1
|
Li YR, Wang G, He WT, Liu T. Application of aggregation-induced emission materials in gastrointestinal diseases. World J Gastroenterol 2025; 31:105378. [PMID: 40308804 PMCID: PMC12038521 DOI: 10.3748/wjg.v31.i16.105378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/12/2025] [Accepted: 04/11/2025] [Indexed: 04/27/2025] Open
Abstract
Aggregation-induced emission (AIE) is a phenomenon characterized by certain fluorescent molecules that exhibit weak or no luminescence in solution but demonstrate significantly enhanced luminescence upon aggregation. Accordingly, AIE materials have successfully addressed the limitations associated with aggregation-caused quenching effects and have made significant progress in the application of various fields of medicine in recent years. At present, the application of AIE materials in gastrointestinal (GI) diseases is mainly in GI imaging, diagnosis and treatment. In this review, we summarize the applications of AIE materials in GI pathogens and GI diseases, including inflammatory bowel disease and GI tumors, and outline combined treatment methods of AIE materials in GI tumor therapy.
Collapse
Affiliation(s)
- Yi-Rong Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Gang Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Wen-Ting He
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University, Lanzhou 730000, Gansu Province, China
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Tao Liu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University, Lanzhou 730000, Gansu Province, China
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu Province, China
| |
Collapse
|
2
|
Li C, Xu T, Hou G, Wang Y, Fu Q. DNA nanotechnology-based strategies for gastric cancer diagnosis and therapy. Mater Today Bio 2025; 30:101459. [PMID: 39866794 PMCID: PMC11762204 DOI: 10.1016/j.mtbio.2025.101459] [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: 11/01/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Gastric cancer (GC) is a formidable adversary in the field of oncology. The low early diagnosis rate of GC results in a low overall survival rate. Therefore, early accurate diagnosis and effective treatment are the key to reduce the mortality of GC. With the advent of nanotechnology, researchers continue to explore new possibilities for accurate diagnosis and effective treatment. One such breakthrough is the application of DNA nanotechnology. In this paper, the application of exciting DNA nanomaterials in the diagnosis and treatment of GC is discussed in depth. Firstly, the biomarkers related to GC and the diagnostic strategies related to DNA nanotechnology are summarized. Second, the latest research progress of DNA nanomaterials in the GC targeted therapy are summarized. Finally, the challenges and opportunities of DNA nanomaterials in the research and clinical application of GC are prospected.
Collapse
Affiliation(s)
- Congcong Li
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Tongyang Xu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Guopeng Hou
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Yin Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Qinrui Fu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| |
Collapse
|
3
|
Zhang H, Liang F, Wang F, Xu Q, Qiu Y, Lu X, Jiang L, Jian K. miR-148-3p inhibits gastric cancer cell malignant phenotypes and chemotherapy resistance by targeting Bcl2. Bioengineered 2024; 15:2005742. [PMID: 34783293 PMCID: PMC10841002 DOI: 10.1080/21655979.2021.2005742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer in the world. This work was designed to explore the biological effects of miR-148-3p on GC. Quantitative reverse transcription-polymerase chain reaction (RT-qPCR) was utilized to analyze the mRNA expression of miR-148-3p in GC cell lines. The mimics and inhibitors of miR-148-3p were carefully transfected into GC cells to up-regulate or down-regulate miR-148-3p expression. Observe the effect on miR-148-3p expression change to GC cell proliferation, colony formation, tumorigenesis, chemotherapy sensitivity, transwell migration, and invasion. Use online database tool to predict the miR-148-3p promising targets, and can be verified via RT-qPCR, Western blot, and luciferase report. We found that miR-148-3p expression level in GC cells was markedly down-regulated (P < 0.05), as compared with human normal gastric mucosal cells GES-1. Otherwise, miR-148-3p overexpression could effectively inhibit the cell proliferation, cell cycle progress, colony formation, anti-apoptosis, anti-migration and anti-invasion in gastric cancer cells, whereas miR-148-3p inhibition exhibited the opposite phenomenon (P < 0.05). Further research revealed that Bcl2 set as a direct downstream target of miR-148-3p. Our study firstly confirmed that, miR-148-3p might play a crucial role in tumorigenesis, as well as development of gastric cancer by targeting Bcl2, and could become a promising target for gastric cancer treatment.
Collapse
Affiliation(s)
- Hongyan Zhang
- Department of Oncology, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Feng Liang
- Department of General Surgery, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Fei Wang
- Department of Oncology, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Qianru Xu
- Department of General Surgery, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Yuxuan Qiu
- Department of General Surgery, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Xin Lu
- Department of General Surgery, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Lin Jiang
- Department of General Surgery, The Seventh Medical Center of Pla General Hospital, Beijing, China
| | - Kaiyu Jian
- Department of General Surgery, The Seventh Medical Center of Pla General Hospital, Beijing, China
| |
Collapse
|
4
|
Xiao S, Mu M, Feng C, Pan S, Chen N. The application of bacteria-nanomaterial hybrids in antitumor therapy. J Nanobiotechnology 2024; 22:536. [PMID: 39227831 PMCID: PMC11373302 DOI: 10.1186/s12951-024-02793-x] [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: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
Adverse effects and multidrug resistance remain significant obstacles in conventional cancer therapy. Nanomedicines, with their intrinsic properties such as nano-sized dimensions and tunable surface characteristics, have the potential to mitigate the side effects of traditional cancer treatments. While nanomaterials have been widely applied in cancer treatment, challenges such as low targeting efficiency and poor tumor penetration persist. Recent research has shown that anaerobic bacteria exhibit high selectivity for primary tumors and metastatic cancers, offering good safety and superior tumor penetration capabilities. This suggests that combining nanomaterials with bacteria could complement their respective limitations, opening vast potential applications in cancer therapy. The use of bacteria in combination with nanomaterials for anticancer treatments, including chemotherapy, radiotherapy, and photothermal/photodynamic therapy, has contributed to the rapid development of the field of bacterial oncology treatments. This review explores the mechanisms of bacterial tumor targeting and summarizes strategies for synthesizing bacterial-nanomaterial and their application in cancer therapy. The combination of bacterial-nanomaterial hybrids with modern therapeutic approaches represents a promising avenue for future cancer treatment research, with the potential to improve treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Susu Xiao
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Mu
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chenqian Feng
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shulin Pan
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nianyong Chen
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Chen X, Li ZW, Duan H, Sun YW, Su Y, Peng S, Guo Y, Xiong Y, Tang BZ, Huang X. A Ligand-Directed Spatial Regulation to Structural and Functional Tunability in Aggregation-Induced Emission Luminogen-Functionalized Organic-Inorganic Nanoassemblies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313381. [PMID: 38647215 DOI: 10.1002/adma.202313381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Aggregation-induced emission luminogen (AIEgen)-functionalized organic-inorganic hybrid nanoparticles (OINPs) are an emerging category of multifunctional nanomaterials with vast potential applications. The spatial arrangement and positioning of AIEgens and inorganic compounds in AIEgen-functionalized OINPs determine the structures, properties, and functionalities of the self-assembled nanomaterials. In this work, a facile and general emulsion self-assembly tactic for synthesizing well-defined AIEgen-functionalized OINPs is proposed by coassembling alkane chain-functionalized inorganic nanoparticles with hydrophobic organic AIEgens. As a proof of concept, the self-assembly and structural evolution of plasmonic-fluorescent hybrid nanoparticles (PFNPs) from concentric circle to core shell and then to Janus structures is demonstrated by using alkane chain-modified AuNPs and AIEgens as building blocks. The spatial position of AuNPs in the signal nanocomposite is controlled by varying the alkane ligand length and density on the AuNP surface. The mechanism behind the formation of various PFNP nanostructures is also elucidated through experiments and theoretical simulation. The obtained PFNPs with diverse structures exhibit spatially tunable optical and photothermal properties for advanced applications in multicolor and multimode immunolabeling and photothermal sterilization. This work presents an innovative synthetic approach of constructing AIEgen-functionalized OINPs with diverse structures, compositions, and functionalities, thereby championing the progressive development of these OINPs.
Collapse
Affiliation(s)
- Xirui Chen
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Zhan-Wei Li
- College of Chemistry and Green Catalysis Center, Zhengzhou University, Zhengzhou, 450001, China
| | - Hong Duan
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University, Beijing, 100048, China
| | - Yu-Wei Sun
- College of Chemistry and Green Catalysis Center, Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Su
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Shiyu Peng
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Yuqian Guo
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| |
Collapse
|
6
|
Li W, Yang Y, Wang J, Ge T, Wan S, Gui L, Tao Y, Song P, Yang L, Ge F, Zhang W. Establishment of bone-targeted nano-platform and the study of its combination with 2-deoxy-d-glucose enhanced photodynamic therapy to inhibit bone metastasis. J Mech Behav Biomed Mater 2024; 150:106306. [PMID: 38091923 DOI: 10.1016/j.jmbbm.2023.106306] [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: 09/07/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024]
Abstract
At present, simple anti-tumor drugs are ineffective at targeting bone tissue and are not purposed to treat patients with bone metastasis. In this study, zoledronic acid (ZOL) demonstrated excellent bone-targeting properties as a bone-targeting ligand. The metal-organic framework (MOF) known as ZIF-90 was modified with ZOL to construct a bone-targeting-based drug delivery system. Chlorin e6 (Ce6) was loaded in the bone-targeted drug delivery system and combined with 2-deoxy-D-glucose (2-DG), which successfully treated bone tumors when enhanced photodynamic therapy was applied. The Ce6@ZIF-PEG-ZOL (Ce6@ZPZ) nanoparticles were observed to have uniform morphology, a particle size of approximately 210 nm, and a potential of approximately -30.4 mV. The results of the bone-targeting experiments showed that Ce6@ZPZ exhibited a superior bone-targeted effect when compared to Ce6@ZIF-90-PEG. The Ce6@ZPZ solution was subjected to 660 nm irradiation and the resulting production of reactive oxygen species increased over time, which could be further increased when Ce6@ZPZ was used in combination with 2-DG. Their combination had a stronger inhibitory capacity against tumor cells than either 2-DG or Ce6@ZPZ alone, increasing the rate of tumor cell apoptosis. The apoptosis rate caused by HGC-27 was 61.56% when 2-DG was combined with Ce6@ZPZ. In vivo results also showed that Ce6@ZPZ combined with 2-DG maximally inhibited tumor growth and prolonged mice survival compared to the other experimental groups. Therefore, the combination of PDT and glycolytic inhibitors serves as a potential option for the treatment of cancer.
Collapse
Affiliation(s)
- Wanzhen Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China
| | - Yongqi Yang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China
| | - Jun Wang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China
| | - Ting Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China
| | - Shuixia Wan
- Soil and Fertilizer Research Institute, Anhui Academy of Agricultural Sciences, Hefei, 230031, People's Republic of China
| | - Lin Gui
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui, 241002, People's Republic of China
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China
| | - Ping Song
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China.
| | - Liangjun Yang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China.
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China.
| | - Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui, 241000, People's Republic of China.
| |
Collapse
|
7
|
Shen Q, Yu C. Advances in superparamagnetic iron oxide nanoparticles modified with branched polyethyleneimine for multimodal imaging. Front Bioeng Biotechnol 2024; 11:1323316. [PMID: 38333548 PMCID: PMC10851169 DOI: 10.3389/fbioe.2023.1323316] [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: 10/17/2023] [Accepted: 12/18/2023] [Indexed: 02/10/2024] Open
Abstract
Multimodal imaging are approaches which combines multiple imaging techniques to obtain multi-aspect information of a target through different imaging modalities, thereby greatly improve the accuracy and comprehensiveness of imaging. Superparamagnetic iron oxide nanoparticles (SPIONs) modified with branched polyethyleneimine have revealed good biocompatibility and stability, high drug loading capacity and nucleic acid transfection efficiency. SPIONs have been developed as functionalized platforms which can be further modified to enhance their functionalities. Those further modifications facilitate the application of SPIONs in multimodal imaging. In this review, we discuss the methods, advantages, applications, and prospects of BPEI-modified SPIONs in multimodal imaging.
Collapse
Affiliation(s)
- Qiaoling Shen
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
8
|
Xie D, Song C, Qin T, Zhai Z, Cai J, Dai J, Sun T, Xu Y. Moschus ameliorates glutamate-induced cellular damage by regulating autophagy and apoptosis pathway. Sci Rep 2023; 13:18586. [PMID: 37903904 PMCID: PMC10616123 DOI: 10.1038/s41598-023-45878-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 10/25/2023] [Indexed: 11/01/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, causes short-term memory and cognition declines. It is estimated that one in three elderly people die from AD or other dementias. Chinese herbal medicine as a potential drug for treating AD has gained growing interest from many researchers. Moschus, a rare and valuable traditional Chinese animal medicine, was originally documented in Shennong Ben Cao Jing and recognized for its properties of reviving consciousness/resuscitation. Additionally, Moschus has the efficacy of "regulation of menstruation with blood activation, relief of swelling and pain" and is used for treating unconsciousness, stroke, coma, and cerebrovascular diseases. However, it is uncertain whether Moschus has any protective effect on AD patients. We explored whether Moschus could protect glutamate (Glu)-induced PC12 cells from cellular injury and preliminarily explored their related action mechanisms. The chemical compounds of Moschus were analyzed and identified by GC-MS. The Glu-induced differentiated PC12 cell model was thought to be the common AD cellular model. The study aims to preliminarily investigate the intervention effect of Moschus on Glu-induced PC12 cell damage as well as their related action mechanisms. Cell viability, lactate dehydrogenase (LDH), mitochondrial reactive oxygen species, mitochondrial membrane potential (MMP), cell apoptosis, autophagic vacuoles, autolysosomes or autophagosomes, proteins related to apoptosis, and the proteins related to autophagy were examined and analyzed. Seventeen active compounds of the Moschus sample were identified based on GC-MS analysis. In comparison to the control group, Glu stimulation increased cell viability loss, LDH release, mitochondrial damage, loss of MMP, apoptosis rate, and the number of cells containing autophagic vacuoles, and autolysosomes or autophagosomes, while these results were decreased after the pretreatment with Moschus and 3-methyladenine (3-MA). Furthermore, Glu stimulation significantly increased cleaved caspase-3, Beclin1, and LC3II protein expression, and reduced B-cell lymphoma 2/BAX ratio and p62 protein expression, but these results were reversed after pretreatment of Moschus and 3-MA. Moschus has protective activity in Glu-induced PC12 cell injury, and the potential mechanism might involve the regulation of autophagy and apoptosis. Our study may promote research on Moschus in the field of neurodegenerative diseases, and Moschus may be considered as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Caiyou Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jie Cai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jingyi Dai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
9
|
Pous A, Notario L, Hierro C, Layos L, Bugés C. HER2-Positive Gastric Cancer: The Role of Immunotherapy and Novel Therapeutic Strategies. Int J Mol Sci 2023; 24:11403. [PMID: 37511163 PMCID: PMC10380453 DOI: 10.3390/ijms241411403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer is an aggressive disease with increasing global incidence in recent years. Human epidermal growth receptor 2 (HER2) is overexpressed in approximately 10-20% of gastric cancers. The implementation of targeted therapy against HER2 as part of the standard of care treatment in metastatic disease has improved the prognosis of this subset of patients. However, gastric cancer still has high mortality rates and urgently requires new treatment strategies. The combination of immunotherapy with HER2-targeted therapies has shown synergistic effects in preclinical models, this being the rationale behind exploring this combination in clinical trials in locally advanced and metastatic settings. Additionally, the irruption of antibody-drug conjugates and other novel HER2-targeted agents has led to the development of numerous clinical trials showing promising results. This review presents the molecular mechanisms supporting the use of HER2-targeted drugs in combination with immunotherapy and provides an overview of the therapeutic scenario of HER2-positive disease. We focus on the role of immunotherapy but also summarize emerging therapies and combinations under clinical research that may change the standard treatment in HER-2 positive disease in the future.
Collapse
Affiliation(s)
- Anna Pous
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Lucía Notario
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Cinta Hierro
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Laura Layos
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Cristina Bugés
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| |
Collapse
|
10
|
Huang L, Zhang Z. Recent Advances in the DNA-Mediated Multi-Mode Analytical Methods for Biological Samples. BIOSENSORS 2023; 13:693. [PMID: 37504092 PMCID: PMC10377368 DOI: 10.3390/bios13070693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023]
Abstract
DNA-mediated nanotechnology has become a research hot spot in recent decades and is widely used in the field of biosensing analysis due to its distinctive properties of precise programmability, easy synthesis and high stability. Multi-mode analytical methods can provide sensitive, accurate and complementary analytical information by merging two or more detection techniques with higher analytical throughput and efficiency. Currently, the development of DNA-mediated multi-mode analytical methods by integrating DNA-mediated nanotechnology with multi-mode analytical methods has been proved to be an effective assay for greatly enhancing the selectivity, sensitivity and accuracy, as well as detection throughput, for complex biological analysis. In this paper, the recent progress in the preparation of typical DNA-mediated multi-mode probes is reviewed from the aspect of deoxyribozyme, aptamer, templated-DNA and G-quadruplex-mediated strategies. Then, the advances in DNA-mediated multi-mode analytical methods for biological samples are summarized in detail. Moreover, the corresponding current applications for biomarker analysis, bioimaging analysis and biological monitoring are introduced. Finally, a proper summary is given and future prospective trends are discussed, hopefully providing useful information to the readers in this research field.
Collapse
Affiliation(s)
- Lu Huang
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhuomin Zhang
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
11
|
Lv QY, Cui HF, Song X. Aptamer-based technology for gastric cancer theranostics. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:2142-2153. [PMID: 37114324 DOI: 10.1039/d3ay00415e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Gastric cancer is one of the most common causes of cancer death worldwide. This cancer exhibits high molecular and phenotype heterogeneity. The overall survival rate for gastric cancer is very low because it is always diagnosed in the advanced stages. Therefore, early detection and treatment are of great significance. Currently, biomedical studies have tapped the potential clinical applicability of aptamer-based technology for gastric cancer diagnosis and targeted therapy. Herein, we summarize the enrichment and evolution of relevant aptamers, followed by documentation of the recent developments in aptamer-based techniques for early diagnosis and precision therapy for gastric cancers.
Collapse
Affiliation(s)
- Qi-Yan Lv
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| | - Hui-Fang Cui
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| | - Xiaojie Song
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| |
Collapse
|
12
|
Dai W, Dai YG, Ren DF, Zhu DW. Dieckol, a natural polyphenolic drug, inhibits the proliferation and migration of colon cancer cells by inhibiting PI3K, AKT, and mTOR phosphorylation. J Biochem Mol Toxicol 2023; 37:e23313. [PMID: 36683349 DOI: 10.1002/jbt.23313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/07/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023]
Abstract
This study investigated that dieckol (DKL), a natural drug, inhibits colon cancer cell proliferation and migration by inhibiting phosphoinositide-3-kinase (PI3K), protein kinase B (AKT), and mammalian target of rapamycin (mTOR) phosphorylation in HCT-116 cells. The cells were treated with DKL in various concentrations (32 and 50 μM) for 24 h and then analyzed for various experiments. MTT (tetrazolium bromide) and crystal violet assay investigated DKL-mediated cytotoxicity. Dichlorodihydrofluorescein diacetate staining was used to assess the reactive oxygen species (ROS) measurement, and apoptotic changes were studied by dual acridine orange and ethidium bromide staining. Protein expression of cell survival, cell cycle, proliferation, and apoptosis protein was evaluated by western blot analysis. Results indicated that DKL produces significant cytotoxicity in HCT-116, and the half-maximal inhibitory concentration was found to be 32 μM for 24-h incubation. Moreover, effective production of ROS and enhanced apoptotic signs were observed upon DKL treatment in HCT-116. DKL induces the expression of phosphorylated PI3K, AKT, and mToR-associated enhanced expression of cyclin-D1, proliferating cell nuclear antigen, cyclin-dependent kinase (CDK)-4, CDK-6, and Bcl-2 in HCT-116. In addition, proapoptotic proteins such as Bax, caspase-9, and caspase-3 were significantly enhanced by DKL treatment in HCT-116. Hence, DKL has been considered a chemotherapeutic drug by impeding the expression of PI3K-, AKT-, and mTOR-mediated inhibition of proliferation and cell cycle-regulating proteins.
Collapse
Affiliation(s)
- Wei Dai
- Department of Clinical Laboratory, Ganzhou People's Hospital, Jiangxi, Ganzhou, China
| | - Yong Gang Dai
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Dong Feng Ren
- Department of Oncology, The First Hospital of Yulin, Shaanxi, Yulin, China
| | - Da Wei Zhu
- Department of Gastroenterology, Hongze District People's Hospital, Jiangsu, Huai'an, China
| |
Collapse
|
13
|
Yin C, Wang Z, Dai C, Yang B, Wang W, Yang E, Guo F, Fan C, Zhang P, Sun J, Sun D. Light-triggered photosynthetic engineered bacteria for enhanced-photodynamic therapy by relieving tumor hypoxic microenvironment. Theranostics 2023; 13:1632-1648. [PMID: 37056566 PMCID: PMC10086205 DOI: 10.7150/thno.81718] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/03/2023] [Indexed: 04/15/2023] Open
Abstract
Background: Singlet oxygen (1O2) has received considerable research attention in photodynamic therapy (PDT) due to its cytotoxic solid features. However, the inherent hypoxic state of the tumor microenvironment (TME) leads to the meager 1O2 quantum yield of inorganic PDT reagents, and their application in vivo remains elusive. Methods: We developed a novel strategy to fabricate active photosynthetic bacteria/photosensitizer/photothermal agent hybrids for photosynthetic tumor oxygenation and PDT and PTT tumor therapy under different laser irradiation sources. Photosynthetic bacteria combined with Ce6 photosensitizer and Au NPs photothermal agent, the obtained Bac@Au-Ce6 effectively targets tumor tissues and further enhances the tumor accumulation of Au-Ce6. Results: The results showed that the Au-Ce6-loaded engineered bacteria (Bac@Au-Ce6) maintained the photosynthetic properties of Syne. After i.v. injection, Bac@Au-Ce6 efficiently aggregates at tumor sites due to the tumor-targeting ability of active Syne. With 660 nm laser irradiation at the tumor site, the photoautotrophic Syne undergoes sustained photosynthetic O2 release and immediately activates O2 to 1O2 via a loaded photosensitizer. PTT was subsequently imparted by 808 laser irradiations to enhance tumor killing further. Conclusions: This work provides a new platform for engineering bacteria-mediated photosynthesis to promote PDT combined with PTT multi-faceted anti-tumor.
Collapse
Affiliation(s)
- Chenyang Yin
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - ZeKun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Chunxue Dai
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Bangjia Yang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Weiyun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Endong Yang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Feng Guo
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Cundong Fan
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, China
- ✉ Corresponding authors: Dongdong Sun, ; Cundong Fan, ; Jikui Sun, ; Pu Zhang,
| | - Pu Zhang
- Department of Cardiology, Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, 271000, China
- ✉ Corresponding authors: Dongdong Sun, ; Cundong Fan, ; Jikui Sun, ; Pu Zhang,
| | - Jikui Sun
- Department of Neurosurgery, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- ✉ Corresponding authors: Dongdong Sun, ; Cundong Fan, ; Jikui Sun, ; Pu Zhang,
| | - Dongdong Sun
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
- ✉ Corresponding authors: Dongdong Sun, ; Cundong Fan, ; Jikui Sun, ; Pu Zhang,
| |
Collapse
|
14
|
Liu H, Miyamoto N, Nguyen MT, Shirato H, Yonezawa T. Injectable Fiducial Marker for Image-Guided Radiation Therapy Based on Gold Nanoparticles and a Body Temperature-Activated Gel-Forming System. ACS APPLIED BIO MATERIALS 2022; 5:4838-4848. [PMID: 36074396 DOI: 10.1021/acsabm.2c00566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Injectable fiducial markers are crucial in image-guided radiation therapy (IGRT) due to their minimally invasive operations and improved patient compliance. This study presents the development of a ready-to-use injectable fiducial marker utilizing alginate stabilized-gold nanoparticles (alg-Au NPs) and a body temperature-activated in situ gel-forming system. Gram-scale alg-Au NPs were prepared in an hour by a green microwave-induced plasma-in-liquid process (MWPLP). Sodium alginate was introduced in this process to avoid aggregation between Au NPs, which ensured their stability and injectability. The gelation behavior of alginate with divalent cations and a temperature-dependent release of calcium source (glucono-delta-lactone (GDL) and CaCO3) served as the foundation of the body temperature-activated in situ gel-forming system. The injectable fiducial marker GDL/CaCO3/alg-Au NPs could maintain a liquid state at a low temperature for a higher injectability. After injection, on the other hand, Ca2+ would be released due to the body temperature-activated hydrolysis of GDL and the subsequent reaction with CaCO3, which would initiate the gelation of alginate. The injectable fiducial marker can be therefore delivered via injection and form gel at target site to avoid marker movement or Au NPs leakage after injection. Rheological measurements demonstrate the stability and gelation behavior of GDL/CaCO3/alg-Au NPs at different temperatures. Furthermore, the injectability and imaging ability of GDL/CaCO3/alg-Au NPs were also examined. In summary, ready-to-use injectable fiducial marker GDL/CaCO3/alg-Au NPs were developed via a green and facile method for IGRT.
Collapse
Affiliation(s)
- Haoran Liu
- Division of Materials Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Naoki Miyamoto
- Division of Quantum Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
- Department of Medical Physics, Hokkaido University Hospital, Kita 14 Nishi 5, Kita-ku, Sapporo, Hokkaido 060-8648, Japan
| | - Mai Thanh Nguyen
- Division of Materials Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Tetsu Yonezawa
- Division of Materials Science and Engineering, Faculty of Engineering, Hokkaido University, Kita 13 Nishi 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| |
Collapse
|
15
|
Li C, Cheng Y, Li D, An Q, Zhang W, Zhang Y, Fu Y. Antitumor Applications of Photothermal Agents and Photothermal Synergistic Therapies. Int J Mol Sci 2022; 23:ijms23147909. [PMID: 35887255 PMCID: PMC9324234 DOI: 10.3390/ijms23147909] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
As a new tumor treatment strategy, photothermal therapy (PTT) has the advantages of accuracy, ease of administration, a high efficiency and low side effects. Photothermal transduction agents (PTAs) are the key factor which play an important role in PTT. The mechanism of PTT is discussed in detail. The photothermal conversion efficiency (PCE) can be improved by increasing the light absorption and reducing the light scattering of photothermal conversion agents. Additionally, non-radiative relaxation path attenuation can also promote energy conversion to obtain a higher value in terms of PCE. The structure and photothermal characteristics of various kinds of PTAs (metal materials, carbon-based nanomaterials, two-dimensional nanomaterials, and organic materials) were compared and analyzed. This paper reviews the antitumor applications of photothermal synergistic therapies, including PTT combined with immunotherapy, chemotherapy, and photodynamic therapy. This review proposes that these PTAs promote the development of photothermal synergistic therapies and have a great potential in the application of tumor treatment.
Collapse
Affiliation(s)
- Chaowei Li
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
| | - Yue Cheng
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
- National & Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Health, Nantong University, Nantong 226019, China
| | - Dawei Li
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
- National & Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Health, Nantong University, Nantong 226019, China
- Correspondence: (D.L.); (Y.F.)
| | - Qi An
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
- National & Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Health, Nantong University, Nantong 226019, China
| | - Wei Zhang
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
- National & Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Health, Nantong University, Nantong 226019, China
| | - Yu Zhang
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
- National & Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Health, Nantong University, Nantong 226019, China
| | - Yijun Fu
- School of Textile and Clothing, Nantong University, Nantong 226019, China; (C.L.); (Y.C.); (Q.A.); (W.Z.); (Y.Z.)
- National & Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Health, Nantong University, Nantong 226019, China
- Correspondence: (D.L.); (Y.F.)
| |
Collapse
|
16
|
Li P, He X, Li Y, Lam JWY, Kwok RTK, Wang CC, Xia LG, Tang BZ. Recent advances in aggregation-induced emission luminogens in photoacoustic imaging. Eur J Nucl Med Mol Imaging 2022; 49:2560-2583. [PMID: 35277741 DOI: 10.1007/s00259-022-05726-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/13/2022] [Indexed: 12/14/2022]
Abstract
Photoacoustic imaging (PAI) is a rapidly emerging modality in biomedical research with the advantages of noncontact operation, high optical resolution, and deep penetration. Great efforts and progress in the development of PAI agents with improved imaging resolution and sensitivity have been made over the past 2 decades. Among them, organic agents are the most promising candidates for preclinical/clinical applications due to their outstanding in vivo properties and facile biofunctionalities. Motivated by the unique properties of aggregation-induced emission (AIE) luminogens (AIEgens), various optical probes have been developed for bioanalyte detection, multimodal bioimaging, photodynamic/photothermal therapy, and imaging-guided therapeutics. In particular, AIE-active contrast agents have been demonstrated in PAI applications with excellent performance in imaging resolution and tissue permeability in vivo. This paper presents a brief overview of recent progress in AIE-based agents in the field of photoacoustic imaging. In particular, we focus on the basic concepts, data sorting and comparison, developing trends, and perspectives of photoacoustic imaging. Through numerous typical examples, the way each system realizes the desired photoacoustic performance in various biomedical applications is clearly illustrated. We believe that AIE-based PAI agents would be promising multifunctional theranostic platforms in clinical fields and will facilitate significant advancements in this research topic.
Collapse
Affiliation(s)
- Pei Li
- Department of Gastrointestinal Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital, Southern University of Science and Technology, 518020, Shenzhen, China
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration & Reconstruction, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
- Department of General Surgery, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Xuewen He
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration & Reconstruction, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yang Li
- Department of Gastrointestinal Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital, Southern University of Science and Technology, 518020, Shenzhen, China
- Department of General Surgery, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Jacky Wing Yip Lam
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration & Reconstruction, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ryan Tsz Kin Kwok
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration & Reconstruction, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| | - Cun Chuan Wang
- The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Li Gang Xia
- Department of Gastrointestinal Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital, Southern University of Science and Technology, 518020, Shenzhen, China.
- Department of General Surgery, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration & Reconstruction, Division of Life Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Shenzhen Institute of Molecular Aggregate Science and Engineering, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen City, 518172, Guangdong, China
| |
Collapse
|
17
|
Zuo Y, Shen H, Sun F, Li P, Sun J, Kwok RTK, Lam JWY, Tang BZ. Aggregation-Induced Emission Luminogens for Cell Death Research. ACS BIO & MED CHEM AU 2022; 2:236-257. [PMID: 37101570 PMCID: PMC10114857 DOI: 10.1021/acsbiomedchemau.1c00066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cell death is closely related to various diseases, and monitoring and controlling cell death is a promising strategy to develop efficient therapy. Aggregation-induced emission luminogens (AIEgens) are ideal candidates for developing novel theranostic agents because of their intriguing properties in the aggregate state. The rational application of AIE materials in cell death-related research is still in its infancy but has shown great clinical potential. This review discussed the research frontier and our understanding of AIE materials in various subroutines of cell death, including apoptosis, necrosis, immunogenic cell death, pyroptosis, autophagy, lysosome-dependent cell death, and ferroptosis. We hope that the new insights can be offered to this growing field and attract more researchers to provide valuable contributions.
Collapse
Affiliation(s)
- Yunfei Zuo
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Hanchen Shen
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Feiyi Sun
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Pei Li
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
- Department
of Gastrointestinal Surgery, The Second Clinical Medical College, Shenzhen People’s Hospital, Jinan University, Shenzhen, 518020, China
| | - Jianwei Sun
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Ryan T. K. Kwok
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Jacky W. Y. Lam
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
| | - Ben Zhong Tang
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, and Guangdong-Hong
Kong-Macau Joint Laboratory of Optoelectronic and Magnetic Functional
Materials, Division of Life Science, and State Key Laboratory of Molecular
Neuroscience, The Hong Kong University of
Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P.R. China
- Shenzhen
Institute of Aggregate Science and Technology, School of Science and
Engineering, The Chinese University of Hong
Kong, Shenzhen, 2001
Longxiang Boulevard, Longgang District, Shenzhen
City, Guangdong 518172, China
| |
Collapse
|
18
|
Recombinant programmed cell death protein 1 functions as an immune check point blockade and enhances anti-cancer immunity. Biomaterials 2022; 285:121550. [DOI: 10.1016/j.biomaterials.2022.121550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/22/2022] [Accepted: 04/25/2022] [Indexed: 11/21/2022]
|
19
|
Zhang W, Kuang Z, Song P, Li W, Gui L, Tang C, Tao Y, Ge F, Zhu L. Synthesis of a Two-Dimensional Molybdenum Disulfide Nanosheet and Ultrasensitive Trapping of Staphylococcus Aureus for Enhanced Photothermal and Antibacterial Wound-Healing Therapy. NANOMATERIALS 2022; 12:nano12111865. [PMID: 35683721 PMCID: PMC9182539 DOI: 10.3390/nano12111865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/25/2022]
Abstract
Photothermal therapy has been widely used in the treatment of bacterial infections. However, the short photothermal effective radius of conventional nano-photothermal agents makes it difficult to achieve effective photothermal antibacterial activity. Therefore, improving composite targeting can significantly inhibit bacterial growth. We inhibited the growth of Staphylococcus aureus (S. aureus) by using an extremely low concentration of vancomycin (Van) and applied photothermal therapy with molybdenum disulfide (MoS2). This simple method used chitosan (CS) to synthesize fluorescein 5(6)-isothiocyanate (FITC)-labeled and Van-loaded MoS2-nanosheet hydrogels (MoS2-Van-FITC@CS). After modifying the surface, an extremely low concentration of Van could inhibit bacterial growth by trapping bacteria synergistically with the photothermal effects of MoS2, while FITC labeled bacteria and chitosan hydrogels promoted wound healing. The results showed that MoS2-Van-FITC@CS nanosheets had a thickness of approximately 30 nm, indicating the successful synthesis of the nanosheets. The vitro antibacterial results showed that MoS2-Van-FITC with near-infrared irradiation significantly inhibited S. aureus growth, reaching an inhibition rate of 94.5% at nanoparticle concentrations of up to 100 µg/mL. Furthermore, MoS2-Van-FITC@CS could exert a healing effect on wounds in mice. Our results demonstrate that MoS2-Van-FITC@CS is biocompatible and can be used as a wound-healing agent.
Collapse
Affiliation(s)
- Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
| | - Zhao Kuang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
| | - Ping Song
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
| | - Wanzhen Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
| | - Lin Gui
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu 241002, China;
| | - Chuchu Tang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
- Correspondence: (Y.T.); (F.G.); (L.Z.)
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
- Correspondence: (Y.T.); (F.G.); (L.Z.)
| | - Longbao Zhu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, China; (W.Z.); (Z.K.); (P.S.); (W.L.); (C.T.)
- Correspondence: (Y.T.); (F.G.); (L.Z.)
| |
Collapse
|
20
|
Zhu X, Su T, Wang S, Zhou H, Shi W. New Advances in Nano-Drug Delivery Systems: Helicobacter pylori and Gastric Cancer. Front Oncol 2022; 12:834934. [PMID: 35619913 PMCID: PMC9127958 DOI: 10.3389/fonc.2022.834934] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/29/2022] [Indexed: 01/07/2023] Open
Abstract
With the development of materials science and biomedicine, the application of nanomaterials in the medical field is further promoted. In the process of the diagnosis and treatment of diseases, a variety of drugs need to be used. It is an ideal state to make these drugs arrive at a specific location at a specific time and release at a specific speed, which can improve the bioavailability of drugs and reduce the adverse effects of drugs on normal tissues. Traditional drug delivery methods such as tablets, capsules, syrups, and ointments have certain limitations. The emergence of a new nano-drug delivery system further improves the accuracy of drug delivery and the efficacy of drugs. It is well known that the development of the cancer of the stomach is the most serious consequence for the infection of Helicobacter pylori. For the patients who are suffering from gastric cancer, the treatments are mainly surgery, chemotherapy, targeted and immune therapy, and other comprehensive treatments. Although great progress has been made, the diagnosis and prognosis of gastric cancer are still poor with patients usually diagnosed with cancer at an advanced stage. Current treatments are of limited benefits for patients, resulting in a poor 5-year survival rate. Nanomaterials may play a critical role in early diagnosis. A nano-drug delivery system can significantly improve the chemotherapy, targeted therapy, and immunotherapy of advanced gastric cancer, reduce the side effects of the original treatment plan and provide patients with better benefits. It is a promising treatment for gastric cancer. This article introduces the application of nanomaterials in the diagnosis and treatment of H. pylori and gastric cancer.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Su
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiqing Zhou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weibin Shi
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Tumor microenvironment pH-responsive pentagonal gold prism-based nanoplatform for multimodal imaging and combined therapy of castration-resistant prostate cancer. Acta Biomater 2022; 141:408-417. [PMID: 35032718 DOI: 10.1016/j.actbio.2022.01.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
Abstract
Given that there is lack of effective therapies for castration-resistant prostate cancer (CRPC), the combination of photothermal (PTT), photodynamic (PDT), and chemical therapy (CT) has emerged as a prominent strategy. Tumor-targeted delivery and controlled release of antitumor drug are key-elements of any combined therapy. Considering these important elements, we designed and constructed tumor microenvironment (TME)-activated nanoprobes (PGP/CaCO3@IR820/DTX-HA). The CaCO3 shell could efficiently entrap the photosensitizer IR820 and the chemotherapeutic docetaxel (DTX) on the surface of pentagonal gold prisms (PGPs) to prevent elimination from the circulation, and it could act as a TME-trigger to achieve TME-responsive drug release. After modification with hyaluronic acid, PGP/CaCO3@IR820/DTX-HA was capable of synergistic TME-triggered PTT/PDT/CT and tumor-targeted delivery. Our in vitro and in vivo studies demonstrate that PGP/CaCO3@IR820/DTX-HA could achieve synergistic antitumor effects following near-infrared (NIR)-light irradiation. In addition, using the NIR fluorescence signal from IR820 and the photoacoustic (PA) signal from PGPs, i.e., through multimodal fluorescence/photoacoustic imaging, we could monitor the in vivo distribution and excretion of PGP/CaCO3@IR820/DTX-HA. Therefore, it can be concluded that PGP/CaCO3@IR820/DTX-HA shows promising clinical translational potential as a treatment for CRPC. STATEMENT OF SIGNIFICANCE: Utilizing pentagonal gold prisms (PGPs), we constructed a multifunctional nanoplatform (PGP/CaCO3@IR820/DTX-HA) for effectively delivering agents into the tumor microenvironment (TME) for the diagnosis and therapy of castration-resistant prostate cancer (CRPC). The synthetic nanoplatform can satisfy TME-activated synergistic photothermal therapy (PTT)/photodynamic therapy (PDT)/chemical therapy (CT) and NIR fluorescence imaging/photoacoustic (PA) imaging. Hyaluronic acid (HA) on the surface of nanoplatform allowed the specific tumor-targeting capacity and biocompatibility. In conclusion, PGP/CaCO3@IR820/DTX-HA could be a promising integrated nanoplatform for CRPC diagnosis and treatment.
Collapse
|
22
|
Ge T, Weiwei Z, Ge F, Zhu L, Song P, Li W, Gui L, Dong W, Tao Y, Yang K. A bone-targeting drug delivery vehicle of a metal-organic framework conjugate with zoledronate combined with photothermal therapy for tumor inhibition in cancer bone metastasis. Biomater Sci 2022; 10:1831-1843. [PMID: 35253030 DOI: 10.1039/d1bm01717a] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chemotherapy is a conventional treatment method for metastatic bone cancer, but it has limitations, such as lower drug-targeting of bone tissues and serious side effects. Bone metastasis almost always occurs in advanced cancer, and most patients in this period have strong drug resistance, which further worsens the curative effect. To address the above-mentioned difficulties, a drug delivery platform is proposed in this paper that accomplishes the bone-targeting of drugs to efficiently inhibit tumors. First, the anti-cancer drugs 5-fluorouracil (5-Fu) and indocyanine green (ICG) were loaded into a zeolitic imidazolate framework (ZIF-90) to form 5-Fu/ICG@ZIF-90. Polyethylene glycol with zoledronic acid (ZOL) was encapsulated using 5-Fu/ICG@ZIF-90 to synthesize 5-Fu/ICG@ZIF-90-PEG-ZOL nanoparticles, which showed dimensional stability, good thermal stability, and bone-targeting ability. Second, the in vitro anti-cancer activity of the designed platform was investigated using cytotoxicity, apoptosis, live-dead staining, cell cycle, and cell ultrathin section analysis. The results indicated that the nanoparticles inhibited MCF-7 cell activity when chemotherapy was combined with PTT. Finally, H&E staining and TUNEL detection were performed in mouse organs and tumors. The nanoparticles combined with photothermal therapy (PTT) and triggered by near-infrared irradiation induce apoptosis of tumor cells in vivo, displaying a better efficacy of combined chemotherapy and photothermal therapy. Experiments conducted on the 5-Fu/ICG@ZIF-90-PEG-ZOL nanoparticles demonstrated their promising performance for cancer bone metastasis inhibition.
Collapse
Affiliation(s)
- Ting Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Zhang Weiwei
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Longbao Zhu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Ping Song
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Wanzheng Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Lin Gui
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, People's Republic of China
| | - Wan Dong
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
23
|
Ning P, Chen Y, Bai Q, Xu C, Deng C, Cheng Q, Cheng Y. Multimodal Imaging-Guided Spatiotemporal Tracking of Photosensitive Stem Cells for Breast Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7551-7564. [PMID: 35107006 DOI: 10.1021/acsami.1c13072] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Stem cell therapy has shown great potential in treating a wide range of diseases including cancer. The real-time tracking of stem cells with high spatiotemporal resolution and stable imaging signals remains the bottleneck to evaluate and monitor therapeutic outcomes once transplanted into patients. Here, we developed a photosensitive mesenchymal stem cell (MSC) loaded with mesoporous silica-coated gold nanostars (MGNSs) integrated with indocyanine green for spatiotemporal tracking and imaging-guided photothermal therapy (PTT) in treating breast cancers. The MGNS served as a stable imaging probe with multifunctional properties for photoacoustic imaging (PAI), fluorescence imaging, and PT imaging. Owing to the excellent PT stability of MGNSs, long-term three-dimensional (3D) PAI was achieved to monitor stem cells in real time at the tumor site, while the tumor structure was imaged using 3D B-mode ultrasound imaging. PAI revealed that the photosensitive stem cells reached the widest distribution area at the tumor site post 24 h of intratumoral injection, which was further confirmed via two-dimensional (2D) PT and fluorescence imaging. With this optimal cell distribution window, in vivo studies showed that the photosensitive stem cells via both intratumoral and intravenous injections successfully inhibited breast cancer cell growth and decreased the tumor recurrence rate post PTT. Our results support that this photo-integrated platform with stable optical properties is promising to achieve real-time tracking and measure the cell distribution quantitatively with high spatiotemporal resolution for stem cell therapy.
Collapse
Affiliation(s)
- Peng Ning
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Yingna Chen
- Institute of Acoustics, School of Physics Science and Engineering, Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, 1239 Siping Road, Shanghai 200092, China
- The Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Qianwen Bai
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Chang Xu
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Cuijun Deng
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, 1239 Siping Road, Shanghai 200092, China
- The Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200065, China
| | - Yu Cheng
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Road, Shanghai 200123, China
| |
Collapse
|
24
|
Ni H, Ruan G, Sun C, Yang X, Miao Z, Li J, Chen Y, Qin H, Liu Y, Zheng L, Xing Y, Xi T, Li X. Tanshinone IIA inhibits gastric cancer cell stemness through inducing ferroptosis. ENVIRONMENTAL TOXICOLOGY 2022; 37:192-200. [PMID: 34661962 DOI: 10.1002/tox.23388] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/27/2021] [Accepted: 10/03/2021] [Indexed: 05/16/2023]
Abstract
Tanshinone IIA is the active constituent extracted from Salvia Miltiorrhza. Numerous studies have shown that Tanshinone IIA could inhibit tumor proliferation and metastasis, including gastric cancer. However, the effect of Tanshinone IIA on gastric cancer cell stemness stays unclear. Here, we found that Tanshinone IIA could reduce gastric cancer cell stemness through detecting spheroid-forming, flow cytometry analysis, and the expression of stemness markers (OCT3/4, ALDH1A1, and CD44). Mechanistically, Tanshinone IIA increased the level of lipid peroxides and decreased glutathione level in gastric cancer cells, both of which are the markers of ferroptosis. Similarly, ferroptosis inducers (erastin, sulfasalazine, and sorafenib) reduced gastric cancer cell stemness. Additionally, the inhibitory effects of Tanshinone IIA on GC cell stemness were reversed by ferroptosis inhibitor (Fer-1) or overexpression of SLC7A11, which is a critical ferroptosis inhibitor. Therefore, we revealed that Tanshinone IIA inhibited the stemness of gastric cancer cells partly through inducing ferroptosis.
Collapse
Affiliation(s)
- Haiwei Ni
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Guojing Ruan
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Cheng Sun
- Department of Pharmacy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Yang
- Department of Pharmacy, Huai'an Third People's Hospital, Huai'an, China
| | - Zhenyan Miao
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Jifei Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Ying Chen
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Hai Qin
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yichen Liu
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
25
|
Liu W, Su J, Shi Q, Wang J, Chen X, Zhang S, Li M, Cui J, Fan C, Sun B, Wang G. RGD Peptide-Conjugated Selenium Nanocomposite Inhibits Human Glioma Growth by Triggering Mitochondrial Dysfunction and ROS-Dependent MAPKs Activation. Front Bioeng Biotechnol 2022; 9:781608. [PMID: 35004643 PMCID: PMC8733670 DOI: 10.3389/fbioe.2021.781608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/10/2021] [Indexed: 01/17/2023] Open
Abstract
Chemotherapy is still one of the most common ways to treat human glioblastoma in clinic. However, severe side effects limited its clinic application. Design of cancer-targeted drugs with high efficiency and low side effect is urgently needed. Herein, silver nanoparticles (Ag NPs) and nano-selenium (Se NPs) conjugated with RGD peptides (Ag@Se@RGD NPs) to target integrin high-expressed glioma were designed. The results found that Ag@Se@RGD NPs displayed stable particle size and morphology in physiological condition, and induced significant integrin-targeted intracellular uptake. Ag@Se@RGD NPs in vitro dose-dependently inhibited U251 human glioma cells growth by induction of cells apoptosis through triggering the loss of mitochondrial membrane potential, overproduction of reactive oxygen species (ROS), and MAPKs activation. However, ROS inhibition dramatically attenuated Ag@Se@RGD NPs-induced MAPKs activation, indicating the significant role of ROS as an early apoptotic event. Importantly, Ag@Se@RGD NPs administration in vivov effectively inhibited U251 tumor xenografts growth by induction of apoptosis through regulation MAPKs activation. Taken together, our findings validated the rational design that Ag-Se NPs conjugated with RGD peptides was a promising strategy to combat human glioma by induction of apoptosis through triggering mitochondrial dysfunction and ROS-dependent MAPKs activation.
Collapse
Affiliation(s)
- Wenjian Liu
- Department of Oncology, Second Affiliated Hospital of Shandong First Medical University, Shandong Academy of Medical Sciences, Taian, China
| | - Jing Su
- Department of Geriatrics, Taian City Central Hospital, Taian, China
| | - Qiang Shi
- Department of Oncology, Second Affiliated Hospital of Shandong First Medical University, Shandong Academy of Medical Sciences, Taian, China
| | - Jinlei Wang
- Department of Internal Medicine, Taian Traffic Hospital, Taian, China
| | - Xiao Chen
- Department of Neurosurgery, Taian City Central Hospital, Taian, China
| | - Shizhong Zhang
- Department of Neurosurgery, Taian City Central Hospital, Taian, China
| | - Mengkao Li
- Department of Neurosurgery, Taian City Central Hospital, Taian, China
| | - Jie Cui
- Department of Oncology, Second Affiliated Hospital of Shandong First Medical University, Shandong Academy of Medical Sciences, Taian, China
| | - Cundong Fan
- Department of Oncology, Second Affiliated Hospital of Shandong First Medical University, Shandong Academy of Medical Sciences, Taian, China
| | - Beibei Sun
- Department of Oncology, Second Affiliated Hospital of Shandong First Medical University, Shandong Academy of Medical Sciences, Taian, China
| | - Guojun Wang
- Department of Neurosurgery, Taian City Central Hospital, Taian, China
| |
Collapse
|
26
|
Liang P, Mao L, Dong Y, Zhao Z, Sun Q, Mazhar M, Ma Y, Yang S, Ren W. Design and Application of Near-Infrared Nanomaterial-Liposome Hybrid Nanocarriers for Cancer Photothermal Therapy. Pharmaceutics 2021; 13:2070. [PMID: 34959351 PMCID: PMC8704010 DOI: 10.3390/pharmaceutics13122070] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/09/2021] [Accepted: 11/26/2021] [Indexed: 01/04/2023] Open
Abstract
Liposomes are attractive carriers for targeted and controlled drug delivery receiving increasing attention in cancer photothermal therapy. However, the field of creating near-infrared nanomaterial-liposome hybrid nanocarriers (NIRN-Lips) is relatively little understood. The hybrid nanocarriers combine the dual superiority of nanomaterials and liposomes, with more stable particles, enhanced photoluminescence, higher tumor permeability, better tumor-targeted drug delivery, stimulus-responsive drug release, and thus exhibiting better anti-tumor efficacy. Herein, this review covers the liposomes supported various types of near-infrared nanomaterials, including gold-based nanomaterials, carbon-based nanomaterials, and semiconductor quantum dots. Specifically, the NIRN-Lips are described in terms of their feature, synthesis, and drug-release mechanism. The design considerations of NIRN-Lips are highlighted. Further, we briefly introduced the photothermal conversion mechanism of NIRNs and the cell death mechanism induced by photothermal therapy. Subsequently, we provided a brief conclusion of NIRNs-Lips applied in cancer photothermal therapy. Finally, we discussed a synopsis of associated challenges and future perspectives for the applications of NIRN-Lips in cancer photothermal therapy.
Collapse
Affiliation(s)
- Pan Liang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Linshen Mao
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yanli Dong
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Zhenwen Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing Mass Spectrum Center, Beijing 100190, China;
| | - Qin Sun
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yining Ma
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (P.L.); (L.M.); (Y.D.); (Q.S.); (M.M.); (Y.M.)
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
27
|
Li X, Ai S, Lu X, Liu S, Guan W. Nanotechnology-based strategies for gastric cancer imaging and treatment. RSC Adv 2021; 11:35392-35407. [PMID: 35493171 PMCID: PMC9043273 DOI: 10.1039/d1ra01947c] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer is the second biggest cause of cancer-related deaths worldwide. Despite the improvement in deciphering molecular mechanisms, advances of detection and imaging, implementation of prevention programs, and personalized treatment, the overall curative rate remains low. In particular, with the emergence of nanomaterials, different imaging modalities can be integrated into one single platform, and combined therapies with synergetic effects against gastric cancer were established. Moreover, the development of theranostic strategies with simultaneous diagnostic and therapeutic ability was boosted by multifunctional nanoparticles. Herein, we present a comprehensive review of major nanotechnology-based breakthroughs for gastric cancer imaging and treatment. We will describe the superiority of nanomaterials used in gastric cancer and summarize nanotechnology applications for the improvement of cancer imaging and therapeutic efficacy.
Collapse
Affiliation(s)
- Xianghui Li
- Affiliated Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing 210008 China +86-25-68182222. ext. 60930, 60931, 60932
| | - Shichao Ai
- Affiliated Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing 210008 China +86-25-68182222. ext. 60930, 60931, 60932
| | - Xiaofeng Lu
- Affiliated Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing 210008 China +86-25-68182222. ext. 60930, 60931, 60932
| | - Song Liu
- Affiliated Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing 210008 China +86-25-68182222. ext. 60930, 60931, 60932
| | - Wenxian Guan
- Affiliated Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing 210008 China +86-25-68182222. ext. 60930, 60931, 60932
| |
Collapse
|
28
|
Diana R, Caruso U, Panunzi B. Stimuli-Responsive Zinc (II) Coordination Polymers: A Novel Platform for Supramolecular Chromic Smart Tools. Polymers (Basel) 2021; 13:3712. [PMID: 34771269 PMCID: PMC8588226 DOI: 10.3390/polym13213712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022] Open
Abstract
The unique role of the zinc (II) cation prompted us to cut a cross-section of the large and complex topic of the stimuli-responsive coordination polymers (CPs). Due to its flexible coordination environment and geometries, easiness of coordination-decoordination equilibria, "optically innocent" ability to "clip" the ligands in emissive architectures, non-toxicity and sustainability, the zinc (II) cation is a good candidate for building supramolecular smart tools. The review summarizes the recent achievements of zinc-based CPs as stimuli-responsive materials able to provide a chromic response. An overview of the past five years has been organised, encompassing 1, 2 and 3D responsive zinc-based CPs; specifically zinc-based metallorganic frameworks and zinc-based nanosized polymeric probes. The most relevant examples were collected following a consequential and progressive approach, referring to the structure-responsiveness relationship, the sensing mechanisms, the analytes and/or parameters detected. Finally, applications of highly bioengineered Zn-CPs for advanced imaging technique have been discussed.
Collapse
Affiliation(s)
- Rosita Diana
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy;
| | - Ugo Caruso
- Department of Chemical Science, University of Naples Federico II, 80126 Napoli, Italy;
| | - Barbara Panunzi
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy;
| |
Collapse
|
29
|
Kayani Z, Islami N, Behzadpour N, Zahraie N, Imanlou S, Tamaddon P, Salehi F, Daneshvar F, Perota G, Sorati E, Mohammadi S, Sattarahmady N. Combating cancer by utilizing noble metallic nanostructures in combination with laser photothermal and X-ray radiotherapy. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Zhang S, Ren H, Sun H, Cao S. Dieckol exerts anticancer activity in human osteosarcoma (MG-63) cells through the inhibition of PI3K/AKT/mTOR signaling pathway. Saudi J Biol Sci 2021; 28:4908-4915. [PMID: 34466065 PMCID: PMC8381078 DOI: 10.1016/j.sjbs.2021.07.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/06/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common malignant bone cancer with more metastasis and increased occurrence in children and teen-agers and being responsible for more number of morbidity and mortality worldwide. Objective The current exploration was planned study the in vitro anticancer actions of dieckol against human OS MG-63 cells via PI3K/AKT/mTOR signaling inhibition. Methodology The cytotoxicity of dieckol was scrutinized by MTT assay. Effects of dieckol on the ROS accumulation, apoptotic cell death, and MMP level in the MG-63 cells were studied by respective fluorescence staining assays. The levels of proliferative, inflammatory, and apoptotic markers in the dieckol treated MG-63 cells were scrutinized by marker specific kits. The expressions of PI3K, AKT, and mTOR was assayed by RT-PCR. Results The MTT assay revealed that the dieckol dose dependently prevented MG-63 cells viability and the IC50 was found at 15 µM. Dieckol treatment effectively reduced the MMP level and improved the ROS generation and apoptosis in MG-63 cells. Dieckol also regulated the proliferative (cyclin D1), inflammatory (COX-2, IL-6, TNF-α, and NF-κB), and apoptotic (caspase-3, Bax, Bcl-2) markers in the MG-63 cells. The PI3K/AKT/mTOR signaling in the MG-63 cells were effectively inhibited by the dieckol treatment. Conclusion In conclusion, our findings from this study recommends that the dieckol could be a talented anticancer candidate for the OS management in the future.
Collapse
Affiliation(s)
- Shouqiang Zhang
- Department of Orthopaedic & Trauma Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, Shandong 250033, China
| | - Hui Ren
- Department of Cardiothoracic Surgery, Xinwen Mining Group Central Hospital, Xintai City, Shandong Province 271200, China
| | - Hanting Sun
- Department of Orthopaedic Surgery, ZouPing Hospital of TCM, ZouPing City, Shandong Province 256200, China
| | - Songhua Cao
- Department of Hand Surgery/Foot & Ankle Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, Shandong 250033, China
| |
Collapse
|
31
|
Zhu L, Kuang Z, Song P, Li W, Gui L, Yang K, Ge F, Tao Y, Zhang W. Gold nanorod-loaded thermosensitive liposomes facilitate the targeted release of ruthenium(II) polypyridyl complexes with anti-tumor activity. NANOTECHNOLOGY 2021; 32:455103. [PMID: 34352731 DOI: 10.1088/1361-6528/ac1afc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Ruthenium(II) polypyridyl complexes (Ru) show high anti-tumor activity, but their poor solubility and low biocompatibility impede their use in anti-tumor therapy. Here,we circumvented the problem of low solubility by encapsulating the Ru in thermosensitive liposomes (LTSLs) and used gold nanorods (Au NRs) modified on the surface of the liposomes to permit the precise release of Ru at the tumor site. A facile and simple method was developed to synthesize Ru-loaded Au NR-decorated LTSL (Au@LTSL-Ru NPs). The loaded Au NRs improved the anti-tumor effect of Ru and enhanced the photothermal therapeutic properties of the nanosystem. A characterization experiment indicated that the average particle size of Au@LTSL-Ru was approximately 300 nm and that the Au NRs were successfully modified on the surface of LTSL. In thein vitroanti-tumor test, Au@LTSL-Ru and NIR significantly inhibited the proliferation of SGC-7901 cells. The IC50value of Au@LTSL-Ru + NIR was 7.1 ± 1.2μM (13μg ml-1), and the inhibition rate was greater than 90% when the concentration reached 30μg ml-1.In vivostudies revealed that Au@LTSL-Ru and NIR had a significant inhibitory effect on subcutaneous tumor tissues derived from SGC-7901 cells. Analysis of histopathology and immunocytotoxicity indicated that Au@LTSL-Ru has fewer side effects and high biocompatibility. Our results confirm that Au@LTSL-Ru can effectively inhibit tumor growth and aid the development of Ru for use in the thermal response in anti-tumor activity research.
Collapse
Affiliation(s)
- Longbao Zhu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Zhao Kuang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Ping Song
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Wanzhen Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Lin Gui
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, Peoples Republic of China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, People's Republic of China
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| |
Collapse
|
32
|
Sun JY, Hou YJ, Cui HJ, Zhang C, Yang MF, Wang FZ, Sun Z, Fan CD, Sun BL, Oh JR. VS-5584 Inhibits Human Osteosarcoma Cells Growth by Induction of G1- phase Arrest through Regulating PI3K/mTOR and MAPK Pathways. Curr Cancer Drug Targets 2021; 20:616-623. [PMID: 32286946 DOI: 10.2174/1568009620666200414150353] [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: 12/05/2019] [Revised: 02/08/2020] [Accepted: 03/03/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Activation of the PI3K/mTOR signaling pathway plays a key role in the progression of human osteosarcoma. Studies have confirmed that VS-5584 was a novel inhibitor of the PI3K/mTOR pathway, and displayed potential anticancer activity. OBJECTIVE To explore the anticancer effect and underlying mechanism of VS-5584 against the growth of human osteosarcoma cells. METHODS U2OS and MG-63 human osteosarcoma cells were cultured and the cytotoxicity, cell apoptosis in VS-5584-treated cells were explored by the CCK8 assay, flow cytometric analysis and western blot. Cell migration and tube formation were also employed to examine the anticancer potential. RESULTS The results showed that VS-5584 treatment dose-dependently inhibited the growth of U2OS and MG-63 cells by induction of G1-phase arrest through regulating p21, p27, Cyclin B1 and Cdc2. Further investigation revealed that VS-5584 treatment effectively inhibited the PI3K/mTOR signaling pathway and triggered MAPK phosphorylation. Moreover, VS-5584 treatment dramatically suppressed cell migration and tube formation of HUVECs, followed by the down-regulation of HIF-1α and VEGF. CONCLUSION Our findings validated that VS-5584 may be a promising anticancer agent with potential application in the chemotherapy and chemoprevention of human osteosarcoma.
Collapse
Affiliation(s)
- Jing-Yi Sun
- Department of Orthopedics, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon, 26426, Korea
| | - Ya-Jun Hou
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Hai-Juan Cui
- Department of Nursing, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, China
| | - Cheng Zhang
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Ming-Feng Yang
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Feng-Ze Wang
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Zheng Sun
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Cun-Dong Fan
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Bao-Liang Sun
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Jin Rok Oh
- Department of Orthopedics, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon, 26426, Korea
| |
Collapse
|
33
|
Mishra SK, Hole A, Reddy BPK, Srivastava R, Chilakapati MK, De A. Raman micro-spectroscopic map estimating in vivo precision of tumor ablative effect achieved by photothermal therapy procedure. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102437. [PMID: 34273597 DOI: 10.1016/j.nano.2021.102437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/04/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022]
Abstract
Photothermal-therapy (PTT) inculcates near-infrared laser guided local heating effect, where high degree of precision is expected, but not well proven to-date. An ex vivo tissue biochemical map with molecular/biochemical response showing the coverage area out of an optimized PTT procedure can reveal precision information. In this work, Raman-microscopic mapping and linear discriminant analysis of spectra of PTT treated and surrounding tissue areas ex vivo are done, revealing three distinct spectral clusters/zones, with minimal overlap between the core treated and adjacent untreated zone. The core treated zone showed intense nucleic-acid, cytochrome/mitochondria and protein damage, an adjacent zone showed lesser degree of damages and far zone showed minimal/no damage. Immunohistochemistry for γH2AX (DNA damage marker protein) in PTT exposed tissue also revealed similar results. Altogether, this study reveals the utility of Raman-microspectroscopy for fine-tuning safety parameters and precision that can be achieved from PTT mediated tumor ablation in preclinical/clinical application.
Collapse
Affiliation(s)
- Sumit K Mishra
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India.
| | - Arti Hole
- Chilakapati Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India.
| | - B Pradeep K Reddy
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | - Rohit Srivastava
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | - Murali Krishna Chilakapati
- Chilakapati Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India.
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India.
| |
Collapse
|
34
|
|
35
|
Diana R, Panunzi B. Zinc (II) and AIEgens: The "Clip Approach" for a Novel Fluorophore Family. A Review. Molecules 2021; 26:4176. [PMID: 34299451 PMCID: PMC8304007 DOI: 10.3390/molecules26144176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022] Open
Abstract
Aggregation-induced emission (AIE) compounds display a photophysical phenomenon in which the aggregate state exhibits stronger emission than the isolated units. The common term of "AIEgens" was coined to describe compounds undergoing the AIE effect. Due to the recent interest in AIEgens, the search for novel hybrid organic-inorganic compounds with unique luminescence properties in the aggregate phase is a relevant goal. In this perspective, the abundant, inexpensive, and nontoxic d10 zinc cation offers unique opportunities for building AIE active fluorophores, sensing probes, and bioimaging tools. Considering the novelty of the topic, relevant examples collected in the last 5 years (2016-2021) through scientific production can be considered fully representative of the state-of-the-art. Starting from the simple phenomenological approach and considering different typological and chemical units and structures, we focused on zinc-based AIEgens offering synthetic novelty, research completeness, and relevant applications. A special section was devoted to Zn(II)-based AIEgens for living cell imaging as the novel technological frontier in biology and medicine.
Collapse
Affiliation(s)
| | - Barbara Panunzi
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy;
| |
Collapse
|
36
|
Sun Y, Wang Z, Zhang P, Wang J, Chen Y, Yin C, Wang W, Fan C, Sun D. Mesoporous silica integrated with Fe 3O 4 and palmitoyl ascorbate as a new nano-Fenton reactor for amplified tumor oxidation therapy. Biomater Sci 2021; 8:7154-7165. [PMID: 33155581 DOI: 10.1039/d0bm01738h] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Co-delivery of H2O2-generating agent and catalyst via a nano-Fenton reactor to the tumor acidic microenvironment for amplified tumor oxidation therapy has been widely studied. However, high side effects and low efficiency remain the limitations of the design and development of this process. Herein, a new nano-Fenton reactor in which mesoporous silica is integrated with Fe3O4 and palmitoyl ascorbate (Fe3O4@SiO2-PA) was designed, with the product exhibiting good dispersion, stability, uniformity and consistent spectral characteristics. The results show that Fe3O4@mSiO2-PA successfully enters cancer cells, significantly inhibits HeLa cells and 3D tumor spheroid growth in vitro via the induction of apoptosis. Meanwhile, Fe3O4@mSiO2-PA administration in vivo markedly suppresses HeLa tumor xenografts growth via the induction of apoptosis, followed by caspase-3 activation and cytochrome C release. Further investigation revealed that Fe3O4@mSiO2-PA causes enhanced production of reactive oxygen species (ROS), which subsequently triggers DNA damage and causes dysfunction of the MAPK and PI3K/AKT pathways. Importantly, Fe3O4@mSiO2-PA shows few side effects and good biocompatibility in vivo. Taken together, these results suggest that Fe3O4@mSiO2-PA inhibits HeLa cell growth in vitro and in vivo by triggering enhanced oxidative damage and regulating multiple signal pathways. Our findings validate the rational design that mesoporous silica integrated with Fe3O4 and palmitoyl ascorbate can act as a new nano-Fenton reactor for amplified tumor oxidation therapy.
Collapse
Affiliation(s)
- Yu Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cannabinoid WIN 55,212-2 Inhibits Human Glioma Cell Growth by Triggering ROS-Mediated Signal Pathways. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6612592. [PMID: 33977107 PMCID: PMC8087470 DOI: 10.1155/2021/6612592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 11/17/2022]
Abstract
Glioblastoma is a highly invasive primary malignant tumor of the central nervous system. Cannabinoid analogue WIN 55,212-2 (WIN) exhibited a novel anticancer effect against human tumors. However, the anticancer potential and underlying mechanism of WIN against human glioma remain unclear. Herein, the anticancer efficiency and mechanism of WIN in U251 human glioma cells were investigated. The results showed that WIN dose-dependently inhibited U251 cell proliferation, migration, and invasion in vitro. WIN treatment also effectively suppressed U251 tumor spheroids growth ex vivo. Further studies found that WIN induced significant apoptosis as convinced by the caspase-3 activation and release of cytochrome C. Mechanism investigation revealed that WIN triggered ROS-mediated DNA damage and caused dysfunction of VEGF-AKT/FAK signal axis. However, ROS inhibition effectively attenuated WIN-induced DNA damage and dysfunction of VEGF-AKT/FAK signal axis and eventually improved U251 cell proliferation, migration, and invasion. Taken together, our findings validated that WIN had the potential to inhibit U251 cell proliferation, migration, and invasion and induce apoptosis by triggering ROS-dependent DNA damage and dysfunction of VEGF-AKT/FAK signal axis.
Collapse
|
38
|
Sun D, Wang Z, Zhang P, Yin C, Wang J, Sun Y, Chen Y, Wang W, Sun B, Fan C. Ruthenium-loaded mesoporous silica as tumor microenvironment-response nano-fenton reactors for precise cancer therapy. J Nanobiotechnology 2021; 19:98. [PMID: 33827604 PMCID: PMC8028739 DOI: 10.1186/s12951-021-00848-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/31/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Nano-Fenton reactors as novel strategy to selectively convert hydrogen peroxide (H2O2) into active hydroxyl radicals in tumor microenvironment for cancer therapy had attracted much attention. However, side effects and low efficiency remain the main drawbacks for cancer precise therapy. RESULTS Here, ruthenium-loaded palmitoyl ascorbate (PA)-modified mesoporous silica (Ru@SiO2-PA) was successfully fabricated and characterized. The results indicated that Ru@SiO2-PA under pH6.0 environment displayed enhanced growth inhibition against human cancer cells than that of pH7.4, which indicated the super selectivity between cancer cells and normal cells. Ru@SiO2-PA also induced enhanced cancer cells apoptosis, followed by caspase-3 activation and cytochrome-c release. Mechanism investigation revealed that Ru@SiO2-PA caused enhanced generation of superoxide anion, which subsequently triggered DNA damage and dysfunction of MAPKs and PI3K/AKT pathways. Moreover, Ru@SiO2-PA effectively inhibited tumor spheroids and tumor xenografts growth in vivo by induction of apoptosis. The real-time imaging by monitoring Ru fluorescence in vitro and in vivo revealed that Ru@SiO2-PA mainly accumulated in cell nucleus and tumor xenografts. Importantly, Ru@SiO2-PA showed no side effects in vivo, predicting the safety and potential application in clinic. CONCLUSIONS Our findings validated the rational design that Ru@SiO2-PA can act as novel tumor microenvironment-response nano-Fenton reactors for cancer precise therapy.
Collapse
Affiliation(s)
- Dongdong Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Zekun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Pu Zhang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, 271000, Shandong, China
| | - Chenyang Yin
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Jingyuan Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Yu Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Ying Chen
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Weiyun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Baoliang Sun
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Cundong Fan
- Department of Neurology, Second Affiliated Hospital; Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| |
Collapse
|
39
|
Zhang Y, Tan J, Zhou L, Shan X, Liu J, Ma Y. Synthesis and Application of AS1411-Functionalized Gold Nanoparticles for Targeted Therapy of Gastric Cancer. ACS OMEGA 2020; 5:31227-31233. [PMID: 33324832 PMCID: PMC7726946 DOI: 10.1021/acsomega.0c04605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/10/2020] [Indexed: 05/09/2023]
Abstract
Gastric cancer therapy is still a big challenge, and nanomedicines bring much more hope. It is essential to develop multifunctional nanoparticles, especially those with high targeted capacity and antitumor effects, to improve gastric cancer therapy. In this study, we constructed AS1411 aptamer-based gold nanoparticles with appropriate size facilitating endocytosis and actively targeted drug delivery for gastric cancer cells via the specific AS1411-nucleolin interaction. The AS1411-based nanoparticles showed obviously increased targeted capacity towards AGS cells compared to random ssDNA-based nanoparticles. Meanwhile, compared to L929 cells, the AS1411-based nanoparticles showed selective drug uptake and delivery for AGS cells. Importantly, the AS1411-based nanoparticles exhibited significantly stronger antitumor effects on AGS cells under laser irradiation compared to chemotherapy alone. Our nanoparticles combined targeted drug delivery and efficient antitumor effects within one single nanoplatform, which are promising to be applied as targeted nanomedicines against gastric cancer.
Collapse
Affiliation(s)
- Yajie Zhang
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
- Department
of Gastroenterology, Shengjing Hospital
of China Medical University, Shenyang 110004, China
| | - Jingwei Tan
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Lu Zhou
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Xiaoqing Shan
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Jianling Liu
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| | - Yong Ma
- Department
of Chemistry, School of Fundamental Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
40
|
Gonçalves ASC, Rodrigues CF, Moreira AF, Correia IJ. Strategies to improve the photothermal capacity of gold-based nanomedicines. Acta Biomater 2020; 116:105-137. [PMID: 32911109 DOI: 10.1016/j.actbio.2020.09.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022]
Abstract
The plasmonic photothermal properties of gold nanoparticles have been widely explored in the biomedical field to mediate a photothermal effect in response to the irradiation with an external light source. Particularly, in cancer therapy, the physicochemical properties of gold-based nanomaterials allow them to efficiently accumulate in the tumor tissue and then mediate the light-triggered thermal destruction of cancer cells with high spatial-temporal control. Nevertheless, the gold nanomaterials can be produced with different shapes, sizes, and organizations such as nanospheres, nanorods, nanocages, nanoshells, and nanoclusters. These gold nanostructures will present different plasmonic photothermal properties that can impact cancer thermal ablation. This review analyses the application of gold-based nanomaterials in cancer photothermal therapy, emphasizing the main parameters that affect its light-to-heat conversion efficiency and consequently the photothermal potential. The different shapes/organizations (clusters, shells, rods, stars, cages) of gold nanomaterials and the parameters that can be fine-tuned to improve the photothermal capacity are presented. Moreover, the gold nanostructures combination with other materials (e.g. silica, graphene, and iron oxide) or small molecules (e.g. indocyanine green and IR780) to improve the nanomaterials photothermal capacity is also overviewed.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Carolina F Rodrigues
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - André F Moreira
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Ilídio J Correia
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CIEPQF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, 3030-790 Coimbra, Portugal.
| |
Collapse
|
41
|
Lin C, Zhang LJ, Li B, Zhang F, Shen QR, Kong GQ, Wang XF, Cui SH, Dai R, Cao WQ, Zhang P. Selenium-Containing Protein From Selenium-Enriched Spirulina platensis Attenuates High Glucose-Induced Calcification of MOVAS Cells by Inhibiting ROS-Mediated DNA Damage and Regulating MAPK and PI3K/AKT Pathways. Front Physiol 2020; 11:791. [PMID: 32733280 PMCID: PMC7363841 DOI: 10.3389/fphys.2020.00791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/15/2020] [Indexed: 01/29/2023] Open
Abstract
Hyperglycemia is the main feature of diabetes and may increase the risk of vascular calcification (VC), which is an independent predictor for cardiovascular and cerebrovascular diseases (CCD). Selenium (Se) may decrease the risk of CCD, and previous studies confirmed that Se-containing protein from Se-enriched Spirulina platensis (Se-SP) exhibited novel antioxidant potential. However, the effect of Se-SP against VC has been not investigated. Herein, the protective effect and underlying mechanism of Se-SP against high glucose-induced calcification in mouse aortic vascular smooth muscle cells (MOVAS) were explored. Inductively coupled plasma atomic emission spectroscopy (ICP-AES) results showed time-dependent uptake of Se-SP in MOVAS cells, which significantly inhibited high glucose-induced abnormal proliferation. Se-SP co-treatment also effectively attenuated high glucose-induced calcification of MOVAS cells, followed by decreased activity and expression of alkaline phosphatase (ALP). Further investigation revealed that Se-SP markedly prevented reactive oxygen species (ROS)-mediated DNA damage in glucose-treated MOVAS cells. ROS inhibition by glutathione (GSH) effectively inhibited high glucose-induced calcification, indicating that Se-SP could act as ROS inhibitor to inhibit high glucose-induced DNA damage and calcification. Moreover, Se-SP dramatically attenuated high glucose-induced dysfunction of mitogen-activated protein kinases (MAPKs) and phosphatidylinositol-3-kinase/AKT (PI3K/AKT) pathways. Se-SP after Se addition achieved enhanced potential in inhibiting high glucose-induced calcification, which validated that Se-SP as a new Se species could be a highly effective treatment for human CCD.
Collapse
Affiliation(s)
- Cong Lin
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Jun Zhang
- Department of Neurology, People's Hospital of Linyi Affiliated to Qingdao University, Linyi, China
| | - Bo Li
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Feng Zhang
- Physical Examination Center, Taian City Central Hospital, Taian, China
| | - Qing-Rong Shen
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Guo-Qing Kong
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Xiao-Fan Wang
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Shou-Hong Cui
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Rong Dai
- Department of Emergency, Taian City Central Hospital, Taian, China
| | - Wen-Qiang Cao
- Department of Biotechnology, Zhuhai Hopegenes Medical and Phamaceutical Institute, Zhuhai, China
| | - Pu Zhang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, China
| |
Collapse
|
42
|
Zhang P, Wang AP, Yang HP, Ai L, Zhang HJ, Wang YM, Bi YL, Fan HH, Gao J, Zhang HY, Liu JZ. Apelin-13 attenuates high glucose-induced calcification of MOVAS cells by regulating MAPKs and PI3K/AKT pathways and ROS-mediated signals. Biomed Pharmacother 2020; 128:110271. [PMID: 32450527 DOI: 10.1016/j.biopha.2020.110271] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 12/29/2022] Open
Abstract
Vascular calcification (VC) is an inducement of many cardiovascular diseases. Clinic evidences have confirmed that diabetes was the independent risk factor for VC, and the mechanism has not been well explored. Apelin as a ligand molecule is widely found in the cardiovascular system and showed potential in inhibiting VC, but the inhibitory effect and mechanism of apelin-13 against high glucose-induced VC have not been investigated yet. Herein, apelin-13 was employed to inhibit high glucose-induced VC in mouse aortic vascular smooth muscle cells (MOVAS), and the underlying mechanism was explored. The results showed that apelin-13 significantly inhibited high glucose-induced cells proliferation, migration and invasion of MOVAS cells. Apelin-13 also effectively attenuated high glucose-induced calcification by inhibiting alkaline phosphatase (ALP) activity and expression. Further investigation revealed that apelin-13 dramatically suppressed high glucose-induced DNA damage through inhibiting reactive oxide species (ROS) generation. Moreover, apelin-13 also effectively improved high glucose-induced dysfunction of MAPKs and PI3K/AKT. Inhibition of ERK by inhibitor (U0126) significantly blocked high glucose-induced calcification, which further confirmed the significance of MAPKs. Taken together, these results suggested that apelin-13 had the potential to attenuate high glucose-induced calcification of MOVAS cells by inhibiting ROS-mediated DNA damage and regulating MAPKs and PI3K/AKT pathways. Our findings validated the strategy of using apelin-13 maybe a novel way in treating high glucose-mediated VC.
Collapse
MESH Headings
- Alkaline Phosphatase/antagonists & inhibitors
- Alkaline Phosphatase/metabolism
- Animals
- Aorta/drug effects
- Aorta/enzymology
- Aorta/pathology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- DNA Damage/drug effects
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Glucose/toxicity
- Intercellular Signaling Peptides and Proteins/pharmacology
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Oxidative Stress/drug effects
- Phosphatidylinositol 3-Kinase/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Vascular Calcification/enzymology
- Vascular Calcification/pathology
- Vascular Calcification/prevention & control
Collapse
Affiliation(s)
- Pu Zhang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Ai-Ping Wang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Hong-Peng Yang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Lei Ai
- Department of Clinical Laboratory, Taishan Coal Sanitarium of Shandong, Taian, Shandong, 271000, China
| | - Hong-Jun Zhang
- Department of Anesthesiology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, 471003, China
| | - Yong-Mei Wang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Yan-Ling Bi
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Huai-Hai Fan
- Department of Intensive Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Jing Gao
- Department of Stomatology, Taian City Central Hospital, Taian, Shandong, 271000, China.
| | - Huan-Yi Zhang
- Department of Cardiovascular Medicine, Taian City Central Hospital, Taian, Shandong, 271000, China.
| | - Jian-Zhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong 271018, China.
| |
Collapse
|
43
|
CCT128930 induces G1-phase arrest and apoptosis and synergistically enhances the anticancer efficiency of VS5584 in human osteosarcoma cells. Biomed Pharmacother 2020; 130:110544. [PMID: 32721630 DOI: 10.1016/j.biopha.2020.110544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/07/2020] [Accepted: 07/20/2020] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma is a highly invasive primary malignant bone tumor. PI3K/mTOR pathway plays a key role in tumor progression, and inhibition of PI3K/mTOR pathway represents a novel strategy in therapy of osteosarcoma. CCT128930 and VS5584 are both inhibitors of PI3K/mTOR, but the anticancer mechanism of CCT128930 or/and VS5584 against human osteosarcoma cells remains unclear. Herein, U2OS and MG63 human osteosarcoma cells were cultured, and the anticancer effects of CCT128930 alone and the combined effect of CCT128930 and VS5584 against human osteosarcoma cells were explored. The results showed that CCT128930 as PI3K/mTOR inhibitor effectively inhibited p-p70 and p-AKT expression and dose-dependently inhibited U2OS cells and MG63 human osteosarcoma cells growth. Further studies found that CCT128930 triggered significant G-1 phase arrest and apoptosis, as convinced by the dysfunction of p27, Cyclin B1, Cyclin D1 and Cdc2, and PARP cleavage and caspase-3 activation. Moreover, CCT128930 treatment obviously enhanced VS5584-induced growth inhibition and apoptosis in human osteosarcoma cells, followed by enhanced PARP cleavage and caspase-3 activation. Taken together, CCT128930 alone or combined treatment with CCT128930 and VS5584 both effectively inhibited human osteosarcoma cells growth by induction of G1-phase arrest and apoptosis through regulating PI3K/mTOR and MAPKs pathways.
Collapse
|
44
|
Wang K, Xiang Y, Pan W, Wang H, Li N, Tang B. Dual-targeted photothermal agents for enhanced cancer therapy. Chem Sci 2020; 11:8055-8072. [PMID: 34123080 PMCID: PMC8163445 DOI: 10.1039/d0sc03173a] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Photothermal therapy, in which light is converted into heat and triggers local hyperthermia to ablate tumors, presents an inherently specific and noninvasive treatment for tumor tissues. In this area, the development of efficient photothermal agents (PTAs) has always been a central topic. Although many efforts have been made on the investigation of novel molecular architectures and photothermal materials over the past decades, PTAs can cause severe damage to normal tissues because of the poor tumor aggregate ability and high irradiation density. Recently, dual-targeted photothermal agents (DTPTAs) provide an attractive strategy to overcome these problems and enhance cancer therapy. DTPTAs are functionalized with two classes of targeting units, including tumor environment targeting sites, tumor targeting sites and organelle targeting sites. In this perspective, typical targeted ligands and representative examples of photothermal therapeutic agents with dual-targeted properties are systematically summarized and recent advances using DTPTAs in tumor therapy are highlighted.
Collapse
Affiliation(s)
- Kaiye Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Yanan Xiang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Hongyu Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
45
|
Yazdian-Robati R, Bayat P, Oroojalian F, Zargari M, Ramezani M, Taghdisi SM, Abnous K. Therapeutic applications of AS1411 aptamer, an update review. Int J Biol Macromol 2020; 155:1420-1431. [PMID: 31734366 DOI: 10.1016/j.ijbiomac.2019.11.118] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
Nucleolin or C23, is one of the most abundant non-ribosomal phosphoproteins of nucleolus. However, in several cancers, nucleolin is highly expressed both intracellularly and on the cell surface. So, it is considered as a potential target for the diagnosis and cancer therapy. Targeting nucleolin by compounds such as AS1411 aptamer can reduce tumor cell growth. In this regard, interest has increased in nucleolin as a molecular target for overcoming cancer therapy challenges. This review paper addressed recent progresses in nucleolin targeting by the G-rich AS1411 aptamer in the field of cancer therapy mainly over the past three years.
Collapse
Affiliation(s)
- Rezvan Yazdian-Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Payam Bayat
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehryar Zargari
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
46
|
Zhang Q, Miao Y, Fu Q, Hu H, Chen H, Zeng A, Jin Y, Jiang Y, Qian L, Wu L, Xu L, Wang G, Qiu L, Huang X, Xia Y. CircRNACCDC66 regulates cisplatin resistance in gastric cancer via the miR-618/BCL2 axis. Biochem Biophys Res Commun 2020; 526:713-720. [PMID: 32253030 DOI: 10.1016/j.bbrc.2020.03.156] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/26/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Qiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China; Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Yongchang Miao
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China; Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qingsheng Fu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hao Hu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hao Chen
- Department of Radiotherapy, The Second People's Hospital of Wuhu, Wuhu, Anhui, China
| | - Ailiang Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Jin
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Yangfan Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Long Qian
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Longchao Wu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Li Xu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Gang Wang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Lei Qiu
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China.
| | - Yabin Xia
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
47
|
Gao B, Xie W, Wu X, Wang L, Guo J. Functionally analyzing the important roles of hepatocyte nuclear factor 3 (FoxA) in tumorigenesis. Biochim Biophys Acta Rev Cancer 2020; 1873:188365. [PMID: 32325165 DOI: 10.1016/j.bbcan.2020.188365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022]
Abstract
Transcriptional factors (TFs) play a central role in governing gene expression under physiological conditions including the processes of embryonic development, metabolic homeostasis and response to extracellular stimuli. Conceivably, the aberrant dysregulations of TFs would dominantly result in various human disorders including tumorigenesis, diabetes and neurodegenerative diseases. Serving as the most evolutionarily reserved TFs, Fox family TFs have been explored to exert distinct biological functions in neoplastic development, by manipulating diverse gene expression. Recently, among the Fox family members, the pilot roles of FoxAs attract more attention due to their functions as both pioneer factor and transcriptional factor in human tumorigenesis, particularly in the sex-dimorphism tumors. Therefore, the pathological roles of FoxAs in tumorigenesis have been well-explored in modulating inflammation, immune response and metabolic homeostasis. In this review, we comprehensively summarize the impressive progression of FoxA functional annotation, clinical relevance, upstream regulators and downstream effectors, as well as valuable animal models, and highlight the potential strategies to target FoxAs for cancer therapies.
Collapse
Affiliation(s)
- Bing Gao
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Wei Xie
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xueji Wu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lei Wang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
48
|
Sharma G, Jagtap JM, Parchur AK, Gogineni VR, Ran S, Bergom C, White SB, Flister MJ, Joshi A. Heritable modifiers of the tumor microenvironment influence nanoparticle uptake, distribution and response to photothermal therapy. Theranostics 2020; 10:5368-5383. [PMID: 32373218 PMCID: PMC7196309 DOI: 10.7150/thno.41171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
We report the impact of notch-DLL4-based hereditary vascular heterogeneities on the enhanced permeation and retention (EPR) effect and plasmonic photothermal therapy response in tumors. Methods: We generated two consomic rat strains with differing DLL4 expression on 3rd chromosome. These strains were based on immunocompromised Salt-sensitive or SSIL2Rγ- (DLL4-high) and SS.BN3IL2Rγ- (DLL4-low) rats with 3rd chromosome substituted from Brown Norway rat. We further constructed three novel SS.BN3IL2Rγ- congenic strains by introgressing varying segments of BN chromosome 3 into the parental SSIL2Rγ- strain to localize the role of SSIL2Rγ- DLL4 on tumor EPR effect with precision. We synthesized multimodal theranostic nanoparticles (TNPs) based on Au-nanorods which provide magnetic resonance imaging (MRI), X-ray, and optical contrasts to assess image guided PTT response and quantify host specific therapy response differences in tumors orthotopically xenografted in DLL4-high and -low strains. We tested recovery of therapy sensitivity of PTT resistant strains by employing anti-DLL4 conjugated TNPs in two triple negative breast cancer tumor xenografts. Results: Host strains with high DLL4 allele demonstrated slightly increased tumor nanoparticle uptake but consistently developed photothermal therapy resistance compared to tumors in host strains with low DLL4 allele. Tumor micro-environment with low DLL4 expression altered the geographic distribution of nanoparticles towards closer proximity with vasculature which improved efficacy of PTT in spite of lower overall TNP uptake. Targeting TNPs to tumor endothelium via anti-DLL4 antibody conjugation improved therapy sensitivity in high DLL4 allele hosts for two triple negative human breast cancer xenografts. Conclusions: Inherited DLL4 expression modulates EPR effects in tumors, and molecular targeting of endothelial DLL4 via nanoparticles is an effective personalized nanomedicine strategy.
Collapse
Affiliation(s)
- Gayatri Sharma
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jaidip M. Jagtap
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Abdul K. Parchur
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Sophia Ran
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sarah B. White
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael J. Flister
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amit Joshi
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
49
|
Liu Y, Li Z, Yin Z, Zhang H, Gao Y, Huo G, Wu A, Zeng L. Amplified Photoacoustic Signal and Enhanced Photothermal Conversion of Polydopamine-Coated Gold Nanobipyramids for Phototheranostics and Synergistic Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:14866-14875. [PMID: 32153178 DOI: 10.1021/acsami.9b22979] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Light-responsive nanoprobes were suffering from the threat of high-dose laser irradiation, and it was important for constructing new nanoprobes for safe and efficient phototheranostics. Here, polydopamine (PDA)-coated gold nanobipyramids (AuNBPs@PDA) were synthesized for amplified photoacoustic (PA) signal and enhanced photothermal conversion with low-dose laser irradiation and then doxorubicin (DOX)-loaded AuNBPs@PDA-DOX nanoprobes were constructed for PA imaging-guided synergistic photothermal therapy (PTT) and chemotherapy. The AuNBPs@PDA nanoparticles possessed higher photothermal conversion efficiency (42.07%) and stronger PA signal than those of AuNBP nanoparticles, and the AuNBPs@PDA-DOX nanoprobes showed dual-responsive DOX release of pH and photothermal stimulation. With low-dose laser irradiation (1.0 W/cm2) and low-concentration AuNBPs@PDA-DOX (60 μg/mL), the 4T1 cell viability was reduced to about 5%, owing to the combination of PTT and chemotherapy, compared with 42.3% of single chemotherapy and 25.3% of single PTT. Moreover, by modeling 4T1 tumor-bearing nude mice, in vivo PA imaging was achieved and the tumors were completely inhibited, demonstrating the excellent synergistic effect of PTT/chemotherapy. Therefore, the developed AuNBPs@PDA-DOX nanoprobes can be used for phototheranostics and synergistic chemotherapy, achieving low-dose laser irradiation and high-efficient visualized theranostics.
Collapse
Affiliation(s)
- Yanhong Liu
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| | - Ziwei Li
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| | - Zhibin Yin
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| | - Hongxin Zhang
- Medical College, Hebei University, Baoding 071002, P. R. China
| | - Yang Gao
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Guoyan Huo
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Leyong Zeng
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| |
Collapse
|
50
|
Moros M, Lewinska A, Merola F, Ferraro P, Wnuk M, Tino A, Tortiglione C. Gold Nanorods and Nanoprisms Mediate Different Photothermal Cell Death Mechanisms In Vitro and In Vivo. ACS APPLIED MATERIALS & INTERFACES 2020; 12:13718-13730. [PMID: 32134240 DOI: 10.1021/acsami.0c02022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Photothermal therapy (PTT) is an efficient method of inducing localized hyperthermia and can be achieved using gold nanoparticles as photothermal agents. However, there are many hurdles to get over before this therapy can safely reach the clinics, including nanoparticles' optimal shape and the accurate prediction of cellular responses. Here, we describe the synthesis of gold nanorods and nanoprisms with similar surface plasmon resonances in the near-infrared (NIR) and comparable photothermal conversion efficiencies and characterize the response to NIR irradiation in two biological systems, melanoma cells and the small invertebrate Hydra vulgaris. By integrating animal, cellular, and molecular biology approaches, we show a diverse outcome of nanorods and nanoprisms on the two systems, sustained by the elicitation of different pathways, from necrosis to programmed cell death mechanisms (apoptosis and necroptosis). The comparative multilevel analysis shows great accuracy of in vivo invertebrate models to predict overall responses to photothermal challenging and superior photothermal performance of nanoprisms. Understanding the molecular pathways of these responses may help develop optimized nanoheaters that, safe by design, may improve PTT efficacy for clinical purposes.
Collapse
Affiliation(s)
- Maria Moros
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Anna Lewinska
- Department of Cell Biochemistry, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Francesco Merola
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Pietro Ferraro
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Maciej Wnuk
- Department of Genetics, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Angela Tino
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Claudia Tortiglione
- Istituto di Scienze Applicate e Sistemi Intelligenti "Eduardo Caianiello", Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| |
Collapse
|