1
|
Kumari M, Sharma A, Tirpude NV. Herbacetin ameliorates lipopolysaccharide-elicited inflammatory response by suppressing NLRP-3/AIM-2 inflammasome activation, PI3K/Akt/MAPKs/NF-κB redox inflammatory signalling, modulating autophagy and macrophage polarization imbalance. Mol Biol Rep 2024; 51:1159. [PMID: 39549151 DOI: 10.1007/s11033-024-10068-9] [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: 09/21/2024] [Accepted: 10/25/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Herbacetin, a flavonol abundant in traditional medicines, is documented as an anti-inflammatory agent. However, information regarding its attributes on lipopolysaccharide (LPS)-induced inflammatory immunopathies has not been delineated yet. The present study aimed to comprehend herbacetin effects on LPS-induced aspects of unwarranted, non-resolving inflammation, particularly via targeting the vicious circle of oxi-inflammatory stress, autophagy-apoptosis, macrophages polarization, impaired inflammasome activation, and inflammatory cascades. METHODS AND RESULTS In-vitro model of LPS-stimulated RAW 264.7 macrophage was recapitulated to investigate different inflammatory anomalies using enzyme-linked immunosorbent assay, qRT-PCR (Real-Time Quantitative Reverse Transcription PCR), immunoblotting. Concanavalin A challenged splenocytes and in silico studies were performed to measure Tregs population and binding affinity, respectively. RESULTS Herbacetin administration caused remarkable reduction in nitric oxide, reactive oxygen species, mitochondrial membrane potential hyperpolarization, tumor necrosis factor-α, interferon-γ, interleukin-6, inducible nitric oxide synthase and ratio of M1/M2 markers (inducible nitric oxide synthase/arginase-1/macrophage scavenger receptor-1/mannose receptor C type-1) in in vitro model of persistent inflammation. Suppression of interleukins-5,17 and matrix metalloproteinases-2,3,9,13 and proliferating cell nuclear antigen, signifies its anti-inflammatory attributes. Noticeable decline in monodansylcadaverine-Lysotracker staining, caspase-6, and enhanced p62, B-cell lymphoma-2 expression indicates apoptosis-autophagosome accumulation inhibition and lysosomal destabilization. These were accompanied by reduced NLRP3 activation, caspase-1, AIM-2 expression, and interleukin-1β release. Subsequently, up-regulated activation of TLR-4, NF-κB, PI3K, Akt, ERK1/2, and JNK was decisively thwarted by herbacetin. In silico investigation signified the interaction of herbacetin with these targets. Decreased cytokines and enhanced Tregs conferred its role in extenuating inflammation facilitated by T-cells depletion. CONCLUSION Collectively, these findings comprehend attributes of herbacetin as an alternative therapeutic strategy in relieving LPS-associated chronic inflammatory disorders.
Collapse
Affiliation(s)
- Monika Kumari
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, P.O. Box No: 6, Palampur, HP, 176061, India
- Academy of Scientific and Innovative Research, Ghaziabad, UP, India
| | - Anamika Sharma
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, P.O. Box No: 6, Palampur, HP, 176061, India
| | - Narendra Vijay Tirpude
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, P.O. Box No: 6, Palampur, HP, 176061, India.
- Academy of Scientific and Innovative Research, Ghaziabad, UP, India.
| |
Collapse
|
2
|
de Souza Goncalves B, Sangani D, Nayyar A, Puri R, Irtiza M, Nayyar A, Khalyfa A, Sodhi K, Pillai SS. COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals. Int J Mol Sci 2024; 25:8481. [PMID: 39126050 PMCID: PMC11312872 DOI: 10.3390/ijms25158481] [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: 07/08/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
The acute manifestations of coronavirus disease 2019 (COVID-19) exhibit the hallmarks of sepsis-associated complications that reflect multiple organ failure. The inflammatory cytokine storm accompanied by an imbalance in the pro-inflammatory and anti-inflammatory host response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to severe and critical septic shock. The sepsis signature in severely afflicted COVID-19 patients includes cellular reprogramming and organ dysfunction that leads to high mortality rates, emphasizing the importance of improved clinical care and advanced therapeutic interventions for sepsis associated with COVID-19. Phytochemicals of functional foods and nutraceutical importance have an incredible impact on the healthcare system, which includes the prevention and/or treatment of chronic diseases. Hence, in the present review, we aim to explore the pathogenesis of sepsis associated with COVID-19 that disrupts the physiological homeostasis of the body, resulting in severe organ damage. Furthermore, we have summarized the diverse pharmacological properties of some potent phytochemicals, which can be used as functional foods as well as nutraceuticals against sepsis-associated complications of SARS-CoV-2 infection. The phytochemicals explored in this article include quercetin, curcumin, luteolin, apigenin, resveratrol, and naringenin, which are the major phytoconstituents of our daily food intake. We have compiled the findings from various studies, including clinical trials in humans, to explore more into the therapeutic potential of each phytochemical against sepsis and COVID-19, which highlights their possible importance in sepsis-associated COVID-19 pathogenesis. We conclude that our review will open a new research avenue for exploring phytochemical-derived therapeutic agents for preventing or treating the life-threatening complications of sepsis associated with COVID-19.
Collapse
Affiliation(s)
- Bruno de Souza Goncalves
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Darshan Sangani
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Aleen Nayyar
- Department of Medicine, Sharif Medical and Dental College, Lahore 55150, Pakistan;
| | - Raghav Puri
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Mahir Irtiza
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Asma Nayyar
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Abdelnaby Khalyfa
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Komal Sodhi
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Sneha S. Pillai
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| |
Collapse
|
3
|
Srdić T, Đurašević S, Lakić I, Ružičić A, Vujović P, Jevđović T, Dakić T, Đorđević J, Tosti T, Glumac S, Todorović Z, Jasnić N. From Molecular Mechanisms to Clinical Therapy: Understanding Sepsis-Induced Multiple Organ Dysfunction. Int J Mol Sci 2024; 25:7770. [PMID: 39063011 PMCID: PMC11277140 DOI: 10.3390/ijms25147770] [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: 05/20/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Sepsis-induced multiple organ dysfunction arises from the highly complex pathophysiology encompassing the interplay of inflammation, oxidative stress, endothelial dysfunction, mitochondrial damage, cellular energy failure, and dysbiosis. Over the past decades, numerous studies have been dedicated to elucidating the underlying molecular mechanisms of sepsis in order to develop effective treatments. Current research underscores liver and cardiac dysfunction, along with acute lung and kidney injuries, as predominant causes of mortality in sepsis patients. This understanding of sepsis-induced organ failure unveils potential therapeutic targets for sepsis treatment. Various novel therapeutics, including melatonin, metformin, palmitoylethanolamide (PEA), certain herbal extracts, and gut microbiota modulators, have demonstrated efficacy in different sepsis models. In recent years, the research focus has shifted from anti-inflammatory and antioxidative agents to exploring the modulation of energy metabolism and gut microbiota in sepsis. These approaches have shown a significant impact in preventing multiple organ damage and mortality in various animal sepsis models but require further clinical investigation. The accumulation of this knowledge enriches our understanding of sepsis and is anticipated to facilitate the development of effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Tijana Srdić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Siniša Đurašević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Iva Lakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Aleksandra Ružičić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Predrag Vujović
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tanja Jevđović
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tamara Dakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Jelena Đorđević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tomislav Tosti
- Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sofija Glumac
- School of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Zoran Todorović
- School of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Nebojša Jasnić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| |
Collapse
|
4
|
Fu CF, Li JL, Chen JW, Liang H, Zhao WR, He SY, Ma XW, Yang XF, Wang HL. Mechanism and therapeutic potential of traditional Chinese medicine extracts in sepsis. Front Pharmacol 2024; 15:1365639. [PMID: 39021837 PMCID: PMC11251979 DOI: 10.3389/fphar.2024.1365639] [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: 01/04/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Sepsis is a complex syndrome characterized by multi-organ dysfunction, due to the presence of harmful microorganisms in blood which could cause mortality. Complications associated with sepsis involve multiple organ dysfunction. The pathogenesis of sepsis remains intricate, with limited treatment options and high mortality rates. Traditional Chinese medicine (TCM) has consistently demonstrated to have a potential on various disease management. Its complements include reduction of oxidative stress, inhibiting inflammatory pathways, regulating immune responses, and improving microcirculation. Traditional Chinese medicine can mitigate or even treat sepsis in a human system. This review examines progress on the use of TCM extracts for treating sepsis through different pharmacological action and its mechanisms. The potential targets of TCM extracts and active ingredients for the treatment of sepsis and its complications have been elucidated through molecular biology research, network pharmacology prediction, molecular docking analysis, and visualization analysis. Our aim is to provide a theoretical basis and empirical support for utilizing TCM in the treatment of sepsis and its complications while also serving as a reference for future research and development of sepsis drugs.
Collapse
Affiliation(s)
- Chen-Fei Fu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jian-Long Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | | | - Hao Liang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wen-Rui Zhao
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Shi-Yu He
- Shenzhen Pingle Orthopedic Hospital, Shenzhen, China
| | - Xiao-Wei Ma
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Xiao-Fan Yang
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - He-Lin Wang
- Donghuashi Community Health Service Center, Beijing, China
| |
Collapse
|
5
|
Xie L, Zhang G, Wu Y, Hua Y, Ding W, Han X, Liu B, Zhou C, Li A. Protective effects of Wenqingyin on sepsis-induced acute lung injury through regulation of the receptor for advanced glycation end products pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155654. [PMID: 38723525 DOI: 10.1016/j.phymed.2024.155654] [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: 10/08/2023] [Revised: 02/06/2024] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Wenqingyin (WQY), an ancient Chinese medicinal agent, has been extensively used in treating infectious ailments throughout history. However, the anti-sepsis mechanism remains unknown. PURPOSE This study investigated the diverse mechanisms of WQY in mitigating sepsis-induced acute lung injury (ALI). Additionally, the effects of WQY were validated using biological experiments. METHODS This study combined UHPLC-Orbitrap-HRMS analysis and network pharmacology to predict the potential anti-sepsis mechanism of WQY. Sepsis-induced ALI models were established in vivo via intraperitoneal lipopolysaccharide (LPS) administration and in vitro by LPS-stimulated RAW 264.7 macrophages. Various techniques, including hematoxylin-eosin staining, TUNEL, qPCR, and ELISA, were used to assess lung damage and quantify inflammatory cytokines. Inflammatory cell infiltration was visualized through immunohistochemistry. Hub targets and signaling pathways were identified using Western blotting, immunohistochemistry, and immunofluorescence staining. RESULTS Seventy-five active components and 237 associated targets were acquired, with 145 of these targets overlapping with processes related to sepsis. Based on the comprehensive protein-protein interaction network analysis, JUN, AKT1, TP53, IL-6, HSP90AA1, CASP3, VEGFA, IL-1β, RELA, and EGFR may be targets of WQY for sepsis. Analysis of the Kyoto Gene and Genome Encyclopedia revealed that WQY is implicated in the advanced glycation end products/receptor for advanced glycation end products (AGE/RAGE) signaling pathway. In vivo, WQY alleviated sepsis-induced ALI, suppressing proinflammatory cytokines and inhibiting macrophage/neutrophil infiltration. In vitro, WQY reduced TNF-α, IL-6, and IL-1β in LPS-induced RAW 264.7 macrophages. Furthermore, we verified that WQY protected against sepsis-induced ALI by regulating the RAGE pathway for the first time. Baicalin, coptisine, and paeoniflorin may be the effective components of WQY that inhibit RAGE. CONCLUSION The primary mechanism of WQY in combating sepsis-induced ALI involves controlling RAGE levels and the PI3K/AKT pathway, suppressing inflammation, and mitigating lung damage. This study establishes a scientific foundation for understanding the mechanism of WQY and its clinical use in treating sepsis.
Collapse
Affiliation(s)
- Lingpeng Xie
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China
| | - Guoyong Zhang
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuting Wu
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Yue Hua
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenjun Ding
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xin Han
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China.
| | - Chuying Zhou
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Aimin Li
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China.
| |
Collapse
|
6
|
Yavuz A, Küçük A, Ergörün Aİ, Dursun AD, Yiğman Z, Alkan M, Arslan M. Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation. Exp Ther Med 2024; 27:242. [PMID: 38655036 PMCID: PMC11036365 DOI: 10.3892/etm.2024.12530] [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: 06/28/2023] [Accepted: 10/27/2023] [Indexed: 04/26/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome that develops in the host against microorganisms. This response develops away from the primary infection site and results in end-organ damage. The present study aimed to investigate the protective and therapeutic effects on lung and kidney tissue of silymarin (S) and dexmedetomidine (DEX) applied 1 h before and after sepsis induced by the cecal ligation and puncture (CLP) method in rats. A total of 62 rats was randomly divided into eight groups: i) Control (n=6); ii) cecal perforation (CLP; n=8); iii) S + CLP (n=8; S + CLP; S administered 1 h before CPL); iv) CLP + S (n=8; S administered 1 h after CLP); v) DEX + CLP (n=8; D + CLP; DEX administered 1 h before CLP); vi) CLP + D (n=8; DEX administered 1 h after CLP); vii) SD + CLP (n=8; S and DEX administered 1 h before CLP) and viii) CLP + SD (n=8; S and DEX administered 1 h after CLP). After the cecum filled with stool, it was tied with 3/0 silk under the ileocecal valve and the anterior surface of the cecum was punctured twice with an 18-gauge needle. A total of 100 mg/kg silymarin and 100 µg/kg DEX were administered intraperitoneally to the treatment groups. Lung and kidney tissue samples were collected to evaluate biochemical and histopathological parameters. In the histopathological examination, all parameters indicating kidney injury; interstitial edema, peritubular capillary dilatation, vacuolization, ablation of tubular epithelium from the basement membrane, loss of brush border in the proximal tubule epithelium, cell swelling and nuclear defragmentation; were increased in the CLP compared with the control group. Silymarin administration increased kidney damage, including ablation of tubular epithelium from the basement membrane, compared with that in the CLP group. DEX significantly reduced kidney damage compared with the CLP and silymarin groups. The co-administration of DEX + silymarin decreased kidney damage, although it was not as effective as DEX-alone. To conclude, intraperitoneal DEX ameliorated injury in CLP rats. DEX + silymarin partially ameliorated injury but silymarin administration increased damage. As a result, silymarin has a negative effects with this dosage and DEX has a protective effect. In the present study, it was determined that using the two drugs together had a greater therapeutic effect than silymarin and no differences in the effects were not observed any when the application times of the agents were changed.
Collapse
Affiliation(s)
- Aydin Yavuz
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ayşegül Küçük
- Department of Physiology, Faculty of Medicine, Kutahya Health Science University, Kutahya 43020, Turkey
| | - Aydan İremnur Ergörün
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ali Doğan Dursun
- Department of Physiology, Faculty of Medicine, Atılım University, Ankara 06830, Turkey
| | - Zeynep Yiğman
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Neuroscience and Neurotechnology Center of Excellence, Gazi University, Ankara 06510, Turkey
| | - Metin Alkan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Life Sciences Application and Research Center, Gazi University, Ankara 06830, Turkey
- Laboratory Animal Breeding and Experimental Research Center, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
7
|
Chen Z, Su Y, Ding J, He J, Lai L, Song Y. Lobetyolin protects mice against LPS-induced sepsis by downregulating the production of inflammatory cytokines in macrophage. Front Pharmacol 2024; 15:1405163. [PMID: 38799158 PMCID: PMC11116692 DOI: 10.3389/fphar.2024.1405163] [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: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction: Sepsis is a clinical syndrome characterized by dysregulation of the host immune response due to infection, resulting in life-threatening organ damage. Despite active promotion and implementation of early preventative measures and bundle treatments, sepsis continues to exhibit high morbidity and mortality rates with no optimal pharmacological intervention available. Lobetyolin (LBT), the crucial component of polyacetylenes found in Codonopsis pilosula, has been scientifically proven to possess potent antioxidant and antitumor properties. However, its therapeutic potential for sepsis remains unknown. Methods: The mice received pretreatment with intraperitoneal injections of LBT, followed by injection with lipopolysaccharide (LPS) to induce sepsis. Peripheral blood samples were collected to detect TNF-α, IL-1β, and IL-6 levels. The survival status of different groups was recorded at various time intervals. RNA-Seq was utilized for the analysis of gene expression in peritoneal macrophages treated with LBT or LPS. Results: In this study, we observed a significant increase in the survival rate of mice pretreated with LBT in LPS induced sepsis mouse model. LBT demonstrated a remarkable reduction in the production of IL-6, TNF-α, and IL-1β in the serum, along with mitigated lung and liver tissue damage characterized by reduced inflammatory cell infiltration. Additionally, through RNA-seq analysis coupled with GO and KEGG analysis, it was revealed that LBT effectively suppressed genes associated with bacterium presence, cellular response to lipopolysaccharide stimulation, as well as cytokine-cytokine receptor interaction involving Cxcl10, Tgtp1, Gbp5, Tnf, Il1b and IRF7 specifically within macrophages. We also confirmed that LBT significantly downregulates the expression of IL-6, TNF-α, and IL-1β in macrophage activation induced by LPS. Discussion: Therefore, our findings demonstrated that LBT effectively inhibits the production of inflammatory cytokines (IL-6, TNF-α, and IL-1β) and mitigates sepsis induced by LPS through modulating macrophages' ability to generate these cytokines. These results suggest that LBT holds promise as a potential therapeutic agent for sepsis treatment.
Collapse
Affiliation(s)
- Zhonghua Chen
- Department of Emergency, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yixin Su
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingtong Ding
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia He
- Teaching Experimental Center of Public Health, Zhejiang University, Hangzhou, China
| | - Lihua Lai
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinjing Song
- Centre of Biomedical Research, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Hwang YS, Lim J, Yoon HR, Park SH, Kim A, Jang JP, Cho HJ, Lee HG. Astragalus Complanatus Ethanol Attenuates Septic Shock by Exerting Anti-Inflammatory Effects on Macrophages. Int J Mol Sci 2023; 25:384. [PMID: 38203555 PMCID: PMC10778658 DOI: 10.3390/ijms25010384] [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: 11/27/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Sepsis is a systemic inflammatory syndrome that results in multiple-organ failure caused by a dysregulated host immune response to microbial infection. Astragali complanati semen extract (ACSE) exhibits pharmacological activities, including antioxidant, anticancer, antiaging, and anti-diabetes effects. It is widely used in traditional medicine to treat liver and kidney diseases; however, the protective effect of ACSE on sepsis and its mechanisms are unknown. In the present study, we investigated the anti-inflammatory effects and potential mechanisms of the action of ACSE on sepsis. We show that ACSE improved survival rates in mouse models of acute sepsis induced by CLP (cecal ligation and puncture) and LPS stimulation. ACSE administration decreased aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in sepsis-induced mice. Furthermore, ACSE reduced the levels of nitric oxide (NO), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the serum of septic mice. ACSE treatment inhibited the expression of these proinflammatory genes in LPS-stimulated J774 macrophages. Moreover, ACSE inhibited the phosphorylation of the IκB kinase (IKK) and the nuclear translocation of p65 NF-κB by LPS stimulation in macrophages. These results reveal the mechanism underlying the protective effect of ACSE against sepsis by inhibiting NF-κB activation and suggest that ACSE could be a potential therapeutic candidate to treat acute inflammatory diseases.
Collapse
Affiliation(s)
- Yo Sep Hwang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
| | - Jeewon Lim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hyang Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea;
| | - Aeyung Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Jun-Pil Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
| | - Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
9
|
de O. Trovão L, dos S. Rodrigues L, Mendes PM, Alves PCS, da S. Oliveira A, Brito JM, Vale AAM, de O. Garbis DV, Simão G, dos Santos APSA, Pereira PVS, Silva LA, Berretta AA, Nascimento FRF, Guerra RNM, Monteiro-Neto V, Fernandes ES, Maciel MCG. The Immunomodulatory Activity of Punica granatum L. Peel Extract Increases the Lifespan of Mice with Lethal Sepsis. J Immunol Res 2023; 2023:2868707. [PMID: 37621924 PMCID: PMC10447006 DOI: 10.1155/2023/2868707] [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: 04/19/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023] Open
Abstract
Sepsis is an organ dysfunction syndrome associated with high mortality. To date, no effective treatment is available to combat this disease. Punica granatum L. is a potential alternative treatment due to its anti-inflammatory, antimicrobial, and antioxidant properties. Thus, this study aimed to evaluate the effects of a hydroalcoholic crude extract from the peels of P. granatum (HCEPg) in mice with lethal sepsis. Lethal polymicrobial sepsis was induced in female Swiss mice via cecal ligation and puncture (CLP). Initially, the animals were divided into three groups: Sham (false-operated), CLP-control (phosphate-buffered saline), and CLP-HCEPg (single dose, 5 mg/kg, subcutaneous administration). Treatment was initiated immediately after the induction of sepsis, and survival was evaluated every 12 hr for 5 days. Those who survived were euthanized. Serum cytokine levels were measured using a cytometric bead array Mouse Inflammatory Cytokine Kit. The number of colony-forming units, as well as the number of cells in the lymphoid organs and their activation markers, were analyzed. Results showed that treatment with HCEPg increased lifespan and reduced bacterial counts in the peritoneum, bloodstream, and spleen. HCEPg also decreased hydrogen peroxide secretion by phagocytes and augmented serum IL-10 levels, indicating its systemic anti-inflammatory effects. Additionally, treatment with HCEPg attenuated infection-induced lung hemorrhage. Overall, P. granatum extract improved the lifespan of septic mice, possibly due to its antimicrobial, anti-inflammatory, and immunomodulatory effects, thereby regulating bacterial load and translocation, as well as controlling the systemic inflammation induced by sepsis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gisele Simão
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe e Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | | | | | | | | | | | | | | | - Elizabeth S. Fernandes
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe e Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | | |
Collapse
|
10
|
Chen Y, Bian H, Lv J, Song W, Xing C, Hui C, Zhang D, Zhang C, Zhao L, Li Y, Su L. Gelsevirine is a novel STING-specific inhibitor and mitigates STING-related inflammation in sepsis. Front Immunol 2023; 14:1190707. [PMID: 37583703 PMCID: PMC10424845 DOI: 10.3389/fimmu.2023.1190707] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/13/2023] [Indexed: 08/17/2023] Open
Abstract
Background Stimulation of IFN genes (STING) is central to the production of interferon and proinflammatory cytokines in response to microbial DNA or self-DNA in the cytosol. The detrimental role of the activation of STING during sepsis has been well documented. Methods Here, we found that gelsevirine (GS) potently inhibit interferon and inflammatory cytokine induction in macrophages exposed to STING agonists (2'3'-cGAMP, IFN stimulatory DNA (ISD), and poly(dA:dT)). I n silico docking analysis and surface plasmon resonance binding study showed that GS bonds with high affinity to the cyclic dinucleotide (CDN)-binding pocket of STING. Biotin pull-down assay also confirmed that GS competitively bonded to STING protein. Furthermore, GS inhibited 2'3'-cGAMP-induced STING dimerization and subsequent activation. In addition, GS induced K48-linked STING ubiquitination and degradation, which was likely through upregulating and recruiting TRIM21. In mice exposed to cecal ligation and puncture (CLP)-induced sepsis, post-operative administration of GS significantly extended the survival period and mitigated acute organ damage. Results Overall, GS inhibited STING signaling by competitively binding to the CDN-binding pocket to lock STING in an inactive open conformation, while also promoting K48-linked STING ubiquitination and degradation. Conclusions Our findings identify a novel STING-specific inhibitor that could be applied in the treatment of sepsis.
Collapse
Affiliation(s)
- Yuhong Chen
- School of Pharmacy, Bengbu Medical College, Bengbu, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Huihui Bian
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Juan Lv
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Wanxue Song
- Department of Anesthesiology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chunlei Xing
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Chunlei Hui
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Dinglei Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Chenxi Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Liang Zhao
- Luodian Clinical Drug Research Center, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Yingke Li
- Department of Anesthesiology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Li Su
- School of Pharmacy, Bengbu Medical College, Bengbu, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Luodian Clinical Drug Research Center, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
| |
Collapse
|
11
|
Jeong SH, Park JY, Ryu YB, Kim WS, Lee IC, Kim JH, Kim D, Ha JH, Lee BW, Nam J, Cho KO, Kwon HJ. Myristica fragrans Extract Inhibits Platelet Desialylation and Activation to Ameliorate Sepsis-Associated Thrombocytopenia in a Murine CLP-Induced Sepsis Model. Int J Mol Sci 2023; 24:ijms24108863. [PMID: 37240208 DOI: 10.3390/ijms24108863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Sepsis, characterized by an uncontrolled host inflammatory response to infections, remains a leading cause of death in critically ill patients worldwide. Sepsis-associated thrombocytopenia (SAT), a common disease in patients with sepsis, is an indicator of disease severity. Therefore, alleviating SAT is an important aspect of sepsis treatment; however, platelet transfusion is the only available treatment strategy for SAT. The pathogenesis of SAT involves increased platelet desialylation and activation. In this study, we investigated the effects of Myristica fragrans ethanol extract (MF) on sepsis and SAT. Desialylation and activation of platelets treated with sialidase and adenosine diphosphate (platelet agonist) were assessed using flow cytometry. The extract inhibited platelet desialylation and activation via inhibiting bacterial sialidase activity in washed platelets. Moreover, MF improved survival and reduced organ damage and inflammation in a mouse model of cecal ligation and puncture (CLP)-induced sepsis. It also prevented platelet desialylation and activation via inhibiting circulating sialidase activity, while maintaining platelet count. Inhibition of platelet desialylation reduces hepatic Ashwell-Morell receptor-mediated platelet clearance, thereby reducing hepatic JAK2/STAT3 phosphorylation and thrombopoietin mRNA expression. This study lays a foundation for the development of plant-derived therapeutics for sepsis and SAT and provides insights into sialidase-inhibition-based sepsis treatment strategies.
Collapse
Affiliation(s)
- Seong-Hun Jeong
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ji-Young Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea
| | - Young Bae Ryu
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea
| | - Ju-Hong Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Dohoon Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Ji-Hye Ha
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Ba-Wool Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Jiyoung Nam
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Kyoung-Oh Cho
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyung-Jun Kwon
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea
| |
Collapse
|
12
|
Yang K, Wu B, Wei W, Li C, Li L, Cong Z, Xiang Q. Curdione ameliorates sepsis-induced lung injury by inhibiting platelet-mediated neutrophil extracellular trap formation. Int Immunopharmacol 2023; 118:110082. [PMID: 36989889 DOI: 10.1016/j.intimp.2023.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Sepsis-associated acute lung injury remains to be a major cause of morbidity and mortality worldwide, and there is a lack of effective therapeutic drugs. Curdione, an activeingredient of Curcuma zedoary, a traditional Chinese medicine (TCM), possesses a variety of pharmacological actions, such as anti-inflammatory, antioxidant and inhibition of platelet aggregation. However, whether curdione protects against sepsis-induced lung injury is still undetermined. In this study, we investigated the effects of curdione on sepsis-induced lung injury. Cecal ligation and puncture (CLP) surgery was performed in mice to establish a model of sepsis. Twenty-four hours after CLP, bronchoalveolar lavage fluid (BALF) and lung tissue samples were harvested for investigation. The protective effects of curdione on acute lung injury and potential mechanisms were explored by detecting pathological sections, exudative proteins, oxidative responses, inflammatory factors, platelet activation, neutrophil infiltration, and neutrophil extracellular trap (NET) formation in the lung and were further verified in vitro. We showed that treatment with curdione clearly relieved histopathological changes, reduced inflammatory cytokine elevation and total protein concentrations in BALF, and decreased oxidative stress responses in lung tissues. In addition, curdione inhibited platelet activation, further blocking the interaction between platelets and neutrophils. Finally, neutrophil infiltration and NET formation was also reduced in mice treated with curdione. In conclusion, curdione alleviates sepsis-induced lung injury by inhibiting platelet-mediated neutrophil recruitment, infiltration, and NET formation as well as its anti-inflammatory and antioxidant properties. Curdione has great therapeutic potential in sepsis.
Collapse
Affiliation(s)
- Kai Yang
- Department of Anesthesiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725, South Wanping Road, Xuhui District, Shanghai 200032, China
| | - Bin Wu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Wu Wei
- Department of Anesthesiology, the Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, No. A17, Heishanhu Road, Haidian District, Beijing 100091, China
| | - Cuiyu Li
- Department of Anesthesiology, the Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, No. A17, Heishanhu Road, Haidian District, Beijing 100091, China
| | - Lu Li
- Department of Infectious Diseases, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhukai Cong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| | - Qian Xiang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| |
Collapse
|
13
|
Usmani J, Wasim M, Ansari MN, Hassan MJ, Sharma M, Ahmad R. Potential therapeutic effect of Carica papaya leaves extract on immune response, biochemical and hematological mechanisms on cecal ligation and puncture model of sepsis in rats: an in vivo study. 3 Biotech 2023; 13:151. [PMID: 37131965 PMCID: PMC10148938 DOI: 10.1007/s13205-023-03567-2] [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: 07/16/2022] [Accepted: 04/19/2023] [Indexed: 05/04/2023] Open
Abstract
Antibiotics and immunotherapies possess unavoidable adverse effects that hinder sepsis management. Herbal drugs have demonstrated potential immunomodulatory properties vital for sepsis treatment. We hypothesized in the present study that the use of Carica papaya leaves extract had the potential to improve survival and modulate immune cytokine release during sepsis. Animals were subjected to cecal ligation and puncture (CLP) to induce sepsis. Septic rats divided into 10 groups received ethanol extract of C. papaya leaves (50 and 100 mg/kg), imipenem (120 mg/kg) and cyclophosphamide (CP, 10 mg/kg). To investigate the immunomodulatory potentials of EE, cytokine levels like interleukin (IL-6), tumor necrosis factor (TNF-α), and IL-10 along with hematological and biochemical parameters were analyzed. Our results exhibited improved survival rates concerning ethanol extract treatment alone and in combination with imipenem and CP (100%) as compared to the CLP group (33.3%) on day 7 post-surgery. The combination treatment of ethanol extract with imipenem and CP significantly (P < 0.001) ameliorated cytokine levels and hematological and biochemical parameters in septic rats. A histopathological examination suggested improved liver and kidney tissue condition after combination treatment as compared to the CLP group. Therefore, it was concluded that combination therapy of extract with imipenem and CP improved survival rates and marked immunomodulatory potential in septic rats compared to monotherapy. The findings suggested the use of a mixture of these drugs in clinical settings to treat sepsis.
Collapse
Affiliation(s)
- Juveria Usmani
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Mohd Wasim
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Mohd Nazam Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942 Kingdom of Saudi Arabia
| | - Mohammed Jaseem Hassan
- Department of Pathology, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Razi Ahmad
- Department of Pharmacology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062 India
| |
Collapse
|
14
|
He S, Zhao C, Guo Y, Zhao J, Xu X, Hu Y, Lian B, Ye H, Wang N, Luo L, Liu Q. Alterations in the gut microbiome and metabolome profiles of septic mice treated with Shen FuHuang formula. Front Microbiol 2023; 14:1111962. [PMID: 36970673 PMCID: PMC10030955 DOI: 10.3389/fmicb.2023.1111962] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
Sepsis has a high mortality rate, and treating sepsis remains a significant challenge worldwide. In former studies, our group found that traditional Chinese medicine, Shen FuHuang formula (SFH), is a promising medicine in treating coronavirus disease 2019 (COVID-19) patients with the septic syndrome. However, the underlying mechanisms remain elusive. In the present study, we first investigated the therapeutic effects of SFH on septic mice. To investigate the mechanisms of SFH-treated sepsis, we identified the gut microbiome profile and exploited untargeted metabolomics analyses. The results demonstrated that SFH significantly enhanced the mice’s 7-day survival rate and hindered the release of inflammatory mediators, i.e., TNF-α, IL-6, and IL-1β. 16S rDNA sequencing further deciphered that SFH decreased the proportion of Campylobacterota and Proteobacteria at the phylum level. LEfSe analysis revealed that the treatment of SFH enriched Blautia while decreased Escherichia_Shigella. Furthermore, serum untargeted metabolomics analysis indicated that SFH could regulate the glucagon signaling pathway, PPAR signaling pathway, galactose metabolism, and pyrimidine metabolism. Finally, we found the relative abundance of Bacteroides, Lachnospiraceae_NK4A136_group, Escherichia_Shigella, Blautia, Ruminococcus, and Prevotella were closely related to the enrichment of the metabolic signaling pathways, including L-tryptophan, uracil, glucuronic acid, protocatechuic acid, and gamma-Glutamylcysteine. In conclusion, our study demonstrated that SFH alleviated sepsis by suppressing the inflammatory response and hence reduced mortality. The mechanism of SFH for treating sepsis may be ascribed to the enrichment of beneficial gut flora and modulation in glucagon signaling pathway, PPAR signaling pathway, galactose metabolism, and pyrimidine metabolism. To sum up, these findings provide a new scientific perspective for the clinical application of SFH in treating sepsis.
Collapse
Affiliation(s)
- Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Chunxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Yuhong Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Yahui Hu
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bo Lian
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Haoran Ye
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Ning Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China
- Lianxiang Luo,
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Institute of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
- *Correspondence: Qingquan Liu,
| |
Collapse
|
15
|
Usmani J, Kausar H, Akbar S, Sartaj A, Mir SR, Hassan MJ, Sharma M, Ahmad R, Rashid S, Ansari MN. Molecular Docking of Bacterial Protein Modulators and Pharmacotherapeutics of Carica papaya Leaves as a Promising Therapy for Sepsis: Synchronising In Silico and In Vitro Studies. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020574. [PMID: 36677632 PMCID: PMC9862608 DOI: 10.3390/molecules28020574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/09/2023]
Abstract
Sepsis is a serious health concern globally, which necessitates understanding the root cause of infection for the prevention of proliferation inside the host's body. Phytochemicals present in plants exhibit antibacterial and anti-proliferative properties stipulated for sepsis treatment. The aim of the study was to determine the potential role of Carica papaya leaf extract for sepsis treatment in silico and in vitro. We selected two phytochemical compounds, carpaine and quercetin, and docked them with bacterial proteins, heat shock protein (PDB ID: 4PO2), surfactant protein D (PDB ID: 1PW9), and lactobacillus bacterial protein (PDB ID: 4MKS) against imipenem and cyclophosphamide. Quercetin showed the strongest interaction with 1PW9 and 4MKS proteins. The leaves were extracted using ethanol, methanol, and water through Soxhlet extraction. Total flavonoid content, DPPH assay, HPTLC, and FTIR were performed. In vitro cytotoxicity of ethanol extract was screened via MTT assay on the J774 cell line. Ethanol extract (EE) possessed the maximum number of phytocomponents, the highest amount of flavonoid content, and the maximum antioxidant activity compared to other extracts. FTIR analysis confirmed the presence of N-H, O-H, C-H, C=O, C=C, and C-Cl functional groups in ethanol extract. Cell viability was highest (100%) at 25 µg/mL of EE. The present study demonstrated that the papaya leaves possessed antibacterial and cytotoxic activity against sepsis infection.
Collapse
Affiliation(s)
- Juveria Usmani
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Hina Kausar
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Saleem Akbar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Ali Sartaj
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Showkat R. Mir
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Mohammed Jaseem Hassan
- Department of Pathology, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Razi Ahmad
- Department of Pharmacology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard University, New Delhi 110062, India
- Correspondence: (R.A.); (M.N.A.)
| | - Summaya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohd Nazam Ansari
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Correspondence: (R.A.); (M.N.A.)
| |
Collapse
|
16
|
Mahomoodally MF, Aumeeruddy MZ, Legoabe LJ, Dall’Acqua S, Zengin G. Plants' bioactive secondary metabolites in the management of sepsis: Recent findings on their mechanism of action. Front Pharmacol 2022; 13:1046523. [PMID: 36588685 PMCID: PMC9800845 DOI: 10.3389/fphar.2022.1046523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis is a severe inflammatory response to systemic infection and is a threatening cause of death in intensive care units. In recent years, a number of studies have been conducted on the protective effect of natural products against sepsis-induced organ injury. However, a comprehensive review of these studies indicating the mechanisms of action of the bioactive compounds is still lacking. In this context, this review aimed to provide an updated analysis of the mechanism of action of plants' secondary metabolites in the management of sepsis. Scopus, Science Direct, Google Scholar, and PubMed were searched from inception to July 2022. A variety of secondary metabolites were found to be effective in sepsis management including allicin, aloin, cepharanthine, chrysin, curcumin, cyanidin, gallic acid, gingerol, ginsenoside, glycyrrhizin, hesperidin, kaempferol, narciclasine, naringenin, naringin, piperine, quercetin, resveratrol, rosmarinic acid, shogaol, silymarin, sulforaphane, thymoquinone, umbelliferone, and zingerone. The protective effects exerted by these compounds can be ascribed to their antioxidant properties as well as induction of endogenous antioxidant mechanisms, and also via the downregulation of inflammatory response and reduction of biochemical and inflammatory markers of sepsis. These findings suggest that these secondary metabolites could be of potential therapeutic value in the management of sepsis, but human studies must be performed to provide strength to their potential clinical relevance in sepsis-related morbidity and mortality reduction.
Collapse
Affiliation(s)
- Mohamad Fawzi Mahomoodally
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam,Faculty of Natural Sciences, Duy Tan University, Da Nang, Vietnam,*Correspondence: Mohamad Fawzi Mahomoodally, ; Stefano Dall’Acqua,
| | | | - Lesetja Jan Legoabe
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen), North West University, Potchefstroom, South Africa
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy,*Correspondence: Mohamad Fawzi Mahomoodally, ; Stefano Dall’Acqua,
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University, Campus, Konya, Turkey
| |
Collapse
|
17
|
Hwang YS, Jang JP, Park SH, Kim A, Jang JH, Yoon HR, Yoon SR, Park JH, Cho HJ, Lee HG. Ponciri Fructus Immaturus ethanol extract attenuates septic shock through inhibition of the STAT1 signaling pathway. Front Nutr 2022; 9:988309. [PMID: 36185646 PMCID: PMC9520981 DOI: 10.3389/fnut.2022.988309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a systemic inflammatory disease to infections and results in tissue damage and multiple organ failure. Ponciri Fructus Immaturus (PFI) is widely used in traditional medicine for allergic inflammation and gastrointestinal disorders. However, the effect of PFI on sepsis is still unknown. This study investigated the anti-inflammatory and antiseptic effects of PFI ethanol extract (PFIE) in LPS-stimulated J774 macrophages and mice with CLP- or LPS-induced sepsis, respectively. PFIE attenuates the LPS-induced production of the proinflammatory mediator NO by inhibiting the expression of iNOS in J774 cells. Real-time RT-PCR data and ELISA showed that the mRNA and protein levels of TNF-α, IL-1β, and IL-6 increased in LPS-stimulated J774 cells. However, this induction was significantly suppressed in PFIE pre-treated J774 cells. We also found that PFIE administration increased the survival rate of mice with LPS- and CLP-induced sepsis. Decreased serum levels of AST, ALT, and CK were observed after administration of PFIE, which was associated with reduced production of proinflammatory factors, such as NO, TNF-α, IL-1β, and IL-6. Moreover, PFIE suppressed the phosphorylation and nuclear translocation of STAT1 in LPS-stimulated J774 cells, suggesting that PFIE can inhibit LPS- and CLP-induced septic shock by suppressing the STAT1 activation. These findings provide the potential therapeutic relevance of PFIE in treating acute inflammatory disease.
Collapse
Affiliation(s)
- Yo Sep Hwang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jun-Pil Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon, South Korea
| | - Aeyung Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, South Korea
| | - Jae-Hyuk Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Hyang Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Suk Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, South Korea
- *Correspondence: Jun Hong Park,
| | - Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- Hee Jun Cho,
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
- Hee Gu Lee,
| |
Collapse
|
18
|
Fu YS, Kang N, Yu Y, Mi Y, Guo J, Wu J, Weng CF. Polyphenols, flavonoids and inflammasomes: the role of cigarette smoke in COPD. Eur Respir Rev 2022; 31:31/164/220028. [PMID: 35705209 PMCID: PMC9648508 DOI: 10.1183/16000617.0028-2022] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
COPD is predicted to become the third leading cause of morbidity and mortality worldwide by 2030. Cigarette smoking (active or passive) is one of its chief causes, with about 20% of cigarette smokers developing COPD from cigarette smoke (CS)-induced irreversible damage and sustained inflammation of the airway epithelium. Inflammasome activation leads to the cleavage of pro-interleukin (IL)-1β and pro-IL-18, along with the release of pro-inflammatory cytokines via gasdermin D N-terminal fragment membrane pores, which further triggers acute phase pro-inflammatory responses and concurrent pyroptosis. There is currently intense interest in the role of nucleotide-binding oligomerisation domain-like receptor family, pyrin domain containing protein-3 inflammasomes in chronic inflammatory lung diseases such as COPD and their potential for therapeutic targeting. Phytochemicals including polyphenols and flavonoids have phyto-medicinal benefits in CS-COPD. Here, we review published articles from the last decade regarding the known associations between inflammasome-mediated responses and ameliorations in pre-clinical manifestations of CS-COPD via polyphenol and flavonoid treatment, with a focus on the underlying mechanistic insights. This article will potentially assist the development of drugs for the prevention and therapy of COPD, particularly in cigarette smokers. This review compiles current investigations into the role of polyphenols/flavonoids in the alleviation of cigarette smoke-induced inflammasome; notably it provides a promising hit for rectifying the treatment of COPD.https://bit.ly/36OcUO9
Collapse
Affiliation(s)
- Yaw-Syan Fu
- Anatomy and Functional Physiology Section, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China.,Institute of Respiratory Disease, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| | - Ning Kang
- Dept of Otorhinolaryngology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China
| | - Yanping Yu
- Institute of Respiratory Disease, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| | - Yan Mi
- Institute of Respiratory Disease, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| | - Jialin Guo
- Anatomy and Functional Physiology Section, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| | - Jingyi Wu
- Anatomy and Functional Physiology Section, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| | - Ching-Feng Weng
- Anatomy and Functional Physiology Section, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China .,Institute of Respiratory Disease, Dept of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, China
| |
Collapse
|
19
|
Ng MY, Lin T, Chao SC, Chu PM, Yu CC. Potential Therapeutic Applications of Natural Compounds in Diabetes-Associated Periodontitis. J Clin Med 2022; 11:jcm11133614. [PMID: 35806899 PMCID: PMC9267692 DOI: 10.3390/jcm11133614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetes mellitus (DM) is a major worldwide health burden. DM is a metabolic disease characterized by chronic hyperglycemia, and if left untreated, can lead to various complications. Individuals with uncontrolled DM are more susceptible to periodontitis due to both a hyper-inflammatory host response and an impaired immune response. Periodontitis, on the other hand, may exacerbate DM by increasing both local and systemic inflammatory components of DM-related complications. The current standard for periodontal treatment in diabetes-associated periodontitis (DP) focuses mostly on reducing bacterial load and less on controlling the excessive host response, and hence, may not be able to resolve DP completely. Over the past decade, natural compounds have emerged as an adjunct approach for modulating the host immune response with the hope of curing DP. The anti-oxidant, anti-inflammatory, and anti-diabetic characteristics of natural substances are well-known, and they can be found in regularly consumed foods and drinks, as well as plants. The pathophysiology of DP and the treatment benefits of various bioactive extracts for DP will be covered in this review.
Collapse
Affiliation(s)
- Min Yee Ng
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan; (M.Y.N.); (T.L.)
| | - Taichen Lin
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan; (M.Y.N.); (T.L.)
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Shih-Chi Chao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research and Education, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yi-lan, Luodong 265501, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404333, Taiwan;
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan; (M.Y.N.); (T.L.)
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan;
- Correspondence: ; Tel.: +886-4-2471-8668
| |
Collapse
|
20
|
Wu Y, Li D, Wang H, Wan X. Protective Effect of Poria Cocos Polysaccharides on Fecal Peritonitis-Induced Sepsis in Mice Through Inhibition of Oxidative Stress, Inflammation, Apoptosis, and Reduction of Treg Cells. Front Microbiol 2022; 13:887949. [PMID: 35694296 PMCID: PMC9184799 DOI: 10.3389/fmicb.2022.887949] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/14/2022] [Indexed: 12/29/2022] Open
Abstract
This study was conducted to investigate the potential pharmacological effects of Poria cocos polysaccharides (PCPs) on fecal-induced peritonitis (FIP) mice. Consequently, the fecal peritonitis (FP)-induced septic mice with the higher levels of tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), IL-1β, malondialdehyde (MDA), myeloperoxidase (MPO), histopathological lesion and bacterial burden, and lower levels of superoxide dismutase (SOD) and glutathione (GSH). Interestingly, PCP pre-treatment reduced inflammatory cytokines and oxidative stress in plasma and spleen and improved the resistance to FIP. Inflammatory infiltration and cell death in thymus or splenic tissue were alleviated with PCP pretreatment. Furthermore, Treg cells were moderated in the spleen with PCP pre-administration. In addition, PCP pretreatment downregulated Annexin-V in the thymus of FP-induced septic mice, and apoptosis of splenic cells was dose-dependent. In conclusion, PCPs have pharmacological and biological effects on FP-induced septic mice, and its molecular mechanism is related to antioxidative, anti-inflammation, anti-apoptosis, and the reduction of Treg activity in splenic cells.
Collapse
Affiliation(s)
- Yu Wu
- Department of Anesthesiology and Intensive Care Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dai Li
- Department of Anesthesiology and Intensive Care Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Han Wang
- Department of Anesthesiology and Intensive Care Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaojian Wan
- Department of Anesthesiology and Intensive Care Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Xiaojian Wan
| |
Collapse
|