1
|
Xiao T, Yu Z, Yang H, You J, Wu X. Marine polysaccharides hydrogel with encapsulated mesalazine for the treatment of ulcerative colitis: Integrative effects on inflammation, microbiota, and mucosal repair. Colloids Surf B Biointerfaces 2025; 253:114722. [PMID: 40262307 DOI: 10.1016/j.colsurfb.2025.114722] [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: 02/24/2025] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
Ulcerative colitis is a chronic non-specific inflammatory disease of the intestine that significantly impacts patient quality of life. This study introduces a OF/CC/SM hydrogel containing oxidized fucoidan (OF), carboxymethyl chitosan (CC), and silk sericin-stabilized mesalazine (SM), designed for rectal administration to target mesalazine delivery specifically to the colon. The OF/CC/SM hydrogel demonstrated good biocompatibility (cell compatibility > 99 %), injectability, and adhesion strength, ensuring effective mesalazine retention and release. In vitro assays confirmed the hydrogel's antioxidant and anti-inflammatory properties, which were further validated in vivo using a mouse model of ulcerative colitis. Rectal administration of OF/CC/SM hydrogel significantly relieved weight loss, lowered disease activity index scores, and prevented intestinal shortening associated with dextran sulfate sodium (DSS) treatment. The hydrogel decreased the expression of proinflammatory cytokines (e.g., tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β)), while normalized the level of biomarkers (e.g., inducible nitric oxide synthase (iNOS), myeloperoxidase (MPO), catalase (CAT), and malondialdehyde (MDA)). Additionally, the OF/CC/SM hydrogel modulated the gut microbiota, increasing beneficial bacteria while decreasing potentially harmful species. Histopathological analysis revealed a reduction in inflammatory infiltration and improved mucosal architecture. Additionally, in vivo imaging studies confirmed sustained presence of OF/CC/SM hydrogel in the intestines following rectal administration, highlighting its potential for enhanced therapeutic efficacy in treating ulcerative colitis.
Collapse
Affiliation(s)
- Teng Xiao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Zhenxin Yu
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Haomin Yang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China
| | - Jun You
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei University, Youyi Road 368, Wuhan 430062, China
| | - Xiaochen Wu
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao 266042, China.
| |
Collapse
|
2
|
Liu MW, Zhang CH, Ma SH, Zhang DQ, Jiang LQ, Tan Y. Protective Effects of Baicalein on Lipopolysaccharide-Induced AR42J PACs through Attenuation of Both Inflammation and Pyroptosis via Downregulation of miR-224-5p/PARP1. Mediators Inflamm 2024; 2024:6618927. [PMID: 39421730 PMCID: PMC11486537 DOI: 10.1155/2024/6618927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 05/22/2024] [Accepted: 08/24/2024] [Indexed: 10/19/2024] Open
Abstract
Background Baicalein has been used to treat inflammation-related diseases; nevertheless, its specific mechanism of action is unclear. Therefore, we examined the protective effects of baicalein on lipopolysaccharide-induced damage to AR42J pancreatic acinar cells (PACs) and determined its mechanism of action for protection. Methods An in vitro cell model of acute pancreatitis (AP) was established using lipopolysaccharide (LPS) (1 mg/L)-induced PACs (AR42J), and the relative survival rate was determined using the 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) technique. Flow cytometry was applied to evaluate the apoptotic rates of AR42J PACs. The RNA and protein expression of miR-224-5p, poly ADP-ribose polymerase-1 (PARP1), nuclear transcription factor-κB65 (NF-κB65), phospho-kappa B alpha(p-IκB-α), interleukin(IL)-18R, NOD-like receptor thermal protein domain-associated protein 3 (NLRP3), gasdermin D (GSDMD), apoptosis-associated speck-like protein containing a CARD (ASC), and caspase-1 was detected based on the WB and RT-PCR assays. IL-1β, IL-6, IL-18, and TNF-α expression levels in AR42J cells were measured via ELISA method. The cell morphology was examined using the AO/EB method. Results The experiment confirmed a significant increase in the activity of AR42J cells treated with various doses of baicalein. Moreover, IL-1β, IL-6, TNF-α, and IL-18 expression levels in AR42J cells were dramatically reduced (P < 0.05), while miR-224-5p level was obviously enhanced. The protein and gene expression of PARP1, NF-κB65, p-IκB-α, IL-18R, GSDMD, ASC, NLRP3, and caspase-1 was obviously decreased (P < 0.05). Apoptosis in AR42J cells was significantly reduced with significant improvement in cell morphology. Conclusion Baicalein may significantly alleviate LPS-induced AR42J PAC damage by inhibiting the inflammatory response and pyroptosis. Its mode of action might be linked to higher miR-224-5p expression, which inhibits the PARP1/NF-κB and NLPR3/ASC/caspase-1/GSDMD pathways.
Collapse
Affiliation(s)
- Ming-Wei Liu
- Department of Emergency, Dali Bai Autonomous Prefecture People's Hospital, Dali 671000, China
| | - Chun-Hai Zhang
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Shou-Hong Ma
- Department of Medical Affairs, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi 653100, China
| | - De-Qiong Zhang
- Department of Medical Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Li-Qiong Jiang
- Physical Examination Center, Yunnan Fuwai Cardiovascular Hospital, Kunming 650032, China
| | - Yang Tan
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| |
Collapse
|
3
|
Yan J, Cai M, Zang C, Li W, Liu Z, Li X, Gao Y, Qi Y. The natural sesquiterpene lactone inulicin suppresses the production of pro-inflammatory mediators via inhibiting NF-κB and AP-1 pathways in LPS-activated macrophages. Immunopharmacol Immunotoxicol 2024:1-36. [PMID: 39048515 DOI: 10.1080/08923973.2024.2384899] [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: 04/10/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Inulicin is a sesquiterpene lactone in Inulae Flos which is clinically used for the treatment of inflammatory diseases, such as cough, sputum production and vomit. This study aimed to demonstrate the anti-inflammatory activity and the underlying mechanism of inulicin by using LPS-induced in vitro and in vivo models. METHODS LPS-stimulated RAW264.7 macrophages and mouse peritoneal macrophages (MPMs) were used for evaluating the in vitro anti-inflammatory activity of inulicin, while endotoxemia mice were used for evaluating its in vivo action. Cytokines' levels were determined by ELISA. RT-qPCR and western blot were used for assaying the mRNA and protein levels of target genes. RAW264.7 macrophages transfected with reporter plasmid pNFκB-TA-luc or pAP1-TA-luc were used for assaying the activation of NF-κB or AP-1 signaling. RESULTS Inulicin significantly inhibited LPS-induced production of NO, IL-6, c-c motif chemokine ligand 2 (CCL2) and IL-1β in both RAW264.7 cells and MPMs. Mechanism study indicated that it could suppress inducible nitric oxide synthase (iNOS), IL-6, CCL2 and IL-1β mRNA levels in LPS-stimulated RAW264.7 cells. Moreover, inulicin inhibited IκBα phosphorylation and prevented the nuclear translocation of p65, thereby inactivating NF-κB signaling. Concurrently, it also inhibited AP-1 signaling through reducing the phosphorylation of JNK and ERK. In endotoxemia mice, a single intraperitoneal administration of inulicin could decrease the production of pro-inflammatory cytokines in serum and peritoneal lavage fluid. CONCLUSIONS The present study demonstrates that inulicin possesses anti-inflammatory effects in vitro and in vivo, which suggests that inulicin might be a promising candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Jingjing Yan
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Min Cai
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Chenchen Zang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Wenjing Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Zhuangzhuang Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Ximeng Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Yuan Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Yun Qi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| |
Collapse
|
4
|
Chen S, Zeng J, Li R, Zhang Y, Tao Y, Hou Y, Yang L, Zhang Y, Wu J, Meng X. Traditional Chinese medicine in regulating macrophage polarization in immune response of inflammatory diseases. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117838. [PMID: 38310986 DOI: 10.1016/j.jep.2024.117838] [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: 09/26/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Numerous studies have demonstrated that various traditional Chinese medicines (TCMs) exhibit potent anti-inflammatory effects against inflammatory diseases mediated through macrophage polarization and metabolic reprogramming. AIM OF THE STUDY The objective of this review was to assess and consolidate the current understanding regarding the pathogenic mechanisms governing macrophage polarization in the context of regulating inflammatory diseases. We also summarize the mechanism action of various TCMs on the regulation of macrophage polarization, which may contribute to facilitate the development of natural anti-inflammatory drugs based on reshaping macrophage polarization. MATERIALS AND METHODS We conducted a comprehensive review of recently published articles, utilizing keywords such as "macrophage polarization" and "traditional Chinese medicines" in combination with "inflammation," as well as "macrophage polarization" and "inflammation" in conjunction with "natural products," and similar combinations, to search within PubMed and Google Scholar databases. RESULTS A total of 113 kinds of TCMs (including 62 components of TCMs, 27 TCMs as well as various types of extracts of TCMs and 24 Chinese prescriptions) was reported to exert anti-inflammatory effects through the regulation of key pathways of macrophage polarization and metabolic reprogramming. CONCLUSIONS In this review, we have analyzed studies concerning the involvement of macrophage polarization and metabolic reprogramming in inflammation therapy. TCMs has great advantages in regulating macrophage polarization in treating inflammatory diseases due to its multi-pathway and multi-target pharmacological action. This review may contribute to facilitate the development of natural anti-inflammatory drugs based on reshaping macrophage polarization.
Collapse
Affiliation(s)
- Shiyu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Rui Li
- The Affiliated Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, PR China
| | - Yingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yiwen Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Ya Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Lu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yating Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiasi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
5
|
Vizzoni L, Migone C, Grassiri B, Zambito Y, Ferro B, Roncucci P, Mori F, Salvatore A, Ascione E, Crea R, Esin S, Batoni G, Piras AM. Biopharmaceutical Assessment of Mesh Aerosolised Plasminogen, a Step towards ARDS Treatment. Pharmaceutics 2023; 15:1618. [PMID: 37376068 PMCID: PMC10300680 DOI: 10.3390/pharmaceutics15061618] [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: 02/27/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe complication of lung injuries, commonly associated with bacterial, fungal and viral infections, including SARS-CoV-2 viral infections. ARDS is strongly correlated with patient mortality and its clinical management is very complex, with no effective treatment presently available. ARDS involves severe respiratory failure, fibrin deposition in both airways and lung parenchyma, with the development of an obstructing hyaline membrane drastically limiting gas exchange. Moreover, hypercoagulation is related to deep lung inflammation, and a pharmacological action toward both aspects is expected to be beneficial. Plasminogen (PLG) is a main component of the fibrinolytic system playing key roles in various inflammation regulatory processes. The inhalation of PLG has been proposed in the form of the off-label administration of an eyedrop solution, namely, a plasminogen-based orphan medicinal product (PLG-OMP), by means of jet nebulisation. Being a protein, PLG is susceptible to partial inactivation under jet nebulisation. The aim of the present work is to demonstrate the efficacy of the mesh nebulisation of PLG-OMP in an in vitro simulation of clinical off-label administration, considering both the enzymatic and immunomodulating activities of PLG. Biopharmaceutical aspects are also investigated to corroborate the feasibility of PLG-OMP administration by inhalation. The nebulisation of the solution was performed using an Aerogen® SoloTM vibrating-mesh nebuliser. Aerosolised PLG showed an optimal in vitro deposition profile, with 90% of the active ingredient impacting the lower portions of a glass impinger. The nebulised PLG remained in its monomeric form, with no alteration of glycoform composition and 94% of enzymatic activity maintenance. Activity loss was observed only when PLG-OMP nebulisation was performed under simulated clinical oxygen administration. In vitro investigations evidenced good penetration of aerosolised PLG through artificial airway mucus, as well as poor permeation across an Air-Liquid Interface model of pulmonary epithelium. The results suggest a good safety profile of inhalable PLG, excluding high systemic absorption but with good mucus diffusion. Most importantly, the aerosolised PLG was capable of reversing the effects of an LPS-activated macrophage RAW 264.7 cell line, demonstrating the immunomodulating activity of PLG in an already induced inflammatory state. All physical, biochemical and biopharmaceutical assessments of mesh aerosolised PLG-OMP provided evidence for its potential off-label administration as a treatment for ARDS patients.
Collapse
Affiliation(s)
- Lucia Vizzoni
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Chiara Migone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Ylenia Zambito
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Research Centre for Nutraceutical and Healthy Foods “NUTRAFOOD”, University of Pisa, 56124 Pisa, Italy
| | - Baldassare Ferro
- Anestesia e Rianimazione, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Paolo Roncucci
- Anestesia e Rianimazione, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Filippo Mori
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Alfonso Salvatore
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Ester Ascione
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Roberto Crea
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| | - Anna Maria Piras
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| |
Collapse
|
6
|
Świerczek A, Jusko WJ. Pharmacokinetic/Pharmacodynamic Modeling of Dexamethasone Anti-Inflammatory and Immunomodulatory Effects in LPS-Challenged Rats: A Model for Cytokine Release Syndrome. J Pharmacol Exp Ther 2023; 384:455-472. [PMID: 36631280 PMCID: PMC9976795 DOI: 10.1124/jpet.122.001477] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/01/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Dexamethasone (DEX) is a potent synthetic glucocorticoid used for the treatment of variety of inflammatory and immune-mediated disorders. The RECOVERY clinical trial revealed benefits of DEX therapy in COVID-19 patients. Severe SARS-CoV-2 infection leads to an excessive inflammatory reaction commonly known as a cytokine release syndrome that is associated with activation of the toll like receptor 4 (TLR4) signaling pathway. The possible mechanism of action of DEX in the treatment of COVID-19 is related to its anti-inflammatory activity arising from inhibition of cytokine production but may be also attributed to its influence on immune cell trafficking and turnover. This study, by means of pharmacokinetic/pharmacodynamic modeling, aimed at the comprehensive quantitative assessment of DEX effects in lipopolysaccharide-challenged rats and to describe interrelations among relevant signaling molecules in this animal model of cytokine release syndrome induced by activation of TLR4 pathway. DEX was administered in a range of doses from 0.005 to 2.25 mg·kg-1 in LPS-challenged rats. Serum DEX, corticosterone (CST), tumor necrosis factor α, interleukin-6, and nitric oxide as well as lymphocyte and granulocyte counts in peripheral blood were quantified at different time points. A minimal physiologically based pharmacokinetic/pharmacodynamic (mPBPK/PD) model was proposed characterizing the time courses of plasma DEX and the investigated biomarkers. A high but not complete inhibition of production of inflammatory mediators and CST was produced in vivo by DEX. The mPBPK/PD model, upon translation to humans, may help to optimize DEX therapy in patients with diseases associated with excessive production of inflammatory mediators, such as COVID-19. SIGNIFICANCE STATEMENT: A mPBPK/PD model was developed to describe concentration-time profiles of plasma DEX, mediators of inflammation, and immune cell trafficking and turnover in LPS-challenged rats. Interrelations among DEX and relevant biomarkers were reflected in the mechanistic model structure. The mPBPK/PD model enabled quantitative assessment of in vivo potency of DEX and, upon translation to humans, may help optimize dosing regimens of DEX for the treatment of immune-related conditions associated with exaggerated immune response.
Collapse
Affiliation(s)
- Artur Świerczek
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
7
|
Duan Z, Xie H, Yu S, Wang S, Yang H. Piperine Derived from Piper nigrum L. Inhibits LPS-Induced Inflammatory through the MAPK and NF-κB Signalling Pathways in RAW264.7 Cells. Foods 2022; 11:foods11192990. [PMID: 36230067 PMCID: PMC9563280 DOI: 10.3390/foods11192990] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 12/28/2022] Open
Abstract
Piperine, an important natural product, has a good anti-inflammatory effect. However, few researchers have studied its mechanism in these pathways. The objective of this research was to evaluate the molecular mechanism underlying the anti-inflammatory responses of piperine in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. The purification and characterization of piperine from Piper nigrum L. were determined by HPLC, UPLC-Q-TOF-MS and 1H NMR. Then, the anti-inflammatory activity was evaluated by a reagent test kit, ELISA kits, RT-PCR and Western blot experiments. The results suggested that piperine (90.65 ± 0.46% purity) at a concentration of 10–20 mg/L attenuated the production of NO and ROS, downregulated the protein and mRNA expression levels of TNF-α, IL-1β and IL-6, and upregulated the protein and mRNA transcription levels of IL-10. Meanwhile, the Western blot results indicated that piperine could inhibit the phosphorylation levels of the ERK, JNK, p38 and p65 proteins. Our findings suggest that piperine is a potential anti-inflammatory substance, whose molecular mechanism may be to regulate the key factors of the NF-κB and MAPK signalling pathways.
Collapse
Affiliation(s)
- Zhouwei Duan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Institute of Agro-Products Processing and Design, Hainan Academy of Agricultural Science, Haikou 571100, China
| | - Hui Xie
- Institute of Agro-Products Processing and Design, Hainan Academy of Agricultural Science, Haikou 571100, China
| | - Shasha Yu
- Institute of Agro-Products Processing and Design, Hainan Academy of Agricultural Science, Haikou 571100, China
- College of Food Science and Technology, Hainan University, Haikou 570228, China
| | - Shiping Wang
- Institute of Agro-Products Processing and Design, Hainan Academy of Agricultural Science, Haikou 571100, China
| | - Hong Yang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
8
|
Larsson J, Hoppe E, Gautrois M, Cvijovic M, Jirstrand M. Optimizing study design in LPS challenge studies for quantifying drug induced inhibition of TNFα response: Did we miss the prime time? Eur J Pharm Sci 2022; 176:106256. [PMID: 35820630 DOI: 10.1016/j.ejps.2022.106256] [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: 02/02/2022] [Revised: 05/30/2022] [Accepted: 07/07/2022] [Indexed: 11/03/2022]
Abstract
In this work we evaluate the study design of LPS challenge experiments used for quantification of drug induced inhibition of TNFα response and provide general guidelines of how to improve the study design. Analysis of model simulated data, using a recently published TNFα turnover model, as well as the optimal design tool PopED have been used to find the optimal values of three key study design variables - time delay between drug and LPS administration, LPS dose, and sampling time points - that in turn could make the resulting TNFα response data more informative. Our findings suggest that the current rule of thumb for choosing the time delay should be reconsidered, and that the placement of the measurements after maximal TNFα response are crucial for the quality of the experiment. Furthermore, a literature study summarizing a wide range of published LPS challenge studies is provided, giving a broader perspective of how LPS challenge studies are usually conducted both in a preclinical and clinical setting.
Collapse
Affiliation(s)
- Julia Larsson
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg 412 88, Sweden; Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg 412 96, Sweden.
| | | | | | - Marija Cvijovic
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg 412 96, Sweden
| | - Mats Jirstrand
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg 412 88, Sweden
| |
Collapse
|
9
|
Liu HT, Lin YN, Tsai MC, Wu YC, Lee MC. Baicalein Exerts Therapeutic Effects against Endotoxin-Induced Depression-like Behavior in Mice by Decreasing Inflammatory Cytokines and Increasing Brain-Derived Neurotrophic Factor Levels. Antioxidants (Basel) 2022; 11:antiox11050947. [PMID: 35624812 PMCID: PMC9137772 DOI: 10.3390/antiox11050947] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 12/07/2022] Open
Abstract
Inflammation plays an important role in the pathophysiology of depression. This study aims to elucidate the antidepressant effect of baicalein, an anti-inflammatory component of a traditional Chinese herbal medicine (Scutellaria baicalensis), on lipopolysaccharide (LPS)-induced depression-like behavior in mice, and to investigate the underlying mechanisms. In vitro, baicalein exhibited antioxidant activity and protected macrophages from LPS-induced damage. The results of the tail suspension test and forced swimming test (tests for despair potential in mice) showed the antidepressant effect of baicalein on LPS-treated mice. It also substantially decreased the production of pro-inflammatory cytokines, including IL-6, TNF-α, MCP-1, and eotaxin, elicited by LPS in the plasma. Baicalein downregulated NF-κB-p65 and iNOS protein levels in the hippocampus, demonstrated its ability to mitigate neuroinflammation. Additionally, baicalein increased the levels of the mature brain-derived neurotrophic factor (mBDNF) in the hippocampus of LPS-treated mice, and elevated the ratio of mBDNF/proBDNF, which regulates neuronal survival and synaptic plasticity. Baicalein also promoted the expression of CREB, which plays a role in a variety of signaling pathways. In summary, the findings of this study demonstrate that the administration of baicalein can attenuate LPS-induced depression-like behavior by suppressing neuroinflammation and inflammation induced by the peripheral immune response.
Collapse
Affiliation(s)
- Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-T.L.); (Y.-C.W.)
| | - Yu-Ning Lin
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan;
| | - Ming-Cheng Tsai
- Department of Neurosurgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
| | - Ya-Chi Wu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-T.L.); (Y.-C.W.)
| | - Ming-Chung Lee
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
- School of Life Science, National Taiwan Normal University, No. 88, Sec. 4, Ting-Zhou Road, Taipei 116, Taiwan
- Correspondence: ; Tel.: +886-2-7749-6254; Fax: +886-2-29312904
| |
Collapse
|
10
|
Baicalein Attenuates Severe Polymicrobial Sepsis via lleviating Immune Dysfunction of T Lymphocytes and Inflammation. Chin J Integr Med 2022; 28:711-718. [PMID: 35355199 DOI: 10.1007/s11655-022-3510-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the effect of baicalein on polymicrobial sepsis-induced immune dysfunction and organ injury. METHODS A sepsis model was induced in Sprague-Dawley rats via caecal ligation and puncture (CLP). Specific pathogen free rats were randomly divided into a sham group, CLP group and CLP + baicalein (Bai) group (n=16 each). Rats in the CLP + Bai group were intravenously injected with baicalein (20 mg/kg) at 1 and 10 h after CLP. Survival rate, bacterial load, and organ damage were assessed. Then each group was evaluated at 6, 12, and 24 h to investigate the effect of baicalein on immune cells and inflammatory cytokines in septic rats. RESULTS Baicalein treatment significantly improved the survival of septic rats, decreased the bacterial burden, and moderated tissue damage (spleen, liver, and lung), as observed by haematoxylin and eosin staining. Septic rats treated with baicalein had strikingly increased proportions of CD3+CD4+ T cells and ratios of CD4+/CD8+ T cells in the peripheral blood and spleen (all P<0.05). Moreover, baicalein treatment decreased the apoptotic rate of whole white blood cells and spleen cells at 24 h after surgery (P<0.05). Baicalein significantly reduced the levels of tumor necrosis factor α and interleukin-6 (IL-6) and increased IL-10, and the expression levels of galectin 9 were also raised in the spleen (P<0.01). CONCLUSION Baicalein may be an effective immunomodulator that attenuates overwhelming inflammatory responses in severe abdominal sepsis.
Collapse
|
11
|
Wu Q, Yin CH, Li Y, Cai JQ, Yang HY, Huang YY, Zheng YX, Xiong K, Yu HL, Lu AP, Wang KX, Guan DG, Chen YP. Detecting Critical Functional Ingredients Group and Mechanism of Xuebijing Injection in Treating Sepsis. Front Pharmacol 2021; 12:769190. [PMID: 34938184 PMCID: PMC8687625 DOI: 10.3389/fphar.2021.769190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a systemic inflammatory reaction caused by various infectious or noninfectious factors, which can lead to shock, multiple organ dysfunction syndrome, and death. It is one of the common complications and a main cause of death in critically ill patients. At present, the treatments of sepsis are mainly focused on the controlling of inflammatory response and reduction of various organ function damage, including anti-infection, hormones, mechanical ventilation, nutritional support, and traditional Chinese medicine (TCM). Among them, Xuebijing injection (XBJI) is an important derivative of TCM, which is widely used in clinical research. However, the molecular mechanism of XBJI on sepsis is still not clear. The mechanism of treatment of "bacteria, poison and inflammation" and the effects of multi-ingredient, multi-target, and multi-pathway have still not been clarified. For solving this issue, we designed a new systems pharmacology strategy which combines target genes of XBJI and the pathogenetic genes of sepsis to construct functional response space (FRS). The key response proteins in the FRS were determined by using a novel node importance calculation method and were condensed by a dynamic programming strategy to conduct the critical functional ingredients group (CFIG). The results showed that enriched pathways of key response proteins selected from FRS could cover 95.83% of the enriched pathways of reference targets, which were defined as the intersections of ingredient targets and pathogenetic genes. The targets of the optimized CFIG with 60 ingredients could be enriched into 182 pathways which covered 81.58% of 152 pathways of 1,606 pathogenetic genes. The prediction of CFIG targets showed that the CFIG of XBJI could affect sepsis synergistically through genes such as TAK1, TNF-α, IL-1β, and MEK1 in the pathways of MAPK, NF-κB, PI3K-AKT, Toll-like receptor, and tumor necrosis factor signaling. Finally, the effects of apigenin, baicalein, and luteolin were evaluated by in vitro experiments and were proved to be effective in reducing the production of intracellular reactive oxygen species in lipopolysaccharide-stimulated RAW264.7 cells, significantly. These results indicate that the novel integrative model can promote reliability and accuracy on depicting the CFIGs in XBJI and figure out a methodological coordinate for simplicity, mechanism analysis, and secondary development of formulas in TCM.
Collapse
Affiliation(s)
- Qi- Wu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuan-Hui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie-Qi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Han-Yun Yang
- The First Clinical Medical College of Southern Medical University, Guangzhou, China
| | - Ying-Ying Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi-Xu Zheng
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke Xiong
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-Lang Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Ai-Ping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong China
| | - Ke-Xin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Neurosurgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dao-Gang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Yu-Peng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Dietary Flavone Baicalein Combinate with Genipin Attenuates Inflammation Stimulated by Lipopolysaccharide in RAW264.7 Cells or Pseudomonas aeruginosa in Mice via Regulating the Expression and Phosphorylation of AKT. Nutrients 2021; 13:nu13124462. [PMID: 34960014 PMCID: PMC8708859 DOI: 10.3390/nu13124462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 01/01/2023] Open
Abstract
Mounting evidence has shown that single-targeted therapy might be inadequate to achieve satisfactory effects. Thus, drug combinations are gaining attention as they can regulate multiple targets to obtain more beneficial effects. Heat shock protein 90 (HSP90) is a molecular chaperone that assists the protein assembly and folding of client proteins and maintains their stability. Interfering with the interaction between HSP90 and its client proteins by inhibiting the latter’s activity may offer a new approach toward combination therapy. The HSP90 client protein AKT plays an important role in the inflammatory response syndrome caused by infections. In this study, the dietary flavone baicalein was identified as a novel inhibitor of HSP90 that targeted the N-terminal ATP binding pocket of HSP90 and hindered the chaperone cycle, resulting in AKT degradation. Combining baicalein with genipin, which was extracted from Gardenia jasminoides, could inhibit the pleckstrin homology domain of AKT, significantly increasing the anti-inflammatory effects both in vitro and in vivo. This synergistic effect was attributed to the reduction in AKT expression and phosphorylation. Thus, elucidating the mechanism underlying this effect will provide a new avenue for the clinical application and development of synergistic anti-inflammatory drugs.
Collapse
|
13
|
Larsson J, Hoppe E, Gautrois M, Cvijovic M, Jirstrand M. Second-generation TNFα turnover model for improved analysis of test compound interventions in LPS challenge studies. Eur J Pharm Sci 2021; 165:105937. [PMID: 34260892 DOI: 10.1016/j.ejps.2021.105937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/31/2021] [Accepted: 07/04/2021] [Indexed: 11/30/2022]
Abstract
This study presents a non-linear mixed effects model describing tumour necrosis factor alpha (TNFα) release after lipopolysaccharide (LPS) provocations in absence or presence of anti-inflammatory test compounds. Inter-occasion variability and the pharmacokinetics of two test compounds have been added to this second-generation model, and the goal is to produce a framework of how to model TNFα response in LPS challenge studies in vivo and demonstrate its general applicability regardless of occasion or type of test compound. Model improvements based on experimental data were successfully implemented and provided a robust model for TNFα response after LPS provocation, as well as reliable estimates of the median pharmacodynamic parameters. The two test compounds, Test Compound A and roflumilast, showed 81.1% and 74.9% partial reduction of TNFα response, respectively, and the potency of Test Compound A was estimated to 0.166 µmol/L. Comparing this study with previously published work reveals that our model leads to biologically reasonable output, handles complex data pooled from different studies, and highlights the importance of accurately distinguishing the stimulatory effect of LPS from the inhibitory effect of the test compound.
Collapse
Affiliation(s)
- Julia Larsson
- Fraunhofer-Chalmers Centre, Chalmers Science Park, 412 88 Gothenburg, Sweden.; Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96 Gothenburg, Sweden..
| | | | | | - Marija Cvijovic
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96 Gothenburg, Sweden
| | - Mats Jirstrand
- Fraunhofer-Chalmers Centre, Chalmers Science Park, 412 88 Gothenburg, Sweden
| |
Collapse
|
14
|
Inhibition of Oxidative Stress and ALOX12 and NF-κB Pathways Contribute to the Protective Effect of Baicalein on Carbon Tetrachloride-Induced Acute Liver Injury. Antioxidants (Basel) 2021; 10:antiox10060976. [PMID: 34207230 PMCID: PMC8235740 DOI: 10.3390/antiox10060976] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
This study investigates the protective effect of baicalein on carbon tetrachloride (CCl4)-induced acute liver injury and the underlying molecular mechanisms. Mice were orally administrated baicalein at 25 and 100 mg/kg/day for 7 consecutive days or ferrostatin-1 (Fer-1) at 10 mg/kg was i.p. injected in mice at 2 and 24 h prior to CCl4 injection or the vehicle. Our results showed that baicalein or Fer-1 supplementation significantly attenuated CCl4 exposure-induced elevations of serum alanine aminotransferase and aspartate aminotransferase, and malondialdehyde levels in the liver tissues and unregulated glutathione levels. Baicalein treatment inhibited the nuclear factor kappa-B (NF-κB) pathway, activated the erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway in liver tissues, and markedly improved CCl4-induced apoptosis, inflammation and ferroptosis in liver tissues exposed with CCl4. In vitro, baicalein treatment improved CCl4 -induced decreases of cell viabilities and knockdown of Nrf2 and arachidonate 12-lipoxygenase (ALOX12) genes partly abolished the protective effect of baicalein on CCl4 -induced cytotoxicity in HepG2 cells. In conclusion, our results reveal that baicalein supplementation ameliorates CCl4-induced acute liver injury in mice by upregulating the antioxidant defense pathways and downregulating oxidative stress, apoptosis, inflammation and ferroptosis, which involved the activation of Nrf2 pathway and the inhibition of ALOX12 and NF-κB pathways.
Collapse
|
15
|
Hu Y, Huang W, Luo Y, Xiang L, Wu J, Zhang Y, Zeng Y, Xu C, Meng X, Wang P. Assessment of the anti-inflammatory effects of three rhubarb anthraquinones in LPS-Stimulated RAW264.7 macrophages using a pharmacodynamic model and evaluation of the structure-activity relationships. JOURNAL OF ETHNOPHARMACOLOGY 2021; 273:114027. [PMID: 33741438 DOI: 10.1016/j.jep.2021.114027] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/10/2021] [Accepted: 03/11/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhubarb (Rhei Radix et Rhizoma) is a traditional Chinese medicine, has been used as a strong astringent in China to treat inflammation-related diseases, such as acute pancreatitis, acute cholecystitis, appendicitis and so on. Rhein, emodin and aloe-emodin are the important active anthraquinone in rhubarb, and are considered to be the main ingredients contributing to anti-inflammatory. AIM OF THE STUDY Rhein, emodin and aloe-emodin, anthraquinones with the same parent structure that are found in rhubarb, have beneficial anti-inflammatory effects in vitro and in vivo. Anthraquinone derivatives also have important clinical roles. However, their pharmacodynamic differences and the structure-activity relationships associated with their anti-inflammatory properties have not been systematically explored. The present study was designed to quantify the effects of three rhubarb anthraquinones on inflammation and to explore the structure-activity relationships of these compounds. MATERIALS AND METHODS In this study, we detected NF-κB phosphorylation, iNOS protein expression, and IL-6 and NO production in LPS-stimulated RAW264.7 cells and then calculated median effect equations and built a dynamic pharmacodynamic model to quantitatively evaluate the efficacy of these three anthraquinones. Additionally, to determine the structure-activity relationships, we investigated the physicochemical properties and molecular electrostatic potentials of the drug molecules. RESULTS We found that rhein, emodin, and aloe-emodin exerted at least dual-target (NF-κB, iNOS) inhibition of LPS-induced inflammatory responses. Compared with rhein and emodin, aloe-emodin had a stronger anti-inflammatory effect, and its inhibition of iNOS protein expression was approximately twice that of NF-κB phosphorylation. In addition, aloe-emodin had the strongest hydrophobic effect among the three anthraquinones. CONCLUSIONS Overall, we concluded that the receptor binding the rhubarb anthraquinones had a hydrophobic pocket. Anthraquinone molecules with stronger hydrophobic effects had higher affinity for the receptor, resulting in greater anti-inflammatory activity. These results suggest that the addition of a hydrophobic group is a potential method for structural modification to design anti-inflammatory anthraquinone derivatives with enhanced potency.
Collapse
Affiliation(s)
- Yingfan Hu
- School of Preclinical Medicine, Chengdu University, Chengdu, 610106, Sichuan, China; Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Wen'ge Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yu Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Li Xiang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Jiasi Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yan Zhang
- School of Preclinical Medicine, Chengdu University, Chengdu, 610106, Sichuan, China
| | - Yong Zeng
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Chensi Xu
- Chengdu Pharmoko Tech Corp., Ltd., Chengdu, 610041, China
| | - Xianli Meng
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| | - Ping Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
16
|
Dexamethasone Creates a Suppressive Microenvironment and Promotes Aspergillus fumigatus Invasion in a Human 3D Epithelial/Immune Respiratory Model. J Fungi (Basel) 2021; 7:jof7030221. [PMID: 33803702 PMCID: PMC8003030 DOI: 10.3390/jof7030221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 11/19/2022] Open
Abstract
Lung immunity and susceptibility to infections is subject to interactions between the epithelial layer and immune cells residing in the pulmonary space. Aspergillus (A.) fumigatus, the most prevalent pathogenic fungus, affects both upper and lower respiratory tracts of immunocompromised hosts. Several reports implicate corticosteroids as a major risk factor due to their anti-inflammatory and immunosuppressive effects, which are exacerbated by long-term treatment regimens. Here we demonstrate for the first time the influence of dexamethasone when it comes to germination and hyphae formation of A. fumigatus in the presence of macrophages within a highly differentiated air–liquid interphase (ALI) epithelial/immune lung model. We illustrate suppressed mucus production within the highly differentiated 3D respiratory model as well as significantly decreased cilia beat frequencies by dexamethasone treatment. This goes along with corticosteroid-mediated macrophage M2 polarization within the epithelial/immune microenvironment. Therefore, we here showed that corticosteroids promote enhanced fungal growth and invasion A. fumigatus by creating a suppressive environment affecting both epithelial as well as immune cells.
Collapse
|
17
|
Rebolledo V, Otero MC, Delgado JM, Torres F, Herrera M, Ríos M, Cabañas M, Martinez JL, Rodríguez-Díaz M. Phytochemical profile and antioxidant activity of extracts of the peruvian peppertree Schinus areira L. from Chile. Saudi J Biol Sci 2021; 28:1052-1062. [PMID: 33424399 PMCID: PMC7785446 DOI: 10.1016/j.sjbs.2020.10.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/17/2020] [Accepted: 10/20/2020] [Indexed: 11/03/2022] Open
Abstract
The Andean tree Schinus areira L. has multiple traditional uses, from the treatment of bronchitis and rheumatic diseases to menstrual cycle regulation and wound healing. With reported hypotensive, analgesic, antitumoral and anti-inflammatory properties, it acts predominantly against diseases related to oxidative stress. This study focuses on the antioxidant activity and phytochemical profile of the extracts of Schinus areira L. Serial extraction of the fruits was performed both by maceration and by Soxhlet. Total phenols and flavonoids were measured using the Folin-Ciocalteu method and AlCl3, respectively. In vitro antioxidant activity was determined by FRAP and DPPH. Results were similar for both extraction methods. Primary metabolites detected included carbohydrates, proteins and amino acids; secondary metabolites included tannins, flavonoids, saponins, steroids and triterpenes. Antioxidant activity was confirmed for ethyl acetate, methanolic and aqueous extracts. The methanolic extract had both the highest polyphenol content (>195 mg GAE/ g dry weight) and the highest antioxidant activity [EC50 > 476 μg/mL; >273 mg AA/g dry weight (DPPH); >301 mg AA/ g dry weight (FRAP)]. The extract does not produce macrophage cytotoxicity in RAW 264.7, which is indicated by an average cytotoxicity of 2% over 24 h. Our study serves as a starting point for future research on the pharmacological properties of Schinus areira L.
Collapse
Affiliation(s)
- Vania Rebolledo
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - María Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - José Manuel Delgado
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Fernando Torres
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Michelle Herrera
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Miguel Ríos
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Mauricio Cabañas
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - José L Martinez
- Vicerrectoria de Investigación, Desarrollo e Innovación, Universidad de Santiago de Chile, Santiago, Chile
| | - Maité Rodríguez-Díaz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
18
|
Anti-Inflammatory Activity and ROS Regulation Effect of Sinapaldehyde in LPS-Stimulated RAW 264.7 Macrophages. Molecules 2020; 25:molecules25184089. [PMID: 32906766 PMCID: PMC7570554 DOI: 10.3390/molecules25184089] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/30/2022] Open
Abstract
We evaluated the anti-inflammatory effects of SNAH in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages by performing nitric oxide (NO) assays, cytokine enzyme-linked immunosorbent assays, Western blotting, and real-time reverse transcription-polymerase chain reaction analysis. SNAH inhibited the production of NO (nitric oxide), reactive oxygen species (ROS), tumor necrosis factor (TNF)-α, and interleukin (IL)-6. Additionally, 100 μM SNAH significantly inhibited total NO and ROS inhibitory activity by 93% (p < 0.001) and 34% (p < 0.05), respectively. Protein expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) stimulated by LPS were also decreased by SNAH. Moreover, SNAH significantly (p < 0.001) downregulated the TNF-α, IL-6, and iNOS mRNA expression upon LPS stimulation. In addition, 3–100 µM SNAH was not cytotoxic. Docking simulations and enzyme inhibitory assays with COX-2 revealed binding scores of −6.4 kcal/mol (IC50 = 47.8 μM) with SNAH compared to −11.1 kcal/mol (IC50 = 0.45 μM) with celecoxib, a known selective COX-2 inhibitor. Our results demonstrate that SNAH exerts anti-inflammatory effects via suppression of ROS and NO by COX-2 inhibition. Thus, SNAH may be useful as a pharmacological agent for treating inflammation-related diseases.
Collapse
|
19
|
Held F, Hoppe E, Cvijovic M, Jirstrand M, Gabrielsson J. Challenge model of TNF α turnover at varying LPS and drug provocations. J Pharmacokinet Pharmacodyn 2019; 46:223-240. [PMID: 30778719 PMCID: PMC6529397 DOI: 10.1007/s10928-019-09622-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/08/2019] [Indexed: 11/28/2022]
Abstract
A mechanism-based biomarker model of TNFα-response, including different external provocations of LPS challenge and test compound intervention, was developed. The model contained system properties (such as kt, kout), challenge characteristics (such as ks, kLPS, Km, LPS, Smax, SC50) and test-compound-related parameters (Imax, IC50). The exposure to test compound was modelled by means of first-order input and Michaelis–Menten type of nonlinear elimination. Test compound potency was estimated to 20 nM with a 70% partial reduction in TNFα-response at the highest dose of 30 mg·kg−1. Future selection of drug candidates may focus the estimation on potency and efficacy by applying the selected structure consisting of TNFα system and LPS challenge characteristics. A related aim was to demonstrate how an exploratory (graphical) analysis may guide us to a tentative model structure, which enables us to better understand target biology. The analysis demonstrated how to tackle a biomarker with a baseline below the limit of detection. Repeated LPS-challenges may also reveal how the rate and extent of replenishment of TNFα pools occur. Lack of LPS exposure-time courses was solved by including a biophase model, with the underlying assumption that TNFα-response time courses, as such, contain kinetic information. A transduction type of model with non-linear stimulation of TNFα release was finally selected. Typical features of a challenge experiment were shown by means of model simulations. Experimental shortcomings of present and published designs are identified and discussed. The final model coupled to suggested guidance rules may serve as a general basis for the collection and analysis of pharmacological challenge data of future studies.
Collapse
Affiliation(s)
- Felix Held
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg, Sweden. .,Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden.
| | | | - Marija Cvijovic
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | - Mats Jirstrand
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg, Sweden
| | - Johan Gabrielsson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, 75007, Uppsala, Sweden
| |
Collapse
|