1
|
Lin Y, Xie R, Yu T. Photodynamic Therapy for Atherosclerosis: Past, Present, and Future. Pharmaceutics 2024; 16:729. [PMID: 38931851 PMCID: PMC11206729 DOI: 10.3390/pharmaceutics16060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
This review paper examines the evolution of photodynamic therapy (PDT) as a novel, minimally invasive strategy for treating atherosclerosis, a leading global health concern. Atherosclerosis is characterized by the accumulation of lipids and inflammation within arterial walls, leading to significant morbidity and mortality through cardiovascular diseases such as myocardial infarction and stroke. Traditional therapeutic approaches have primarily focused on modulating risk factors such as hypertension and hyperlipidemia, with emerging evidence highlighting the pivotal role of inflammation. PDT, leveraging a photosensitizer, specific-wavelength light, and oxygen, offers targeted treatment by inducing cell death in diseased tissues while sparing healthy ones. This specificity, combined with advancements in nanoparticle technology for improved delivery, positions PDT as a promising alternative to traditional interventions. The review explores the mechanistic basis of PDT, its efficacy in preclinical studies, and the potential for enhancing plaque stability and reducing macrophage density within plaques. It also addresses the need for further research to optimize treatment parameters, mitigate adverse effects, and validate long-term outcomes. By detailing past developments, current progress, and future directions, this paper aims to highlight PDT's potential in revolutionizing atherosclerosis treatment, bridging the gap from experimental research to clinical application.
Collapse
Affiliation(s)
- Yanqing Lin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China;
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI 53705, USA;
| | - Tao Yu
- Department of Cardiac Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
2
|
Mahdizade Ari M, Amirmozafari N, Atieh Darbandi, Afifirad R, Asadollahi P, Irajian G. Effectiveness of photodynamic therapy on the treatment of chronic periodontitis: a systematic review during 2008-2023. Front Chem 2024; 12:1384344. [PMID: 38817441 PMCID: PMC11138352 DOI: 10.3389/fchem.2024.1384344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/10/2024] [Indexed: 06/01/2024] Open
Abstract
Objective This study investigated the effect of photodynamic therapy on chronic periodontitis patients and then evaluated the microbial, immunological, periodontal, and clinical outcomes. The significant effects of photodynamic therapy obtained by in vitro and in vivo studies have made it a popular treatment for periodontal diseases in recent years. Photodynamic therapy is a novel bactericidal strategy that is stronger, faster, and less expensive than scaling and root planing. Method This study registered on PROSPERO (CRD42021267008) and retrieved fifty-three randomized controlled trials by searching nine databases (Medline, Embase, Scopus, Open Gray, Google Scholar, ProQuest, the Cochrane Library, Web of Science, and ClinicalTrials.gov) from 2008 to 2023. Of 721 records identified through database searches following title and full-text analysis, and excluding duplicate and irrelevant publications, 53 articles were included in this systematic review. Fifty of the 53 eligible studies fulfilled all the criteria in the Joanna Briggs Institute's (JBI's) Checklist for RCTs; the remaining articles met 9-12 criteria and were considered high quality. Results The present study showed that photodynamic therapy in adjunct to scaling and root planing has the potential to improve periodontal parameters such as clinical attachment loss or gain, decrease in bleeding on probing, and probing pocket depth. In addition, photodynamic therapy decreases the rate of periodontal pathogens and inflammation markers, which, in turn, reduces the progression of periodontitis. Conclusion Photodynamic therapy is considered a promising, adjunctive, and low-cost therapeutic method that is effective in tissue repair, reducing chronic periodontitis, reducing inflammation, and well-tolerated by patients.
Collapse
Affiliation(s)
- Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, University of Medical Sciences, Tehran, Iran
| | - Nour Amirmozafari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atieh Darbandi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, University of Medical Sciences, Tehran, Iran
| | - Roghayeh Afifirad
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Asadollahi
- Department of Microbiology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Center, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Mytych W, Bartusik-Aebisher D, Łoś A, Dynarowicz K, Myśliwiec A, Aebisher D. Photodynamic Therapy for Atherosclerosis. Int J Mol Sci 2024; 25:1958. [PMID: 38396639 PMCID: PMC10888721 DOI: 10.3390/ijms25041958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis, which currently contributes to 31% of deaths globally, is of critical cardiovascular concern. Current diagnostic tools and biomarkers are limited, emphasizing the need for early detection. Lifestyle modifications and medications form the basis of treatment, and emerging therapies such as photodynamic therapy are being developed. Photodynamic therapy involves a photosensitizer selectively targeting components of atherosclerotic plaques. When activated by specific light wavelengths, it induces localized oxidative stress aiming to stabilize plaques and reduce inflammation. The key advantage lies in its selective targeting, sparing healthy tissues. While preclinical studies are encouraging, ongoing research and clinical trials are crucial for optimizing protocols and ensuring long-term safety and efficacy. The potential combination with other therapies makes photodynamic therapy a versatile and promising avenue for addressing atherosclerosis and associated cardiovascular disease. The investigations underscore the possibility of utilizing photodynamic therapy as a valuable treatment choice for atherosclerosis. As advancements in research continue, photodynamic therapy might become more seamlessly incorporated into clinical approaches for managing atherosclerosis, providing a blend of efficacy and limited invasiveness.
Collapse
Affiliation(s)
- Wiktoria Mytych
- Students English Division Science Club, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland; (W.M.); (A.Ł.)
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| | - Aleksandra Łoś
- Students English Division Science Club, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland; (W.M.); (A.Ł.)
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland; (K.D.); (A.M.)
| | - Angelika Myśliwiec
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland; (K.D.); (A.M.)
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland
| |
Collapse
|
4
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [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: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
5
|
Nie M, Zhang P, Pathak JL, Wang X, Wu Y, Yang J, Shen Y. Photodynamic therapy in periodontitis: A narrative review. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2024; 40:e12946. [PMID: 38288767 DOI: 10.1111/phpp.12946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Periodontitis, a chronic infectious disease, is primarily caused by a dysbiotic microbiome, leading to the destruction of tooth-supporting tissues and tooth loss. Photodynamic therapy (PDT), which combines excitation light with photosensitizers (PS) and oxygen to produce antibacterial reactive oxygen species, is emerging as a promising adjuvant treatment for periodontitis. METHODS This review focuses on studies examining the antibacterial effects of PDT against periodontal pathogens. It also explores the impact of PDT on various aspects of periodontal health, including periodontal immune cells, human gingival fibroblasts, gingival collagen, inflammatory mediators, cytokines in the periodontium, vascular oxidative stress, vascular behavior, and alveolar bone health. Clinical trials assessing the types of PSs and light sources used in PDT, as well as its effects on clinical and immune factors in gingival sulcus fluid and the bacterial composition of dental plaque, are discussed. RESULTS The findings indicate that PDT is effective in reducing periodontal pathogens and improving markers of periodontal health. It has shown positive impacts on periodontal immune response, tissue integrity, and alveolar bone preservation. Clinical trials have demonstrated improvements in periodontal health and alterations in the microbial composition of dental plaque when PDT is used alongside conventional treatments. CONCLUSIONS PDT offers a promising adjunctive treatment for periodontitis, with benefits in bacterial reduction, tissue healing, and immune modulation. This article highlights the potential of PDT in periodontal therapy and emphasizes the need for further research to refine its clinical application and efficacy.
Collapse
Affiliation(s)
- Min Nie
- Department of Periodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Periodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peipei Zhang
- Department of Oral Medicine, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, China
| | - Janak Lal Pathak
- Department of Periodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyu Wang
- Department of Periodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yafei Wu
- Department of Periodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingmei Yang
- Department of Periodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqin Shen
- Department of Periodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Wu G, Yu G, Zheng M, Peng W, Li L. Recent Advances for Dynamic-Based Therapy of Atherosclerosis. Int J Nanomedicine 2023; 18:3851-3878. [PMID: 37469455 PMCID: PMC10352141 DOI: 10.2147/ijn.s402678] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 05/06/2023] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease, which may lead to high morbidity and mortality. Currently, the clinical treatment strategy for AS is administering drugs and performing surgery. However, advanced therapy strategies are urgently required because of the deficient therapeutic effects of current managements. Increased number of energy conversion-based organic or inorganic materials has been used in cancer and other major disease treatments, bringing hope to patients with the development of nanomedicine and materials. These treatment strategies employ specific nanomaterials with specific own physiochemical properties (external stimuli: light or ultrasound) to promote foam cell apoptosis and cholesterol efflux. Based on the pathological characteristics of vulnerable plaques, energy conversion-based nano-therapy has attracted increasing attention in the field of anti-atherosclerosis. Therefore, this review focuses on recent advances in energy conversion-based treatments. In addition to summarizing the therapeutic effects of various techniques, the regulated pathological processes are highlighted. Finally, the challenges and prospects for further development of dynamic treatment for AS are discussed.
Collapse
Affiliation(s)
- Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
| | - Guanye Yu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Meiling Zheng
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, 101121, People’s Republic of China
| | - Wenhui Peng
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Lei Li
- National Clinical Research Center for Obstetric & Gynecologic Diseases, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People’s Republic of China
| |
Collapse
|
7
|
Zou L, Zhang Y, Cheraga N, Abodunrin OD, Qu KY, Qiao L, Ma YQ, Chen LJ, Huang NP. Chlorin e6 (Ce6)-loaded plaque-specific liposome with enhanced photodynamic therapy effect for atherosclerosis treatment. Talanta 2023; 265:124772. [PMID: 37327664 DOI: 10.1016/j.talanta.2023.124772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023]
Abstract
Recently, photodynamic therapy (PDT) has been considered as a new strategy for atherosclerosis treatment. Targeted delivery of photosensitizer could significantly reduce its toxicity and enhance its phototherapeutic efficiency. CD68 is an antibody that can be conjugated to nano-drug delivery systems to actively target plaque sites, owing to its specific binding to CD68 receptors that are highly expressed on the surfaces of macrophage-derived foam cells. Liposomes are very popular nanocarriers due to their ability to encapsulate a wide range of therapeutic compounds including drugs, microRNAs and photosensitizers, and their ability to be surface-modified with targeting moieties leading to the development of nanocarriers with an improved targeted ability. Hence, we designed a Ce6-loaded liposomes using the film dispersion method, followed by the conjugation of CD68 antibody on the liposomal surface through a covalent crosslinking reaction, forming CD68-modified Ce6-loaded liposomes (CD68-Ce6-mediated liposomes). Flow cytometry results indicated that Ce6-containing liposomes were more effective in promoting intracellular uptake after laser irradiation. Furthermore, CD68-modified liposomes significantly strengthened the cellular recognization and thus internalization. Different cell lines have been incubated with the liposomes, and the results showed that CD68-Ce6-mediated liposomes had no significant cytotoxicity to coronary artery endothelial cells (HCAEC) under selected conditions. Interestingly, they promoted autophagy in foam cells through the increase in LC3-Ⅰ, LC3-Ⅱ expression and the decrease in p62 expression, and restrained the migration of mouse aortic vascular smooth muscle cells (MOVAS) in vitro. Moreover, the enhancement of atherosclerotic plaque stability and the reduction in the cholesterol content by CD68-Ce6-mediated liposomes were dependent on transient reactive oxygen species (ROS) generated under laser irradiation. In summary, we demonstrated that CD68-Ce6-mediated liposomes, as a photosensitizer nano-drug delivery system, have an inhibitory effect on MOVAS migration and a promotion of cholesterol efflux in foam cells, and thereby, represent promising nanocarriers for atherosclerosis photodynamic therapy.
Collapse
Affiliation(s)
- Lin Zou
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Nihad Cheraga
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Oluwatosin David Abodunrin
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Kai-Yun Qu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Li Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu-Qing Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Li-Juan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Nanjing, 211200, China.
| | - Ning-Ping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
8
|
Meng F, Ke J, Guo F, Yan J, Wang L. DhHP-6 alleviates inflammation and reduces vascular permeability by eliminating reactive oxygen species. Free Radic Res 2023; 57:325-337. [PMID: 37533406 DOI: 10.1080/10715762.2023.2243030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
Inflammation is a defensive immune response to external stimuli. However, uncontrolled inflammation may cause potential damage to the host. Therefore, timely control of uncontrolled inflammation is particularly important. Previous studies have found that small molecules with antioxidant activity, such as peroxidase mimic enzymes, can inhibit the development of inflammation. DhHP-6 is a new peptide mimic of peroxidase previously designed by our laboratory. Here, we explored its anti-inflammatory activity in vitro and in vivo. Our results showed that treatment with DhHP-6 significantly reduced the production of reactive oxygen species (ROS), NO, IL-6, and TNF-α in RAW264.7 cells induced by lipopolysaccharides (LPS); in addition, it also blocked the phosphorylation of extracellularly regulated kinase 1 and 2 (ERK1/2) and ribosomal s6 kinase 1 (RSK1), thereby blocking the phosphorylation and degradation of IκBα, and inhibiting the nuclear translocation of p65. Interestingly, treatment with DhHP-6 blocked the phosphorylation of ERK1/2 and myosin light chain kinase (MLCK) in HUVECs induced by LPS. Finally, we found that DhHP-6 treatment significantly reduced the infiltration of immune cells in balloon model rats. Therefore, we believe that DhHP-6 is a potent inhibitor of inflammation.
Collapse
Affiliation(s)
- Fanwei Meng
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Junfeng Ke
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Feng Guo
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Jiaqing Yan
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Liping Wang
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, China
| |
Collapse
|
9
|
Dai T, He W, Tu S, Han J, Yuan B, Yao C, Ren W, Wu A. Black TiO2 nanoprobe-mediated mild phototherapy reduces intracellular lipid levels in atherosclerotic foam cells via cholesterol regulation pathways instead of apoptosis. Bioact Mater 2022; 17:18-28. [PMID: 35386468 PMCID: PMC8958315 DOI: 10.1016/j.bioactmat.2022.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
Given that apoptosis increases the risk of plaque rupture, strategies that reduce intracellular lipid levels without killing foam cells are warranted for safe and effective treatment of atherosclerosis. In this study, a mild phototherapy strategy is carried out to achieve the hypothesis. Foam cell-targeted nanoprobes that allow photothermal therapy (PTT) and/or photodynamic therapy (PDT) were prepared by loading hyaluronan and porphine onto black TiO2 nanoparticles. The results showed that when temperatures below 45 °C, PTT alone and PTT + PDT significantly reduced the intracellular lipid burden without inducing evidently apoptosis or necrosis. In contrast, the use of PDT alone resulted in only a slight reduction in lipid levels and induced massive apoptosis or necrosis. The protective effect against apoptosis or necrosis after mild-temperature PTT and PTT + PDT was correlated with the upregulation of heat shock protein 27. Further, mild-temperature PTT and PTT + PDT attenuated intracellular cholesterol biosynthesis and excess cholesterol uptake via the SREBP2/LDLR pathway, and also triggered ABCA1-mediated cholesterol efflux, ultimately inhibiting lipid accumulation in foam cells. Our results offer new insights into the mechanism of lipid regulation in foam cells and indicate that the black TiO2 nanoprobes could allow safer and more effective phototherapy of atherosclerosis. Mild phototherapy reduced the intracellular lipid in foam cells without inducing obvious apoptosis or necrosis. HSP27 was upregulated in foam cells treated by mild phototherapy, which could protect cells against apoptosis or necrosis. Mild phototherapy attenuated intracellular cholesterol biosynthesis and excess uptake, also boosted cholesterol efflux.
Collapse
Affiliation(s)
- Ting Dai
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province, 315020, China
| | - Wenming He
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province, 315020, China
| | - Shuangshuang Tu
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province, 315020, China
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
| | - Jinru Han
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No. 1 Yanqihu East Road, Huairou District, Beijing, 101408, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
| | - Bo Yuan
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
| | - Chenyang Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
| | - Wenzhi Ren
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
- Corresponding author. Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China.
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
- Corresponding author. Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China.
| |
Collapse
|
10
|
Pu W, Su Z, Wazir J, Zhao C, Wei L, Wang R, Chen Q, Zheng S, Zhang S, Wang H. Protective effect of α7 nicotinic acetylcholine receptor activation on experimental colitis and its mechanism. Mol Med 2022; 28:104. [PMID: 36058917 PMCID: PMC9441089 DOI: 10.1186/s10020-022-00532-2] [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: 11/30/2021] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Background Inflammatory bowel disease (IBD) is a common chronic remitting disease with no satisfactory treatment. The aim of this study was to investigate the protective effect of α7 nicotinic acetylcholine receptor (α7nAChR), and to determine the underlying mechanism of its activity. Methods The expression and distribution of α7nAChR in the intestinal tissue of patients with ulcerative colitis and Crohn’s disease were analyzed. The effects of vagal excitation on murine experimental colitis were investigated. The colitis model was induced in C57BL/6 mice by the administration of 3% dextran sulfate sodium (DSS). The therapeutic group received treatment with the α7nAChR agonist PNU-282987 by intraperitoneal injection. Results Our results showed that there was significantly increased expression of α7nAChR in colitis and Crohn’s disease intestinal tissue, and its expression was mainly located in macrophages and neutrophils, which were extensively infiltrated in the disease status. Treatment with an α7nAChR agonist potently ameliorated the DSS-induced illness state, including weight loss, stool consistency, bleeding, colon shortening, and colon histological injury. α7nAChR agonist exerted anti-inflammatory effects in DSS colitis mice by suppressing the secretion of multiple types of proinflammatory factors, such as IL6, TNFα, and IL1β, and it also inhibited the colonic infiltration of inflammatory cells by blocking the DSS-induced overactivation of the NF-κB and MAPK signaling pathways. Mechanistically, activation of α7nAChR decreased the number of infiltrated M1 macrophages in the colitis intestine and inhibited the phagocytosis ability of macrophages, which were activated in response to LPS stimulation. Conclusion Thus, an α7nAChR agonist ameliorated colonic pathology and inflammation in DSS-induced colitis mice by blocking the activation of inflammatory M1 macrophages.
Collapse
Affiliation(s)
- Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Zhenzi Su
- The Affiliated Suqian Hospital of Xuzhou Medical University and Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, 223800, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Ranran Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China
| | - Qiyi Chen
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Saifang Zheng
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Shaoyi Zhang
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
11
|
Effects of ALA-PDT on the macrophages in wound healing and its related mechanisms in vivo and in vitro. Photodiagnosis Photodyn Ther 2022; 38:102816. [PMID: 35378277 DOI: 10.1016/j.pdpdt.2022.102816] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND Several studies have suggested the effectiveness of photodynamic therapy (PDT) for wound healing. Macrophages are critical immune cells necessary for regulated inflammation during wound repair. However, the available information regarding the effects of PDT on macrophages during cutaneous wound healing remains insufficient. This study aimed to further investigate these aspects in vivo and in vitro. METHODS Mouse full-thickness wound models were used as the study samples to investigate the therapeutic effects and mechanisms of 5-aminolevulinic acid (ALA) PDT. Wound healing rate, granulation tissue formation, local inflammation, M1/M2 macrophages differentiation, were measured at different time points treated by ALA-PDT. The polarization of macrophages induced by ALA-PDT was further evaluated in vitro using PCR and western blot analysis. RESULTS ALA-PDT could promote formation of granulation tissue, increase inflammatory infiltration and activate M1 macrophages in the early stage of injury. While, ALA-PDT could also facilitate absorption of granulation tissue, inhibit inflammatory infiltration and enhance M2 macrophages polarization in the later stage of wound repair. In vitro, ALA-PDT could modulate the ratio of M2 polarization to M1 polarization via NF-κB signaling pathway. CONCLUSIONS ALA-PDT topical application stimulates wound healing by regulating formation of granulation tissue, inflammatory process and M1/M2 macrophages differentiation. The study places a preliminary theoretical basis for topical ALA-PDT to be administered clinically in cutaneous wounds healing.
Collapse
|
12
|
Zuo Q, Ou Y, Zhong S, Yu H, Zhan F, Zhang M. Targeting GRP78 enhances the sensitivity of HOS osteosarcoma cells to pyropheophorbide-α methyl ester-mediated photodynamic therapy via the Wnt/β-catenin signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1387-1397. [PMID: 34494093 PMCID: PMC8507956 DOI: 10.1093/abbs/gmab115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Indexed: 12/20/2022] Open
Abstract
Photodynamic therapy (PDT), which is a new method for treating tumors, has been used in the treatment of cancer. In-depth research has shown that PDT cannot completely kill tumor cells, indicating that tumor cells are resistant to PDT. Glucose regulatory protein 78 (GRP78), which is a key regulator of endoplasmic reticulum stress, has been confirmed to be related to tumor resistance and recurrence, but there are relatively few studies on the further mechanism of GRP78 in PDT. Our experiment aimed to observe the role of GRP78 in HOS human osteosarcoma cells treated with pyropheophorbide-α methyl ester-mediated photodynamic therapy (MPPα-PDT) and to explore the possible mechanism by which the silencing of GRP78 expression enhances the sensitivity of HOS osteosarcoma cells to MPPα-PDT. HOS osteosarcoma cells were transfected with siRNA-GRP78. Apoptosis and reactive oxygen species (ROS) levels were detected by Hoechst staining and flow cytometry, cell viability was detected by Cell Counting Kit-8 assay, GRP78 protein fluorescence intensity was detected by immunofluorescence, and apoptosis-related proteins, cell proliferation-related proteins, and Wnt pathway-related proteins were detected by western blot. The results showed that MPPα-PDT can induce HOS cell apoptosis and increase GRP78 expression. After successful siRNA-GRP78 transfection, HOS cell proliferation was decreased, and apoptosis-related proteins expressions was increased, Wnt/β-catenin-related proteins expressions was decreased, and ROS levels was increased. In summary, siRNA-GRP78 enhances the sensitivity of HOS cells to MPPα-PDT, the mechanism may be related to inhibiting Wnt pathway activation and increasing ROS levels.
Collapse
Affiliation(s)
- Qiang Zuo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shenxi Zhong
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haoyang Yu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fangbiao Zhan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Muzi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
13
|
Zhou X, Zheng M, Zou Y, Wang J, Zhang L, Yin R. 5-Aminolevulinic acid induced photodynamic therapy (ALA-PDT) for erosive adenomatosis of the nipple: A case report. Photodiagnosis Photodyn Ther 2021; 35:102387. [PMID: 34107318 DOI: 10.1016/j.pdpdt.2021.102387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/27/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022]
Abstract
Erosive adenomatosis of the nipple (EAN) is an uncommon, benign neoplasm that involves the nipple. Traditional treatments include complete surgical excision, limited forms of complete surgical excision and Mohs micrographic surgery. Here, we report a case of a 40-year-old woman with a 2-year history of asymptomatic erosion, papillomatous hyperplasia and intermittent serosanguineous discharge on her right nipple. Histopathological examination confirmed the diagnosis of EAN. She was treated with 5-aminolevulinic acid induced photodynamic therapy (ALA-PDT) for 2 sessions with 2 weeks intervals. No recurrence occurred within 6 months. Therefore, our report suggested that ALA-PDT is a possible method to treat EAN, especially in patients who have the need of breastfeeding and cosmetic appearance, but this needs to be examined in a larger clinical trial.
Collapse
Affiliation(s)
- Xiaoqing Zhou
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mengxue Zheng
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yongzhen Zou
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Juan Wang
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Lian Zhang
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rui Yin
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
14
|
Huang J, Wu S, Wu M, Zeng Q, Wang X, Wang H. Efficacy of the therapy of 5-aminolevulinic acid photodynamic therapy combined with human umbilical cord mesenchymal stem cells on methicillin-resistant Staphylococcus aureus-infected wound in a diabetic mouse model. Photodiagnosis Photodyn Ther 2021; 36:102480. [PMID: 34375775 DOI: 10.1016/j.pdpdt.2021.102480] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND A distressing issue of diabetic ulcer (DU) is its poor healing feature with limited clinical solutions. We have previously shown that 5-aminolevulinic acid photodynamic therapy (ALA-PDT) is a promising alternative to the currently limited measures for DU. Mesenchymal stem cells (MSCs) transplantation has been believed to impose certain therapeutic effect on restoration of injury. Thus, this study aims to explore whether the combination of MSCs and ALA-PDT will exert a more advanced curative effect on DU. METHODS Diabetic mice were induced by intraperitoneal injection of streptozotocin (STZ, 60 mg/kg/d) for consecutive 5 days. A full-thickness skin injury (diameter 6 mm) was created in the center of the back of each mouse, and then 10 μl of methicillin-resistant Staphylococcus aureus (MRSA) suspension was added to establish an infected DU model. All DU models were randomly divided into four groups: Untreated group, MSCs group, ALA-PDT group, and ALA-PDT combined with human umbilical cord mesenchymal stem cells (hUC-MSCs) (ALA-PDT + MSCs) group. The wound sizes were recorded by a digital camera, and the healing rates were calculated using Image J software. Bacterial loads on wounds were measured using CFU (Colony forming units) analysis. The epithelialization, inflammatory cells infiltration and granulation tissue formation were monitored by Haematoxylin and eosin (H&E) staining, and the corresponding semi-quantitative score was matched. Growth and pro-inflammatory cytokines were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS Either ALA-PDT or injection of hUC-MSCs resulted in a rapid wound closure compared with the untreated, while their combination brought about the most prominent healing. On day 12, healing rates of the untreated, MSCs, ALA-PDT and ALA-PDT + MSCs were 40.56% ± 7.06%, 74.23 ± 4.83%, 84.03 ± 3.53%, 99.67 ± 0.49%, respectively. The bacterial burden reductions were approximately 1.58 logs (97.36%, P < 0.05), 2.34 logs (99.54%, P < 0.01), 4.50 logs (nearly 100%, P < 0.001) for MSCs, ALA-PDT and ALA-PDT + MSCs, respectively. Histology revealed reduced inflammatory cells and improved collagen precipitation and angiogenesis after hUC-MSCs and ALA-PDT treatment compared to the untreated. The combined therapy leaded to a more intact epithelium, similar to the healthy. Finally, ELISA revealed that the property of ALA-PDT to stimulate transforming growth factor-β1 (TGF-β1) and vascular endothelial growth factor (VEGF) and inhibit IL (interleukin) -1β and IL-6 outweighed that of hUC-MSCs, and this function of the combination overwhelmed that of any single therapy. CONCLUSIONS Our findings indicated that the strategy of combining ALA-PDT with hUC-MSCs possessed a significantly enhanced therapeutic effect over either single therapy, providing a promising innovative therapeutic candidate for refractory wounds.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, PR China.
| | - Shutian Wu
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, PR China.
| | - Minfeng Wu
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, PR China.
| | - Qingyu Zeng
- Shanghai Skin Disease Hospital, Institute of Photomedicine, Tongji University School of Medicine, Shanghai, PR China.
| | - Xiuli Wang
- Shanghai Skin Disease Hospital, Institute of Photomedicine, Tongji University School of Medicine, Shanghai, PR China.
| | - Hongwei Wang
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, PR China.
| |
Collapse
|
15
|
Feng Y, Mei L, Wang M, Huang Q, Huang R. Anti-inflammatory and Pro-apoptotic Effects of 18beta-Glycyrrhetinic Acid In Vitro and In Vivo Models of Rheumatoid Arthritis. Front Pharmacol 2021; 12:681525. [PMID: 34381358 PMCID: PMC8351798 DOI: 10.3389/fphar.2021.681525] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/23/2021] [Indexed: 12/29/2022] Open
Abstract
18β-Glycyrrhetinic acid (18β-GA), an active component from Glycyrrhiza glabra L. root (licorice), has been demonstrated to be able to protect against inflammatory response and reduce methotrexate (MTX)-derived toxicity. This study was therefore designed to test the therapeutic possibility of 18β-GA on rheumatoid arthritis (RA) and to explore the underlying mechanism. LPS or TNF-α-induced inflammatory cell models and collagen-induced arthritis (CIA) animal models were applied in this study. Real-time quantitative PCR (RT-qPCR) was used to measure the mRNA levels of various cytokines and FOXO family members. The protein levels of molecules in the MAPK/NF-κB signaling pathway were analyzed using western blot. The cell proliferation assay and colony-forming assay were used to test the influence of 18β-GA on cell viability. The cell apoptosis assay and cell cycle assay were performed to detect the effect of 18β-GA on cell proliferative capacity by using flow cytometry. Hematoxylin and eosin (H&E) staining was performed to evaluate pathological changes after drug administration. The enzyme-linked immunosorbent assay (ELISA) was carried out for the detection of cytokines in serum. In vitro, we found that 18β-GA decreased the mRNA levels of IL-1β, IL-6, and COX-2 by inhibiting the MAPK/NF-κB signaling pathway in MH7A and RAW264.7 cell lines. Moreover, 18β-GA was able to suppress cell viability, trigger cell apoptosis, and G1 phase cell cycle arrest in our in vitro studies. 18β-GA dramatically enhanced the mRNA level of FOXO3 in both TNF-α- and LPS-induced inflammation models in vitro. Interestingly, after analyzing GEO datasets, we found that the FOXO3 gene was significantly decreased in the RA synovial tissue as compared to healthy donors in multiple microarray studies. In vivo, 18β-GA exhibited a promising therapeutic effect in a collagen-induced arthritis mouse model by alleviating joint pathological changes and declining serum levels of TNF-α, IL-1β, and IL-6. Finally, we observed that 18β-GA administration could mitigate liver damage caused by collagen or MTX. Collectively, the current study demonstrates for the first time that 18β-GA can inhibit inflammation and proliferation of synovial cells, and the underlying mechanism may be associated with its inhibition of MAPK/NF-κB signaling and promotion of FOXO3 signaling. Therefore, 18β-GA is expected to be a new drug candidate for RA therapy.
Collapse
Affiliation(s)
- Yunhui Feng
- College of Physical Education, Guangzhou University, Guangzhou, China
| | - Liyan Mei
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Maojie Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Qingchun Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Runyue Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine (The Second Affiliated Hospital of Guangzhou University of Chinese Medicine), Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Curcumin-mediated photodynamic therapy inhibits the phenotypic transformation, migration, and foaming of oxidized low-density lipoprotein-treated vascular smooth muscle cells by promoting autophagy. J Cardiovasc Pharmacol 2021; 78:308-318. [PMID: 34091481 PMCID: PMC8340951 DOI: 10.1097/fjc.0000000000001069] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/02/2021] [Indexed: 02/05/2023]
Abstract
Supplemental Digital Content is Available in the Text. Vascular smooth muscle cells (VSMCs) are becoming a hot spot and target of atherosclerosis research. This study aimed to observe the specific effects of curcumin (CUR)-mediated photodynamic therapy (CUR-PDT) on oxidized low-density lipoprotein (ox-LDL)-treated VSMCs and confirm whether these effects are mediated by autophagy. In this study, the mouse aortic smooth muscle cell line and A7r5 cell lines were used for parallel experiments. VSMC viability was evaluated by Cell Counting Kit-8 assay. VSMCs were treated with ox-LDL to establish a model of atherosclerosis in vitro. The autophagy level and the expression of proteins related to phenotypic transformation were detected by western blotting. The migration ability of the cells was detected by using transwell assay. The presence of intracellular lipid droplets was detected by Oil Red O staining. The results showed that VSMCs transformed from the contraction phenotype to the synthetic phenotype when stimulated by ox-LDL, during which autophagy was inhibited. However, CUR-PDT treatment significantly promoted the level of autophagy and inhibited the process of phenotypic transformation induced by ox-LDL. In addition, ox-LDL significantly promoted VSMC migration and increased the number of lipid droplets, whereas CUR-PDT treatment significantly reduced the ox-LDL-induced increase in the migration ability of, and lipid droplet numbers in, VSMCs. When the VSMCs were pretreated with the autophagy inhibitor 3-methyladenine for 24 hours, the effects of CUR-PDT were reversed. Therefore, our study indicated that CUR-PDT can inhibit the phenotypic transformation, migration, and foaming of ox-LDL–treated VSMCs by inducing autophagy.
Collapse
|
17
|
Zhang M, Qin X, Xu W, Wang Y, Song Y, Garg S, Luan Y. Engineering of a dual-modal phototherapeutic nanoplatform for single NIR laser-triggered tumor therapy. J Colloid Interface Sci 2021; 594:493-501. [PMID: 33774405 DOI: 10.1016/j.jcis.2021.03.050] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/27/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
Theranostic nanoplatforms integrating simultaneously photodynamic therapy (PDT) and photothermal therapy (PTT) exhibit intrinsic advantages in tumor therapy due to distinct mechanisms of action. However, it is challenging to achieve PDT and PTT under single near-infrared (NIR) laser irradiation with a nanoplatform utilizing conventional organic photodynamic agent and inorganic photothermal agent owing to the difference in inherent excitation wavelengths. Particularly, the single NIR light (660 nm)-triggered PTT and PDT nanoplatform, constructed from chlorin e6 (Ce6) and copper sulfide (CuS) nanoparticles (NPs), has never been reported. Herein, we, for the first time, designed and established a dual-modal phototherapeutic nanoplatform that achieved both PTT and PDT under single NIR laser (660 nm) irradiation for Ce6 and CuS NPs with the strategy of core-shell structured CuS@Carbon integrated with Ce6. Introducing of carbon shell not only endows small CuS NPs with excellent tumor accumulation, but also significantly strengthens the photothermal performance of CuS NPs, realizing efficient photothermal performance under 660 nm laser irradiation. Moreover, Ce6 in carbon shell endowed the nanoplatform with photodynamic effect under 660 nm laser irradiation. The as-prepared Ce6/CuS@Carbon nanoplatform thus achieved dual-modal phototherapy under single NIR laser irradiation, significantly inhibiting tumor growth with minimal adverse effects and superior biosafety.
Collapse
Affiliation(s)
- Mengzhu Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaohan Qin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wei Xu
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250014, China
| | - Yibing Wang
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250014, China.
| | - Yunmei Song
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Yuxia Luan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
18
|
Effect of photodynamic therapy on expression of HRAS, NRAS and caspase 3 genes at mRNA levels, apoptosis of head and neck squamous cell carcinoma cell line. Photodiagnosis Photodyn Ther 2020; 33:102142. [PMID: 33307231 DOI: 10.1016/j.pdpdt.2020.102142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 11/14/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVES This study aimed to assess the effect of photodynamic therapy (PDT) on expression of CASP3, NRAS and HRAS genes at mRNA levels, and apoptosis of head and neck squamous cell carcinoma (HNSCC) cell line. MATERIALS AND METHODS In order to complete the present in vitro study, HNSCC cell line (NCBI C196 HN5) purchased from Pasteur Institute. Cells were divided into four groups; Group 1: photodynamic treatment (laser + methylene blue (MB) as photosensitizer), group 2: MB, group 3: laser (with 660 nm wavelength), and group 4: control (without any treatment). To determine the optimal concentration of MB, in a pilot study, toxicity of MB in different concentration was assessed using MTT assay. Cells in group 1, 2 and 3 was treated at optimal concentration of MB (1.6 μg/mL). Gene expression at mRNA levels was assessed after 24 h incubation, using real-time (qRT)-PCR. The expression of BAX and BCL2 genes at the mRNA levels was analyzed to evaluate apoptosis. 2-ΔΔCt values of BCL2, BAX, CASP3, NRAS, and HRAS in groups was analyzed using ANOVA. Tukey's HSD and Games Howell test was used to compare between two groups. RESULTS Over-expression of BAX (p < 0.001), CASP3 (p < 0.001) and down-regulation of BCL2 (p = 0.004), HRAS (p = 0.023) and NRAS (p = 0.045) were noted in group 1 (PDT), compared with the control group. Treatment by laser alone induce down-regulation of CASP3 (p < 0.05), BAX (p < 0.05), BCL2 (p > 0.05), HRAS (p > 0.05) and NRAS (p > 0.05). CONCLUSION PDT caused down-regulation of NRAS, HRAS and BCL2 and over-expression of CASP3 and BAX genes at mRNA levels in HNSCC cell line. The present study raises the possibility that the role of MB on BCL2 down-regulation and BAX and CASP3 over-expression was higher than laser alone while it seems that laser alone was more effective than MB in HRAS and NRAS down-regulation.
Collapse
|
19
|
Wang Y, Liu Y, Li J, Xu X, Li X. Zinc ferrate nanoparticles for applications in medicine: synthesis, physicochemical properties, regulation of macrophage functions, and in vivo safety evaluation. Nanotoxicology 2020; 14:1381-1398. [PMID: 33075238 DOI: 10.1080/17435390.2020.1831094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Zinc ferrate nanoparticles (ZnFe2O4 NPs) have attracted enormous interest as potential nanomaterials. The purpose of this study was to examine the in vitro macrophages toxicity, in vivo safety, and immunogenicity. Three kinds of ZnFe2O4 NPs with different shapes (round, litchi, and raspberry), nano-sizes, and pores were successfully prepared. In vitro experiments showed that ZnFe2O4 NPs caused no cytotoxicity against the RAW 264.7 cells up to administered dose of 200 μg/mL, enhanced proinflammatory cytokine TNF-α, and costimulatory marker CD86 expression in the RAW 264.7 cells. Interestingly, ZnFe2O4 NPs reduced ROS expression, which was inconsistent with common metal oxide NPs such as iron oxide (Fe3O4) NPs and zinc oxide (ZnO) NPs. ZnFe2O4 NPs improved the RAW 264.7 cells phagocytosed more neutral red. There was no obvious difference in body weight, the number of immune cells, organ index, and expression of inflammatory factors in serum of rats administrated intravenously and subcutaneously on day 21 after treatment by ZnFe2O4 NPs in comparison with the blank control. These results demonstrated that ZnFe2O4 NPs slightly enhanced the function of the RAW 264.7 cells in vitro but caused no obvious toxicity to macrophages as well as rat blood cells, and low immunogenicity in rats, suggesting that ZnFe2O4 NPs as a biocompatible nanomaterials achieved potential for bioapplication in the future.
Collapse
Affiliation(s)
- Yu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Yajie Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiajia Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoqing Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Xinru Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
20
|
Lau H, Khosrawipour T, Mikolajczyk A, Frelkiewicz P, Nicpon J, Arafkas M, Pigazzi A, Knoefel WT, Khosrawipour V. Intraperitoneal chemotherapy of the peritoneal surface using high-intensity ultrasound (HIUS): investigation of technical feasibility, safety and possible limitations. J Cancer 2020; 11:7209-7215. [PMID: 33193884 PMCID: PMC7646163 DOI: 10.7150/jca.48519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/11/2020] [Indexed: 01/25/2023] Open
Abstract
Introduction: The penetration of chemotherapeutic drugs into peritoneal nodules remains at levels well below 1 mm, thus significantly limiting the antitumor effect of intraperitoneal chemotherapy (IPC). Recently, high-Intensity ultrasound (HIUS) has been discovered as a potential tool to significantly improve peritoneal diffusion rates. Despite promising preliminary data, basic aspects regarding its technical feasibility, safety and possible limitations remain unclear. This study aims to enhance our current understanding of HIUS and test its applicability using an ex-vivo swine model. Methods: Three postmortem swine were subject to laparotomy and consecutive lavage with 0.9%NaCl saline and HIUS application. For this purpose, a large HIUS radiating pen was introduced into the abdominal cavity and HIUS was applied on two of the four abdominal quadrants for 300 seconds each at an output power of 70 W, 50 % amplitude and 20 kHz frequency. Following the procedure, small intestinal tissue samples were retrieved for further analyses. Results: Peritoneal and subperitoneal layers showed structural changes only visible on a microscopic level. The peritoneal layer was transformed into a mesh-like structure while the subperitoneal layer (depth of 142 +/- 28 µm) exhibited microcavities and vascular detachment from surrounding tissues. No bowel rupture or vascular perforations were observed. Conclusions: Our data indicate that HIUS is a technically feasible and safe add-on procedure for intraperitoneal chemotherapy (IPC) with measurable microscopic changes on the peritoneal surface. Pretreatment of the abdominal cavity with HIUS could significantly improve IPC efficacy. Further studies are required to optimize and evaluate this novel approach.
Collapse
Affiliation(s)
- Hien Lau
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine, Orange, USA
| | - Tanja Khosrawipour
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine, Orange, USA.,Department of Surgery (A), University-Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Germany
| | - Agata Mikolajczyk
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Piotr Frelkiewicz
- The Center of Experimental Diagnostics and Innovative Biomedical Technology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Jakub Nicpon
- The Center of Experimental Diagnostics and Innovative Biomedical Technology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Mohamed Arafkas
- Department of Plastic Surgery, Ortho-Clinic Dortmund, Dortmund, Germany
| | - Alessio Pigazzi
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine, Orange, USA
| | - Wolfram Trudo Knoefel
- Department of Surgery (A), University-Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Germany
| | - Veria Khosrawipour
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine, Orange, USA.,Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| |
Collapse
|
21
|
Khanahmadi M, Manafi B, Tayebinia H, Karimi J, Khodadadi I. Downregulation of Sirt1 is correlated to upregulation of p53 and increased apoptosis in epicardial adipose tissue of patients with coronary artery disease. EXCLI JOURNAL 2020; 19:1387-1398. [PMID: 33250679 PMCID: PMC7689241 DOI: 10.17179/excli2020-2423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022]
Abstract
The higher expression level of p53 in epithelial adipose tissue (EAT) has previously been reported in atherosclerosis. Since we hypothesized that the expression of p53 is modulated by Sirt1, the aim of this study was to determine the expression levels of Sirt1 and p53 and to investigate their correlation to apoptosis in EAT of patients with coronary artery disease (CAD). Thirty-five patients with more than 50 % stenosis in at least one of the main coronary arteries were considered as CAD group while 29 patients with no clinical signs of atherosclerosis who underwent open-heart surgery for valve replacement were classified as control group. EAT biopsy samples were collected from all participants during surgery. Sirt1, p53, Bax, and Bcl-2 gene expression levels were determined in EAT by qRT-PCR and Western blotting was carried out to assess Sirt1 and p53 protein levels. Hematoxylin and eosin staining was used for histopathological analysis. mRNA and protein levels of Sirt1 in EAT were significantly lower in patients with CAD compared with control group, whereas CAD patients showed greater p53 gene and protein expressions. In addition, inverse correlations were observed between Sirt1 and p53 at both mRNA and protein levels. The Bax and ratio of Bax/Bcl-2 gene expressions were higher in CAD group, but no difference was observed in Bcl-2 expression. Histopathological analysis showed apoptotic bodies and infiltrated immune cells in EAT of CAD group. Our results suggest that the Sirt1-p53 axis may involve in atherosclerosis by promotion of apoptosis.
Collapse
Affiliation(s)
- Mahdieh Khanahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Babak Manafi
- Department of Surgery, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Heidar Tayebinia
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
22
|
Yu Z, Yang L, Deng S, Liang M. Daidzein ameliorates LPS-induced hepatocyte injury by inhibiting inflammation and oxidative stress. Eur J Pharmacol 2020; 885:173399. [PMID: 32712091 DOI: 10.1016/j.ejphar.2020.173399] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023]
Abstract
Endotoxin-induced acute liver injury (ALI) is a severe disease associated with a poor prognosis. Therefore, it is urgent to discover new effective therapies to prevent ALI. Daidzein, extracted from leguminous plants, possess anti-inflammatory and antioxidative bioactivities. However, little is known about whether daidzein could attenuate lipopolysaccharide (LPS)-induced ALI. We investigated the effects of daidzein on hepatocyte injury and its underlying mechanisms. In LPS-induced hepatocyte supernatant, 100 μM daidzein decreased ALT and AST expression levels by 49.3% ± 5.6% and 39.3% ± 3.5%, respectively, with no cytotoxicity. In addition, the expression of inflammatory factors, including interleukin-1β (IL-lβ), interleukin-6 (IL-6) and tumor necrosis factor α (TNF-α) were decreased by 100 μM daidzein (73.8% ± 5.3%, 58.8 ± 9.0% and 55.5% ± 7.2%, respectively) in LPS-treated hepatocytes. Western blot analysis showed that daidzein inhibited LPS-induced p-ERK1/2, p-IκBα and p-p65 expression levels. Moreover, 100 μM daidzein reduced the LPS-induced production of Reactive oxygen species by 23.9 ± 7.8% and increased SOD activity by 88.4% ± 18.9% by downregulating Keap-1 and upregulating Nrf2 expression. In conclusion, these data indicate that daidzein ameliorates LPS-induced hepatocyte injury by inhibiting inflammation and oxidative stress.
Collapse
Affiliation(s)
- Zuying Yu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Deng
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Dai T, He W, Yao C, Ma X, Ren W, Mai Y, Wu A. Applications of inorganic nanoparticles in the diagnosis and therapy of atherosclerosis. Biomater Sci 2020; 8:3784-3799. [PMID: 32469010 DOI: 10.1039/d0bm00196a] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a chronic progressive disease, which may result in serious clinical outcomes, such as acute heart events or stroke with high mortality. At present, the clinical problems of atherosclerosis mainly consist of the difficulty in confirming the plaques or identifying the stability of the plaques in the early phase and the shortage of valid treatments. Fortunately, with the development of nanotechnology, various inorganic nanoparticles with imaging enhancement and noninvasive therapy functions have been studied in the imaging and treatment of atherosclerosis, which has brought new hope to patients. This review focuses on the recent progress in the use of inorganic nanoparticles in the diagnosis and therapy of atherosclerosis, including the key processes in the development of atherosclerosis and the mainly involved cells, inorganic nanoparticle-based dual-mode imaging methods classified by the types of targeting cells, and inorganic nanoparticle-based therapeutic approaches, such as photothermal therapy (PTT), photodynamic therapy (PDT), sonodynamic therapy (SDT), drug delivery, gene therapy and imaging-guided therapy for atherosclerosis. Finally, this review discusses the challenges and directions of inorganic nanoparticles in potential clinical translation of anti-atherosclerosis in future. We believe this review will enable readers to systematically understand the progress of the inorganic nanoparticle-based imaging and therapy of atherosclerosis and therefore promote the further development of anti-atherosclerosis.
Collapse
Affiliation(s)
- Ting Dai
- Department of Cardiology, The Affiliated Hospital of Medical school of Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province 315020, P.R. China.
| | | | | | | | | | | | | |
Collapse
|
24
|
Khosrawipour V, Reinhard S, Martino A, Khosrawipour T, Arafkas M, Mikolajczyk A. Increased Tissue Penetration of Doxorubicin in Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC) after High-Intensity Ultrasound (HIUS). Int J Surg Oncol 2019; 2019:6185313. [PMID: 31915548 PMCID: PMC6930754 DOI: 10.1155/2019/6185313] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/23/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND High-intensity ultrasound (HIUS) has been studied for the past two decades as a new therapeutic option for solid tumor direct treatment and a method for better chemotherapy delivery and perfusion. This treatment approach has not been tested to our knowledge in peritoneal metastatic therapy, where limited tissue penetration of intraperitoneal chemotherapy has been a main problem. Both liquid instillations and pressurized aerosols are affected by this limitation. This study was performed to evaluate whether HIUS improves chemotherapy penetration rates. METHODS High-intensity ultrasound (HIUS) was applied for 0, 5, 30, 60, 120, and 300 seconds on the peritoneal tissue samples from fresh postmortem swine. Samples were then treated with doxorubicin via pressurized intraperitoneal aerosol chemotherapy (PIPAC) under 12 mmHg and 37°C temperature. Tissue penetration of doxorubicin was measured using fluorescence microscopy on frozen thin sections. RESULTS Macroscopic structural changes, identified by swelling of the superficial layer of the peritoneal surface, were observed after 120 seconds of HIUS. Maximum doxorubicin penetration was significantly higher in peritoneum treated with HIUS for 300 seconds, with a depth of 962.88 ± 161.4 μm (p < 0.05). Samples without HIUS had a penetration depth of 252.25 ± 60.41. Tissue penetration was significantly increased with longer HIUS duration, with up to 3.8-fold increased penetration after 300 sec of HIUS treatment. CONCLUSION Our data indicate that HIUS may be used as a method to prepare the peritoneal tissue for intraperitoneal chemotherapy. Higher tissue penetration rates can be achieved without increasing chemotherapy concentrations and preventing structural damage to tissue using short time intervals. More studies need to be performed to analyze the effect of HIUS in combination with intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Veria Khosrawipour
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine (UCI), Irvine, CA, USA
| | - Sören Reinhard
- Department of Bioengineering, University of California, Berkeley (UC Berkeley), Oakland, CA, USA
| | - Alice Martino
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine (UCI), Irvine, CA, USA
| | - Tanja Khosrawipour
- Division of Colorectal Surgery, Department of Surgery, University of California Irvine (UCI), Irvine, CA, USA
- Department of Surgery, University-Hospital Düsseldorf, Düsseldorf, North-Rhein Westfalia, Germany
| | - Mohamed Arafkas
- Department of Plastic Surgery, Ortho-Klinik Dortmund, Dortmund, North-Rhein Westfalia, Germany
| | - Agata Mikolajczyk
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Lower Silesia, Poland
| |
Collapse
|
25
|
Gerola AP, Costa PFA, de Morais FAP, Tsubone TM, Caleare AO, Nakamura CV, Brunaldi K, Caetano W, Kimura E, Hioka N. Liposome and polymeric micelle-based delivery systems for chlorophylls: Photodamage effects on Staphylococcus aureus. Colloids Surf B Biointerfaces 2019; 177:487-495. [PMID: 30807963 DOI: 10.1016/j.colsurfb.2019.02.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/22/2019] [Accepted: 02/17/2019] [Indexed: 01/25/2023]
Abstract
Chlorophyll derivatives (Chls), loaded in F-127 polymeric micelles and DPPC liposomes as drug delivery systems (DDS), have been shown to be remarkable photosensitizers for photodynamic inactivation (PDI). Assays of photoinactivation of Staphylococcus aureus bacteria (as biological models) showed that the effectiveness of Chls in these nanocarriers is dependent on photobleaching processes, photosensitizer locations in DDS, singlet oxygen quantum yields, and Chl uptake to bacteria. These are factors related to changes in Chl structure, such as the presence of metals, charge, and the phytyl chain. The photodynamic activity was significantly greater for Chls without the phytyl chain, i.e., phorbides derivatives. Furthermore, the inactivation of S. aureus was increased by the use of liposomes compared to micelles. Therefore, this research details and shows the high significance of the Chl structure and delivery system to enhance the photodynamic activity. It also highlights the chlorophylls (particularly phorbides) in liposomes as promising photosensitizers for PDI.
Collapse
Affiliation(s)
- Adriana P Gerola
- Chemistry Department, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil; Chemistry Department, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil.
| | - Paulo F A Costa
- Chemistry Department, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil; Chemistry Department, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Flávia A P de Morais
- Chemistry Department, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Tayana M Tsubone
- Institute of Chemistry, Universidade de São Paulo, São Paulo, São Paulo, 05508-000, Brazil
| | - Angelo O Caleare
- Department of Clinical Analyzes and Biomedicine, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Celso V Nakamura
- Department of Physiological Sciences, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Kellen Brunaldi
- Department of Pharmacy and Pharmacology, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Wilker Caetano
- Chemistry Department, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Elza Kimura
- Department of Pharmacy and Pharmacology, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Noboru Hioka
- Chemistry Department, Universidade Estadual de Maringá, Maringá, Paraná, 87020-900, Brazil
| |
Collapse
|