokines/cytokine, such as IL-6, IL-8, TARC, CTACK, and RANTES in keratinocytes stimulated by tumor necrosis factor-α/interferon-γ (T + I). These anti-inflammatory properties of BT-AuNPs were regulated by inhibiting the NF-κB and MAPKs signaling pathways. Furthermore, BT-AuNPs greatly promoted hyaluronic acid (HA) production by enhancing the expression of hyaluronic acid synthase genes (HAS1, HAS2, and HAS3) and suppressing the expression of hyaluronidase genes (HYAL1 and HYAL2) in HaCaT cells. Discussion: These results suggest that BT-AuNPs can be used as a promising therapeutic alternative for treating skin inflammation. Our findings provide a potential platform for the use of BT-AuNPs as candidates for treating inflammatory skin diseases and promoting skin health.
Collapse
Affiliation(s)
- Thi Hoa My Tran
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Yongin, Republic of Korea
| | - Rongbo Wang
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Yongin, Republic of Korea
| | - Hoon Kim
- Department of Food and Nutrition, Chung Ang University, Anseong, Republic of Korea
| | - Yeon-Ju Kim
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
4
|
Aili M, Zhou K, Zhan J, Zheng H, Luo F. Anti-inflammatory role of gold nanoparticles in the prevention and treatment of Alzheimer's disease. J Mater Chem B 2023; 11:8605-8621. [PMID: 37615596 DOI: 10.1039/d3tb01023f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that causes memory and cognitive dysfunction and reduces a person's decision-making and reasoning functions. AD is the leading cause of dementia in the elderly. Patients with AD have increased expression of pro-inflammatory cytokines in the nervous system, and the sustained inflammatory response impairs neuronal function. Meanwhile, long-term use of anti-inflammatory drugs can reduce the incidence of AD to some extent. This confirms that anti-neuroinflammation may be an effective treatment for AD. Gold nanoparticles (AuNPs) are an emerging nanomaterial with promising physicochemical properties, anti-inflammatory and antioxidant. AuNPs reduce neuroinflammation by inducing macrophage polarization toward the M2 phenotype, reducing pro-inflammatory cytokine expression, blocking leukocyte adhesion, and decreasing oxidative stress. Therefore, AuNPs are gradually attracting the interest of scholars and are used for treating inflammatory diseases and drug delivery. Herein, we explored the role and mechanism of AuNPs in treating neuroinflammation in AD. The use of AuNPs for treating AD is a topic worth exploring in the future, not only to help solve a global public health problem but also to provide a reference for treating other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Munire Aili
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Kebing Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jun Zhan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Huaping Zheng
- Department of Dermatology, Rare Diseases Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China
| |
Collapse
|
5
|
Jin Y, Guo Y, Yang J, Chu X, Huang X, Wang Q, Zeng Y, Su L, Lu S, Wang C, Yang J, Qu J, Yang Y, Wang B. A Novel "Inside-Out" Intraocular Nanomedicine Delivery Mode for Nanomaterials' Biological Effect Enhanced Choroidal Neovascularization Occlusion and Microenvironment Regulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209690. [PMID: 36527723 DOI: 10.1002/adma.202209690] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/08/2022] [Indexed: 06/17/2023]
Abstract
Photodynamic therapy (PDT) is commonly used in choroidal neovascularization (CNV) treatment due to the superior light transmittance of the eye. However, PDT often leads to surrounding tissue damage and further microenvironmental deterioration, including exacerbated hypoxia, inflammation, and secondary neovascularization. In this work, Pt nanoparticles (NPs) and Au NPs decorated zeolitic imidazolate framework-8 nanoplatform is developed to load indocyanine green for precise PDT and microenvironment amelioration, which can penetrate the internal limiting membrane through Müller cells endocytosis and target to CNV by surface-grafted cyclo(Arg-Gly-Asp-d-Phe-Lys) after intravitreal injection. The excessive H2 O2 in the CNV microenvironment is catalyzed by catalase-like Pt NPs for hypoxia relief and enhanced PDT occlusion of neovascular. Meanwhile, Au NPs show significant anti-inflammatory and anti-angiogenesis properties in regulating macrophages and blocking vascular endothelial growth factor (VEGF). Compared with verteporfin treatment, the mRNA expressions of hypoxia-inducible factor-1α and VEGF in the nanoplatform group are downregulated by 90.2% and 81.7%, respectively. Therefore, the nanoplatform realizes a comprehensive CNV treatment effect based on the high drug loading capacity and biosafety. The CNV treatment mode developed in this work provides a valuable reference for treating other diseases with similar physiological barriers that limit drug delivery and similar microenvironment.
Collapse
Affiliation(s)
- Yingying Jin
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Yishun Guo
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Jianhua Yang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Xiaoying Chu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Xiaomin Huang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Qingying Wang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Yanlin Zeng
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Lili Su
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Si Lu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Chenyang Wang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
| | - Jie Yang
- College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Jia Qu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
- National Engineering Research Center of Ophthalmology and Optometry, Wenzhou, 325027, P. R. China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, P. R. China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Wenzhou, 325027, P. R. China
| | - Yingwei Yang
- College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130012, P. R. China
| | - Bailiang Wang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China
- National Engineering Research Center of Ophthalmology and Optometry, Wenzhou, 325027, P. R. China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, P. R. China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Wenzhou, 325027, P. R. China
| |
Collapse
|
6
|
Comparison of In Vitro Estrogenic Activity of Polygoni multiflori Radix and Cynanchi wilfordii Radix via the Enhancement of ERα/β Expression in MCF7 Cells. Molecules 2023; 28:molecules28052199. [PMID: 36903444 PMCID: PMC10005224 DOI: 10.3390/molecules28052199] [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: 11/22/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Postmenopausal women experience several symptoms, including inflammation and a sharp rise in oxidative stress caused by estrogen deprivation. Although estrogen replacement therapy (ERT) is generally regarded as an effective treatment for menopause, it has been used less frequently due to some adverse effects and high costs. Therefore, there is an immediate need to develop an effective herbal-based treatment that is affordable for low-income populations. Acordingly, this study explored the estrogen-like properties of methanol extracts from Cynanchum wilfordii (CW) and Poligonum multiflorum (PM), two important medicinal plants in Republic of Korea, Japan, and China. Due to the similar names and morphologies of these two radixes, they are frequently confused in the marketplace. Our previous colleagues discriminated between these two plants. In this study, we investigated the estrogenic activity of PM and CW using several in vitro assays with their possible mechanism of action. First, their phytochemical contents, such as gallic acid, 2,3,5,4'-tetrahydroxystilbene-2-O-glucoside (TSG) and emodin, were quantified using high-performance liquid chromatography (HPLC). Secondly, estrogen-like activity was assessed utilizing the well-known E-screen test and gene expression analysis in estrogen receptor (ER)-positive MCF7 cells. ROS inhibition and anti-inflammatory effects were analyzed using HaCaT and Raw 264.7 cells, respectively. Our findings demonstrate that PM extracts significantly increased the expression of the estrogen-dependent genes (ERα, ERβ, pS2) and boosted MCF7 cell proliferation in comparison to CW extracts. Additionally, PM extract demonstrated a significant reduction in reactive oxygen species (ROS) production as well as an enhanced antioxidant profile compared to the CW extract. Further, the PM extract treatment significantly reduced the generation of nitric oxide (NO) in RAW 264.7 cells, a murine macrophage cell line, demonstrating the anti-inflammatory properties of the extract. Finally, this research offers an experimental foundation for the use of PM as a phytoestrogen to minimize menopausal symptoms.
Collapse
|
7
|
Farhadi R, Aroon MA, Ebrahimian Pirbazari A, Safarpour M, Matsuura T, Seirafi P. Simultaneous separation and degradation of methylene blue by a thin film nanocomposite membrane containing TiO 2/MWCNTs nanophotocatalyst. ENVIRONMENTAL TECHNOLOGY 2023; 44:670-685. [PMID: 34606412 DOI: 10.1080/09593330.2021.1982019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/31/2021] [Indexed: 06/13/2023]
Abstract
ABSTRACTAn innovative photocatalytic thin film nanocomposite (TFN) membrane was prepared and used for the simultaneous separation/degradation of methylene blue (MB) under UV irradiation. For this purpose, we used the sol-gel method to prepare the TiO2/MWCNTs nanophotocatalyst and added to the 1,3-phenylenediamine (MPD) solution during interfacial polymerization (IP) of 1,3,5-benzenetricarbonyl chloride (TMC) and 1,3-phenylenediamine monomers on the polysulfone (PSF) support. Using scanning electron microscopy (SEM) analysis and studying the cross-sectional images of membrane samples, it was revealed that the polyamide (PA) thin layer was well fabricated over the support membrane. The attendance of the TiO2/MWCNTs nanophotocatalyst in the PA thin layer of TFN samples was also proved using EDX (energy-dispersive X-ray) analysis. According to the results of the contact angle (CA), it is clear that the hydrophilicity of membrane samples first increased and then decreased by enhancing the TiO2/MWCNTs nanophotocatalyst content in the PA thin layer. In comparison with the pristine thin film composite (TFC) membrane, TFN samples showed higher water flux and MB removal when they were exposed to UV light. Finally, it turned out that the TFN membrane comprising 0.2 wt. % TiO2/MWCNTs nanophotocatalyst (TFN 0.2) had the foremost efficiency among TFNs with the water flux of 13 L/m2·hr and dye separation/degradation of almost 100% under UV irradiation.
Collapse
Affiliation(s)
- Roya Farhadi
- Membrane Research Laboratory, Caspian Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Ali Aroon
- Membrane Research Laboratory, Caspian Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Azadeh Ebrahimian Pirbazari
- Hybrid Nanomaterials & Environment Lab, Fouman Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mahdie Safarpour
- Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Takeshi Matsuura
- Industrial Membrane Research Institute, Department of Chemical Engineering, University of Ottawa, Ottawa, Canada
| | - Pouya Seirafi
- Membrane Research Laboratory, Caspian Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
8
|
Sun L, Wu J, Wang K, Liang T, Liu Q, Yan J, Yang Y, Qiao K, Ma S, Wang D. Comparative Analysis of Acanthopanacis Cortex and Periplocae Cortex Using an Electronic Nose and Gas Chromatography-Mass Spectrometry Coupled with Multivariate Statistical Analysis. Molecules 2022; 27:molecules27248964. [PMID: 36558097 PMCID: PMC9781861 DOI: 10.3390/molecules27248964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Chinese Herbal Medicines (CHMs) can be identified by experts according to their odors. However, the identification of these medicines is subjective and requires long-term experience. The samples of Acanthopanacis Cortex and Periplocae Cortex used were dried cortexes, which are often confused in the market due to their similar appearance, but their chemical composition and odor are different. The clinical use of the two herbs is different, but the phenomenon of being confused with each other often occurs. Therefore, we used an electronic nose (E-nose) to explore the differences in odor information between the two species for fast and robust discrimination, in order to provide a scientific basis for avoiding confusion and misuse in the process of production, circulation and clinical use. In this study, the odor and volatile components of these two medicinal materials were detected by the E-nose and by gas chromatography-mass spectrometry (GC-MS), respectively. An E-nose combined with pattern analysis methods such as principal component analysis (PCA) and partial least squares (PLS) was used to discriminate the cortex samples. The E-nose was used to determine the odors of the samples and enable rapid differentiation of Acanthopanacis Cortex and Periplocae Cortex. GC-MS was utilized to reveal the differences between the volatile constituents of Acanthopanacis Cortex and Periplocae Cortex. In all, 82 components including 9 co-contained components were extracted by chromatographic peak integration and matching, and 24 constituents could be used as chemical markers to distinguish these two species. The E-nose detection technology is able to discriminate between Acanthopanacis Cortex and Periplocae Cortex, with GC-MS providing support to determine the material basis of the E-nose sensors' response. The proposed method is rapid, simple, eco-friendly and can successfully differentiate these two medicinal materials by their odors. It can be applied to quality control links such as online detection, and also provide reference for the establishment of other rapid detection methods. The further development and utilization of this technology is conducive to the further supervision of the quality of CHMs and the healthy development of the industry.
Collapse
|
9
|
Zhu J, Liu Z, Pu Y, Xu J, Zhang S, Bao Y. Green synthesized gold nanoparticles from Pseudobulbus Cremastrae seu Pleiones show efficacy against hepatic carcinoma potentially through immunoregulation. Drug Deliv 2022; 29:1983-1993. [PMID: 35762637 PMCID: PMC9246265 DOI: 10.1080/10717544.2022.2092238] [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] [Indexed: 11/11/2022] Open
Abstract
Nanobiotechnology, the interface between biology and nanotechnology, has recently emerged in full bloom in the medical field due to its minimal side-effects and high efficiency. To broaden the application of nanobiotechnology, we composed gold nanoparticles from the extract of Pseudobulbus Cremastrae seu Pleiones (PCSP) using an efficient and green procedure. The biosynthesized Au nanoparticles containing PCSP (PCSP-AuNPs) were characterized by UV-vis spectroscopic, transmission electron microscopy (TEM), atomic force microscopy (AFM), dynamic light scattering (DLS), Fourier transform infrared spectroscopy (FT-IR), and Energy Dispersive X-ray (EDAX). After verifying the stability of PCSP-AuNPs, we detected its biosafety and immune-modulatory effects on RAW264.7 in vitro using NO assay, ELISA (TNF-α, IL-12p70, and IL-1β), and CCK-8 test. Furthermore, we examined the direct in vitro effects of PCSP-AuNPs on hepatocellular carcinomas (HCCs). Finally, we evaluated the immune regulation of PCSP-AuNPs using a mouse model with H22-tumor by testing the index of immune organs, splenic lymphocyte proliferation, cytokines levels (TNF-α and IL-10), and the CD4+/CD8+ cell ratio in the peripheral blood. Immunohistochemical analyses including H&E and PCNA staining were performed to investigate the anti-cancer efficacy and biocompatibility of PCSP-AuNPs. We found that PCSP-AuNPs not just possessed low toxicity, but also improved the immune-mediated antitumor response as compared to PCSP alone, suggesting its potential as a novel and efficient drug for liver cancer therapy.
Collapse
Affiliation(s)
- Junmo Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijing Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Youwei Pu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sitong Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Mulens-Arias V, Nicolás-Boluda A, Carn F, Gazeau F. Cationic Polyethyleneimine (PEI)–Gold Nanocomposites Modulate Macrophage Activation and Reprogram Mouse Breast Triple-Negative MET-1 Tumor Immunological Microenvironment. Pharmaceutics 2022; 14:pharmaceutics14102234. [PMID: 36297669 PMCID: PMC9607133 DOI: 10.3390/pharmaceutics14102234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Nanomedicines based on inorganic nanoparticles have grown in the last decades due to the nanosystems’ versatility in the coating, tuneability, and physical and chemical properties. Nonetheless, concerns have been raised regarding the immunotropic profile of nanoparticles and how metallic nanoparticles affect the immune system. Cationic polymer nanoparticles are widely used for cell transfection and proved to exert an adjuvant immunomodulatory effect that improves the efficiency of conventional vaccines against infection or cancer. Likewise, gold nanoparticles (AuNPs) also exhibit diverse effects on immune response depending on size or coatings. Photothermal or photodynamic therapy, radiosensitization, and drug or gene delivery systems take advantage of the unique properties of AuNPs to deeply modify the tumoral ecosystem. However, the collective effects that AuNPs combined with cationic polymers might exert on their own in the tumor immunological microenvironment remain elusive. The purpose of this study was to analyze the triple-negative breast tumor immunological microenvironment upon intratumoral injection of polyethyleneimine (PEI)–AuNP nanocomposites (named AuPEI) and elucidate how it might affect future immunotherapeutic approaches based on this nanosystem. AuPEI nanocomposites were synthesized through a one-pot synthesis method with PEI as both a reducing and capping agent, resulting in fractal assemblies of about 10 nm AuNPs. AuPEI induced an inflammatory profile in vitro in the mouse macrophage-like cells RAW264.7 as determined by the secretion of TNF-α and CCL5 while the immunosuppressor IL-10 was not increased. However, in vivo in the mouse breast MET-1 tumor model, AuPEI nanocomposites shifted the immunological tumor microenvironment toward an M2 phenotype with an immunosuppressive profile as determined by the infiltration of PD-1-positive lymphocytes. This dichotomy in AuPEI nanocomposites in vitro and in vivo might be attributed to the highly complex tumor microenvironment and highlights the importance of testing the immunogenicity of nanomaterials in vitro and more importantly in vivo in relevant immunocompetent mouse tumor models to better elucidate any adverse or unexpected effect.
Collapse
Affiliation(s)
- Vladimir Mulens-Arias
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Pompeu Fabra University, PRBB, Carrer Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Alba Nicolás-Boluda
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
| | - Florent Carn
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
| | - Florence Gazeau
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
- Correspondence:
| |
Collapse
|
11
|
Lee S, Shim HS, Park HJ, Chang Y, Han YE, Oh SJ, Lee W, Im H, Seol Y, Ryu H, Kang H, Lee YK, Park S, Yoo J. Elongated nanoporous Au networks improve somatic cell direct conversion into induced dopaminergic neurons for Parkinson's disease therapy. Acta Biomater 2022; 151:561-575. [PMID: 35931279 DOI: 10.1016/j.actbio.2022.07.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022]
Abstract
The efficient production of dopaminergic neurons via the direct conversion of other cell types is of interest as a potential therapeutic approach for Parkinson's disease. This study aimed to investigate the use of elongated porous gold nanorods (AuNpRs) as an enhancer of cell fate conversion. We observed that AuNpRs promoted the direct conversion of fibroblasts into dopaminergic neurons in vivo and in vitro. The extent of conversion of fibroblasts into dopaminergic neurons depended on the porosity of AuNpRs, as determined by their aspect ratio. The mechanism underlying these results involves specific AuNpR-induced transcriptional changes that altered the expression of antioxidant-related molecules. The generation of dopaminergic neurons via the direct conversion method will open a new avenue for developing a therapeutic platform for Parkinson's disease treatment. STATEMENT OF SIGNIFICANCE: In this study, we applied modified gold nanoporous materials (AuNpRs) to the direct lineage reprogramming of dopaminergic neurons. The cell reprogramming process is energy-intensive, resulting in an excess of oxidative stress. AuNpRs facilitated the direct conversion of dopaminergic neurons by ameliorating oxidative stress during the reprogramming process. We have found this mechanistic clue from high throughput studies in this research work.
Collapse
Affiliation(s)
- Sungwoo Lee
- Department of Chemistry, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon-si, Gyeongki-do, 16419, Republic of Korea
| | - Hyun Soo Shim
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea
| | - Hyeok Ju Park
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Database Laboratory, Department of Computer Science and Engineering, Dongguk University-Seoul, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Yujung Chang
- Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wonwoong Lee
- College of Pharmacy, Woosuk University, 443, Samnye-ro, Samnye-eup, Wanju_Gun, Jeollabuk-do, 55338, Republic of Korea
| | - Hyeonjoo Im
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Anatomy, College of Medicine, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 20841, Republic of Korea Seoul
| | - YunHee Seol
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hoon Kang
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea
| | - Yong Kyu Lee
- Database Laboratory, Department of Computer Science and Engineering, Dongguk University-Seoul, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Sungho Park
- Department of Chemistry, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon-si, Gyeongki-do, 16419, Republic of Korea.
| | - Junsang Yoo
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea.
| |
Collapse
|
12
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
13
|
Binary Effects of Gynostemma Gold Nanoparticles on Obesity and Inflammation via Downregulation of PPARγ/CEPBα and TNF-α Gene Expression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092795. [PMID: 35566145 PMCID: PMC9104634 DOI: 10.3390/molecules27092795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/23/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023]
Abstract
Nanoscience is a multidisciplinary skill with elucidated nanoscale particles and their advantages in applications to various fields. Owing to their economical synthesis, biocompatible nature, and widespread biomedical and environmental applications, the green synthesis of metal nanoparticles using medicinal plants has become a potential research area in biomedical research and functional food formulations. Gynostemma pentaphyllum (GP) has been extensively used in traditional Chinese medicine to cure several diseases, including diabetes mellitus (DM). This is the first study in which we examined the efficacy of G. pentaphyllum gold nanoparticles (GP-AuNPs) against obesity and related inflammation. GP extract was used as a capping agent to reduce Au2+ to Au0 to form stable gold nanoparticles. The nanoparticles were characterized by using UV–VIS spectroscopy, and TEM images were used to analyze morphology. In contrast, the existence of the functional group was measured using FTIR, and size and shape were examined using XRD analysis. In vitro analysis on GP-AuNPs was nontoxic to RAW 264.7 cells and 3T3-L1 cells up to a specific concentration. It significantly decreased lipid accumulation in 3T3-L1 obese and reduced NO production in Raw 264.7 macrophage cells. The significant adipogenic genes PPARγ and CEPBα and a major pro-inflammatory cytokine TNF-α expression were quantified using RT-PCR. The GP-AuNPs decreased the face of these genes remarkably, revealing the antiadipogenic and anti-inflammatory activity of our synthesized GP-AuNPs. This study represents thorough research on the antiobesity effect of Gynostemma pentaphyllum gold nanoparticles synthesized using a green approach and the efficacy instead of related inflammatory responses.
Collapse
|
14
|
Abdallah BM, Ali EM. Therapeutic Potential of Green Synthesized Gold Nanoparticles Using Extract of Leptadenia hastata against Invasive Pulmonary Aspergillosis. J Fungi (Basel) 2022; 8:jof8050442. [PMID: 35628698 PMCID: PMC9146234 DOI: 10.3390/jof8050442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 12/10/2022] Open
Abstract
Gold nanoparticles are widely used in the biomedical field for the treatment of several diseases, including cancer, inflammatory diseases, and immune system disorders, due to their distinctive physicochemical characteristics. In this study, we investigated the therapeutic potential of green synthesized gold nanoparticles using ethanolic leaf extract of Leptadenia hastata (LH-AuNPs) against invasive pulmonary aspergillosis (IPA) in mice. UV/visible spectroscopy, Fourier transform infrared spectroscopy (FTIR), transmission electron microscopy (TEM), X-ray diffraction (XRD), energy-dispersive X-ray spectroscopy (EDX), and zeta potential were used to characterize the biofabricated LH-AuNPs. Antifungal activity of LH-AuNPs was determined by MTT assay, (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide), time-kill assay, and radial growth inhibition. TEM and SEM were used to examine the mode of the antifungal action of LH-AuNPs. The in vivo activity of LH-AuNPs against IPA was studied using a well-established IPA mouse model. LH-AuNPs excreted antifungal activity against Aspergillus fumigatus with MIC 64 µg/mL and inhibited the radial growth of A. fumigatus by 30% compared to the control. LH-AuNPs caused distortion and collapse of fungal hyphae and deterioration of cell walls. Interestingly, LH-AuNPs did not display any cytotoxicity on cultured primary bone marrow stem cells (BMSCs) or A549 human lung cell line in vitro at MIC concentration. IPA mice treated with LH-AuNPs displayed significant lung tissue repair without any in vivo cytotoxicity. LH-AuNPs administration showed significant suppression of fungal burden and gliotoxin production in the lung. In addition, LH-AuNPs inhibited IPA-induced pro-inflammatory cytokines production, including interleukin-1 (IL-1), interleukin-17 (IL-17), and tumor necrosis factor-alpha (TNF-α), and reduced oxidative stress in lung. In conclusion, our data provide LH-AuNPs as a novel nanoparticle therapy for IPA.
Collapse
Affiliation(s)
- Basem M Abdallah
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Enas M Ali
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
15
|
Xiao Y, Ren C, Chen G, Shang P, Song X, You G, Yan S, Yao Y, Zhou H. Neutrophil membrane-mimicking nanodecoys with intrinsic anti-inflammatory properties alleviate sepsis-induced acute liver injury and lethality in a mouse endotoxemia model. Mater Today Bio 2022; 14:100244. [PMID: 35345558 PMCID: PMC8956822 DOI: 10.1016/j.mtbio.2022.100244] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/01/2022] [Accepted: 03/14/2022] [Indexed: 11/28/2022] Open
Abstract
Sepsis-induced acute liver injury often develops in the early stages of sepsis and can exacerbate the pathology by contributing to multiple organ dysfunction and increasing lethality. No specific therapies for sepsis-induced liver injury are currently available; therefore, effective countermeasures are urgently needed. Considering the crucial role of neutrophils in sepsis-induced liver injury, herein, neutrophil membrane-mimicking nanodecoys (NM) were explored as a biomimetic nanomedicine for the treatment of sepsis-associated liver injury. NM administration exhibited excellent biocompatibility and dramatically decreased the plasma levels of inflammatory cytokines and liver injury biomarkers, including aspartate aminotransferase, alanine aminotransferase, and direct bilirubin, in a sepsis mouse model. NM treatment also reduced hepatic malondialdehyde content, myeloperoxidase activity, and histological injury, and ultimately improved survival in the septic mice. Further in vitro studies showed that NM treatment neutralized the neutrophil chemokines and inflammatory mediators and directly mitigated neutrophil chemotaxis and adhesion. Additionally, NM also markedly weakened lipopolysaccharide-induced reactive oxygen species generation, cyclooxygenase-2 expression, nitric oxide secretion, and subsequent hepatocyte injury. Thus, this study provides a promising therapeutic strategy for the management of sepsis-induced acute liver injury.
Collapse
Affiliation(s)
- Yao Xiao
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Chao Ren
- Translational Medicine Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Gan Chen
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Pan Shang
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Xiang Song
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Shaoduo Yan
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Yongming Yao
- Translational Medicine Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, China
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Beijing, China
| |
Collapse
|
16
|
LUO J, LIU XQ, LEE GH, YOOK CS. Inhibition of LPS-induced expression of iNOS and COX-2 on extracts of Acanthopanax leucorrhizus (Oliv.) Harms stems. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.06122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Jiao LUO
- Hunan University of Chinese Medicine, China
| | | | | | | |
Collapse
|
17
|
LUO J, LI XJ, LEE GH, HUANG JJ, WHANG WK, ZHANG XD, YOOK CS, LIU XQ. Anti-inflammatory effects of two lupane-type triterpenes from leaves of Acanthopanax gracilistylus on LPS-induced RAW264.7 macrophages. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.89721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Jiao LUO
- Hunan University of Chinese Medicine, China
| | - Xiao-jun LI
- Hunan University of Chinese Medicine, China; Gannan Medical University, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Mikhailova EO. Gold Nanoparticles: Biosynthesis and Potential of Biomedical Application. J Funct Biomater 2021; 12:70. [PMID: 34940549 PMCID: PMC8708476 DOI: 10.3390/jfb12040070] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
Gold nanoparticles (AuNPs) are extremely promising objects for solving a wide range of biomedical problems. The gold nanoparticles production by biological method ("green synthesis") is eco-friendly and allows minimization of the amount of harmful chemical and toxic byproducts. This review is devoted to the AuNPs biosynthesis peculiarities using various living organisms (bacteria, fungi, algae, and plants). The participation of various biomolecules in the AuNPs synthesis and the influence of size, shapes, and capping agents on the functionalities are described. The proposed action mechanisms on target cells are highlighted. The biological activities of "green" AuNPs (antimicrobial, anticancer, antiviral, etc.) and the possibilities of their further biomedical application are also discussed.
Collapse
Affiliation(s)
- Ekaterina O Mikhailova
- Institute of Innovation Management, Kazan National Research Technological University, K. Marx Street 68, 420015 Kazan, Russia
| |
Collapse
|
19
|
Cai F, Li S, Huang H, Iqbal J, Wang C, Jiang X. Green synthesis of gold nanoparticles for immune response regulation: Mechanisms, applications, and perspectives. J Biomed Mater Res A 2021; 110:424-442. [PMID: 34331516 DOI: 10.1002/jbm.a.37281] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
Immune responses are involved in the pathogenesis of many diseases, including cancer, autoimmune diseases, and chronic inflammation. These responses are attributed to immune cells that produce cytokines, mediate cytotoxicity, and synthesize antibodies. Gold nanoparticles (GNPs) are novel agents that intervene with immune responses because of their unique physical-chemical properties. In particular, GNPs enhance anti-tumour activity during immunotherapy and eliminate excessive inflammation in autoimmune diseases. However, GNPs synthesized by conventional methods are toxic to living organisms. Green biosynthesis provides a safe and eco-friendly method to obtain GNPs from microbes or plant extracts. In this review, we describe several patterns for green GNP biosynthesis. The applications of GNPs to target immune cells and modulate the immune response are summarized. In particular, we elaborate on how GNPs regulate innate immunity and adaptive immunity, including inflammatory signaling and immune cell differentiation. Finally, perspectives and challenges in utilizing green biosynthesized GNPs for novel therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Feiyang Cai
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China.,School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyi Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Huang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Javed Iqbal
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Canran Wang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Jiang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Wahab S, Khan T, Adil M, Khan A. Mechanistic aspects of plant-based silver nanoparticles against multi-drug resistant bacteria. Heliyon 2021; 7:e07448. [PMID: 34286126 PMCID: PMC8273360 DOI: 10.1016/j.heliyon.2021.e07448] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/05/2021] [Accepted: 06/28/2021] [Indexed: 12/23/2022] Open
Abstract
Resistance among pathogenic bacteria to the existing antibiotics is one of the most alarming problems of the modern world. Alongwith reducing the use of antibiotics, and antibiotic stewardship, an alternative to antibiotics is much needed in the current scenario to combact infectious diseases. One alternative is to produce nanomaterials, especially, silver nanoparticles (AgNPs) against antibiotic-resistant bacteria. AgNPs are the most vital and fascinating nanoparticles because of their unique structural and functional properties and application against pathogenic bacteria. However, the synthesis of AgNPs remains a problem because of the chemicals and energy requirements and the byproducts of the reactions. Concerns have been raised about using chemically and physically synthesized nanoparticles because of their potential risks to the human body, animals, and environment. Green synthesis of these nanoparticles is a better alternative to physical and chemical approaches. Plant-based synthesis in turn is a method which can provide AgNPs that are cost-effective and eco-friendly as well as biocompatible. The specific features of size, morphology and shape of plant-based AgNPs give them the potency to fight multi-drug resistant bacteria. A detailed look into mechanistic aspects of the action of AgNPs against resistant bacteria with a focus on characteristic properties of AgNPs is required. This review discusses in detail these aspects and the potential of plant-based AgNPs as a solution to antibiotic resistance.
Collapse
Affiliation(s)
- Shahid Wahab
- Department of Biotechnology, University of Malakand, Chakdara Dir Lower, Pakistan
| | - Tariq Khan
- Department of Biotechnology, University of Malakand, Chakdara Dir Lower, Pakistan
| | - Muhammad Adil
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Centre for Nanoscience and Technology (NCNST), China
| | - Ajmal Khan
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| |
Collapse
|
21
|
Xu XY, Tran THM, Perumalsamy H, Sanjeevram D, Kim YJ. Biosynthetic gold nanoparticles of Hibiscus syriacus L. callus potentiates anti-inflammation efficacy via an autophagy-dependent mechanism. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112035. [PMID: 33947536 DOI: 10.1016/j.msec.2021.112035] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Biological applications of gold nanoparticles (AuNps) have potentially explored an efficient agent attributed to their biocompatibility and high efficiency in drug delivery. Our study applied an extract of Hibiscus syriacus L. callus (HCE) with a pioneer implementation on the induction of mass production. Bioactive compounds present in HCE were identified by Gas chromatography-mass spectrometry (GC-MS) and Liquid chromatography MS (LC-MS), wherein, the Denatonium was exclusively identifiable in HCE. Next, AuNps were synthesized and optimized using HCE (HCE-AuNps), and the comparison was conducted to evaluate the anti-inflammatory effect in lipopolysaccharide (LPS)-stimulated macrophages. As per result, HCE-AuNps was reported to show a prominent reduction of pro-inflammatory cytokines and renovate the mitochondrial function through restoring the mitochondrial membrane potential changes, decreasing reactive oxygen species (ROS) accumulation, and recovering ATP contents, respectively. Furthermore, the immunoblotting of LC3b/a accumulation, and p62 rapid degradation revealed that HCE-AuNps could induce the autophagy as an intracellular response to reinforce alleviation of pro-inflammatory cytokines and mitochondria dysfunction. Besides, 740 Y-P (PI3K agonist) was used to verify that inhibiting autophagy could partially reverse HCE-AuNps suppressed mitochondrial dysfunction, and thus exacerbated inflammation, supporting a causal role for autophagy in the anti-inflammatory effect of HCE-AuNps. Taken together, we strongly anticipate that HCE-AuNps would act as a potential autophagy inducer for LPS-triggered macrophage's inflammation, providing a novel insight for biosynthetic nanoparticles in the treatment of mitochondria dysfunction and inflammation related diseases.
Collapse
Affiliation(s)
- Xing Yue Xu
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Thi Hoa My Tran
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Haribalan Perumalsamy
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Dhandapani Sanjeevram
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Yeon-Ju Kim
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
22
|
Ali MR, Bacchu MS, Al-Mamun MR, Ahommed MS, Saad Aly MA, Khan MZH. N-Hydroxysuccinimide crosslinked graphene oxide–gold nanoflower modified SPE electrode for sensitive detection of chloramphenicol antibiotic. RSC Adv 2021; 11:15565-15572. [PMID: 35481161 PMCID: PMC9029409 DOI: 10.1039/d1ra02450g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/21/2021] [Indexed: 11/27/2022] Open
Abstract
Here we introduce a composite material that consists of graphene oxide (GO) sheets crosslinked with N-hydroxysuccinimide (NHS) and functionalized with gold nanoflowers (AuNFs). Furthermore, a screen printed electrode (SPE) modified with the introduced composite is electrochemically reduced to obtain an SPE/rGO–NHS–AuNFs electrode for sensitive and selective determination of chloramphenicol (CAP) antibiotic drug. The morphological structure of the as-prepared nanocomposite was characterized by scanning electron microscopy, energy-dispersive X-ray spectroscopy, cyclic voltammetry, Fourier-transform infrared spectroscopy and electrochemical impedance spectroscopy. The proposed sensor demonstrated excellent performance with a linear concentration range of 0.05 to 100 μM and a detection limit of 1 nM. The proposed electrode offers a high level of selectivity, stability, reproducibility and a satisfactory recovery rate for electrochemical detection of CAP in real samples such as blood serum, poultry feed, milk, eggs, honey and powdered milk samples. This further demonstrates the practical feasibility of the proposed sensor in food analysis. Here we introduce a composite material that consists of graphene oxide (GO) sheets crosslinked with N-hydroxysuccinimide (NHS) and functionalized with gold nanoflowers (AuNFs).![]()
Collapse
Affiliation(s)
- M. R. Ali
- Dept of Chemical Engineering
- Jashore University of Science and Technology
- Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME)
- Jashore University of Science and Technology
| | - M. S. Bacchu
- Dept of Chemical Engineering
- Jashore University of Science and Technology
- Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME)
- Jashore University of Science and Technology
| | - M. R. Al-Mamun
- Dept of Chemical Engineering
- Jashore University of Science and Technology
- Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME)
- Jashore University of Science and Technology
| | - M. S. Ahommed
- Department of Chemistry
- Graduate School of Science
- Tohoku University
- Sendai 980-8578
- Japan
| | - M. Aly Saad Aly
- Department of Electronics and Information Science
- Miami College of Henan University
- Kaifeng 475000
- China
| | - M. Z. H. Khan
- Dept of Chemical Engineering
- Jashore University of Science and Technology
- Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME)
- Jashore University of Science and Technology
| |
Collapse
|
23
|
Huo S, Chen C, Lyu Z, Zhang S, Wang Y, Nie B, Yue B. Overcoming Planktonic and Intracellular Staphylococcus aureus-Associated Infection with a Cell-Penetrating Peptide-Conjugated Antimicrobial Peptide. ACS Infect Dis 2020; 6:3147-3162. [PMID: 33236626 DOI: 10.1021/acsinfecdis.0c00264] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Staphylococcus aureus is a primary pathogen responsible for causing postoperative infections as it survives and persists in host cells, including osteoblasts and macrophages. These cells then serve as reservoirs resulting in chronic infections. Most traditional antibiotics have poor effects on intracellular S. aureus because they cannot enter the cell. Herein, a cell-penetrating peptide TAT-KR-12 was derived from the trans-activating transcription (TAT) peptide and KR-12 (residues 18-29 of human cathelicidin LL-37). The TAT acts as a "trojan horse" to deliver KR-12 peptide into the cells to kill S. aureus. Moreover, effective antibacterial properties and biocompatibility were observed in vitro, demonstrating that TAT-KR-12 is effective not only in eliminating planktonic S. aureus, but also in eliminating intracellular S. aureus cells in vitro. TAT-KR-12, as with LL-37, also elicits strong anti-inflammatory activities in LPS-stimulated macrophages, as demonstrated by significant inhibition of NO, TNF-α, and IL-1β expression and secretion from LPS-stimulated RAW264.7 cells. In the subcutaneous infection mouse model of planktonic and intracellular infections, the growth of S. aureus in vivo is evidently inhibited without cytotoxicity. These results suggest that the novel antimicrobial TAT-KR-12 may prove to be an effective treatment option to overcome antibiotic resistance caused by intracellular bacterial infections.
Collapse
Affiliation(s)
- Shicheng Huo
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Chi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, China
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Bin’en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| |
Collapse
|
24
|
Exploring the physicochemical and antimicrobial properties of gold-chitosan hybrid nanoparticles composed of varying chitosan amounts. Int J Biol Macromol 2020; 162:1760-1769. [DOI: 10.1016/j.ijbiomac.2020.08.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022]
|
25
|
Mioc A, Mioc M, Ghiulai R, Voicu M, Racoviceanu R, Trandafirescu C, Dehelean C, Coricovac D, Soica C. Gold Nanoparticles as Targeted Delivery Systems and Theranostic Agents in Cancer Therapy. Curr Med Chem 2019; 26:6493-6513. [PMID: 31057102 DOI: 10.2174/0929867326666190506123721] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
Cancer is still a leading cause of death worldwide, while most chemotherapies induce nonselective toxicity and severe systemic side effects. To address these problems, targeted nanoscience is an emerging field that promises to benefit cancer patients. Gold nanoparticles are nowadays in the spotlight due to their many well-established advantages. Gold nanoparticles are easily synthesizable in various shapes and sizes by a continuously developing set of means, including chemical, physical or eco-friendly biological methods. This review presents gold nanoparticles as versatile therapeutic agents playing many roles, such as targeted delivery systems (anticancer agents, nucleic acids, biological proteins, vaccines), theranostics and agents in photothermal therapy. They have also been outlined to bring great contributions in the bioimaging field such as radiotherapy, magnetic resonance angiography and photoacoustic imaging. Nevertheless, gold nanoparticles are therapeutic agents demonstrating its in vitro anti-angiogenic, anti-proliferative and pro-apoptotic effects on various cell lines, such as human cervix, human breast, human lung, human prostate and murine melanoma cancer cells. In vivo studies have pointed out data regarding the bioaccumulation and cytotoxicity of gold nanoparticles, but it has been emphasized that size, dose, surface charge, sex and especially administration routes are very important variables.
Collapse
Affiliation(s)
- Alexandra Mioc
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Marius Mioc
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Roxana Ghiulai
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Mirela Voicu
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Roxana Racoviceanu
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Cristina Trandafirescu
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Cristina Dehelean
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Dorina Coricovac
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| | - Codruta Soica
- Faculty of Pharmacy, 'Victor Babes' University of Medicine and Pharmacy, Timisoara 300041, Romania
| |
Collapse
|
26
|
Shcharbin D, Halets-Bui I, Abashkin V, Dzmitruk V, Loznikova S, Odabaşı M, Acet Ö, Önal B, Özdemir N, Shcharbina N, Bryszewska M. Hybrid metal-organic nanoflowers and their application in biotechnology and medicine. Colloids Surf B Biointerfaces 2019; 182:110354. [DOI: 10.1016/j.colsurfb.2019.110354] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/07/2019] [Indexed: 12/11/2022]
|
27
|
Ramalingam V. Multifunctionality of gold nanoparticles: Plausible and convincing properties. Adv Colloid Interface Sci 2019; 271:101989. [PMID: 31330396 DOI: 10.1016/j.cis.2019.101989] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/17/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022]
Abstract
In a couple of decades, nanotechnology has become a trending area in science due to it covers all subject that combines diverse range of fields including but not limited to chemistry, physics and medicine. Various metal and metal oxide nanomaterials have been developed for wide range applications. However, the application of gold nanostructures and nanoparticles has been received more attention in various biomedical applications. The unique property of gold nanoparticles (AuNPs) is surface plasmon resonance (SPR) that determine the size, shape and stability. The wide surface area of AuNPs eases the proteins, peptides, oligonucleotides, and many other compounds to tether and enhance the biological activity of AuNPs. AuNPs have multifunctionality including antimicrobial, anticancer, drug and gene delivery, sensing applications and imaging. This state-of-the-art review is focused on the role of unique properties of AuNPs in multifunctionality and its various applications.
Collapse
|
28
|
Yin X, Li Y, Yang C, Weng J, Wang J, Zhou J, Feng B. Alginate/chitosan multilayer films coated on IL-4-loaded TiO2 nanotubes for modulation of macrophage phenotype. Int J Biol Macromol 2019; 133:503-513. [DOI: 10.1016/j.ijbiomac.2019.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 01/21/2023]
|
29
|
Alginate/chitosan multilayer films coated on IL-4-loaded TiO2 nanotubes for modulation of macrophage phenotype. Int J Biol Macromol 2019; 132:495-505. [DOI: 10.1016/j.ijbiomac.2019.03.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/08/2019] [Accepted: 03/25/2019] [Indexed: 11/24/2022]
|
30
|
Ni C, Zhou J, Kong N, Bian T, Zhang Y, Huang X, Xiao Y, Yang W, Yan F. Gold nanoparticles modulate the crosstalk between macrophages and periodontal ligament cells for periodontitis treatment. Biomaterials 2019; 206:115-132. [DOI: 10.1016/j.biomaterials.2019.03.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/23/2019] [Indexed: 12/12/2022]
|
31
|
Ke Y, Al Aboody MS, Alturaiki W, Alsagaby SA, Alfaiz FA, Veeraraghavan VP, Mickymaray S. Photosynthesized gold nanoparticles from Catharanthus roseus induces caspase-mediated apoptosis in cervical cancer cells (HeLa). ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1938-1946. [DOI: 10.1080/21691401.2019.1614017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yang Ke
- Department of Oncology, Zhengzhou Central Hospital Affiliated To Zhengzhou University, Zhengzhou City, China
| | - Mohammed Saleh Al Aboody
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Majmaah, Kingdom of Saudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratories, College of Applied Medical Science, Majmaah University, Majmaah, Kingdom of Saudi Arabia
| | - Suliman A. Alsagaby
- Department of Medical Laboratories, College of Applied Medical Science, Majmaah University, Majmaah, Kingdom of Saudi Arabia
| | - Faiz Abdulaziz Alfaiz
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Majmaah, Kingdom of Saudi Arabia
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Suresh Mickymaray
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Majmaah, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
Kim K, Shin KM, Hunt CL, Wang Z, Bauer BA, Kwon O, Lee JH, Seo BN, Jung SY, Youn Y, Lee SH, Choi JC, Jung JE, Kim J, Qu W, Kim TH, Eldrige JS. Nonsurgical integrative inpatient treatments for symptomatic lumbar spinal stenosis: a multi-arm randomized controlled pilot trial. J Pain Res 2019; 12:1103-1113. [PMID: 30992679 PMCID: PMC6445233 DOI: 10.2147/jpr.s173178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background Lumbar spinal stenosis (LSS) is a chronic condition that causes low back pain and neurogenic claudication, often resulting in significant limitation of daily activities. In this open-label randomized controlled pilot study, we assessed the safety and feasibility of 4-week novel integrative inpatient treatments for LSS. Methods Thirty-six symptomatic LSS patients were randomly and equally allocated to one of the three groups: Mokhuri Chuna treatment 1 (MT1) group, Mokhuri Chuna treatment 2 (MT2) group, or conventional management treatment (CMT) group. MT1 patients were treated with herbal medication, Mokhuri Chuna, and acupuncture, and received daily physician consultation; MT2 patients were treated with Mokhuri Chuna and acupuncture without any herbal medication, and received daily physician consultation; and CMT patients received conventional pain management therapy that included epidural steroid injection, oral NSAID, and muscle relaxant medication, along with daily physiotherapy. The primary outcome of this pilot study was safety as measured by the type and incidence of adverse events (AEs). The secondary outcome measures included VAS score for low back pain and leg pain, Oswestry Disability Index, Oxford Claudication Score (OCS), walking capacity on a 50 m flat track and treadmill, and EuroQol-5D score. Magnetic resonance imaging was also performed up to 6 months after treatment cessation. Results Thirty-four treated patients were included in the analysis, based on the modified intention-to-treat principle. No serious AEs were observed or reported. Compared to the CMT group, the MT1 and MT2 groups did show significant improvement at 3 and 6 months in various domains, including pain (VAS score for leg and back pain) and function (OCS and treadmill walking). Conclusion These novel multimodal integrative treatments for LSS are both clinically safe and logistically feasible. Larger, adequately powered randomized controlled trials will be necessary to assess comparative efficacy and thoroughly analyze the cost-effectiveness of each treatment approach. Clinical trial registration number (CRIS) KCT0001218.
Collapse
Affiliation(s)
- Kiok Kim
- Department of Spine Center, Mokhuri Neck & Back Hospital, Seoul, South Korea
| | - Kyung-Min Shin
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Christy L Hunt
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Zhen Wang
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Brent A Bauer
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ojin Kwon
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Jun-Hwan Lee
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,Korean Medicine Life Science, Campus of Korea Institute of Oriental Medicine, University of Science & Technology (UST), Daejeon, South Korea
| | - Bok-Nam Seo
- Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - So-Young Jung
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Yousuk Youn
- Department of Spine Center, Mokhuri Neck & Back Hospital, Seoul, South Korea
| | - Sang Ho Lee
- Department of Spine Center, Mokhuri Neck & Back Hospital, Seoul, South Korea
| | - Jung Chul Choi
- Department of Spine Center, Mokhuri Neck & Back Hospital, Seoul, South Korea
| | - Jae Eun Jung
- Hongik Neurosurgery Hospital, Seongnam, South Korea
| | - Jaehong Kim
- Department of Spine Center, Mokhuri Neck & Back Hospital, Seoul, South Korea
| | - Wenchun Qu
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA.,Department of Anesthesiology, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA,
| | - Tae-Hun Kim
- Korean Medicine Clinical Trial Center, Korean Medicine Hospital, Kyung Hee University, Seoul, South Korea
| | - Jason S Eldrige
- Department of Anesthesiology, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA,
| |
Collapse
|
33
|
Dexmedetomidine protects against lipopolysaccharide-induced early acute kidney injury by inhibiting the iNOS/NO signaling pathway in rats. Nitric Oxide 2019; 85:1-9. [PMID: 30659917 DOI: 10.1016/j.niox.2019.01.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
Increasing evidence has demonstrated that dexmedetomidine (DEX) possesses multiple pharmacological actions. Herein, we explored the protective effect and potential molecular mechanism of DEX on lipopolysaccharide (LPS)-induced early acute kidney injury (AKI) from the perspective of antioxidant stress. We found that DEX (30 μg/kg, i.p.) ameliorated the renal dysfunction and histopathological damage (tubular necrosis, vacuolar degeneration, infiltration of inflammatory cells and cast formation) induced by LPS (10 mg/kg). DEX also attenuated renal oxidative stress remarkably in LPS-induced early AKI, as evidenced by reduction in production of reactive nitrogen species, decreasing malondialdehyde levels, as well as increasing superoxide dismutase activity and glutathione content. DEX prevented activator protein-1 translocation, inhibited phosphorylation of I-kappa B (IκB) and activation of nuclear factor kappa B (NF-κB) in LPS-induced early AKI, as assessed by real-time quantitative polymerase chain reaction and protein levels of c-Jun, c-Fos, IκB and NF-κB. Notably, DEX pretreatment had the same effect as intraperitoneal injection of an inhibitor of inducible nitric oxide synthase inhibitor (1400W; 15 mg/kg), and inhibited the activity of renal inducible nitric oxide synthase (iNOS) and decreased the expression of iNOS mRNA and NO production. However, the protective effect of DEX on LPS-induced early AKI was reversed by the alpha 2 adrenal receptor (α2-AR) inhibitor atipamezole, whereas the imidazoline receptor inhibitor idazoxan did not. Taken together, DEX protects against LPS-induced early AKI in rats by inhibiting the iNOS/NO signaling pathway, mainly by acting on α2-ARs instead of IRs.
Collapse
|
34
|
Singh P, Garg A, Pandit S, Mokkapati VRSS, Mijakovic I. Antimicrobial Effects of Biogenic Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E1009. [PMID: 30563095 PMCID: PMC6315689 DOI: 10.3390/nano8121009] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/23/2022]
Abstract
Infectious diseases pose one of the greatest health challenges in the medical world. Though numerous antimicrobial drugs are commercially available, they often lack effectiveness against recently developed multidrug resistant (MDR) microorganisms. This results in high antibiotic dose administration and a need to develop new antibiotics, which in turn requires time, money, and labor investments. Recently, biogenic metallic nanoparticles have proven their effectiveness against MDR microorganisms, individually and in synergy with the current/conventional antibiotics. Importantly, biogenic nanoparticles are easy to produce, facile, biocompatible, and environmentally friendly in nature. In addition, biogenic nanoparticles are surrounded by capping layers, which provide them with biocompatibility and long-term stability. Moreover, these capping layers provide an active surface for interaction with biological components, facilitated by free active surface functional groups. These groups are available for modification, such as conjugation with antimicrobial drugs, genes, and peptides, in order to enhance their efficacy and delivery. This review summarizes the conventional antibiotic treatments and highlights the benefits of using nanoparticles in combating infectious diseases.
Collapse
Affiliation(s)
- Priyanka Singh
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Abhroop Garg
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Santosh Pandit
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, 41296 Chalmers, Sweden.
| | - V R S S Mokkapati
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, 41296 Chalmers, Sweden.
| | - Ivan Mijakovic
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, 41296 Chalmers, Sweden.
| |
Collapse
|
35
|
Shin NR, Park SH, Ko JW, Cho YK, Lee IC, Kim JC, Shin IS, Kim JS. Lobeglitazone Attenuates Airway Inflammation and Mucus Hypersecretion in a Murine Model of Ovalbumin-Induced Asthma. Front Pharmacol 2018; 9:906. [PMID: 30135657 PMCID: PMC6092601 DOI: 10.3389/fphar.2018.00906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/24/2018] [Indexed: 01/13/2023] Open
Abstract
Lobeglitazone (LB) is a novel agonist of peroxisome proliferator-activated receptor (PPAR)-α and γ that was developed as a drug to treat diabetes mellitus. We explored the ameliorative effects of LB on allergic asthma using a murine model of ovalbumin (OVA)-induced asthma. To boost the immune response of animals, OVA sensitization was performed on days 0 and 14. LB (250 or 500 μg/kg) was administered by oral gavage on days 18 to 23, and the OVA challenge was performed using an ultrasonic nebulizer on days 21 to 23. Plethysmography showed airway hyperresponsiveness (AHR) on day 24. LB treatment effectively decreased inflammatory cell recruitment, T-helper type 2 cytokines in the bronchoalveolar lavage fluid, and immunoglobulin (Ig) E in the serum of the animals with OVA-induced asthma, which was accompanied by a marked reduction in AHR. It also decreased airway inflammation, mucus hypersecretion, phosphorylation of nuclear transcription factor-kappa-B (NF-κB), and expression of activating protein (AP)-1 and mucin 5AC (MUC5AC). Overall, LB effectively attenuated the pathophysiological changes of asthma and its effects appear related to a reduction in the phosphorylation of NF-κB and the expression of AP-1. Thus, our results suggest that LB has a potential to treat allergic asthma.
Collapse
Affiliation(s)
- Na-Rae Shin
- College of Veterinary Medicine BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Sung-Hyeuk Park
- College of Veterinary Medicine BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Je-Won Ko
- College of Veterinary Medicine BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, Cheongju, South Korea
| | - In-Chul Lee
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, South Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - In-Sik Shin
- College of Veterinary Medicine BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Joong-Sun Kim
- K-herb Research Center, Korea Institute of Oriental Medicine, Daejeon, South Korea
| |
Collapse
|
36
|
Marslin G, Siram K, Maqbool Q, Selvakesavan RK, Kruszka D, Kachlicki P, Franklin G. Secondary Metabolites in the Green Synthesis of Metallic Nanoparticles. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E940. [PMID: 29865278 PMCID: PMC6024997 DOI: 10.3390/ma11060940] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 05/25/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022]
Abstract
The ability of organisms and organic compounds to reduce metal ions and stabilize them into nanoparticles (NPs) forms the basis of green synthesis. To date, synthesis of NPs from various metal ions using a diverse array of plant extracts has been reported. However, a clear understanding of the mechanism of green synthesis of NPs is lacking. Although most studies have neglected to analyze the green-synthesized NPs (GNPs) for the presence of compounds derived from the extract, several studies have demonstrated the conjugation of sugars, secondary metabolites, and proteins in these biogenic NPs. Despite several reports on the bioactivities (antimicrobial, antioxidant, cytotoxic, catalytic, etc.) of GNPs, only a handful of studies have compared these activities with their chemically synthesized counterparts. These comparisons have demonstrated that GNPs possess better bioactivities than NPs synthesized by other methods, which might be attributed to the presence of plant-derived compounds in these NPs. The ability of NPs to bind with organic compounds to form a stable complex has huge potential in the harvesting of precious molecules and for drug discovery, if harnessed meticulously. A thorough understanding of the mechanisms of green synthesis and high-throughput screening of stabilizing/capping agents on the physico-chemical properties of GNPs is warranted to realize the full potential of green nanotechnology.
Collapse
Affiliation(s)
- Gregory Marslin
- Ratnam Institute of Pharmacy and Research, Nellore 524346, India.
| | - Karthik Siram
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore 641004, India.
| | - Qaisar Maqbool
- Institute of Plant Genetics of the Polish Academy of Sciences, Poznan 60479, Poland.
| | | | - Dariusz Kruszka
- Institute of Plant Genetics of the Polish Academy of Sciences, Poznan 60479, Poland.
| | - Piotr Kachlicki
- Institute of Plant Genetics of the Polish Academy of Sciences, Poznan 60479, Poland.
| | - Gregory Franklin
- Institute of Plant Genetics of the Polish Academy of Sciences, Poznan 60479, Poland.
| |
Collapse
|
37
|
Yao Y, Liu K, Zhao Y, Hu X, Wang M. Pterostilbene and 4'-Methoxyresveratrol Inhibited Lipopolysaccharide-Induced Inflammatory Response in RAW264.7 Macrophages. Molecules 2018; 23:molecules23051148. [PMID: 29751609 PMCID: PMC6100408 DOI: 10.3390/molecules23051148] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 01/20/2023] Open
Abstract
Pterostilbene (Pte) and 4′-Methoxyresveratrol (4MR) are methylated derivatives of resveratrol. We investigated the anti-inflammatory effect of Pte and 4MR in lipopolysaccharide (LPS)-stimulated RAW264.7 murine macrophages. Both Pte and 4MR significantly reduced LPS-induced nitric oxide release by inhibiting the inducible nitric oxide synthase mRNA expression. Moreover, both of them inhibited LPS-induced mRNA expression of inflammatory cytokines including monocyte chemoattractant protein (MCP)-1, interleukin (IL)-6 and IL-1β, and tumor necrosis factor α (TNF-α), and attenuated LPS-induced nuclear factor-κB (NF-κB) activation by decreasing p65 phosphorylation. In addition, 4MR but not Pte inhibited LPS-induced the activator protein (AP)-1 pathway in RAW 264.7 macrophages. Further study suggested that Pte had an inhibitory effect on extracellular regulated protein kinases (ERK) and p38 activation, but not on c-Jun N-terminal kinase (JNK), while 4MR had an inhibitory effect on JNK and p38 activation, but not on ERK. Taken together, our data suggested that Pte induced anti-inflammatory activity by blocking mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways, while 4MR showed anti-inflammatory activity through suppression of MAPK, AP-1, and NF-κB signaling pathways in LPS-treated RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Yun Yao
- College of Food Science and Technology, Shanghai Ocean University, No.999 Hu-Cheng-Huan Road, Shanghai 201306, China.
| | - Kehai Liu
- College of Food Science and Technology, Shanghai Ocean University, No.999 Hu-Cheng-Huan Road, Shanghai 201306, China.
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China.
| | - Yueliang Zhao
- College of Food Science and Technology, Shanghai Ocean University, No.999 Hu-Cheng-Huan Road, Shanghai 201306, China.
| | - Xiaoqian Hu
- College of Food Science and Technology, Shanghai Ocean University, No.999 Hu-Cheng-Huan Road, Shanghai 201306, China.
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China.
| | - Mingfu Wang
- College of Food Science and Technology, Shanghai Ocean University, No.999 Hu-Cheng-Huan Road, Shanghai 201306, China.
| |
Collapse
|
38
|
Borah D, Hazarika M, Tailor P, Silva AR, Chetia B, Singaravelu G, Das P. Starch-templated bio-synthesis of gold nanoflowers for in vitro antimicrobial and anticancer activities. APPLIED NANOSCIENCE 2018. [DOI: 10.1007/s13204-018-0793-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Abstract
We describe an in situ method of synthesizing highly branched gold nanoflower (AuNFs) using aqueous seed extract of Syzygium cumini (L.) Skeels as reductant in the presence of 0.3% starch. Surprisingly, when the same reaction was carried out in the absence of starch or with starch at a lower concentration (0.15%), instead of flower-like morphology quasi-spherical or polyhedral nanoparticles (AuNPs) are obtained. The nanomaterials were extensively characterized by HRTEM, FESEM, UV–Vis, FTIR, XRD, XPS and TGA analysis. The biological activities of the materials were investigated for antimicrobial activities against four bacterial strains that include one Gram positive (Staphylococcus aureus MTCC 121), two Gram negative (Escherichia coli MTCC 40 and Pseudomonas aeruginosa MTCC 4673) and one fungi (Candida albicans MTCC 227). The nanoparticles functioned as effective antimicrobial and anti-biofilm agents against all the strains under study. Controlled study revealed that, the AuNFs showed improved efficacy over conventional polyhedral AuNPs against all the microbes under study which might be attributed to the larger surface-to-volume ratio of the nanoflowers. The AuNFs also showed effective in vitro anticancer activity against a human liver cancer cell line (HepG2) with no significant cytotoxicity. Our data suggest that the AuNFs can significantly reduce the cancer cell growth with IC50 value of 20 µg mL−1.
Collapse
|