1. Self-delivery of a metal-coordinated anti-angiogenic nanodrug with GSH depleting ability for synergistic chemo-phototherapy.
期刊:Biomaterials science
日期:2023-10-24
DOI :10.1039/d3bm00994g
Synergistic chemo-phototherapy has offered tremendous potential in cancer treatment. Nevertheless, nanosystems usually suffer from the complexity of multicomponents (polymeric or inorganic materials), which results in carrier-related toxicity issues. Moreover, the GSH over-expression of tumor cells seriously compromises ROS therapeutic efficiency. Herein, we designed a self-delivered nanodrug Cu(II) coordination-driven co-self-assembly of celastrol (CST, a chemo-drug with anti-angiogenesis activity) and indocyanine green (ICG, a photosensitizer) for synergistic chemo-phototherapy with GSH depletion. The nanodrug was further cloaked by an erythrocyte membrane (RBC) to prolong the circulation time. Within the tumor microenvironment, the nanodrug would be disassembled upon intracellular GSH triggering. Moreover, the released Cu(II) could efficiently deplete the GSH, thus damaging the ROS-scavenging system and amplifying the phototherapeutic efficiency upon laser irradiation. The experiments validated the highly effective accumulation at tumor sites, potent tumor growth inhibition, and inappreciable systemic toxicity. The tumor microenvironment-responsive coordination-driven self-assembled biomimetic nanodrug may hold potential applications in tumor theranostics.
添加收藏
创建看单
引用
1区Q1影响因子: 9.7
跳转PDF
登录
英汉
2. A celastrol-based nanodrug with reduced hepatotoxicity for primary and metastatic cancer treatment.
期刊:EBioMedicine
日期:2023-07-20
DOI :10.1016/j.ebiom.2023.104724
BACKGROUND:Cancer is the world's leading cause of death and a key hindrance to extending life expectancy. Celastrol, a bioactive compound derived from Tripterygium wilfordii, has been shown to have excellent antitumor activity, but its poor solubility and severe organ toxicity side effects have hampered its clinical application. METHODS:In this study, a self-assembled nanodrug (PLC-NP) was designed to deliver celastrol to tumor sites while efficiently reducing its side effects by conjugating celastrol with the bioactive material LMWH and P-selectin targeting peptide (PSN). Extensive in vitro and in vivo experiments were performed to investigate both therapeutic efficacy and adverse effects. Furthermore, the specific mechanism of the antitumor activity has also been explored. FINDING:The PLC-NP nanodrugs were spherical in shape, with a mean particle size of 115.83 ± 6.93 nm. PLC-NP was sufficiently stable during blood circulation, with a selective target to P-selectin-highly expressed tumor cells, followed by releasing the containing celastrol under acidic environment and high levels of esterase in tumor cells. Both in vitro and in vivo results confirmed that celastrol's antitumor and anti-metastatic abilities were not attenuated and were actually strengthened after being formed into nanodrugs. More importantly, the organ toxicities of the modified celastrol nanodrug were dramatically reduced. Mechanistic study indicated that the inactivation of PI3K/Akt/mTOR signaling pathway and ROS-mediated mitochondrial dysfunction play critical roles in celastrol-mediated autophagy and apoptosis. INTERPRETATION:Our findings could offer a potential strategy for the translation of toxic compounds into clinical therapeutic nanomedicine. FUNDING:See a detailed list of funding bodies in the Acknowledgements section at the end of the manuscript.
添加收藏
创建看单
引用
2区Q1影响因子: 6
英汉
3. Development of novel celastrol-ligustrazine hybrids as potent peroxiredoxin 1 inhibitors against lung cancer.
期刊:European journal of medicinal chemistry
日期:2023-07-17
DOI :10.1016/j.ejmech.2023.115656
The disruption of oxidation-reduction equilibrium through inhibiting reactive oxygen species (ROS) clearance or enhancing ROS production has emerged as a novel and promising strategy for cancer therapy. Herein, a series of celastrol-ligustrazine hybrids were designed and synthesized as effective ROS promoters, and their biological activities were further evaluated. Among them, compound 7e stood out as the most potent peroxiredoxin 1 (PRDX1) inhibitor (IC = 0.164 μM), which was significant super to the recognized PRDX1 inhibitor Conoidin A (IC = 14.80 μM) and the control compound celastrol (IC = 1.622 μM). Furthermore, 7e dramatically promoted intracellular ROS accumulation, and inhibited the proliferation, invasion and migration of cancer cells besides inducing apoptosis in vitro. Additionally, 7e suppressed the key signaling pathways (AKT and ERK) and promoted the expression of apoptosis-related proteins (cleaved caspase-3/8 and cleaved PARP) in A549 cells, which resulted in the prevention of tumor progression. Most importantly, compound 7e (TGI = 77.47%) showed more considerable in vivo antitumor efficacy and less toxicity than celastrol (TGI = 71.00%). Overall, this work indicates 7e as the most potential PRDX1 inhibitor and may be a promising candidate for the therapy of lung cancer.
添加收藏
创建看单
引用
1区Q1影响因子: 9.6
打开PDF
登录
英汉
4. Inflammation-Targeted Celastrol Nanodrug Attenuates Collagen-Induced Arthritis through NF-κB and Notch1 Pathways.
作者:An Lemei , Li Zhanrong , Shi Liuqi , Wang Liujun , Wang Yong , Jin Lin , Shuai Xintao , Li Jingguo
期刊:Nano letters
日期:2020-09-30
DOI :10.1021/acs.nanolett.0c03279
Rheumatoid arthritis (RA) is a systemic inflammatory disorder which can cause bone and cartilage damage leading to disability, yet the treatment remains unsatisfactory nowadays. Celastrol (Cel) has shown antirheumatic activity against RA. However, the frequent parenteral delivery and poor water solubility of Cel restrict its further therapeutic applications. Here, aiming at effectively overcoming the poor water solubility and short half-life of Cel to boost its beneficial effects for treating RA, we developed a polymeric micelle for Cel delivery based on a reactive oxygen species (ROS) sensitive polymer. Our results demonstrated that Cel may inhibit the repolarization of macrophages toward the pro-inflammatory M1 pheno-type via regulating the NF-κB and Notch1 pathways, which resulted in significantly decreased secretion of multiple pro-inflammatory cytokines to suppress the RA progression. Consequently, the Cel-loaded micelle effectively alleviated the major RA-associated symptoms including articular scores, ankle thickness, synovial inflammation, bone erosion, and cartilage degradation.
添加收藏
创建看单
引用
3区Q2影响因子: 3.8
打开PDF
登录
英汉
5. Celastrol Loaded Nanoparticles With ROS-Response and ROS-Inducer for the Treatment of Ovarian Cancer.
作者:Niu Weina , Wang Jianguo , Wang Qinyao , Shen Jianjun
期刊:Frontiers in chemistry
日期:2020-10-30
DOI :10.3389/fchem.2020.574614
Ovarian cancer is a gynecological cancer from which it is difficult to be completely cured. It is common to use regimens as an effective treatment for ovarian cancer, but these inevitably bring serious side effects. New treatment strategies and special drugs are needed to improve the prognosis of patients. Celastrol is a natural product, isolated from traditional medicine, that has been proven to be curative for inflammation and cancers. However, the non-targeting and low solubility of celastrol limit its clinical application. We prepared celastrol-loaded nanoparticles for the efficient treatment of ovarian cancer via oxidative stress amplification. In this work, a tumor-targeted, ROS-sensitive nanoparticle was designed, synthesized, and assembled into a drug delivery system that used celastrol. Folic acid (FA) groups on the surface of nanoparticles guide them to actively target the surface of the tumor cell membrane. Thioketal (TK) bonds in nanoparticles can be oxidized and broken into -SH within the ROS level of tumor tissues, which causes the breaking of the PEG hydrophilic shell layer of nanoparticles and promotes the release of celastrol. The released celastrol further stimulated the production of ROS and amplified the intracellular ROS level to promote the apoptosis of tumor cells, thus achieving a therapeutic effect on the celastrol treated ovarian cancer.
添加收藏
创建看单
引用
2区Q1影响因子: 4.8
打开PDF
登录
英汉
6. Celastrol inhibits rheumatoid arthritis through the ROS-NF-κB-NLRP3 inflammasome axis.
作者:Jing Ming , Yang Junjie , Zhang Lirong , Liu Jing , Xu Sen , Wang Meiling , Zhang Leiming , Sun Yue , Yan Weibin , Hou Guige , Wang Chunhua , Xin Wenyu
期刊:International immunopharmacology
日期:2021-06-17
DOI :10.1016/j.intimp.2021.107879
Emerging evidence indicates that NOD-like receptor protein 3 (NLRP3) inflammasome-induced inflammation plays a critical role in the pathogenesis of rheumatoid arthritis (RA). Celastrol (Cel) is a quinone-methylated triterpenoid extracted from Tripterygium wilfordii that is used to treat RA. However, researchers have not determined whether Cel exerts anti-RA effects by regulating the activation of the NLRP3 inflammasome. In the present study, complete Freund's adjuvant (CFA)- induced rats and human mononuclear macrophages (THP-1 cells) were used to explore the anti-RA effects of Cel and its underlying mechanism. Joint swelling, the arthritis index score, inflammatory cell infiltration, and synovial hyperplasia in CFA-induced rats were correspondingly reduced after Cel treatment. The secretion of interleukin (IL)-1β and IL-18 in the serum of CFA-induced rats and supernatants of THP-1 cells exposed to Cel was significantly decreased. These inhibitory effects occurred because Cel blocked the nuclear factor-kappa B (NF-κB) signaling pathway and inhibited the activation of the NLRP3 inflammasome. Furthermore, Cel inhibited reactive oxygen species (ROS) production induced by lipopolysaccharide (LPS) and adenosine triphosphate (ATP). We speculated that Cel relieves RA symptoms and inhibits inflammation by inhibiting the ROS-NF-κB-NLRP3 axis.