1
|
Zhang J, Jiang B, Yun X, Gai C, Wang Z, Zou Y, Yang J, Song Y, Meng Q, Zhao Q, Chai X. Discovery of novel N-(5-chloro-2,4-dimethoxyphenyl)-N-heterocyclic ketone analogs as potent anti-inflammatory agents against ulcerative colitis. Bioorg Chem 2025; 161:108576. [PMID: 40373559 DOI: 10.1016/j.bioorg.2025.108576] [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: 12/09/2024] [Revised: 04/12/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025]
Abstract
As the incidence of ulcerative colitis (UC) has increased globally, there is a great unmet clinical need for efficacious, tolerable, and economical, orally administered drugs for its treatment. To help meet this need, we investigated anti-inflammatory small-molecule drugs with a novel structure, high activity, and high selectivity for the treatment of UC. Here, we designed and synthesized a series of novel anti-inflammatory compounds based on the molecular hybridization strategy by merging fragments from anti-inflammatory drugs. Among them, compound 11a best-exhibited lipopolysaccharide (LPS)-induced inflammation in RAW264.7 cells in vitro. Anti-inflammatory mechanism studies showed that compound 11a inhibited the release of pro-inflammatory cytokines and alleviated the inflammatory process by blocking the activation of the ASK1/p38 MAPKs/NF-κB signaling pathway in LPS-stimulated RAW264.7 cells. Analysis of the in vivo biological activity showed that compound 11a significantly alleviated dextran sodium sulfate-induced ulcerative colitis in mice while demonstrating an excellent safety in acute toxicity tests. Our study provides a novel compound for the treatment of UC that is worthy of further investigation and structural optimization.
Collapse
Affiliation(s)
- Juan Zhang
- Naval Medical Center of PLA, Naval Medical University, Shanghai 200433, China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Boye Jiang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Xiaoqing Yun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Conghao Gai
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zhen Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yan Zou
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Jishun Yang
- Naval Medical Center of PLA, Naval Medical University, Shanghai 200433, China
| | - Yan Song
- Naval Medical Center of PLA, Naval Medical University, Shanghai 200433, China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Qingjie Zhao
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Xiaoyun Chai
- Department of Organic Chemistry, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
2
|
Zeng F, Zhu Y, Li R, Chen M, Liang Y, Zhang L, Li Y, Peng S, Li J, Zhou W. Phenolic profiles in the peels of three wampee (Clausena lansium (Lour.) Skeels) fruit cultivars and immune-modulatory activity through the cluster of differentiation 14-mitogen activated protein kinase/nuclear factor kappa B pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119924. [PMID: 40324700 DOI: 10.1016/j.jep.2025.119924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/08/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The peels of wampee (Clausena lansium (Lour.) Skeels) fruits have been ethnomedicinally used to cure malarial fever, coughs, bronchitis, viral hepatitis, and gastrointestinal disorders, suggesting potential immunoprotective and anti-inflammatory properties. AIM OF THE STUDY This study aimed to compare the phenolic profiles in the peels of three wampee fruit cultivars and initially reveal the comprehensive immune-modulatory mechanisms of the phenolic fraction. MATERIALS AND METHODS The fruits of Golden wampee (GLW), Jixin wampee (JXW) and Guifei wampee (GFW) were collected. Their peels (GLP, JXP, GFP) were used to extract crude phenols (GLPE, JXPE, GFPE). JXPF was a phenolic fraction purified from JXPE using preparative high-performance liquid chromatography, which contained the mixture of various phenolic compounds in JXP. Phenolic contents were quantified using HPLC. The binding receptors and immune-modulatory pathways were investigated in LPS-induced RAW 264.7 cells. RESULTS JXP exhibited higher proportions of phenolic compounds, including myricetin-3-O-galactoside, nicotiflorin, rutin, isorhamnetin-3-O-neohesperidoside, isoquercitrin, quercitrin, and 8-hydroxypsoralen. GLPE, JXPE and GFPE reduced LPS-induced overexpression of NO, IL-6 and TNF-α, maintaining these mediators at moderate levels, which indicated immune-modulatory activity. The phenolic compounds in JXPF could occupy the CD14 receptor on macrophage surfaces and finally suppress the phosphorylation of ERK 1/2, p38, JNK, IκBα, and p65 in the MAPK and NF-κB signaling pathways. CONCLUSIONS JXW was identified as a superior cultivar with a higher abundance of phenolic compounds in its peels. The phenols derived from wampee peels demonstrated potential as immune-modulatory agents.
Collapse
Affiliation(s)
- Fanke Zeng
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Yuxiang Zhu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
| | - Ruyi Li
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China.
| | - Mianhong Chen
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Yuwei Liang
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Li Zhang
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Yingying Li
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Shaodan Peng
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Jihua Li
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Wei Zhou
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
3
|
Park S, Cho S, Shin HJ, Baek S, Gwon HI, Lee J, Yoo DS, Park HW, Seo DB, Bae S. Pharmacological Evaluation of Araliadiol as a Novel Anti-Inflammatory Agent in LPS-Induced RAW 264.7 Cells. Biomedicines 2025; 13:1408. [PMID: 40564127 DOI: 10.3390/biomedicines13061408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Revised: 06/03/2025] [Accepted: 06/04/2025] [Indexed: 06/28/2025] Open
Abstract
Background/Objectives: Inflammatory disorders contribute to the pathogenesis of numerous diseases and are known to markedly reduce quality of life. Although anti-inflammatory drugs approved by the Food and Drug Administration are available, their prolonged use is frequently associated with adverse effects. In this study, we evaluated the pharmacological properties of araliadiol, a naturally occurring polyacetylene compound, as a novel anti-inflammatory agent. Methods: An in vitro hyperinflammatory model was established by stimulating RAW 264.7 cells with lipopolysaccharide (LPS). Dexamethasone (DEX) was used as a positive control to compare anti-inflammatory efficacy. The protective effects of araliadiol against LPS-induced cytotoxicity were assessed using adenosine triphosphate content and crystal violet staining assays. The anti-inflammatory activity was further examined by quantitative reverse transcriptase-polymerase chain reaction, Western blotting, cell fractionation, immunofluorescence staining, a nitric oxide assay, and an enzyme-linked immunosorbent assay. Results: Araliadiol significantly attenuated cytotoxicity and cell death in LPS-stimulated RAW 264.7 cells. It suppressed the expression of cell death markers Cleaved caspase-3 and Cleaved PARP-1. In addition, araliadiol downregulated key pro-inflammatory mediators, including inflammasome-related genes, cytokines, chemokines, and inducible nitric oxide synthase. It also reduced the expression of Cox-2 and PGE2, indicating potential anti-hyperalgesic effects. Moreover, araliadiol inhibited the activation of Nfκb and Stat1 signaling pathways in LPS-stimulated macrophages. Conclusions: Araliadiol demonstrated robust anti-cytotoxic, anti-inflammatory, and anti-hyperalgesic activities in LPS-induced RAW 264.7 cells, with efficacy comparable to DEX. These findings support its potential as a plant-derived therapeutic candidate for the management of inflammatory conditions.
Collapse
Affiliation(s)
- Seokmuk Park
- Department of Biological Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Suhyeon Cho
- Department of Biological Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hee-Jae Shin
- Department of Biological Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seyeol Baek
- Department of Biological Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hye-In Gwon
- Department of Biological Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jungmin Lee
- Department of Bio-Cosmetics Engineering, Sungkyul University, 53 Seonggyeoldaehak-ro, Manan-gu, Anyang-si 14097, Republic of Korea
| | - Dae Sung Yoo
- ASK Company Co., Ltd., 86 Dongdaegu-ro, Suseong-gu, Daegu 706841, Republic of Korea
| | - Han Woong Park
- ASK Company Co., Ltd., 86 Dongdaegu-ro, Suseong-gu, Daegu 706841, Republic of Korea
| | - Dae Bang Seo
- ASK Company Co., Ltd., 86 Dongdaegu-ro, Suseong-gu, Daegu 706841, Republic of Korea
| | - Seunghee Bae
- Department of Biological Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
4
|
Liu ML, Liu PY, Chao LK, Yang TJ, Li LH, Weng YM, Sommano SR, Unpaprom Y, Ramaraj R, Ho CL, Hua KF. Anti-Inflammatory Potential of Essential Oil from the Heart-Wood of the Folk Medicinal Tree Cinnamomum kanehirai Hayata in Macrophages. Int J Mol Sci 2025; 26:5419. [PMID: 40508226 PMCID: PMC12156951 DOI: 10.3390/ijms26115419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2025] [Revised: 05/30/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Inflammation is a vital physiological response that plays a crucial role in regulating host defense against pathogens while maintaining tissue homeostasis. Inflammasomes, a family of protein complexes, are responsible for controlling the expression of pro-inflammatory cytokines IL-1β and IL-18, and they play significant roles in inflammatory responses. However, dysregulated inflammation can become a risk factor for the pathogenesis of various diseases. The discovery of anti-inflammatory substances derived from natural products represents an important strategy for new drug development. In this study, we found that the essential oil derived from the heartwood of Cinnamomum kanehirai Hayata (EOC) exhibits anti-inflammatory activities by inhibiting the NLRP3, NLRP1, NLRC4, AIM2, and non-canonical inflammasomes in macrophages. EOC also suppresses the expression of NLRP3, TNF-α, IL-6, and NO in LPS-activated macrophages. The mechanisms underlying the anti-inflammatory activity of EOC were shown to involve a reduction in reactive oxygen species production and NF-κB activation. Furthermore, terpinen-4-ol may be the key anti-inflammatory compound present in EOC. These results suggest that EOC has potential as an anti-inflammatory agent for future development.
Collapse
Affiliation(s)
- May-Lan Liu
- Department of Food Science, National Chiayi University, Chiayi 600355, Taiwan; (M.-L.L.); (Y.-M.W.)
- Department of Early Childhood Educare, WuFeng University, Chiayi 621303, Taiwan
| | - Pang-Yen Liu
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan;
| | - Louis Kuoping Chao
- Department of Cosmeceutics, China Medical University, Taichung 406040, Taiwan;
| | - Tzu-Jung Yang
- Division of Wood Cellulose, Taiwan Forestry Research Institute, Taipei 10066, Taiwan;
| | - Lan-Hui Li
- Department of Laboratory Medicine, Linsen Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 108203, Taiwan;
| | - Yih-Ming Weng
- Department of Food Science, National Chiayi University, Chiayi 600355, Taiwan; (M.-L.L.); (Y.-M.W.)
| | - Sarana Rose Sommano
- Plant Bioactive Compound Laboratory (BAC), Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Yuwalee Unpaprom
- Sustainable Resources and Sustainable Engineering Research Lab, Maejo University, Chiang Mai 50290, Thailand; (Y.U.); (R.R.)
- Program in Biotechnology, Faculty of Science, Maejo University, Chiang Mai 50290, Thailand
| | - Rameshprabu Ramaraj
- Sustainable Resources and Sustainable Engineering Research Lab, Maejo University, Chiang Mai 50290, Thailand; (Y.U.); (R.R.)
- School of Renewable Energy, Maejo University, Chiang Mai 50290, Thailand
| | - Chen-Lung Ho
- Division of Wood Cellulose, Taiwan Forestry Research Institute, Taipei 10066, Taiwan;
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260007, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404328, Taiwan
| |
Collapse
|
5
|
Xing H, Pan J, Liu H, Wang Y, Chang Z. Preliminary research indicates that mechanical force through Pioze1 enhances local immunity during NPWT treatment for spinal infections. Front Immunol 2025; 16:1600194. [PMID: 40529353 PMCID: PMC12170304 DOI: 10.3389/fimmu.2025.1600194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 05/14/2025] [Indexed: 06/20/2025] Open
Abstract
Objective This study aims to investigate the mechanism by which Negative Pressure Wound Therapy (NPWT) regulates local immune responses in spinal infection through Piezo1-mediated mechanical stress, and elucidate its potential role in the treatment of spinal infections. Methods From July 2021 to April 2022, a total of 7 patients with spinal infection treated with NPWT at our department were included in the study. The study analyzed clinical outcomes of spinal infection surgeries, including operative duration, intraoperative blood loss, postoperative drainage, improvements in pain levels as measured by the Visual Analogue Scale (VAS), and inflammatory markers such as C-reactive protein (CRP) and Erythrocyte Sedimentation Rate (ESR) measured one week before and after the procedure. Additionally, healing times and recurrence rates within two years post-surgery were assessed. In addition, lesion specimens were retained during surgery and changes in Piezo1, Interleukin-1β (IL-1β), IL-6, IL-8, and Tumor Necrosis Factor-α (TNF-α) in lesion tissues were observed before and after immunohistochemical analysis. Results All 7 patients with spinal infections successfully underwent NPWT treatment and were ultimately cured. The average healing time was 45.71 ± 9.49 days, and there were no cases of recurrence or death during the two-year follow-up period. Surgical data showed a surgery duration of 96.57 ± 13.31 minutes, intraoperative blood loss of 65.71 ± 29.36 milliliters, and postoperative drainage of 163.57 ± 11.07 milliliters. Postoperatively, CRP, ESR, and VAS all significantly improved compared to preoperative levels (all p<0.05), which was superior to traditional treatment methods. Following NPWT intervention, the expression of Piezo1 protein at the lesion site significantly increased (0.03 ± 0.11 vs. 0.27 ± 0.22; p<0.05), while the expression levels of IL-1β, IL-6, IL-8, and TNF-α in the local immune microenvironment of the infected lesion significantly decreased (0.26 ± 0.11 vs. 0.16 ± 0.09, 0.27 ± 0.12 vs. 0.15 ± 0.67, 0.26 ± 0.18 vs. 0.10 ± 0.12, 0.35 ± 0.21 vs. 0.15 ± 0.11; p<0.05). Conclusion Clinical results demonstrate that NPWT treatment for spinal infections exhibits remarkable efficacy, accompanied by a notable augmentation in local Piezo1 protein consistency. It is hypothesized that the mechanical force employed in NPWT treatment stimulates the Piezo1 protein, thereby modulating local immune cells and factors, ultimately bolstering local immunity. This study not only provides a molecular biology basis for a deeper understanding of the therapeutic effects of NPWT, but also offers new insights for optimizing treatment strategies for spinal infections.
Collapse
Affiliation(s)
- Hao Xing
- Department of Orthopaedics, The 960th Hospital of People’s Liberation Army of China (PLA), Jinan, China
| | - Junlin Pan
- Department of Reproductive Medicine, The 960th Hospital of People's Liberation Army of China (PLA), Jinan, China
| | - Huan Liu
- Department of Orthopaedics, The 960th Hospital of People’s Liberation Army of China (PLA), Jinan, China
| | - Yanan Wang
- Department of Orthopaedics, The 960th Hospital of People’s Liberation Army of China (PLA), Jinan, China
| | - Zhengqi Chang
- Department of Orthopaedics, The 960th Hospital of People’s Liberation Army of China (PLA), Jinan, China
| |
Collapse
|
6
|
da Silva Mendonça L, Rocha KAD, das Chagas Lima Pinto F, da Silva LR, Santos FA, Tavares JF, Silveira ER, Pessoa ODL. Anti-inflammatory glycoside withanolides from leaves of Athenaea velutina. Fitoterapia 2025; 183:106522. [PMID: 40204049 DOI: 10.1016/j.fitote.2025.106522] [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: 07/30/2024] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
The phytochemical study of the CH2Cl2 and EtOAc soluble fractions from the leaf ethanol extract of Athenaea velutina resulted in the isolation of the new withanolides 1-3, two of which substituted at the C-3 by a disaccharide 3β-O-[α-L-rhamnopyranosyl-(1 → 4)-β-d-glucopyranosyl], and four others previously reported (4-7). The structures of the new withanolides were elucidated by detailed interpretation of their 1D- and 2D-NMR spectra and the HRESIMS analysis. An anti-inflammatory screening was performed using the two major withanolides (1 and 2), analyzing their effects on the viability of RAW 264.7 cells and their ability to inhibit cellular nitric oxide production. While withanolide 1 exhibited pro-inflammatory activity by increasing nitrite production in LPS-stimulated macrophages, compound 2, in opposition, demonstrated anti-inflammatory action by reducing those levels.
Collapse
Affiliation(s)
- Lavosyer da Silva Mendonça
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, 60021-970 Fortaleza, CE, Brazil
| | - Késya Amanda Dantas Rocha
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, 60021-970 Fortaleza, CE, Brazil
| | | | - Lidiana Rabelo da Silva
- Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-270 Fortaleza, CE, Brazil
| | - Flavia Almeida Santos
- Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-270 Fortaleza, CE, Brazil
| | - Josean Fechine Tavares
- Centro de Ciências da Saúde, Universidade Federal da Paraíba, 58051-900 João Pessoa, PB, Brazil
| | - Edilberto Rocha Silveira
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, 60021-970 Fortaleza, CE, Brazil
| | | |
Collapse
|
7
|
Choo MZY, Chua JAT, Lee SXY, Ang Y, Wong WSF, Chai CLL. Privileged natural product compound classes for anti-inflammatory drug development. Nat Prod Rep 2025; 42:856-875. [PMID: 40066695 DOI: 10.1039/d4np00066h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Covering: up to early 2025Privileged compound classes of anti-inflammatory natural products are those where there are many reported members that possess anti-inflammatory properties. The identification of these classes is of particular relevance to drug discovery, as they could serve as valuable starting points in developing effective and safe anti-inflammatory agents. The privileged compound classes of natural products include the polyphenols, coumarins, labdane diterpenoids, sesquiterpene lactones, isoquinoline and indole alkaloids, each offering a variety of molecular scaffolds and functional groups that enable diverse interactions with biological targets. From a medicinal chemistry point of view, natural products are both a boon and a bane. The multi-targeting nature of natural products is a boon in the treatment of multi-factorial diseases such as inflammation, but promiscuity, poor potency and pharmacokinetic properties are significant hurdles that must be addressed to ensure these compounds can be effectively used as therapeutics. In addition, there are continued controversies regarding the efficacies of some of these natural products that will continue to polarise their use. In this review, examples of natural products of six privileged compound classes will be discussed for their potential use and possible further development as anti-inflammatory drugs.
Collapse
Affiliation(s)
- Malcolm Z Y Choo
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543, Singapore.
| | - Julian A T Chua
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543, Singapore.
| | - Sean X Y Lee
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543, Singapore.
| | - Yuet Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600, Singapore.
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600, Singapore.
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore
| | - Christina L L Chai
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543, Singapore.
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore
| |
Collapse
|
8
|
Cao J, Zhong M, Wang S, Li Q, Ma W, Yan X, Wang J, Zhao Y. A Stable and Local H 2 Nanogenerator for Antifading Responses. ACS APPLIED BIO MATERIALS 2025. [PMID: 40389364 DOI: 10.1021/acsabm.5c00533] [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: 05/21/2025]
Abstract
Persistent inflammation effects existed in the senescence microenvironment, and hydrogen (H2) was an antioxidant stress agent and could solve the aging and inflammation. In view of the issue of low solubility of H2 and conversion efficiency of traditional hydrogen therapy, an in-site hydrogen production system (HPS) was designed by using liposome as a compartment, emodin as a photosensitizer, l-ascorbic acid-2-phosphate trisodium salt (AAP) as hydrogen sources, and Au NPs as photocatalysts. Different from the commonly used chlorophyll A, emodin with strong absorption in the wide range of 200-500 nm could absorb more photons to form electrons for the participation of H2 generation. Emodin- and AAP-embedded HPS largely increased stability at 50 °C for 28 days. HPS facilitated the decrease of inflammatory factors of TNF-α, IL-6, and ROS in RAW264.7 cells, as well as the elimination of senescence-associated β-galactosidase, realizing convenient and efficient antifading responses. But strikingly, the constructed HPS can penetrate the pig skin in transdermal experiments and showed huge prospects for the application of antifading cosmetics.
Collapse
Affiliation(s)
- Jiaying Cao
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, 214122 Wuxi, Jiangsu, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Mingwei Zhong
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Siyu Wang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Qibin Li
- Zhejiang KylinWay Medical Technology Co., Ltd, 310011 Hangzhou, Zhejiang, China
| | - Wei Ma
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Xiaodong Yan
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Jing Wang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Yuan Zhao
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, School of Chemical and Material Engineering, Jiangnan University, 214122 Wuxi, Jiangsu, China
| |
Collapse
|
9
|
Qin X, Hou R, Qu Z, Yu J, Zhang W, Ma H, Zhuang C. Structure-based molecular hybridization design and synthesis of Keap1-Nrf2 inhibitors for anti-inflammatory treatment. Bioorg Chem 2025; 158:108350. [PMID: 40073597 DOI: 10.1016/j.bioorg.2025.108350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
The Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway stands as a pivotal mechanism in defending against oxidative stress damage and related inflammation. Blocking the Keap1-Nrf2 protein-protein interaction (PPI) offers a promising therapeutic approach for treating diseases related to oxidative stress and inflammation. Our group previously reported NXPZ-2, a naphthalene sulfonamide derivative targeting Keap1, which effectively inhibits the Keap1-Nrf2 PPI, thereby releasing Nrf2 to exert its anti-inflammatory and antioxidant effects. In the present work, we employed a structure-based molecular hybridization strategy to design a series of novel naphthalene sulfonamides by combining NXPZ-2 with the Nrf2 activator dimethyl fumarate (DMF) or its analogues. Among these new derivatives, compound 1c, specifically (Z)-4-((4-(N-(2-amino-2-oxoethyl)-N-(4-((N-(2-amino-2-oxoethyl)-4-methoxyphenyl)sulfonamide)naphthalen-1-yl) sulfamoyl)phenyl)amino)-4-oxobut-2-enoic acid, exhibited the highest PPI inhibitory activity, with a KD2 value of 0.119 μM. In an LPS-induced RAW264.7 cell model, this compound mitigated LPS-induced cellular damage, suppressed the expression of pro-inflammatory cytokine TNF-α and IL-6, and significantly elevated the intracellular GSH and SOD enzyme activities. Furthermore, in an LPS-induced acute lung injury (ALI) mouse model, the compound demonstrated a remarkable ability to alleviate oxidative damage and inflammation in the lungs. In conclusion, this novel naphthalene sulfonamide represents a promising drug candidate for Keap1-targeting therapy in ALI. Molecular docking analysis revealed that the amide and maleic acid groups of 1c facilitate strong interactions with the Kelch domain of Keap1, explaining the compound's preference for binding through hydrogen bonding and π-π stacking interactions.
Collapse
Affiliation(s)
- Xiuting Qin
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Ruilin Hou
- Department of Pharmacy, Drug/Medical Device Clinical Trial Institution Office, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia 750003, China
| | - Zhuo Qu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China; The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Hao Ma
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China.
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China; The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
10
|
Bramorski Mohr ET, Lubschinski TL, Oliveira JMDD, Giarola Fragoso de Oliveira P, Garcia Mendes Borba B, Demarchi IG, Dalmarco EM. Thiazolidines derivatives and their anti-inflammatory activity in LPS-induced RAW 264.7 macrophages: a systematic review and meta-analysis. Nat Prod Res 2025; 39:2895-2911. [PMID: 39165196 DOI: 10.1080/14786419.2024.2394103] [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: 03/02/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Thiazolidine scaffolds have been investigated for decades, due to their wide range of biological activity. In this way, the main objective of this systematic review was to elucidate the anti-inflammatory activity of thiazolidine derivatives against nitric oxide (NO) production in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages. From 9718 identified registers, 13 articles were included, where 11 studies evaluated thiazolidinediones. The summary of relevance demonstrated that seven studies (53.8%) were relevant without restrictions, and 6 (46.2%) were relevant with restrictions. The certainty in cumulative evidence was considered moderate and the six studies included in the meta-analysis demonstrated the positive activity of thiazolidinediones against NO production when compared to the negative LPS control.
Collapse
Affiliation(s)
- Eduarda Talita Bramorski Mohr
- Postgraduate Program in Pharmacy (PPGFar), Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
| | - Tainá Larissa Lubschinski
- Postgraduate Program in Pharmacy (PPGFar), Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
| | - Júlia Meller Dias de Oliveira
- Department of Dentistry, Brazilian Centre for Evidence-Based Research (COBE), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Paula Giarola Fragoso de Oliveira
- Postgraduate Program in Pharmacy (PPGFar), Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
| | - Beatriz Garcia Mendes Borba
- Postgraduate Program in Pharmacy (PPGFar), Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
- Department of Clinical Analysis, Center of Health Sciences, Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
| | - Izabel Galhardo Demarchi
- Postgraduate Program in Pharmacy (PPGFar), Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
- Department of Clinical Analysis, Center of Health Sciences, Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
| | - Eduardo Monguilhott Dalmarco
- Postgraduate Program in Pharmacy (PPGFar), Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
- Department of Clinical Analysis, Center of Health Sciences, Universidade Federal de Santa Catarina, Campus Universitário-Trindade, Florianópolis, Brazil
| |
Collapse
|
11
|
Wang L, Li ZY, Zhong CL, Teng ZY, Wang B, Rehman A, Han LW, Zeng KW, Zhang JG, Lu ZY. Therapeutic potential of naturally derived carbon dots in sepsis-associated acute kidney injury. Chin Med 2025; 20:49. [PMID: 40217355 PMCID: PMC11992765 DOI: 10.1186/s13020-025-01103-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Sepsis is a life-threatening infectious disease characterized by an uncontrolled inflammatory response and consequent multi-organ dysfunction. The kidneys, as primary excretory organs with high blood flow, are particularly susceptible to damage during sepsis. Nonetheless, the existing treatment options for sepsis-associated acute kidney injury (SA-AKI) are still restricted. Nanomedicine, especially carbon dots (CDs), has attracted considerable interest lately for outstanding biomedical characteristics. METHODS To avoid the generation of toxic effects, the natural CDs derived from Ziziphi Spinosae Semen (Z-CDs) were synthesized employing a hydrothermal method. The free radical scavenging capabilities of Z-CDs were evaluated by utilizing ABTS assay, NBT method, and Fenton reaction. A lipopolysaccharide (LPS)-stimulated RAW 264.7 cell model was used to explore the therapeutic potential of Z-CDs on cellular oxidative stress and inflammation. The CuSO4-induced zebrafish inflammation model and LPS-exposed SA-AKI mouse model were employed to assess the therapeutic efficacy of Z-CDs in vivo. RESULTS The synthesized Z-CDs exhibited distinctive unsaturated surface functional groups, which confer exceptional biocompatibility and the ability to scavenge free radicals. Moreover, Z-CDs were particularly effective in eliminating excess reactive oxygen species (ROS) in cells, thus protecting mitochondrial function from oxidative damage. Notably, Z-CDs have demonstrated significant therapeutic benefits in protecting kidney tissue in SA-AKI mouse model with minimizing side effects. In mechanism, Z-CDs effectively reduced ROS production, thereby alleviating inflammatory responses in macrophages through the suppression of the NF-κB pathway. CONCLUSIONS This study developed a multifunctional nanomedicine derived from traditional medicinal herb, providing a promising pathway for the advancement of innovative drug therapies to treat SA-AKI.
Collapse
Affiliation(s)
- Lei Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Zhong-Yao Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Chong-Lei Zhong
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Zi-Yang Teng
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Bin Wang
- Department of Andrology, Guang'Anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Asma Rehman
- National Institute for Biotechnology & Genetic Engineering College Pakistan Institute of Engineering & Applied Sciences (NIBGE-C, PIEAS), Faisalabad, 38000, Pakistan
| | - Li-Wen Han
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Ji-Guo Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Zhi-Yuan Lu
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
12
|
Luo X, Zhang X, Wen X, Wang R, Zhang Q, Luo P, Yu F, Cao H. Valence State Hydrogen Channel Enhances Sustained and Controllable Electrocatalytic Hydrogen Evolution in Diabetic Skin Wound Healing. Angew Chem Int Ed Engl 2025; 64:e202422091. [PMID: 39846755 DOI: 10.1002/anie.202422091] [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: 11/13/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 01/24/2025]
Abstract
Diabetes significantly increases the risk of serious health issues, including prolonged skin inflammation and delayed wound healing, owing to inferior glucose control and suppression of the immune system. Although traditional hydrogen (H2) therapy is slightly effective, its ability to tailor the release of H2 on the skin is limited. Accordingly, this study proposed a novel strategy for electrocatalytic H2 release under neutral conditions to promote wound healing in diabetic mice and rabbit. Herein, a defect-engineered cobalt phosphide (CoP) catalyst was designed by introducing a neutral single-metal electrocatalytic Hydrogen valence state channel into CoP. By effectively regulating the formation and transfer of *H active species during the CoP catalytic process, a considerable enhancement in neutral electrocatalytic H2 evolution performance was achieved (-78.0 mV@-10.0 mA cm-2). Based on this superior catalytic performance, we developed a flexible electrode (namely, CoP/flexible gold electrode made by screen printing (FGSP) by combining a convenient electrolysis platform with continuous electrolyte supply and FGSP, enabling customized H2 release and accelerating wound healing in diabetic mice and rabbits. Notably, the designed flexible electrode features adjustable dimensions, interchangeable substrates, and material adaptability, meeting the diverse needs of clinical and basic research and demonstrating significant potential for applications in clinical medicine.
Collapse
Affiliation(s)
- Xianzhu Luo
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| | - Xiangcheng Zhang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xue Wen
- School of Chemistry and Chemical Engineering, School of Electronics, Information and Electrical Engineering, Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03706, Republic of Korea
| | - Qingguo Zhang
- Department of General Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100006, China
| | - Pan Luo
- Department of General Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100006, China
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| | - Hongshuai Cao
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
- School of Chemistry and Chemical Engineering, School of Electronics, Information and Electrical Engineering, Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
13
|
Wang M, Yu Z, Li X, Li J, Li J, Luo J, Li J, Xiong Y, Yang J. In situ dual-targeted drug delivery system for alleviating imaging and pathological damage in septic arthritis. Acta Biomater 2025; 195:363-377. [PMID: 39922516 DOI: 10.1016/j.actbio.2025.02.014] [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: 10/16/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Septic arthritis is a severe disease that damages articular cartilage and triggers a strong inflammatory response. Current treatments mainly depend on systemic antibiotics and lack effective intra-articular therapies, as well as standardized animal models, and precise detection methods. In this study, we present a drug delivery system responsive to the bacterial microenvironment for targeted inflammation control, along with an effective method for monitoring changes in septic arthritis in SD rats. This system consists a core with pH-sensitive metal-organic frameworks ZIF-8 loading anti-inflammatory drugs indomethacin and a shell with hybrid cell membranes from macrophages (MM) and platelets (PM), refer as MP@ZIF-8@IN. This system, which diverges from traditional treatments, enhances drug utilization, prolongs local retention, and allows for spontaneous release at the treatment site, thereby enabling the exclusive intra-articular treatment of septic arthritis. The drug delivery system inhibits the NF-κB pathway, reduces oxidative stress, and regulates macrophage polarization, preventing cartilage destruction. Additionally, in this standardized animal model utilizing the knee joints of SD rats, we have developed musculoskeletal ultrasound and magnetic resonance imaging for time-based monitoring, thus overcoming the limitation of conventional methods, which are unsuitable for soft tissue analysis. Our findings advance therapeutic strategies for septic arthritis and encourage further application of visualization techniques in related fields. STATEMENT OF SIGNIFICANCE: This study presents significant advancements in the treatment and understanding of septic arthritis. Our customized drug delivery system targets bacteria and macrophages, ensuring long-time drug retention and enhanced inflammation control, all while reducing reliance on antibiotics-an important step toward addressing antibiotic resistance. Additionally, we have refined septic arthritis animal models to establish clearer guidelines for intervention timing, grounded in clinical symptoms and imaging data. This addresses a critical gap in current research and offers a practical framework for future therapeutic approaches.
Collapse
Affiliation(s)
- Mengxian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zeping Yu
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlong Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Junqiao Li
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianshu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Jun Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Yan Xiong
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Orthopedics and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Erana-Perez Z, Igartua M, Santos-Vizcaino E, Hernandez RM. Differential protein and mRNA cargo loading into engineered large and small extracellular vesicles reveals differences in in vitro and in vivo assays. J Control Release 2025; 379:951-966. [PMID: 39892179 DOI: 10.1016/j.jconrel.2025.01.085] [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: 09/28/2024] [Revised: 01/14/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Extracellular vesicles (EV) represent an advanced platform for genetic material and protein delivery, particularly when they are loaded through the so-called endogenous loading method. This study investigates the differences between large EV (lEV) and small EV (sEV) obtained from genetically engineered C2C12 myoblasts overexpressing two different model biomolecules. Erythropoietin (EPO) is a secretory protein with anti-inflammatory, angiogenic and hematopoietic effects, while TGL is a chimeric cytosolic protein containing green fluorescent protein (GFP) and luciferase, used for imaging. We compared these EV subtypes in terms of protein and nucleic acid loading, intercellular cargo transfer capacity, and subsequent functional effects both in vitro and in vivo. Our results demonstrated that lEV exhibited higher protein and mRNA cargo content than sEV, which also translated into increased intercellular cargo transfer capacity, even when dosing according to the constitutive sEV and lEV secretion ratio (10,1). Indeed, we showed that, although receptor cells successfully internalized both EV subtypes, cells treated with lEV displayed stronger intracellular luciferase signals and higher EPO protein secretion compared to those treated with sEV. In terms of functional effects, both EV subtypes exerted anti-inflammatory and antioxidant effects in lipopolysaccharide-activated macrophages, as well as angiogenic effects in human umbilical vein endothelial cells. Finally, in vivo studies evidenced that subcutaneously administered lEV led to a more significant increase in hematocrit levels and red blood cell counts than sEV. Taken together, these findings suggest that the protein and mRNA cargo differ between endogenously loaded EV subtypes, and that this variation in cargo loading leads to differences in their functional outcomes. Therefore, the choice of EV subtype could be critical for optimizing EV-based delivery strategies for biologic drugs.
Collapse
Affiliation(s)
- Zuriñe Erana-Perez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
15
|
Han HJ, Hyun CG. Anti-Inflammatory Effects and Human Skin Safety of the Eastern Traditional Herb Mosla japonica. Life (Basel) 2025; 15:418. [PMID: 40141763 PMCID: PMC11943674 DOI: 10.3390/life15030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Traditional knowledge has long provided natural solutions for disease prevention and treatment, complementing modern medicine. Mosla japonica (Korean mint) has been traditionally valued for its pesticidal, dehumidifying, anti-swelling, and detoxifying properties. This study explores its anti-inflammatory potential using M. japonica extract (MJE) in LPS-stimulated RAW 264.7 macrophages and evaluates its safety for human skin applications. MJE significantly reduced inflammatory mediators such as nitric oxide (NO), prostaglandin E2 (PGE2), and key cytokines (IL-1β, IL-6, TNF-α) in a dose-dependent manner. It also suppressed the expression of iNOS and COX-2, enzymes crucial for inflammation. Mechanistically, MJE inhibited NF-κB activation by stabilizing IκBα, thereby reducing inflammation-related gene expression. Additionally, it downregulated ERK, JNK, and p38 in the MAPK signaling pathway, further contributing to its anti-inflammatory effects. A primary skin irritation test confirmed MJE's safety, showing no significant skin reactions at 100 μg/mL. These findings highlight MJE's strong anti-inflammatory properties and potential for dermatological applications. This study underscores the pharmacological value of M. japonica and its integration into modern scientific research, aligning with global biodiversity frameworks such as the Nagoya Protocol. Future research may further expand its applications in medicine and skincare.
Collapse
Affiliation(s)
| | - Chang-Gu Hyun
- Jeju Inside Agency and Cosmetic Science Center, Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Republic of Korea;
| |
Collapse
|
16
|
Wang Q, Im Y, Park J, Lee HL, Ryu DG, Kim H. Eisenia bicyclis Extract Ameliorates Colitis in In Vitro and In Vivo Models Through Modulation of mTOR Axis and Gut Microbiota Composition. Foods 2025; 14:714. [PMID: 40077417 PMCID: PMC11899094 DOI: 10.3390/foods14050714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon that is associated with dysbiosis in the gut microbiota. Eisenia bicyclis, a marine alga, is known for its anti-inflammatory, antioxidant, and gut microbiota-modulating properties. This study explored the mechanisms by which a 70% ethanol extract of E. bicyclis may alleviate UC, through both in vitro and in vivo experiments. LC-MS/MS analysis revealed eckol, 7-phloroeckol, dieckol, phlorofucofuroeckol A, and fucofuroeckol as key phenolic compounds present in the extract. The administration of E. bicyclis significantly improved symptoms in a dextran sulfate sodium (DSS)-induced colitis mouse model by reducing intestinal shortening, splenomegaly, and histological scores. Both cell and animal studies demonstrated that E. bicyclis suppressed the release of inflammatory cytokines, downregulated the mRNA expression of genes related to the mTOR pathway, and reduced the p-mTOR/mTOR ratio. Microbiota analysis revealed that, while the Firmicutes/Bacteroidetes ratio was elevated in UC mice, E. bicyclis administration normalized this imbalance, with a notable increase in the abundance of beneficial probiotics such as Bifidobacterium bifidum. In conclusion, a phenolic-rich extract of E. bicyclis demonstrates significant potential as a dietary supplement to prevent and mitigate UC by modulating both the mTOR signaling pathway and gut microbiota composition.
Collapse
Affiliation(s)
- Qunzhe Wang
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea; (Q.W.); (Y.I.); (J.P.)
- BK21 FOUR Program: Precision Nutrition Program for Future Global Leaders, Pusan National University, Busan 46241, Republic of Korea
| | - Yuri Im
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea; (Q.W.); (Y.I.); (J.P.)
| | - Jumin Park
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea; (Q.W.); (Y.I.); (J.P.)
- BK21 FOUR Program: Precision Nutrition Program for Future Global Leaders, Pusan National University, Busan 46241, Republic of Korea
| | - Hye Lim Lee
- Department of Internal Medicine, Medical Research Institute, Pusan National University School of Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Dae Gon Ryu
- Department of Internal Medicine, Medical Research Institute, Pusan National University School of Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Hyemee Kim
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea; (Q.W.); (Y.I.); (J.P.)
| |
Collapse
|
17
|
Wei YH, Zhou SM, Zhao W, Chen Q, Wang QH, Yu ML, Wei YY, Hu TJ. Normal butanol fraction of Polygonum hydropiper L. flavonoids reduces inflammation caused by PCV2 infections in cell and mouse models. Front Vet Sci 2025; 12:1539448. [PMID: 39968105 PMCID: PMC11833999 DOI: 10.3389/fvets.2025.1539448] [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: 12/04/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction The normal butanol fraction of Polygonum hydropiper L. flavonoids (FNB) exhibits significant anti-inflammatory effects. This study investigated FNB's impact on inflammatory responses induced by Porcine circovirus type 2 (PCV2) in cell and mouse models. Methods An inflammatory model was established in RAW264.7 cells infected with varying PCV2 concentrations. And assigning both RAW264.7 cells and 108 SPF-grade KM mice to Control, PCV2, Rutin, and various dosages of FNB groups. Inflammatory factors such as Monocyte Chemoattractant Protein-1 (MCP-1), interleukin-6 (IL-6), IL-8, IL-10, Tumor Necrosis Factor-alpha (TNF-α), Reactive Oxygen Species (ROS), and Nitric Oxide (NO) were quantified using ELISA, RT-qPCR and immunohistochemistry. Results Results showed that a PCV2 titer of 104.5 TCID50/0.1 mL when applied to RAW264.7 cells effectively established an in vitro inflammatory model at 12 and 24 h post-infection. Following PCV2 infection, all the inflammatory factors displayed a significant increased both in culture supernatant and intracellular mRNA expression levels (p < 0.05 or p < 0.01), but these levels were reduced by FNB treatment (p < 0.05 or p < 0.01). In mouse sera post-PCV2 infection also showed elevated levels of IL-6, IL-8 IL-10, TNF-α, and MCP-1 (p < 0.05 or p < 0.01). Additionally, mRNA and protein levels for TNF-α, IL-8, IL-10, IL-6, and iNOS rose significantly in lung tissues (p < 0.01) but decreased with FNB treatment (p < 0.05 or p < 0.01). Discussion These findings suggest that FNB reduces inflammatory factor production and modulates the inflammatory response triggered by PCV2 infection, potentially enhancing host resistance against it.
Collapse
Affiliation(s)
- Yu-heng Wei
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Shu-mian Zhou
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, China
| | - Wen Zhao
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Qi Chen
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Qiu-hua Wang
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, China
| | - Mei-ling Yu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ying-yi Wei
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ting-jun Hu
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, China
| |
Collapse
|
18
|
Yang L, Hu Y, Deng H, Li Y, Zhang R, Zhang Q, Yang L, Pang H, Liu F, Fu C. Water-soluble polysaccharides from Torreya grandis nuts: Structural characterization and anti-inflammatory activity. Int J Biol Macromol 2025; 291:138935. [PMID: 39701235 DOI: 10.1016/j.ijbiomac.2024.138935] [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: 09/09/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
Torreya grandis (T. grandis) nuts are widely consumed as a functional food in China. In this study, we investigated the structural characteristics of T. grandis nuts polysaccharides and evaluated their potential biological functions with anti-inflammatory activities. Polysaccharides (TGP) were extracted from T. grandis nuts using water extraction and alcohol precipitation methods. Through a series of purification steps, three heteropolysaccharides (TGP-0a, TGP-2a, and TGP-3a) with distinct molecular weights, monosaccharide compositions, and surface morphologies were isolated. Their anti-inflammatory activities were screened, and TGP-0a was shown to be the most effective component. By combining NMR and methylation studies, TGP-0a was predominantly composed of linear α-1,4-glucan region and linear β-1,4-(gluco)mannan region. In cellular anti-inflammatory assays, TGP-0a significantly diminished the release of pro-inflammatory cytokines. Furthermore, by lowering the levels of iNOS and COX-2, TGP-0a decreased the release of inflammatory mediators (NO and ROS), thereby reducing oxidative stress and inflammatory response. In conclusion, T. grandis nut polysaccharides, particularly TGP-0a, show strong potential as natural anti-inflammatory agents for functional foods and pharmaceutical applications.
Collapse
Affiliation(s)
- Luping Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yunjie Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hongdan Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruiyuan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huiwen Pang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
19
|
Wu XY, Dong QW, Zhang YB, Li JX, Zhang MQ, Zhang DQ, Cui YL. Cimicifuga heracleifolia kom. Attenuates ulcerative colitis through the PI3K/AKT/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118892. [PMID: 39395768 DOI: 10.1016/j.jep.2024.118892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cimicifuga heracleifolia Kom. (C. heracleifolia) has demonstrated efficacy in treating gastrointestinal disorders, including splenasthenic diarrhea. Ulcerative colitis (UC), a chronic inflammatory bowel disease, shares similarities with splenasthenic diarrhea. However, the pharmacological effects of C. heracleifolia on UC and the underlying mechanisms remain unexplored. AIM OF THE STUDY The present study investigates the therapeutic potential and mechanisms of C. heracleifolia in UC. METHODS Initially, network pharmacology analysis, encompassing ingredient screening, target prediction, protein-protein interaction (PPI) network analysis, and enrichment analysis, was employed to predict the mechanisms of C. heracleifolia. The findings were further validated using transcriptomics and functional assays in a dextran sulfate sodium (DSS)-induced UC model. Additionally, bioactive compounds were identified through surface plasmon resonance (SPR) analysis, molecular docking, and cell-based assays. RESULTS A total of 52 ingredients of C. heracleifolia were screened, and 32 key targets were identified within a PPI network comprising 285 potential therapeutic targets. Enrichment analysis indicated that the anti-UC effects of C. heracleifolia are mediated through immune response modulation and the inhibition of inflammatory signaling pathways. In vivo experiments showed that C. heracleifolia mitigated histological damage in the colon, reduced the expression of phosphorylated Akt1, nuclear factor-kappa B (NF-κB) p65, and inhibitor of Kappa B kinase α/β (IKKα/β), suppressed the content of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), and enhanced the expression of tight junction proteins. Moreover, cimigenoside, caffeic acid, and methyl caffeate were identified as the bioactive constituents responsible for the UC treatment effects of C. heracleifolia. CONCLUSIONS In summary, this study is the first to demonstrate that C. heracleifolia exerts therapeutic effects on UC by enhancing the intestinal mucosal barrier and inhibiting the phosphatidylinositol 3-kinase (PI3K)/AKT/NF-κB signaling pathway. These findings offer valuable insights into the clinical application of C. heracleifolia for UC management.
Collapse
Affiliation(s)
- Xue-Yi Wu
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yong-Bo Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Jia-Xin Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Mei-Qing Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - De-Qin Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
20
|
Bae SM, Hyun CG. Insights on the Anti-Inflammatory and Anti-Melanogenic Effects of 2'-Hydroxy-2,6'-dimethoxychalcone in RAW 264.7 and B16F10 Cells. Curr Issues Mol Biol 2025; 47:85. [PMID: 39996806 PMCID: PMC11854153 DOI: 10.3390/cimb47020085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Chalcones are recognized for their diverse pharmacological properties, including anti-inflammatory and anti-melanogenic effects. However, studies on 2'-hydroxy-2-methoxychalcone derivatives remain limited. This study investigated the anti-inflammatory and melanin synthesis-inhibitory effects of three derivatives: 2'-hydroxy-2,4-dimethoxychalcone (2,4-DMC), 2'-hydroxy-2,5'-dimethoxychalcone (2,5'-DMC), and 2'-hydroxy-2,6'-dimethoxychalcone (2,6'-DMC). In lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages, 2,6'-DMC demonstrated a superior inhibition of nitric oxide (NO) production, pro-inflammatory cytokines, and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) compared to the other derivatives. A mechanistic analysis revealed that 2,6'-DMC modulates the NF-κB and MAPK signaling pathways to attenuate inflammation. Additionally, 2,6'-DMC exhibited a significant inhibition of α-melanocyte-stimulating hormone (α-MSH)-induced melanin synthesis in B16F10 melanoma cells by downregulating tyrosinase, TRP-1, TRP-2, and MITF expression. This regulation was achieved through the suppression of the Wnt/β-catenin, PI3K/AKT, MAPK, and PKA/CREB pathways. Compared to 2,4-DMC and 2,5'-DMC, 2,6'-DMC's structural configuration, characterized by methoxy groups at the 2- and 6'-positions, contributed to its enhanced molecular stability and binding affinity, amplifying its inhibitory effects. A primary skin irritation test confirmed that 2,6'-DMC exhibited minimal irritation, demonstrating its safety for dermal applications. These findings suggest that 2,6'-DMC holds promise as a dual-function agent for managing inflammatory conditions and hyperpigmentation-related disorders.
Collapse
Affiliation(s)
| | - Chang-Gu Hyun
- Department of Chemistry and Cosmetics, Jeju Inside Agency and Cosmetic Science Center, Jeju National University, Jeju 63243, Republic of Korea;
| |
Collapse
|
21
|
Lu Z, Li H, Song N, Wang Z, Wang H, Rehman A, Han L, Zeng KW. Therapeutic Potential of Carbon Dots Derived from Phytochemicals as Nanozymes Exhibiting Superoxide Dismutase Activity for Anemia. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4562-4578. [PMID: 39792367 DOI: 10.1021/acsami.4c17885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Anemia is a potentially life-threatening blood disorder caused by an insufficient erythroblast volume in the circulatory system. Self-renewal failure of erythroblast progenitors is one of the key pathological factors leading to erythroblast deficiency. However, there are currently no effective drugs that selectively target this process. In this work, we present a carbon dot (CP-CDs) derived from phytochemicals that significantly promotes the self-renewal of erythroblast progenitors for anemia therapy. As a superoxide dismutase (SOD)-like nanozyme, CP-CDs significantly activate the hypoxia response and JAK/STAT3 pathways in erythroid cells by reprogramming the oxidative stress state. This results in unique erythropoiesis-enhancing properties by promoting the generation of erythroid progenitor cells. Moreover, CP-CDs protect mature red blood cells by inhibiting oxidative stress-induced damage and improving the immune-inflammatory microenvironment. In vivo, CP-CDs showed a promising therapeutic effect in mouse and zebrafish models of anemia with minimal adverse effects, indicating significant translational medical value. Collectively, this study not only illustrates a successful approach for nanomedicine-enhanced anemia therapy but also enhances our understanding of the interaction between nanomedicine and the self-renewal of erythroblast progenitors.
Collapse
Affiliation(s)
- Zhiyuan Lu
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Haojia Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Nannan Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Zhiwei Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, State Key Laboratory of Advanced Drug Delivery and Release Systems, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Hua Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Asma Rehman
- National Institute for Biotechnology & Genetic Engineering College Pakistan Institute of Engineering & Applied Sciences (NIBGE-C, PIEAS), Faisalabad 38000, Pakistan
| | - Liwen Han
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
22
|
Fujii A, Onishi S, Watanabe N, Kajimura M, Ito K, Minamisaka K, Nishidono Y, Shirako S, Ikeya Y, Nishizawa M. Crude Drugs for Clearing Heat Contain Compounds Exhibiting Anti-Inflammatory Effects in Interleukin-1β-Treated Rat Hepatocytes. Molecules 2025; 30:416. [PMID: 39860285 PMCID: PMC11768083 DOI: 10.3390/molecules30020416] [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: 12/12/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Traditional Japanese medicines, i.e., Kampo medicines, consist of crude drugs (mostly plants) that have empirical pharmacological functions ('Yakuno' in Japanese), such as clearing heat. Crude drugs with cold properties, such as Phellodendron bark, have the empirical function of clearing heat as they cool the body. Because we found that anti-inflammatory compounds were present in several crude drugs for clearing heat, it is speculated that the empirical function of clearing heat may be linked to anti-inflammatory activities. When 10 typical crude drugs were selected from 22 herbal crude drugs for clearing heat, we identified anti-inflammatory compounds in five crude drugs, including Phellodendron bark. In this study, the other crude drugs were extracted and partitioned with ethyl acetate (EtOAc) and n-butanol to obtain three crude fractions. All the EtOAc-soluble fractions, except that from Forsythia fruits, inhibited interleukin (IL)-1β-induced nitric oxide (NO) production in primary-cultured rat hepatocytes. Anti-inflammatory compounds were identified from these EtOAc-soluble fractions: baicalein from Scutellaria roots, (-)-nyasol from Anemarrhena rhizomes, and loniflavone from Lonicera leaves and stems. (+)-Phillygenin was purified from Forsythia fruits by removing cytotoxic oleanolic and betulinic acids. These compounds suppressed the production of NO and cytokines in hepatocytes. Anti-inflammatory compounds were not purified from the EtOAc-soluble fraction of Rehmannia roots because of their low abundance. Collectively, these findings indicate that anti-inflammatory compounds are present in all 10 crude drugs for clearing heat, confirming that these anti-inflammatory compounds in crude drugs provide the empirical functions for clearing heat. Other empirical functions of Kampo medicine can also be explained by modern pharmacological activities.
Collapse
Affiliation(s)
- Airi Fujii
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Saki Onishi
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Nodoka Watanabe
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Mizuki Kajimura
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Kentaro Ito
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Keita Minamisaka
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Yuto Nishidono
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (Y.N.); (Y.I.)
| | - Saki Shirako
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
| | - Yukinobu Ikeya
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (Y.N.); (Y.I.)
- Faculty of Pharmacy, Daiichi University of Pharmacy, Fukuoka 815-8511, Fukuoka, Japan
| | - Mikio Nishizawa
- Department of Medical Biosciences, Faculty of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (A.F.); (S.S.)
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Brawijaya, Malang 65113, East Java, Indonesia
| |
Collapse
|
23
|
Guo J, Zhang Y, Du Y, Chen Y, Zhao X, Yu B, Cui T, Mao H, Lv B, Wang X, Gao X. Perilla frutescens leaf extracts alleviate acute lung injury in mice by inhibiting KAT2A. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118730. [PMID: 39181280 DOI: 10.1016/j.jep.2024.118730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) can lead to respiratory failure and even death. KAT2A is a key target to suppress the development of inflammation. A herb, perilla frutescens, is an effective treatment for pulmonary inflammatory diseases with anti-inflammatory effects; however, its mechanism of action remains unclear. AIM OF THE STUDY The purpose of this study was to investigate the therapeutic effect and underlying mechanism of perilla frutescens leaf extracts (PLE), in the treatment of ALI by focusing on its ability to treat inflammation. MATERIALS AND METHODS In vivo and in vitro models of ALI induced by LPS. Respiratory function, histopathological changes of lung, and BEAS-2B cells damage were assessed upon PLE. This effect is also tested under conditions of KAT2A over expression and KAT2A silencing. RESULTS PLE significantly attenuated LPS-induced histopathological changes in the lungs, improved respiratory function, and increased survival rate from LPS stimuation background in mice. PLE remarkably suppressed the phosphorylation of STAT3, AKT, ERK (1/2) and the release of cytokines (IL-6, TNF-α, and IL-1β) induced by LPS via inhibiting the expression of KAT2A. CONCLUSIONS PLE has a dose-dependent anti-inflammatory effect by inhibiting KAT2A expression to suppress LPS-induced ALI n mice. Our study expands the clinical indications of the traditional medicine PLE and provide a theoretical basis for clinical use of acute lung injury.
Collapse
Affiliation(s)
- Jinhe Guo
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuqi Zhang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yaodong Du
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yang Chen
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xin Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tianyi Cui
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haoping Mao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Lv
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xiaoying Wang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xiumei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
24
|
Jia L, Ma M, Xiong W, Zhu J, Cai Y, Chen Y, Jin J, Gao M. Evaluating the Anti-inflammatory Potential of JN-KI3: The Therapeutic Role of PI3Kγ-Selective Inhibitors in Asthma Treatment. Inflammation 2025:10.1007/s10753-024-02180-6. [PMID: 39776396 DOI: 10.1007/s10753-024-02180-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/16/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025]
Abstract
Asthma is a chronic airway inflammatory disease of the airways characterized by the involvement of numerous inflammatory cells and factors. Therefore, targeting airway inflammation is one of the crucial strategies for developing novel drugs in the treatment of asthma. Phosphoinositide 3-kinase gamma (PI3Kγ) has been demonstrated to have a significant impact on inflammation and immune responses, thus emerging as a promising therapeutic target for airway inflammatory disease, including asthma. There are few studies reporting on the therapeutic effects of PI3Kγ-selective inhibitors in asthma disease. In this study, we investigated the anti-inflammatory and therapeutic effects of PI3Kγ-selective inhibitor JN-KI3 for treating asthma by utilizing both in vivo and in vitro approaches, thereby proving that PI3Kγ-selective inhibitors could be valuable in the treatment of asthma. In RAW264.7 macrophages, JN-KI3 effectively suppressed C5a-induced Akt phosphorylation in a concentration-dependent manner, with no discernible toxicity observed in RAW264.7 cells. Furthermore, JN-KI3 can inhibit the PI3K/Akt signaling pathway in lipopolysaccharide-induced RAW264.7 cells, leading to the suppression of transcription and expression of the classical inflammatory cytokines in a concentration-dependent manner. Finally, an ovalbumin-induced murine asthma model was constructed to evaluate the initial therapeutic effect of JN-KI3 for treating asthma. Oral administration of JN-KI3 inhibited the infiltration of inflammatory cells and the expression of T-helper type 2 cytokines in bronchoalveolar lavage fluid, which was associated with the suppression of the PI3K signaling pathway. Lung tissue and immunohistochemical studies demonstrated that JN-KI3 inhibited the accumulation of inflammatory cells around the bronchus and blood vessels, as well as the secretion of mucus and excessive deposition of collagen around the airway. In addition, it reduced the infiltration of white blood cells into the lungs. In summary, JN-KI3 shows promise as a candidate for the treatment of asthma. Our study also suggests that the inhibitory effects of PI3Kγ on inflammation could offer an additional therapeutic strategy for pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Lei Jia
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Mengyun Ma
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wendian Xiong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| | - Mingzhu Gao
- Department of Clinical Research Center for Wuxi No.2 People's Hospital, Jiangnan University Medical Center, Wuxi, 214000, Jiangsu, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
25
|
Kvetkina AN, Klimovich AA, Deriavko YV, Pislyagin EA, Menchinskaya ES, Bystritskaya EP, Isaeva MP, Lyukmanova EN, Shenkarev ZO, Aminin DL, Leychenko EV. Sea Anemone Kunitz Peptide HCIQ2c1 Reduces Histamine-, Lipopolysaccharide-, and Carrageenan-Induced Inflammation via the Suppression of Pro-Inflammatory Mediators. Int J Mol Sci 2025; 26:431. [PMID: 39796283 PMCID: PMC11721031 DOI: 10.3390/ijms26010431] [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: 11/09/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025] Open
Abstract
Inflammation is a physiological response of the immune system to infectious agents or tissue injury, which involves a cascade of vascular and cellular events and the activation of biochemical pathways depending on the type of harmful agent and the stimulus generated. The Kunitz peptide HCIQ2c1 of sea anemone Heteractis magnifica is a strong protease inhibitor and exhibits neuroprotective and analgesic activities. In this study, we investigated the anti-inflammatory potential of HCIQ2c1 in histamine- and lipopolysaccharide (LPS)-activated RAW 264.7 macrophages as well as in LPS-induced systemic inflammation and carrageenan-induced paw edema models in CD-1 mice. We found that 10 μM HCIQ2c1 dramatically decreases histamine-induced intracellular Ca2+ release and LPS-induced reactive oxygen species (ROS) production in RAW 264.7 macrophages. Moreover, HCIQ2c1 significantly inhibited the production of LPS-induced tumor necrosis factor α (TNF-α), inducible NO-synthase (iNOS), and 5-lipoxygenase (5-LO) but slightly influenced the IL-1β and cyclooxygenase-2 (COX-2) expression level in macrophages. Furthermore, intravenous administration by HCIQ2c1 at 0.1 mg/kg dose reduced LPS-induced TNF-α, IL-1β, COX-2, and iNOS gene expression in CD-1 mice. The subplantar administration of HCIQ2c1 at 0.1 mg/kg dose to mice significantly reduced carrageenan-induced paw edema by a factor of two, which is comparable to the effect of diclofenac at 1 mg/kg dose. Thus, peptide HCIQ2c1 has a strong anti-inflammatory potential by the attenuation of systemic and local inflammatory effects through the inhibition of intracellular Ca2+ release, the production of ROS and pro-inflammatory cytokines, and enzymes involved in arachidonic acid metabolism.
Collapse
Affiliation(s)
- Aleksandra N. Kvetkina
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
- Laboratory of Structural Biology of Ion Channels, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| | - Anna A. Klimovich
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
| | - Yulia V. Deriavko
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
| | - Evgeniy A. Pislyagin
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
| | - Ekaterina S. Menchinskaya
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
| | - Evgenia P. Bystritskaya
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
| | - Marina P. Isaeva
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
| | - Ekaterina N. Lyukmanova
- Biological Department, Shenzhen MSU-BIT University, Shenzhen 518172, China;
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University “Molecular Technologies of the Living Systems and Synthetic Biology” Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Zakhar O. Shenkarev
- Laboratory of Structural Biology of Ion Channels, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Dmitriy L. Aminin
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Elena V. Leychenko
- Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (A.N.K.); (A.A.K.); (Y.V.D.); (E.A.P.); (E.S.M.); (E.P.B.); (M.P.I.); (D.L.A.)
- Laboratory of Structural Biology of Ion Channels, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 119997 Moscow, Russia
| |
Collapse
|
26
|
Shen Y, Nakajima H, Zhu J, Wu W. Integrin β2 regulates titanium particle‑induced inflammation in macrophages: In vitro aseptic loosening model. Mol Med Rep 2025; 31:25. [PMID: 39540364 DOI: 10.3892/mmr.2024.13390] [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: 06/15/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
Aseptic loosening is a major complication of joint replacement surgery, characterized by periprosthetic osteolysis and chronic inflammation at the bone‑implant interface. Cells release chemokines, cytokines and other pro‑inflammatory substances that perpetuate inflammation reactions, while other particle‑stimulated macrophages promote osteoclastic bone resorption and impair bone formation. The present study investigated integrin and inflammatory cytokine expression patterns in RAW 264.7 cells treated with titanium (Ti) particles to elucidate the role of integrins in Ti particle‑mediated inflammatory osteolysis. Assessment was performed by reverse transcription‑quantitative PCR, western blotting, confocal immunofluorescence, flow cytometry and enzyme‑linked immunosorbent assays. Cell migration was evaluated by wound healing assay. It was found that Ti particles significantly induced integrin expression in RAW 264.7 cells, including upregulation of integrins β2 (CD18), aL (CD11a), aM (CD11b) and aX (CD11c). Ti particles also enhanced the expression of Toll‑like receptors (TLRs; TLR1, TLR2, TLR3 and TLR4) and triggered the release of inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)‑1β, IL‑8 and IL‑12. Proteomics showed higher expression and activity levels of TLR2 and TLR4, along with their downstream signaling adaptors myeloid differentiation primary response protein 88 (MyD88) and Mal/TIR‑domain‑containing adapter protein (TIRAP), following Ti treatment. Additionally, Ti treatment significantly enhanced the migration rate of RAW 264.7 cells. The present findings indicated that Ti particles regulate the inflammatory response of RAW 264.7 cells in an in vitro aseptic loosening model by activating the TLR/TIRAP/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Yue Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Haruna Nakajima
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113‑8654, Japan
| | - Junfeng Zhu
- Department of Orthopedics Surgery, Suichang Branch of The Second Affiliated Hospital, Zhejiang University School of Medicine (Suichang County People's Hospital in Zhejiang), Lishui, Zhejiang 323300, P.R. China
| | - Weigang Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
27
|
Mao N, Yu Y, Lu X, Yang Y, Liu Z, Wang D. Preventive effects of matrine on LPS-induced inflammation in RAW 264.7 cells and intestinal damage in mice through the TLR4/NF-κB/MAPK pathway. Int Immunopharmacol 2024; 143:113432. [PMID: 39447411 DOI: 10.1016/j.intimp.2024.113432] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Matrine is a tetracyclic quinolizidine alkaloid with diverse bioactive properties, including anti-inflammatory and neuroprotective properties. However, the underlying anti-inflammatory mechanisms remain unclear. PURPOSE This study aimed to explore how matrine reduces Lipopolysaccharide (LPS)-induced inflammation in RAW 264.7 cells and to assess its protective effects against LPS-induced intestinal damage. METHODS The effect of matrine on cell viability was assessed using the cell counting kit-8 (CCK-8) assay. Additionally, its impact on inflammatory cytokines and macrophage polarization was assessed using enzyme-linked immunosorbent assay (ELISA), flow cytometry, and quantitative real-time polymerase chain reaction (qRT-PCR) analyses. The effects on intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP), nitric oxide (NO) production, and oxidative stress were evaluated using 2',7'-dichlorodihydrofluorescein diacetate staining and JC-1 and Griess assays. Immunofluorescence staining was used to observe the translocation of the NF-κB p65 subunit. Western blotting (WB) and qRT-PCR were employed to analyze the expression levels of proteins related to the toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB)/mitogen-activated protein kinase (MAPK) pathway. An LPS-induced mouse model was established to study the intestinal inflammation and barrier injury. Mouse feces characteristics, colon length, and disease activity index (DAI) were recorded. Hematoxylin-eosin (H&E) and alcian blue/periodic acid schiff (AB/PAS) staining were used to observe morphological changes and barrier damage in the duodenum, jejunum, ileum, and colon and to measure villus length, crypt depth, goblet cell count, and positive areas in the duodenum, jejunum, and ileum. The content of short-chain fatty acids (SCFAs) in the colon was determined using gas chromatography (GC). RESULTS Matrine inhibited LPS-induced inflammatory cytokine levels, suppressed macrophage M1 polarization, and promoted M2 macrophage polarization. Matrine reduced LPS-induced increases in ROS and NO levels and regulates oxidative stress. Additionally, matrine inhibited the nuclear translocation of the NF-κB p65 subunit and exerted anti-inflammatory effects by suppressing the activation of the TLR4/NF-κB/MAPK pathway. In vivo experiments indicated that matrine significantly alleviated LPS-induced diarrhea, increased DAI, and shortened the colon. Matrine reduced the production of the pro-inflammatory cytokine interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α and the pro-inflammatory mediator NO in mouse intestinal tissues while promoting the content of the anti-inflammatory cytokine IL-10. Furthermore, it improved intestinal tissue structure and alleviated LPS-induced intestinal barrier damage. Finally, matrine increased the SCFA levels in the intestine. CONCLUSION Matrine exerted its anti-inflammatory effects and protects against intestinal injury through the TLR4/NF-κB/MAPK signaling pathway.
Collapse
Affiliation(s)
- Ningning Mao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaming Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
28
|
Hou B, Li C, Yang F, Deng W, Hu C, Liu C, Chen Y, Xiao X, Huang X, Deng J, Xie S. Ultrasmall Antioxidant Copper Nanozyme to Enhance Stem Cell Microenvironment for Promoting Diabetic Wound Healing. Int J Nanomedicine 2024; 19:13563-13578. [PMID: 39720217 PMCID: PMC11668326 DOI: 10.2147/ijn.s487647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Purpose Stem cell therapy is a promising approach for treating chronic diabetic wounds. However, its effectiveness is significantly limited by the high oxidative stress environment and persistent inflammation induced by diabetes. Strategies to overcome these challenges are essential to enhance the therapeutic potential of stem cell therapy. Methods Cu5.4O ultrasmall nanoparticles (Cu5.4O-USNPs), known for their excellent reactive oxygen species (ROS) scavenging properties, were utilized to protect adipose-derived stem cells (ADSCs) from oxidative stress injury. In vitro experiments were conducted to evaluate the viability, paracrine activity, and anti-inflammatory capabilities of ADSCs loaded with Cu5.4O-USNPs under oxidative stress conditions. In vivo experiments in diabetic mice were performed to assess the therapeutic effects of Cu5.4O-USNP-loaded ADSCs on wound healing, including their impact on inflammation, collagen synthesis, angiogenesis, and wound closure. Results ADSCs treated with Cu5.4O-USNPs showed significantly enhanced viability, paracrine activity, and anti-inflammatory properties under oxidative stress conditions in vitro. In diabetic mice, Cu5.4O-USNP-loaded ADSCs reduced inflammatory responses in wound tissues, promoted collagen synthesis and angiogenesis, and accelerated diabetic wound healing. These findings suggest that Cu5.4O-USNPs effectively mitigate the adverse effects of oxidative stress and inflammation, enhancing the therapeutic efficacy of ADSCs. Conclusion This study presents a simple and effective approach to improve the therapeutic potential of stem cell therapy for diabetic wounds. By incorporating Cu5.4O-USNPs, the antioxidative and anti-inflammatory capabilities of ADSCs are significantly enhanced, offering a promising strategy for ROS-related tissue repair and chronic wound healing.
Collapse
Affiliation(s)
- Biao Hou
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chengyuan Li
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, People’s Republic of China
| | - Fen Yang
- Department of Infectious Diseases, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Wanjun Deng
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chao Hu
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Changxiong Liu
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Yanming Chen
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Xiangjun Xiao
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Xiongjie Huang
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Jun Deng
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Medical University, Chongqing, People’s Republic of China
| | - Songlin Xie
- Department of Hand and Foot Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
29
|
Wang J, An W, Wang Z, Zhao Y, Han B, Tao H, Wang J, Wang X. Vanillin Has Potent Antibacterial, Antioxidant, and Anti-Inflammatory Activities In Vitro and in Mouse Colitis Induced by Multidrug-Resistant Escherichia coli. Antioxidants (Basel) 2024; 13:1544. [PMID: 39765873 PMCID: PMC11673545 DOI: 10.3390/antiox13121544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/08/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
A large number of cases of infectious colitis caused by multidrug-resistant (MDR) bacteria, such as Escherichia coli, can result in colon damage and severe inflammation. Vanilla, a widely utilized flavor and fragrance compound, is extensively used in various food. However, the effect of vanilla on MDR E. coli-induced infectious colitis has received less attention. In this study, the antibacterial activity of vanillin against MDR E. coli and other bacteria was determined by the microtiter broth dilution method. The antioxidant and anti-inflammatory capacity of vanillin was assessed in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells and MDR E. coli-induced mouse colitis. The results demonstrated that vanillin exhibited potent antibacterial activity against various strains of MDR E. coli, Salmonella, and Staphylococcus aureus, with a minimal inhibitory concentration (MIC) of 1.25-2.5 mg/mL and a minimum bactericidal concentration (MBC) of 5-10 mg/mL; it effectively inhibited cell division in E. coli. Vanillin also displayed remarkable antioxidant activity by suppressing the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and reactive oxygen species (ROS) in LPS-stimulated RAW 264.7 cell; it significantly reduced the production of inflammatory mediators including nitroxide (NO), tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and interleukin 1β (IL-1β), while increasing interleukin 10 (IL-10). In an MDR E. coli-induced mouse colitis model, vanillin effectively inhibited inflammation by suppressing inflammatory cytokines, mitogen-activated protein kinase (MAPK), and nuclear factor κ-B (NF-κB) cell signaling pathway activation; it ameliorated changes in intestinal microflora characterized by decreased Firmicutes richness alongside increased Bacteroides richness, rebalancing the dysbiosis caused by E. coli. These findings highlight the potential pharmacological applicability of vanillin as a promising bioactive molecule for treating infectious colitis.
Collapse
Affiliation(s)
- Jiaxue Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
- College of Grassland Agriculture, Northwest A&F University, Yangling 712100, China
| | - Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ya Zhao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Bing Han
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Hui Tao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (J.W.); (W.A.); (Y.Z.); (B.H.); (H.T.); (J.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
30
|
Qin Y, Li K, Zhang Q, Liu J, Xie Y, Zhang T, Wang X, Zhang L, Jiang Y, Liu G. Linoleic acid inhibits lipopolysaccharide-induced inflammation by promoting TLR4 regulated autophagy in murine RAW264.7 macrophages. J Appl Biomed 2024; 22:185-196. [PMID: 40033806 DOI: 10.32725/jab.2024.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/21/2024] [Indexed: 03/05/2025] Open
Abstract
Linoleic acid (LA), an essential fatty acid, has emerged as a pivotal regulator in disorders associated with inflammation in recent years; however, the underlying mechanisms are still not completely understood. We utilized network pharmacology and experimental methodologies to elucidate the mechanisms underlying the anti-inflammatory effects of LA. Our network pharmacology analysis revealed that LA shares common targets with sepsis. These targets are enriched in various pathways comprising C-type signaling pathway, PI3K-Akt signaling pathway, toll-like receptor signaling pathway, neutrophil extracellular trap formation, AMPK signaling pathway, and autophagy-animal. These findings suggest that LA may exert regulatory effects on inflammation and autophagy during sepsis. Subsequently, we established in vivo and ex vivo models of sepsis using lipopolysaccharide (LPS) in experimental study. Treatment with LA reduced lung damage in mice with LPS-induced lung injury, and reduced tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in plasma, bronchoalveolar lavage fluid (BALF), and peritoneal lavage fluid (PLF). LA also decreased the production of TNF-α and IL-6 in RAW264.7 macrophages exposed to LPS. In LPS-induced RAW264.7 macrophages, LA induced an elevation in LC3-II while causing a reduction in p62, which was associated with downregulation of toll-like receptor 4 (TLR4). We utilized 3-methyladenine (3-MA) to inhibit the autophagic activity, which reversed the modulatory effects of LA on LC3-II and p62. 3-MA also prevented the decline in TLR4 expression along with reduction in pro-inflammatory cytokines secretion. Our findings suggest that the activation of autophagy by LA may lead to the downregulation of TLR4, thereby exerting its anti-inflammatory effects.
Collapse
Affiliation(s)
- Yin Qin
- University-Town Hospital of Chongqing Medical University, Department of Emergency and Critical Care Medicine, Chongqing 401331, China
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Kexin Li
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Qiuhong Zhang
- University-Town Hospital of Chongqing Medical University, Department of Emergency and Critical Care Medicine, Chongqing 401331, China
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Jie Liu
- University-Town Hospital of Chongqing Medical University, Department of Emergency and Critical Care Medicine, Chongqing 401331, China
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Yu Xie
- University-Town Hospital of Chongqing Medical University, Department of Emergency and Critical Care Medicine, Chongqing 401331, China
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Tingting Zhang
- University-Town Hospital of Chongqing Medical University, Department of Emergency and Critical Care Medicine, Chongqing 401331, China
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Xiaoliang Wang
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| | - Li Zhang
- Chongqing Medical University, Department of Pathophysiology, Chongqing 400016, China
| | - Yu Jiang
- University-Town Hospital of Chongqing Medical University, Department of Respiratory and Critical Care Medicine, Chongqing 401331, China
| | - Gang Liu
- University-Town Hospital of Chongqing Medical University, Department of Emergency and Critical Care Medicine, Chongqing 401331, China
- University-Town Hospital of Chongqing Medical University, Medical Sciences Research Center, Chongqing 401331, China
| |
Collapse
|
31
|
Song M, Lim KM, Song K, Kang GH, Kim SJ, Lee Y, Yu S, Jeong KH, Cho SG. Efficient Treatment of Psoriasis Using Conditioned Media from Mesenchymal Stem Cell Spheroids Cultured to Produce Transforming Growth Factor- β1-Enriched Small-Sized Extracellular Vesicles. Int J Stem Cells 2024; 17:407-417. [PMID: 39396918 PMCID: PMC11612221 DOI: 10.15283/ijsc24089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 10/15/2024] Open
Abstract
Psoriasis is a common chronic inflammatory disease in which keratinocytes proliferate abnormally due to excessive immune action. Psoriasis can be associated with various comorbidities and has a significant impact on health-related quality of life. Although many systemic treatments, including biologic agents, have been developed, topical treatment remains the main option for psoriasis management. Consequently, there is an urgent need to develop topical treatments with minimal side effects and high efficacy. Mesenchymal stem cells (MSCs) exhibit excellent immune regulation, anti-inflammatory activities, and therapeutic effects, and MSC-derived extracellular vesicles (EVs) can serve as crucial mediators of functional transfer from MSCs. Therefore, this study aimed to develop a safe and easy-to-use emulsion cream for treating psoriasis using MSC conditioned media (CM) containing EVs. We developed an enhanced Wharton's jelly MSC (WJ-MSC) culture method through a three-dimensional (3D) culture containing exogenous transforming growth factor-β3. Using the 3D culture system, we obtained CM from WJ-MSCs, which yielded a higher EV production compared to that of conventional WJ-MSC culture methods, and investigated the effect of EV-enriched 3D-WJ-MSC-CM cream on psoriasis-related inflammation. Administration of the EV-enriched 3D-WJ-MSC-CM cream significantly reduced erythema, thickness, and scaling of skin lesions, alleviated imiquimod-induced psoriasiform lesions in mice, and ameliorated histopathological changes in mouse skin. The upregulated mRNA expression of inflammatory cytokines, including IL-17a, IL-22, IL-23, and IL-36, decreased in the lesions. In conclusion, we present here a new topical treatment for psoriasis using an MSC EV-enriched cream.
Collapse
Affiliation(s)
- Myeongjin Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Kwonwoo Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Geun-Ho Kang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Se Jong Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Youngseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Sujin Yu
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Ki-Heon Jeong
- Department of Dermatology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| |
Collapse
|
32
|
Tolle G, Serreli G, Deiana M, Moi L, Zavattari P, Pantaleo A, Manis C, El Faqir MA, Caboni P. Lipidomics of Caco-2 Cells Under Simulated Microgravity Conditions. Int J Mol Sci 2024; 25:12638. [PMID: 39684348 DOI: 10.3390/ijms252312638] [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: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Microgravity may profoundly impact the cardiovascular system, skeletal muscle system, and immune system of astronauts. At the cellular level, microgravity may also affect cell proliferation, differentiation, and growth, as well as lipid metabolism. In this work, we investigated lipid changes in Caco-2 cells cultured in a clinostat for 24 h under simulated microgravity conditions (SMC). Complex lipids were measured using a UHPLC-QTOF/MS platform, and the data were subjected to multivariate analysis. Under SMC, levels of ceramides Cer 18:0;O2/16:0, Cer 18:1;O2/16:0, Cer 18:1; O2/22:0, Cer 18:1;O2/24:0, and Cer 18:2;O2/24:0 were found to be upregulated, while sphingomyelins SM 16:1;O2/16:0, SM 16:1;O2/18:1, SM 18:1;O2/24:0, and SM 18:2;O2/24:0 were found to be downregulated. On the other hand, considering that sphingolipids are involved in the process of inflammation, we also treated Caco-2 cells with dextran sodium sulfate (DSS) to induce cell inflammation and lipopolysaccharide (LPS) to induce cell immune responses. As a result, we observed similar lipid dysregulation, indicating that SMC may exert a condition similar to inflammation. Our lipidomics strategy provides new insights into the altered metabolic pathway of ceramides and sphingomyelins of Caco-2 cells under SMC.
Collapse
Affiliation(s)
- Giulia Tolle
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Gabriele Serreli
- Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Monica Deiana
- Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Loredana Moi
- Unit of Biology and Genetics, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Patrizia Zavattari
- Unit of Biology and Genetics, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Cristina Manis
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Mohammed Amine El Faqir
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
33
|
Rahmawati SI, Indriani DW, Ningsih FN, Hardhiyuna M, Firdayani F, Ahmadi P, Rosyidah A, Septiana E, Dharmayanti NLPI, Bayu A, Putra MY. Dual anti-inflammatory activities of COX-2/5-LOX driven by kratom alkaloid extracts in lipopolysaccharide-induced RAW 264.7 cells. Sci Rep 2024; 14:28993. [PMID: 39578527 PMCID: PMC11584675 DOI: 10.1038/s41598-024-79229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Cyclooxygenase (COX) and lipoxygenase (LOX) enzymes play a pivotal role in producing pro-inflammatory eicosanoids, including prostaglandins (PGs) and leukotrienes (LTs), in the inflammation process. Mitragynine is a primary alkaloid contained in the kratom's leaves and has been reported to show anti-inflammatory activity by suppressing COX-2 mRNA translation to lowering PGs synthesis. In this study, the Kratom's alkaloid extract containing ~ 46% mitragynine was found to exhibit dual inhibition activity towards COX-2/5-LOX enzymes at concentrations below 25 ppm in the LPS-induced RAW 264.7 macrophage cells. At these levels, no cell toxicity was observed while the cells became death (e.g., 10-46% viability at 50-100 ppm) and only COX-2 inhibition activity was observed after exposed with more than 25 ppm of alkaloid extract. In contrast, the methanolic-crude extract of Kratom's leaf containing ~ 5% mitragynine showed no inhibition toward COX-2/5-LOX enzymes and did not toxic onto the cells, even after treated at 100 ppm. The alkaloid extract suppressed several antiinflammation parameters, including ROS (64% reduction at 25 ppm), NO (30% reduction at 25 ppm), TNF-α (~ 50% reduction at 25 ppm), and IL-6 production (60% reduction at 6.25 ppm). In silico molecular studies indicated strong binding affinity of Kratom alkaloids to COX-2 and 5-LOX active sites, supporting the Kratom's alkaloids to have great potential dual inhibition activity towards COX-2/5-LOX enzymes and to be developed as a safer NSAIDs with fewer side effects.
Collapse
Affiliation(s)
- Siti Irma Rahmawati
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia.
| | - Dwi Wahyu Indriani
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia.
| | - Febby Nurdiya Ningsih
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Mutia Hardhiyuna
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Firdayani Firdayani
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Peni Ahmadi
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - A'liyatur Rosyidah
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Eris Septiana
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Ni Luh Putu Indi Dharmayanti
- Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Asep Bayu
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia
| | - Masteria Yunovilsa Putra
- Research Center for Vaccine and Drugs, Research Organization for Health, National Research and Innovation Agency (BRIN), Jalan Raya Jakarta-Bogor Km. 46, 16911, Jakarta, West Java, Indonesia.
- Faculty of Pharmacy, Universitas Indonesia, Jalan Prof. DR. Mahar Mardjono, Pondok Cina, Beji, Depok, 16424, Jakarta, West Java, Indonesia.
- National Metabolomics Collaborative Research Center, Universitas Indonesia, Kampus UI, Depok, 16424, Jakarta, West Java, Indonesia.
| |
Collapse
|
34
|
Martínez Fajardo C, Morote L, Moreno-Giménez E, López-López S, Rubio-Moraga Á, Díaz-Guerra MJM, Diretto G, López Jiménez AJ, Ahrazem O, Gómez-Gómez L. Exosome-like nanoparticles from Arbutus unedo L. mitigate LPS-induced inflammation via JAK-STAT inactivation. Food Funct 2024; 15:11280-11290. [PMID: 39474636 DOI: 10.1039/d4fo03646h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
Exosomes have garnered attention as a potential cell-free therapy for inflammatory diseases due to their immunomodulatory and anti-inflammatory properties. Exosome-like nanoparticles isolated from Arbutus unedo were characterized and analyzed for their anti-inflammatory potential. The results revealed that the isolated exosomes exhibited a spheroid morphology, with an approximate modal size of 190 nm. Exposure to these exosomes significantly reduced the mRNA expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), the glycoprotein CD80, the transcription factor STAT1, and pro-inflammatory cytokine genes like IL-1b and IL-6, in lipopolysaccharide (LPS)-induced protein RAW264.7 cells. In addition, exosomes reduced the LPS-induced protein levels of PSTAT1 and STAT1, IRF1 and RelB, which are key transcription factors in the control of proinflammatory gene expression. LC-MS analysis identified the presence of carotenoids, mainly β-carotene, with known anti-inflammatory activity, related to its ROS-scavenging activity, suggesting its potential contribution to the anti-inflammatory activity of the exosomes isolated from A. unedo fruits.
Collapse
Affiliation(s)
- Cristian Martínez Fajardo
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
| | - Lucía Morote
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
| | - Elena Moreno-Giménez
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
| | - Susana López-López
- Unidad de Investigación, Complejo Hospitalario Universitario de Albacete, C/Laurel, s/n, 02008 Albacete, Spain
- Facultad de Medicina, Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha, C/Almansa 14, 02008 Albacete, Spain
| | - Ángela Rubio-Moraga
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
- Escuela Técnica Superior de Ingenieros Agrónomos, Montes y Biotecnología, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain
| | - María José M Díaz-Guerra
- Facultad de Medicina, Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla-La Mancha, C/Almansa 14, 02008 Albacete, Spain
| | - Gianfranco Diretto
- Italian National Agency for New Technologies, Energy, and Sustainable Development, Casaccia Research Centre, 00123 Rome, Italy
| | - Alberto J López Jiménez
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
- Escuela Técnica Superior de Ingenieros Agrónomos, Montes y Biotecnología, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain
| | - Oussama Ahrazem
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
- Escuela Técnica Superior de Ingenieros Agrónomos, Montes y Biotecnología, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain
| | - Lourdes Gómez-Gómez
- Instituto Botánico, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain.
- Facultad de Farmacia, Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, Campus Universitario s/n, 02071 Albacete, Spain
| |
Collapse
|
35
|
Fu X, Huang X, Tan H, Huang X, Nie S. Regulatory Effect of Fucoidan Hydrolysates on Lipopolysaccharide-Induced Inflammation and Intestinal Barrier Dysfunction in Caco-2 and RAW264.7 Cells Co-Cultures. Foods 2024; 13:3532. [PMID: 39593947 PMCID: PMC11592468 DOI: 10.3390/foods13223532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Fucoidan, a sulfated polysaccharide rich in fucose, is derived from brown algae and marine invertebrates. Multiple bioactivities have been shown with fucoidan, while growing attraction has emerged in its low-molecular-weight (Mw) hydrolysates. Here, the anti-inflammatory effect of fucoidan, low-Mw acidolyzed fucoidan (LMAF, <1.5 kDa), and high-Mw acidolyzed fucoidan (HMAF, 1.5-20 kDa) were investigated in vitro using lipopolysaccharide (LPS)-stimulated Caco-2 and RAW264.7 co-cultures. Fucoidan, LMAF, and HMAF with different structures exhibited varied anti-inflammatory effects. LMAF and HMAF effectively decreased the nitric oxide release of RAW264.7 cells. LMAF exhibited a competitive effect in reducing tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 levels compared to HMAF and fucoidan. Transcriptome of RAW264.7 revealed that LPS and LMAF mainly regulated the transcriptional expression of genes, including Tnf, Il6, Il1b, Junb, and Nfkb1 in the TNF signaling pathway, NF-kappa B signaling pathway, and cytokine-cytokine receptor interaction. RT-PCR results indicated that LMAF markedly reduced the LPS-elevated expression of Cxcl2, Tnf, Ccl2, Il1b, and Csf2. Moreover, LMAF effectively increased the proteins expression of Claudin-1, Occludin, and Zonula occluden-1 in Caco-2 cells. This study highlights the potential of LMAF to improve inflammation and intestinal barrier integrity, offering a foundation for further application of low-Mw fucoidan hydrolysates.
Collapse
Affiliation(s)
- Xiaodan Fu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.F.); (X.H.); (H.T.); (X.H.)
- China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang 330047, China
- Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, China
| | - Xinru Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.F.); (X.H.); (H.T.); (X.H.)
- China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang 330047, China
- Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, China
| | - Huizi Tan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.F.); (X.H.); (H.T.); (X.H.)
- China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang 330047, China
- Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, China
| | - Xiaojun Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.F.); (X.H.); (H.T.); (X.H.)
- China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang 330047, China
- Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.F.); (X.H.); (H.T.); (X.H.)
- China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang 330047, China
- Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang 330047, China
| |
Collapse
|
36
|
Zhen X, Kim J, Kang JS, Choi BJ, Park KH, Lee DS, Hong SH, Lee JH. Homology-independent targeted insertion-mediated derivation of M1-biased macrophages harbouring Megf10 and CD3ζ from human pluripotent stem cells. EBioMedicine 2024; 109:105390. [PMID: 39383607 PMCID: PMC11497429 DOI: 10.1016/j.ebiom.2024.105390] [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: 04/29/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Macrophages engineered with chimeric antigen receptors (CAR) are suitable for immunotherapy based on their immunomodulatory activity and ability to infiltrate solid tumours. However, the production and application of genetically edited, highly effective, and mass-produced CAR-modified macrophages (CAR-Ms) are challenging. METHODS Here, we used homology-independent targeted insertion (HITI) for site-directed CAR integration into the safe-harbour region of human pluripotent stem cells (hPSCs). This approach, together with a simple differentiation protocol, produced stable and highly effective CAR-Ms without heterogeneity. FINDINGS These engineered cells phagocytosed cancer cells, leading to significant inhibition of cancer-cell proliferation in vitro and in vivo. Furthermore, the engineered CARs, which incorporated a combination of CD3ζ and Megf10 (referred to as FRP5Mζ), markedly enhanced the antitumour effect of CAR-Ms by promoting M1, but not M2, polarisation. FRP5Mζ promoted M1 polarisation via nuclear factor kappa B (NF-κB), ERK, and STAT1 signalling, and concurrently inhibited STAT3 signalling even under M2 conditions. These features of CAR-Ms modulated the tumour microenvironment by activating inflammatory signalling, inducing M1 polarisation of bystander non-CAR macrophages, and enhancing the infiltration of T cells in cancer spheroids. INTERPRETATION Our findings suggest that CAR-Ms have promise as immunotherapeutics. In conclusion, the guided insertion of CAR containing CD3ζ and Megf10 domains is an effective strategy for the immunotherapy of solid tumours. FUNDING This work was supported by KRIBB Research Initiative Program Grant (KGM4562431, KGM5282423) and a Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korean government (Ministry of Science and ICT,Ministry of Health and Welfare) (22A0304L1-01).
Collapse
Affiliation(s)
- Xing Zhen
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea; Department of Nanoscience and Nanotechnology, Graduate School, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Jieun Kim
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea; Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Jong Soon Kang
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Byeong Jo Choi
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Ki Hwan Park
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea.
| | - Dong-Seok Lee
- Department of Nanoscience and Nanotechnology, Graduate School, Kyungpook National University, Daegu, 41566, Republic of Korea; School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea; KW-Bio Co., Ltd, Chuncheon, 24252, South Korea.
| | - Jong-Hee Lee
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, 28116, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
37
|
Xu G, Tian C, Li Y, Fang L, Wang J, Jing Z, Li S, Chen P. Inhibition of BCAT1 expression improves recurrent miscarriage by regulating cellular dysfunction and inflammation of trophoblasts. Cell Tissue Res 2024; 398:111-121. [PMID: 39356334 DOI: 10.1007/s00441-024-03921-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Sustained or chronic inflammation in the placenta can result in placental insufficiency, leading to adverse reproductive outcomes such as pregnancy loss. Branched-chain amino acid transaminase 1 (BCAT1) expresses in the placenta and is involved in the pathological inflammatory response, but its role in recurrent miscarriage (RM) has not been fully investigated. In the present study, we delved into the effects of BCAT1 on trophoblast inflammation induced by lipopolysaccharide (LPS) and a mouse model of pregnancy loss induced by LPS. In vitro, after the HTR-8/SVneo cells were treated with LPS and BCATc inhibitor 2 (a selective BCAT inhibitor), the cell apoptosis was verified by TUNEL assay, and the activity of caspase-3 and caspase-9 was detected. Real-time PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence (IF) were used to determine the expression of inflammatory cytokines (TNF-α, IL-6, and IL-1β) and inflammasomes (NLRP3 and ASC) in LPS-treated trophoblast cells. Western blot analysis was performed to verify the expression of phospho-IκBα (p-IκBα) in cells and NF-κB p65 in the nuclei. IF staining was used to detect the nuclear translocation of NF-κB p65. The DNA binding activity of NF-κB was detected by an electrophoretic mobility shift assay (EMSA). The results demonstrated that inhibition of BCAT1 reduced trophoblast apoptosis, suppressed the release of proinflammatory cytokines, and prevented NLRP3 inflammasome activation in response to LPS. Additionally, BCAT1 inhibition blocked the activation of the NF-κB pathway in trophoblasts. This study highlights the potential therapeutic role of targeting BCAT1 in preventing adverse reproductive outcomes associated with chronic placental inflammation.
Collapse
Affiliation(s)
- Guangli Xu
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| | - Chao Tian
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Yanru Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Lei Fang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Jing Wang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Zhuqing Jing
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Simeng Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Ping Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| |
Collapse
|
38
|
Park BJ, Dhong KR, Park HJ. Cordyceps militaris Grown on Germinated Rhynchosia nulubilis (GRC) Encapsulated in Chitosan Nanoparticle (GCN) Suppresses Particulate Matter (PM)-Induced Lung Inflammation in Mice. Int J Mol Sci 2024; 25:10642. [PMID: 39408971 PMCID: PMC11477187 DOI: 10.3390/ijms251910642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Cordyceps militaris grown on germinated Rhynchosia nulubilis (GRC) exerts various biological effects, including anti-allergic, anti-inflammatory, and immune-regulatory effects. In this study, we investigated the anti-inflammatory effects of GRC encapsulated in chitosan nanoparticles (CN) against particulate matter (PM)-induced lung inflammation. Optimal CN (CN6) (CHI: TPP w/w ratio of 4:1; TPP pH 2) exhibited a zeta potential of +22.77 mV, suitable for GRC encapsulation. At different GRC concentrations, higher levels (60 and 120 mg/mL) led to increased negative zeta potential, enhancing stability. The optimal GRC concentration for maximum entrapment (31.4 ± 1.35%) and loading efficiency (7.6 ± 0.33%) of GRC encapsulated in CN (GCN) was 8 mg/mL with a diameter of 146.1 ± 54 nm and zeta potential of +30.68. In vivo studies revealed that administering 300 mg/kg of GCN significantly decreased the infiltration of macrophages and T cells in the lung tissues of PM-treated mice, as shown by immunohistochemical analysis of CD4 and F4/80 markers. Additionally, GCN ameliorated PM-induced lung tissue damage, inflammatory cell infiltration, and alveolar septal hypertrophy. GCN also decreased total cells and neutrophils, showing notable anti-inflammatory effects in the bronchoalveolar lavage fluid (BALF) from PM-exposed mice, compared to GRC. Next the anti-inflammatory properties of GCN were further explored in PM- and LPS-exposed RAW264.7 cells; it significantly reduced PM- and LPS-induced cell death, NO production, and levels of inflammatory cytokine mRNAs (IL-1β, IL-6, and COX-2). GCN also suppressed NF-κB/MAPK signaling pathways by reducing levels of p-NF-κB, p-ERK, and p-c-Jun proteins, indicating its potential in managing PM-related inflammatory lung disease. Furthermore, GCN significantly reduced PM- and LPS-induced ROS production. The enhanced bioavailability of GRC components was demonstrated by an increase in fluorescence intensity in the intestinal absorption study using FITC-GCN. Our data indicated that GCN exhibited enhanced bioavailability and potent anti-inflammatory and antioxidant effects in cells and in vivo, making it a promising candidate for mitigating PM-induced lung inflammation and oxidative stress.
Collapse
Affiliation(s)
- Byung-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
| | - Kyu-Ree Dhong
- Magicbullettherapeutics Inc., 150 Yeongdeungpo-ro, Yeongdeungpo-gu, Seoul 07292, Republic of Korea;
| | - Hye-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
| |
Collapse
|
39
|
Luo W, Zhang H, Zhang H, Xu Y, Liu X, Xu S, Wang P. Reposition: Focalizing β-Alanine Metabolism and the Anti-Inflammatory Effects of Its Metabolite Based on Multi-Omics Datasets. Int J Mol Sci 2024; 25:10252. [PMID: 39408583 PMCID: PMC11476852 DOI: 10.3390/ijms251910252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
The incorporation of multi-omics data methodologies facilitates the concurrent examination of proteins, metabolites, and genes associated with inflammation, thereby leveraging multi-dimensional biological data to achieve a comprehensive understanding of the complexities involved in the progression of inflammation. Inspired by ensemble learning principles, we implemented ID normalization preprocessing, categorical sampling homogenization, and pathway enrichment across each sample matrix derived from multi-omics datasets available in the literature, directing our focus on inflammation-related targets within lipopolysaccharide (LPS)-stimulated RAW264.7 cells towards β-alanine metabolism. Additionally, through the use of LPS-treated RAW264.7 cells, we tentatively validated the anti-inflammatory properties of the metabolite Ureidopropionic acid, originating from β-alanine metabolism, by evaluating cell viability, nitric oxide production levels, and mRNA expression of inflammatory biomarkers. In conclusion, our research represents the first instance of an integrated analysis of multi-omics datasets pertaining to LPS-stimulated RAW264.7 cells as documented in the literature, underscoring the pivotal role of β-alanine metabolism in cellular inflammation and successfully identifying Ureidopropionic acid as a novel anti-inflammatory compound. Moreover, the findings from database predictions and molecular docking studies indicated that the inflammatory-related pathways and proteins may serve as potential mechanistic targets for Ureidopropionic acid.
Collapse
Affiliation(s)
- Wenjun Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Haijun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Hao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yixi Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiao Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Ping Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (W.L.); (H.Z.); (H.Z.); (Y.X.); (S.X.)
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
40
|
Schulz M, Gonzaga LV, Antunes ACN, Lubschinski T, Mohr ETB, Dalmarco EM, Deolindo CTP, Hoff RB, Zambonim FM, Costa ACO, Fett R. The Protective Effect of Juçara Fruit (Euterpe edulis Martius) Extracts on LPS-Activated J774 Macrophages. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2024; 79:677-684. [PMID: 38976202 DOI: 10.1007/s11130-024-01204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/09/2024]
Abstract
This study investigated the anti-inflammatory effect of hydrophilic and lipophilic extracts from juçara fruits (Euterpe edulis Martius) through measurement of nitric oxide (NOx) and cytokines (IL-12p70, TNF-α, INF-γ, MCP-1, IL-6, and IL-10). J774 macrophages were stimulated with lipopolysaccharides (1 µg/mL) and treated with various concentrations (1-100 µg/mL) of juçara fruits extracts from crude extracts, and hexane, dichloromethane, ethyl acetate, and butanol fractions. Potential relationships between the phenolic composition of the extracts determined by LC-ESI-MS/MS and their anti-inflammatory capacity were also evaluated. Hexane and dichloromethane fractions inhibited NOx and IL-12p70 while increased IL-10. Hexane fractions also decreased IL-6 and IFN-γ production. Hexane and dichloromethane fractions showed a higher number of phenolic compounds (32 and 34, respectively) than the other extracts tested and were also the only ones that presented benzoic acid and pinocembrin. These results suggest juçara fruits compounds as potential anti-inflammatory agents, especially those of a more apolar nature.
Collapse
Affiliation(s)
- Mayara Schulz
- Department of Food Science and Technology, Federal University of Santa Catarina, Admar Gonzaga 1346, Itacorubi, Florianopolis, SC, 88034-001, Brazil.
| | - Luciano Valdemiro Gonzaga
- Department of Food Science and Technology, Federal University of Santa Catarina, Admar Gonzaga 1346, Itacorubi, Florianopolis, SC, 88034-001, Brazil
| | - Ana Clara Nascimento Antunes
- Department of Food Science and Technology, Federal University of Santa Catarina, Admar Gonzaga 1346, Itacorubi, Florianopolis, SC, 88034-001, Brazil
| | - Tainá Lubschinski
- Department of Clinical Analysis, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | | | | | - Carolina Turnes Pasini Deolindo
- Federal Agricultural Defense Laboratory, Brazilian Ministry of Agriculture, Livestock and Food Supply, São José, SC, 91780-580, Brazil
| | - Rodrigo Barcellos Hoff
- Federal Agricultural Defense Laboratory, Brazilian Ministry of Agriculture, Livestock and Food Supply, São José, SC, 91780-580, Brazil
| | - Fábio Martinho Zambonim
- Agricultural Research and Rural Extension Company of Santa Catarina (Epagri)/ Environmental Resources and Hydrometeorology Information Center (Ciram), Florianópolis, SC, 88034-901, Brazil
| | - Ana Carolina Oliveira Costa
- Department of Food Science and Technology, Federal University of Santa Catarina, Admar Gonzaga 1346, Itacorubi, Florianopolis, SC, 88034-001, Brazil
| | - Roseane Fett
- Department of Food Science and Technology, Federal University of Santa Catarina, Admar Gonzaga 1346, Itacorubi, Florianopolis, SC, 88034-001, Brazil.
| |
Collapse
|
41
|
Fernando L, Echesabal-Chen J, Miller M, Powell RR, Bruce T, Paul A, Poudyal N, Saliutama J, Parman K, Paul KS, Stamatikos A. Cholesterol Efflux Decreases TLR4-Target Gene Expression in Cultured Macrophages Exposed to T. brucei Ghosts. Microorganisms 2024; 12:1730. [PMID: 39203572 PMCID: PMC11357207 DOI: 10.3390/microorganisms12081730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Trypanosoma brucei causes African trypanosomiasis in humans. Infection with T. brucei elicits a potent pro-inflammatory immune response within infected human hosts, and this response is thought to at least be partially due to Toll-like receptor (TLR) activation. In response to stimulation by lipopolysaccharide and other pathogen antigens, TLR4 translocates to lipid rafts, which induces the expression of pro-inflammatory genes. However, cholesterol efflux is acknowledged as anti-inflammatory due to promoting lipid raft disruption. In this study, we wanted to assess the impact of T. brucei "ghosts", which are non-viable T. brucei essentially devoid of intracellular contents, in stimulating macrophage TLR4 translocation to lipid rafts, and whether promoting cholesterol efflux in macrophages incubated with T. brucei ghosts attenuates TLR4-target gene expression. When cultured macrophages were exposed to T. brucei ghosts, we observed an increase in lipid raft TLR4 protein content, which suggests certain surface molecules of T. brucei serve as ligands for TLR4. However, pretreating macrophages with cholesterol acceptors before T. brucei ghost exposure decreased lipid raft TLR4 protein content and the expression of pro-inflammatory TLR4-target genes. Taken together, these results imply that macrophage cholesterol efflux weakens pro-inflammatory responses which occur from T. brucei infection via increasing macrophage lipid raft disruption.
Collapse
Affiliation(s)
- Lawrence Fernando
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (L.F.); (J.E.-C.)
| | - Jing Echesabal-Chen
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (L.F.); (J.E.-C.)
| | - Murphy Miller
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29605, USA;
| | - Rhonda Reigers Powell
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Terri Bruce
- Clemson Light Imaging Facility, Clemson University, Clemson, SC 29634, USA; (R.R.P.); (T.B.)
| | - Apurba Paul
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Nava Poudyal
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Joshua Saliutama
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Kristina Parman
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Kimberly S. Paul
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA; (N.P.); (K.S.P.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (L.F.); (J.E.-C.)
| |
Collapse
|
42
|
Pitchakarn P, Buacheen P, Taya S, Karinchai J, Temviriyanukul P, Inthachat W, Chaipoot S, Wiriyacharee P, Phongphisutthinant R, Ounjaijean S, Boonyapranai K. Anti-Inflammatory, Cytotoxic, and Genotoxic Effects of Soybean Oligopeptides Conjugated with Mannose. Foods 2024; 13:2558. [PMID: 39200485 PMCID: PMC11353420 DOI: 10.3390/foods13162558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Soy protein is considered to be a high-quality protein with a range of important biological functions. However, the applications of soy protein are limited due to its poor solubility and high level of allergenicity. Its peptides have been of interest because they exert the same biological functions as soy protein, but are easier to absorb, more stable and soluble, and have a lower allergenicity. Moreover, recent research found that an attachment of chemical moieties to peptides could improve their properties including their biodistribution, pharmacokinetic, and biological activities with lower toxicity. This study therefore aimed to acquire scientific evidence to support the further application and safe use of the soybean oligopeptide (OT) conjugated with allulose (OT-AL) or D-mannose (OT-Man). The anti-inflammation, cytotoxicity, and genotoxicity of OT, OT-AL, and OT-Man were investigated. The results showed that OT, AL, Man, OT-AL, and OT-Man at doses of up to 1000 µg/mL were not toxic to HepG2 (liver cancer cells), HEK293 (kidney cells), LX-2 (hepatic stellate cells), and pre- and mature-3T3-L1 (fibroblasts and adipocytes, respectively), while slightly delaying the proliferation of RAW 264.7 cells (macrophages) at high doses. In addition, the oligopeptides at up to 800 µg/mL were not toxic to isolated human peripheral blood mononuclear cells (PBMCs) and did not induce hemolysis in human red blood cells (RBCs). OT-Man (200 and 400 µg/mL), but not OT, AL, Man, and OT-AL, significantly reduced the production of NO and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) stimulated by lipopolysaccharide (LPS) in RAW 264.7 cells, suggesting that the mannose conjugation of soy peptide had an inhibitory effect against LPS-stimulated inflammation. In addition, the secretion of interleukin-6 (IL-6) stimulated by LPS was significantly reduced by OT-AL (200 and 400 µg/mL) and OT-Man (400 µg/mL). The tumor necrosis factor-α (TNF-α) level was significantly decreased by OT (400 µg/mL), AL (400 µg/mL), OT-AL (200 µg/mL), and OT-Man (200 and 400 µg/mL) in the LPS-stimulated cells. The conjugation of the peptides with either AL or Man is likely to be enhance the anti-inflammation ability to inhibit the secretion of cytokines. As OT-Man exhibited a high potential to inhibit LPS-induced inflammation in macrophages, its mutagenicity ability was then assessed in bacteria and Drosophila. These findings showed that OT-Man did not trigger DNA mutations and was genome-safe. This study provides possible insights into the health advantages and safe use of conjugated soybean peptides.
Collapse
Affiliation(s)
- Pornsiri Pitchakarn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Muang Chiang Mai, Chiang Mai 50200, Thailand; (P.P.); (P.B.); (J.K.)
| | - Pensiri Buacheen
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Muang Chiang Mai, Chiang Mai 50200, Thailand; (P.P.); (P.B.); (J.K.)
| | - Sirinya Taya
- Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (S.C.); (R.P.)
| | - Jirarat Karinchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Muang Chiang Mai, Chiang Mai 50200, Thailand; (P.P.); (P.B.); (J.K.)
| | - Piya Temviriyanukul
- Institute of Nutrition, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand; (P.T.); (W.I.)
| | - Woorawee Inthachat
- Institute of Nutrition, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand; (P.T.); (W.I.)
| | - Supakit Chaipoot
- Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (S.C.); (R.P.)
| | - Pairote Wiriyacharee
- Processing and Product Development Factory, The Royal Project Foundation, Chiang Mai 50100, Thailand;
| | - Rewat Phongphisutthinant
- Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (S.C.); (R.P.)
| | - Sakaewan Ounjaijean
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Kongsak Boonyapranai
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
| |
Collapse
|
43
|
Lee HD, Tonog G, Uy NP, Lee Y, Kim KY, Kim H, Lee S. Phytochemical Profile, Antioxidant, Anti-Atopic, and Anti-Inflammatory Activities of Filipendula glaberrima Nakai at Different Growth Stages. Pharmaceuticals (Basel) 2024; 17:928. [PMID: 39065778 PMCID: PMC11279925 DOI: 10.3390/ph17070928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Since atopic dermatitis is an inflammatory skin disease, natural remedies, such as Filipendula glaberrima Nakai (FG), with anti-inflammatory properties are possible promising therapeutic options. This study aimed to investigate the therapeutic potential of FG extracts at different growth stages. Seven compounds were isolated from the FG leaf extracts using open-column chromatography, and they were analyzed using HPLC. The extracts were further evaluated for their total polyphenol and flavonoid content (TPC and TFC). The in vitro antioxidant properties of the FG extracts were evaluated using radical scavenging assays, whereas their anti-inflammatory activities were assessed by evaluating their ability to inhibit the production of inflammation-associated biomarkers using the Griess assay and ELISA, respectively. The MTT assay was used to evaluate the viability and cytotoxicity of the FG extracts in keratinocyte cell lines. The results showed that the full-flowering stage exhibited the highest TPC, TFC, and antioxidant activities, thus suggesting a positive correlation between these properties. All FG extracts showed significant anti-inflammatory activity by inhibiting the production of pro-inflammatory biomarkers in lipopolysaccharide-stimulated macrophages. Additionally, the FG extracts suppressed the production of cytokines and chemokines in keratinocytes, indicating their anti-atopic potential. HPLC analysis revealed that the full-flowering stage had the highest content of all the analyzed phytochemicals (gallic acid, (+)-catechin, hyperin, miquelianin, astragalin, afzelin, and quercetin). These results suggest that the full-flowering stage of FG is the most promising source for therapeutic applications owing to its superior phytochemical profile and biological activities. This study highlights the potential of FG extracts, particularly in its full-flowering stage, as a natural therapeutic agent for the management of inflammation-related diseases, and it can also serve as a reference for further research on FG.
Collapse
Affiliation(s)
- Hak-Dong Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea; (H.-D.L.); (N.P.U.)
- Natural Product Institute of Science and Technology, Anseong 17546, Republic of Korea
| | - Genevieve Tonog
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, Republic of Korea;
| | - Neil Patrick Uy
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea; (H.-D.L.); (N.P.U.)
| | - Yunji Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Eumseong 27709, Republic of Korea;
| | - Ki-Young Kim
- Department of Genetics and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea;
| | - Hangeun Kim
- Research and Development Center, Skin Biotechnology Center Inc., Yongin 17104, Republic of Korea;
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea; (H.-D.L.); (N.P.U.)
- Natural Product Institute of Science and Technology, Anseong 17546, Republic of Korea
| |
Collapse
|
44
|
Wang W, Li X, Wu H, Shi F, Zhang Z, Lv H. Explore the underlying oral efficacy of α-, β-, γ-Cyclodextrin against the ulcerative colitis using in vitro and in vivo studies assisted by network pharmacology. J Biomol Struct Dyn 2024; 42:4985-5000. [PMID: 37517028 DOI: 10.1080/07391102.2023.2239901] [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: 02/21/2023] [Accepted: 06/05/2023] [Indexed: 08/01/2023]
Abstract
The incidence of ulcerative colitis (UC) is rising worldwide. As a refractory and recurrent disease, UC could seriously affect the patients' quality of life. However, current clinical medical treatments for UC are accompanied by various side effects, especially for long-term applications. Here, the underlying efficacy of cyclodextrins (CDs) was studied. As common excipients, CDs endow proven safety for long-term applications. Results of predictive methods derived from network pharmacology prompted the potential anti-inflammatory effects of CDs by oral administration. RAW264.7 cell experiments verified that CDs could inhibit the excessive secretion of TNF-α (β-CD > α-CD ≈ γ-CD), IL-6, and NO (α-CD > β-CD ≈ γ-CD) as predicted. In mice with DSS-induced acute UC, oral administration of CDs could effectively mitigate the pathological damage of colon tissue and reduce the level of inflammatory mediators. Moreover, 16S rRNA sequencing displayed that gut microbes disturbed by DSS were significantly regulated by CDs. Conclusively, the study showed the therapeutic application prospects of CDs in UC treatment and indicated the feasibility and advantages of developing 'new' therapeutic activities of 'old' ingredients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Weiqin Wang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuefeng Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hangyi Wu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fanli Shi
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Huixia Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
45
|
Jiang L, Sun XY, Wang SQ, Liu YL, Lu LJ, Wu WH, Zhi H, Wang ZY, Liu XD, Liu L. Indoxyl sulphate-TNFα axis mediates uremic encephalopathy in rodent acute kidney injury. Acta Pharmacol Sin 2024; 45:1406-1424. [PMID: 38589687 PMCID: PMC11192958 DOI: 10.1038/s41401-024-01251-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/26/2024] [Indexed: 04/10/2024]
Abstract
Acute kidney injury (AKI) is often accompanied by uremic encephalopathy resulting from accumulation of uremic toxins in brain possibly due to impaired blood-brain barrier (BBB) function. Anionic uremic toxins are substrates or inhibitors of organic anionic transporters (OATs). In this study we investigated the CNS behaviors and expression/function of BBB OAT3 in AKI rats and mice, which received intraperitoneal injection of cisplatin 8 and 20 mg/kg, respectively. We showed that cisplatin treatment significantly inhibited the expressions of OAT3, synaptophysin and microtubule-associated protein 2 (MAP2), impaired locomotor and exploration activities, and increased accumulation of uremic toxins in the brain of AKI rats and mice. In vitro studies showed that uremic toxins neither alter OAT3 expression in human cerebral microvascular endothelial cells, nor synaptophysin and MAP2 expressions in human neuroblastoma (SH-SY5Y) cells. In contrast, tumour necrosis factor alpha (TNFα) and the conditioned medium (CM) from RAW264.7 cells treated with indoxyl sulfate (IS) significantly impaired OAT3 expression. TNFα and CM from IS-treated BV-2 cells also inhibited synaptophysin and MAP2 expressions in SH-SY5Y cells. The alterations caused by TNFα and CMs in vitro, and by AKI and TNFα in vivo were abolished by infliximab, a monoclonal antibody designed to intercept and neutralize TNFα, suggesting that AKI impaired the expressions of OAT3, synaptophysin and MAP2 in the brain via IS-induced TNFα release from macrophages or microglia (termed as IS-TNFα axis). Treatment of mice with TNFα (0.5 mg·kg-1·d-1, i.p. for 3 days) significantly increased p-p65 expression and reduced the expressions of Nrf2 and HO-1. Inhibiting NF-κB pathway, silencing p65, or activating Nrf2 and HO-1 obviously attenuated TNFα-induced downregulation of OAT3, synaptophysin and MAP2 expressions. Significantly increased p-p65 and decreased Nrf2 and HO-1 protein levels were also detected in brain of AKI mice and rats. We conclude that AKI inhibits the expressions of OAT3, synaptophysin and MAP2 due to IS-induced TNFα release from macrophages or microglia. TNFα impairs the expressions of OAT3, synaptophysin and MAP2 partly via activating NF-κB pathway and inhibiting Nrf2-HO-1 pathway.
Collapse
Affiliation(s)
- Ling Jiang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xue-Ying Sun
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Si-Qian Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan-Lin Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ling-Jue Lu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wen-Han Wu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao Zhi
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhong-Yan Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiao-Dong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
46
|
Yang B, Hu C, Zhang Y, Jiang D, Lin P, Qiu S, Shi J, Wang L. Biomimetic-Structured Cobalt Nanocatalyst Suppresses Aortic Dissection Progression by Catalytic Antioxidation. J Am Chem Soc 2024; 146:17201-17210. [PMID: 38874405 DOI: 10.1021/jacs.4c03344] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
As one of the most lethal cardiovascular diseases, aortic dissection (AD) is initiated by overexpression of reactive oxygen species (ROS) in the aorta that damages the vascular structure and finally leads to massive hemorrhage and sudden death. Current drugs used in clinics for AD treatment fail to efficiently scavenge ROS to a large extent, presenting undesirable therapeutic effect. In this work, a nanocatalytic antioxidation concept has been proposed to elevate the therapeutic efficacy of AD by constructing a cobalt nanocatalyst with a biomimetic structure that can scavenge pathological ROS in an efficient and sustainable manner. Theoretical calculations demonstrate that the antioxidation reaction is catalyzed by the redox transition between hydroxocobalt(III) and oxo-hydroxocobalt(V) accompanied by inner-sphere proton-coupled two-electron transfer, forming a nonassociated activation catalytic cycle. The efficient antioxidation action of the biomimetic nanocatalyst in the AD region effectively alleviates oxidative stress, which further modulates the aortic inflammatory microenvironment by promoting phenotype transition of macrophages. Consequently, vascular smooth muscle cells are also protected from inflammation in the meantime, suppressing AD progression. This study provides a nanocatalytic antioxidation approach for the efficient treatment of AD and other cardiovascular diseases.
Collapse
Affiliation(s)
- Bowen Yang
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Chengkai Hu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yuchong Zhang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Di Jiang
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Peng Lin
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Shouji Qiu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Jianlin Shi
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of Vascular Surgery, Zhongshan Xiamen Hospital, Fudan University, 668 JinhuRoad, Xiamen 361015, China
| |
Collapse
|
47
|
Frusciante L, Geminiani M, Olmastroni T, Mastroeni P, Trezza A, Salvini L, Lamponi S, Spiga O, Santucci A. Repurposing Castanea sativa Spiny Burr By-Products Extract as a Potentially Effective Anti-Inflammatory Agent for Novel Future Biotechnological Applications. Life (Basel) 2024; 14:763. [PMID: 38929746 PMCID: PMC11205080 DOI: 10.3390/life14060763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The concept of a "circular bioeconomy" holds great promise for the health, cosmetic, and nutrition sectors by re-using Castanea sativa (Mill.) by-products. This sustainable resource is rich in bioactive secondary metabolites with antioxidant and anti-inflammatory properties. By transforming these by-products into high-value products for human health, we can promote sustainable economic growth and reduce the environmental impact of traditional waste disposal, adding value to previously underutilized resources. In the present study, we investigated the antioxidant capacity, phytochemical composition, and in vitro antioxidant and anti-inflammatory activity of C. sativa burr (CSB) aqueous extract. The spectrophotometric study revealed high total phenolic content (TPC) values with significant antioxidant and anti-radical properties. Using UPLC-MS/MS techniques, the phytochemical investigation identified 56 metabolites, confirming the presence of phenolic compounds in CSBs. In addition, CSBs significantly downregulated pro-inflammatory mediators in LPS-stimulated RAW 264.7 macrophage cells without significant cell toxicity. Lastly, in silico studies pinpointed three kinases from RAW 264.7 cells as binding partners with ellagic acid, the predominant compound found in our extract. These findings strongly advocate for the recycling and valorization of C. sativa by-products, challenging their conventional classification as mere "waste".
Collapse
Affiliation(s)
- Luisa Frusciante
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
| | - Michela Geminiani
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Tommaso Olmastroni
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
| | - Pierfrancesco Mastroeni
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
| | - Alfonso Trezza
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
| | - Laura Salvini
- Fondazione Toscana Life Sciences, Strada del Petriccio e Belriguardo, 53100 Siena, Italy;
| | - Stefania Lamponi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| | - Annalisa Santucci
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (T.O.); (P.M.); (A.T.); (S.L.); (O.S.); (A.S.)
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| |
Collapse
|
48
|
Ji Y, Li H, Li J, Yang G, Zhang W, Shen Y, Xu B, Liu J, Wen J, Song W. Hair Follicle-Targeted Delivery of Azelaic Acid Micro/Nanocrystals Promote the Treatment of Acne Vulgaris. Int J Nanomedicine 2024; 19:5173-5191. [PMID: 38855733 PMCID: PMC11162231 DOI: 10.2147/ijn.s459788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024] Open
Abstract
Purpose Acne vulgaris is a chronic inflammatory skin disorder centered on hair follicles, making hair follicle-targeted delivery of anti-acne drugs a promising option for acne treatment. However, current researches have only focused on the delivering to healthy hair follicles, which are intrinsically different from pathologically clogged hair follicles in acne vulgaris. Patients and Methods Azelaic acid (AZA) micro/nanocrystals with different particle sizes were prepared by wet media milling or high-pressure homogenization. An experiment on AZA micro/nanocrystals delivering to healthy hair follicles was carried out, with and without the use of physical enhancement techniques. More importantly, it innovatively designed an experiment, which could reveal the ability of AZA micro/nanocrystals to penetrate the constructed clogged hair follicles. The anti-inflammatory and antibacterial effects of AZA micro/nanocrystals were evaluated in vitro using a RAW264.7 cell model stimulated by lipopolysaccharide and a Cutibacterium acnes model. Finally, both the anti-acne effects and skin safety of AZA micro/nanocrystals and commercial products were compared in vivo. Results In comparison to commercial products, 200 nm and 500 nm AZA micro/nanocrystals exhibited an increased capacity to target hair follicles. In the combination group of AZA micro/nanocrystals and ultrasound, the ability to penetrate hair follicles was further remarkably enhanced (ER value up to 9.6). However, toward the clogged hair follicles, AZA micro/nanocrystals cannot easily penetrate into by themselves. Only with the help of 1% salicylic acid, AZA micro/nanocrystals had a great potential to penetrate clogged hair follicle. It was also shown that AZA micro/nanocrystals had anti-inflammatory and antibacterial effects by inhibiting pro-inflammatory factors and Cutibacterium acnes. Compared with commercial products, the combination of AZA micro/nanocrystals and ultrasound exhibited an obvious advantage in both skin safety and in vivo anti-acne therapeutic efficacy. Conclusion Hair follicle-targeted delivery of AZA micro/nanocrystals provided a satisfactory alternative in promoting the treatment of acne vulgaris.
Collapse
Affiliation(s)
- Yan Ji
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Haorong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Jiguo Li
- Nanjing Miaobang Meiye Enterprise Management Co, LTD, Nanjing, People’s Republic of China
| | - Guangqiang Yang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Wenli Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yan Shen
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Bohui Xu
- School of Pharmacy, Nantong University, Nantong, 226001, People’s Republic of China
| | - Jianping Liu
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Jingyuan Wen
- School of Pharmacy, Faculty of Medical and Health Sciences, the University of Auckland, Auckland, New Zealand
| | - Wenting Song
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, People’s Republic of China
| |
Collapse
|
49
|
Zhu SL, Zhang HT, Du YY, Jiang Y, Wang SS, Ding WC, Feng L. Histological Features of Uterine Myometrial Dysfunction: Possible Involvement of Localized Inflammation. Curr Med Sci 2024; 44:633-641. [PMID: 38789820 DOI: 10.1007/s11596-024-2873-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE The latest perspective suggests that elevated levels of inflammation and cytokines are implicated in atonic postpartum hemorrhage. Lipopolysaccharide (LPS) has been widely used to induce inflammation in animal models. Therefore, this study aimed to induce uterine inflammation using LPS to investigate whether local inflammation triggers dysfunction and atrophy in the myometrium, as well as the potential underlying molecular mechanisms involved. METHODS In vivo, an animal model was established by intraperitoneal injection of 300 μg/ kg LPS in rats on gestational day 21. Hematoxylin-eosin (H&E) staining and Masson staining were employed to determine morphological changes in the rat uterine smooth muscle. Enzyme-linked immunosorbent assay (ELISA) was used to detect inflammatory cytokines. Immunohistochemistry, tissue fluorescence, and Western blotting were conducted to assess the expression levels of the uterine contraction-related proteins Toll-like receptor 4 (TLR4) and the nuclear factor kappa-B (NF-κB) signaling pathway. In vitro, human uterine smooth muscle cells (HUtSMCs) were exposed to 2 μg/mL LPS to further elucidate the involvement of the TLR4/NF-κB signaling pathway in LPS-mediated inflammation. RESULTS In this study, LPS induced uterine myometrial dysfunction in rats, leading to a disorganized arrangement, a significant increase in collagen fiber deposition, and widespread infiltration of inflammatory cells. In both in vivo animal models and in vitro HUtSMCs, LPS elevated IL-6, IL-1β, and TNF-α levels while concurrently suppressing the expression of connexin 43 (Cx43) and oxytocin receptor (OXTR). Mechanistically, the LPS-treated group exhibited TLR4 activation, and the phosphorylation levels of p65 and IκBα were notably increased. CONCLUSION LPS triggered the TLR4/NF-κB signaling pathway, inducing an inflammatory response in the myometrium and leading to uterine myometrial dysfunction and uterine atony.
Collapse
Affiliation(s)
- Sheng-Lan Zhu
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Ting Zhang
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan-Yuan Du
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Jiang
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shao-Shuai Wang
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Cheng Ding
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Ling Feng
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
50
|
Cicio A, Aloi N, Sut S, Longo V, Terracina F, Dall’Acqua S, Zizzo MG, Bruno M, Ilardi V, Colombo P, Luparello C, Serio R. Chemical Characterization, Free Radical Scavenging, and Cellular Antioxidant Properties of the Egadi Island Endemic Brassica macrocarpa Guss Leaf Extract. Biomolecules 2024; 14:636. [PMID: 38927041 PMCID: PMC11201591 DOI: 10.3390/biom14060636] [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: 03/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
The genus Brassica is an important source of food in the Mediterranean diet with documented nutritional and medicinal properties. However, few studies have investigated the phytochemical composition and the biological activity of wild Sicilian taxa. Thus, we aimed to study the chemical profile and the antioxidant potential, in vitro and in LPS-stimulated RAW 264.7 cells, of a methanolic extract of leaves of wild Brassica macrocarpa Guss (B. macrocarpa) (Egadi Islands; Sicily-Italy). B. macrocarpa methanolic extract showed a large amount of glucosinolates and different phenolic compounds. It exhibited antioxidant activity in the DPPH assay and in LPS-stimulated RAW 264.7 cells, being able to reduce NO and ROS levels and NOS2 mRNA expression. Our study demonstrated that Sicilian B. macrocarpa methanolic extract, in LPS-stimulated macrophages, efficiently counteracts oxidative stress and displays radical scavenging activity. Future studies are required to identify the contribution of the single phytocomponents, to characterize the action mechanism, and to reveal possible applications in human health.
Collapse
Affiliation(s)
- Adele Cicio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
| | - Noemi Aloi
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy; (N.A.); (V.L.); (P.C.)
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (S.S.); (S.D.)
| | - Valeria Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy; (N.A.); (V.L.); (P.C.)
| | - Francesca Terracina
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (S.S.); (S.D.)
| | - Maria Grazia Zizzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
- ATeN (Advanced Technologies Network) Center, Viale delle Scienze, University of Palermo, 90128 Palermo, Italy
- NBFC—National Biodiversity Future Center, University of Palermo, 90133 Palermo, Italy
| | - Maurizio Bruno
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
- NBFC—National Biodiversity Future Center, University of Palermo, 90133 Palermo, Italy
| | - Vincenzo Ilardi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
| | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo la Malfa 153, 90146 Palermo, Italy; (N.A.); (V.L.); (P.C.)
| | - Claudio Luparello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
- NBFC—National Biodiversity Future Center, University of Palermo, 90133 Palermo, Italy
| | - Rosa Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy (F.T.); (M.B.); (C.L.); (R.S.)
| |
Collapse
|