1
|
Zhang J, Huang FY, Dai SZ, Wang L, Zhou X, Zheng ZY, Li Q, Tan GH, Wang CC. Toxicarioside H-mediated modulation of the immune microenvironment attenuates ovalbumin-induced allergic airway inflammation by inhibiting NETosis. Int Immunopharmacol 2024; 136:112329. [PMID: 38815351 DOI: 10.1016/j.intimp.2024.112329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024]
Abstract
PURPOSE Our team identified a new cardiac glycoside, Toxicarioside H (ToxH), in a tropical plant. Previous research has indicated the potential of cardenolides in mitigating inflammation, particularly in the context of NETosis. Therefore, this study sought to examine the potential of ToxH in attenuating allergic airway inflammation by influencing the immune microenvironment. METHODS An OVA-induced airway inflammation model was established in BALB/c mice. After the experiment was completed, serum, bronchoalveolar lavage fluid (BALF), and lung tissue samples were collected and further examined using H&E and PAS staining, flow cytometry, immunofluorescence observation, and Western blot analysis. RESULTS Treatment with ToxH was found to be effective in reducing airway inflammation and mucus production. This was accompanied by an increase in Th1 cytokines (IFN-γ, IL-2, and TNF-β), and the Th17 cytokine IL-17, while levels of Th2 cytokines (IL-4, IL-5, and IL-13) and Treg cytokines (IL-10 and TGF-β1) were decreased in both the bronchoalveolar lavage fluid (BALF) and the CD45+ immune cells in the lungs. Additionally, ToxH inhibited the infiltration of inflammatory cells and decreased the number of pulmonary CD44+ memory T cells, while augmenting the numbers of Th17 and Treg cells. Furthermore, the neutrophil elastase inhibitor GW311616A was observed to suppress airway inflammation and mucus production, as well as alter the secretion of immune Th1, Th2, Th17, and Treg cytokines in the lung CD45+ immune cells. Moreover, our study also demonstrated that treatment with ToxH efficiently inhibited ROS generation, thereby rectifying the dysregulation of immune cells in the immune microenvironment in OVA-induced allergic asthma. CONCLUSIONS Our findings indicate that ToxH could serve as a promising therapeutic intervention for allergic airway inflammation and various other inflammatory disorders. Modulating the balance of Th1/Th2 and Treg/Th17 cells within the pulmonary immune microenvironment may offer an effective strategy for controlling allergic airway inflammation.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Feng-Ying Huang
- Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 571199, China
| | - Shu-Zhen Dai
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Lin Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China; Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 571199, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Zhen-You Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China.
| | - Guang-Hong Tan
- Key Laborato1y of Tropical Translational Medicine of Ministry of Education & School of Tropical Medicine, Hainan Medical University, Haikou 571199, China.
| | - Cai-Chun Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China.
| |
Collapse
|
2
|
Shim D, Bak Y, Choi HG, Lee S, Park SC. Effects of Panax species and their bioactive components on allergic airway diseases. J Ginseng Res 2024; 48:354-365. [PMID: 39036733 PMCID: PMC11258390 DOI: 10.1016/j.jgr.2024.04.003] [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: 12/28/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 07/23/2024] Open
Abstract
Panax species include Panax ginseng Meyer, Panax quinquefolium L., Panax notoginseng, Panax japonicum, Panax trifolium, and Panax pseudoginseng, which contain bioactive components (BCs) such as ginsenosides and polysaccharides. Recently, growing evidence has revealed the pharmacological effects of Panax species and their BCs on allergic airway diseases (AADs), including allergic asthma (AA) and allergic rhinitis (AR). AADs are characterized by damaged epithelium, sustained acquired immune responses with enforced Th2 responses, allergen-specific IgE production, and enhanced production of histamine and leukotrienes by activated mast cells and basophils. In this review, we summarize how Panax species and their BCs modulate acquired immune responses involving interactions between dendritic cells and T cells, reduce the pro-inflammatory responses of epithelial cells, and reduce allergenic responses from basophils and mast cells in vitro. In addition, we highlight the current understanding of the alleviative effects of Panax species and their BCs against AA and AR in vivo. Moreover, we discuss the unmet needs of research and considerations for the treatment of patients to provide basic scientific knowledge for the treatment of AADs using Panax species and their BCs.
Collapse
Affiliation(s)
- Dahee Shim
- Industry-Academic Cooperation Foundation, Hallym University, Chuncheon, Republic of Korea
| | - Yeeun Bak
- Department of Biomedical Science, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Han-Gyu Choi
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seunghyun Lee
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Chul Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Yang C, Qu L, Wang R, Wang F, Yang Z, Xiao F. Multi-layered effects of Panax notoginseng on immune system. Pharmacol Res 2024; 204:107203. [PMID: 38719196 DOI: 10.1016/j.phrs.2024.107203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Recent research has demonstrated the immunomodulatory potential of Panax notoginseng in the treatment of chronic inflammatory diseases and cerebral hemorrhage, suggesting its significance in clinical practice. Nevertheless, the complex immune activity of various components has hindered a comprehensive understanding of the immune-regulating properties of Panax notoginseng, impeding its broader utilization. This review evaluates the effect of Panax notoginseng to various types of white blood cells, elucidates the underlying mechanisms, and compares the immunomodulatory effects of different Panax notoginseng active fractions, aiming to provide the theory basis for future immunomodulatory investigation.
Collapse
Affiliation(s)
- Chunhao Yang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China
| | - Liping Qu
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China; Innovation Materials Research and Development Center, Botanee Research Institute, Shanghai Jiyan Biomedical Development Co., Ltd., Shanghai 201702, China
| | - Rui Wang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China
| | - Feifei Wang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China; Innovation Materials Research and Development Center, Botanee Research Institute, Shanghai Jiyan Biomedical Development Co., Ltd., Shanghai 201702, China
| | - Zhaoxiang Yang
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China
| | - Fengkun Xiao
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd., Kunming 650106, China; Yunnan Botanee Bio-Technology Group Co., Ltd., Kunming 650106, China.
| |
Collapse
|
4
|
Yuan L, Sun C. The protective effects of Arctiin in asthma by attenuating airway inflammation and inhibiting p38/NF-κB signaling. Aging (Albany NY) 2024; 16:5038-5049. [PMID: 38546350 PMCID: PMC11006498 DOI: 10.18632/aging.205584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/27/2023] [Indexed: 04/06/2024]
Abstract
Asthma is a common chronic inflammatory disease of the airways, which affects millions of people worldwide. Arctiin, a bioactive molecule derived from the traditional Chinese Burdock, has not been previously reported for its effects on asthma in infants. In this study, an asthma model was established in mice by stimulation with ovalbumin (OVA). Bronchoalveolar lavage (BALF) was collected from OVA-challenged mice and the cells were counted. Lung tissue was harvested for hematoxylin-eosin (HE) staining and measurement of Wet/Dry weight ratios. The expressions of proteins were detected using enzyme-linked immunosorbent assay (ELISA) and Western blots. The superoxide dismutase (SOD) activity in lung tissue was measured using a commercial kit. We found that Arctiin had beneficial effects on asthma treatment. Firstly, it attenuated OVA-challenged lung pathological alterations. Secondly, it ameliorated pro-inflammatory response by reducing the number of inflammatory cells and mitigating the imbalance of Th1/Th2 factors in the bronchoalveolar lavage (BALF) of OVA-challenged mice. Importantly, Arctiin ameliorated OVA-induced lung tissue impairment and improved lung function. Additionally, we observed that oxidative stress (OS) in the pulmonary tissue of OVA-challenged mice was ameliorated by Arctiin. Mechanistically, Arctiin prevented OVA-induced activation of p38 and nuclear factor-κB (NF-κB). Based on these findings, we conclude that Arctiin might serve as a promising agent for the treatment of asthma.
Collapse
Affiliation(s)
- Lang Yuan
- Department of Respiratory Medicine, Children's Hospital of Shanghai, Shanghai Jiaotong University, Shanghai 200062, China
| | - Chao Sun
- Department of Respiratory Medicine, Children's Hospital of Shanghai, Shanghai Jiaotong University, Shanghai 200062, China
| |
Collapse
|
5
|
Park JM, Park JW, Lee J, Kim SH, Seo DY, Ahn KS, Han SB, Lee JW. Aromadendrin inhibits PMA-induced cytokine formation/NF-κB activation in A549 cells and ovalbumin-induced bronchial inflammation in mice. Heliyon 2023; 9:e22932. [PMID: 38125474 PMCID: PMC10730751 DOI: 10.1016/j.heliyon.2023.e22932] [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: 07/24/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Hyperproduction of immune cell-derived inflammatory molecules and recruitment of immune cells promote the development of allergic asthma (AA). Aromadendrin (ARO) has various biological properties including anti-inflammatory effects. In this study, we evaluated the ameliorative effects of ARO on the development of AA in vitro and in vivo. Phorbol 12-myristate 13-acetate (PMA, 100 nM) was used to induce inflammation in A549 airway epithelial cells. The cohesion of A549 and eosinophil EOL-1 cells was studied. Ovalbumin (30 or 60 μg)/Alum (3 mg) mixture was adapted for AA induction in mice. ARO (5 or 10 mg/kg, p. o.) was administered to mice to investigate its ameliorative effect on AA development. Enzyme-linked immunosorbent assay, western blotting, and hematoxylin and eosin/periodic acid Schiff staining were performed to study the ameliorative effect of ARO on bronchial inflammation. In PMA-stimulated A549 cells, the upregulation of cytokines (interleukin [IL]-1β/IL-6/tumor necrosis factor alpha [TNF-α]/monocyte chemoattractant protein [MCP]-1]) and nuclear factor kappa B (NF-κB) activation was effectively reduced by ARO pretreatment. ARO suppressed the adhesion of A549 cells and eosinophils. In ovalbumin-induced AA mice, the levels of cells, such as eosinophils, Th2 cytokines, MCP-1 in bronchoalveolar lavage fluid, IgE in serum, and inducible nitric oxide synthase/cyclooxygenase-2 expression in the lung tissue were upregulated, which were all suppressed by ARO. In addition, the increase in cell inflow and mucus formation in the lungs of AA mice was reversed by ARO as per histological analysis. ARO also modulated NF-κB activation in the lungs of AA mice. Overall, the anti-inflammatory properties of ARO in vitro/in vivo studies of AA were notable. Thus, ARO has a modulatory effect on bronchial inflammation and may be a potential adjuvant for AA treatment.
Collapse
Affiliation(s)
- Jin-Mi Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Ji-Won Park
- Practical Research Division, Honam National Institute of Biological Resources (HNIBR), 99, Gohadoan-gil, Mokpo-si, Jeollanam-do, 58762, Republic of Korea
| | - Juhyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Seung-Ho Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Da-Yun Seo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| |
Collapse
|
6
|
Liu P, Zhang Z, Cai Y, Yang Y, Yuan J, Chen Q. Inhibition of the pyroptosis-associated inflammasome pathway: The important potential mechanism of ginsenosides in ameliorating diabetes and its complications. Eur J Med Chem 2023; 253:115336. [PMID: 37031528 DOI: 10.1016/j.ejmech.2023.115336] [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: 01/30/2023] [Revised: 04/01/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023]
Abstract
Diabetes mellitus (DM) and its complications have become an important global public health issue, affecting human health and negatively impacting life and lifespan. Pyroptosis is a recently discovered form of pro-inflammatory programmed cell death (PCD). To date, pyroptosis-associated inflammasome pathways have been identified primarily in the canonical and non-canonical inflammasome pathway, apoptotic caspase-mediated pathway, granzyme-mediated pathway, and streptococcal pyrogenic exotoxin B (SpeB)-mediated pathway. The activation of diabetes-mediated pyroptosis-associated factors play an important role in the pathophysiology of DM and its complications. Studies have shown that ginsenosides exert significant protective effects on DM and its complications. Through inhibiting the activation of pyroptosis-associated inflammasome pathways, and then the DM and its complications are improved. This review summarizes the subtypes of ginsenosides and their chemical characteristics, pharmacokinetics and side effects, the main pyroptosis-associated inflammasome pathways that have been discovered to date, and the potential mechanism of different subtypes of ginsenosides in the treatment of DM and its complications (such as diabetic cardiomyopathy, diabetic nephropathy, diabetic liver injury, diabetic retinopathy, and diabetic ischemic stroke) via anti-pyroptosis-associated inflammasome pathways. These findings may provide ideas for further research to explore ginsenoside mechanism in improving DM and its complications. However, many pyroptosis-associated inflammasome pathways and targets involved in the occurrence and development of DM and its complications are still unknown. In the future, further studies using in vitro cell models, in vivo animal models, and human disease models can be used to further elucidate the mechanism of ginsenosides in the treatment of DM and its complications.
Collapse
Affiliation(s)
- Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, PR China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, Sichuan Province, PR China; Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan Province, PR China
| | - Yichen Cai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, PR China
| | - Yunjiao Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, PR China
| | - Jun Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, PR China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, PR China.
| |
Collapse
|
7
|
Jin Y, Tangchang W, Kwon OS, Lee JY, Heo KS, Son HY. Ginsenoside Rh1 ameliorates the asthma and allergic inflammation via inhibiting Akt, MAPK, and NF-κB signaling pathways in vitro and in vivo. Life Sci 2023; 321:121607. [PMID: 36958436 DOI: 10.1016/j.lfs.2023.121607] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
AIMS Overproduction of pro-inflammatory cytokines and its-mediated immune cell infiltration play a crucial role in asthma progression. In this study, we investigated the role of ginsenoside Rh1 (Rh1) in ovalbumin (OVA)/lipopolysaccharide (LPS)-induced allergic asthma both in vitro and in vivo. MATERIALS AND MAIN METHODS The phorbol ester (PMA) and LPS were used to induce inflammation in lung airway cells and macrophage activation, respectively. Western blotting, quantitative reverse transcription-PCR, and immunofluorescence (IF) assays were performed to elucidate the underlying molecular mechanisms. To evaluating the effects of Rh1 in vivo, OVA and LPS were used to establish allergic asthma models. KEY FINDINGS Rh1 significantly suppressed PMA-induced lung inflammation and macrophage activation by suppressing pro-inflammatory cytokines (TNF-α, IL-1β, MCP-1), ICMA-1, and matrix metallopeptidase 9 (MMP9) in A549 cells. Rh1 abolished the PMA-induced inflammation by suppressing MAPK, Akt, and NF-κB p65. Pretreatment with Rh1 blocked PMA-mediated translocation of NF-κB, a key marker of pro-inflammatory cytokine release, into the nucleus. Similar to PMA-induced lung inflammation, Rh1 suppressed LPS-induced macrophage activation by suppressing NF-κB p65 activation and inducible nitric oxide synthase protein and mRNA expression. Consistent with in vitro data, LPS injection enhanced the number of immune cells induced by OVA in bronchoalveolar lavage fluid, whereas 20 mg/kg Rh1 significantly decreased OVA/LPS-mediated immune cell induction. In addition, Rh1 inhibited eosinophil, macrophage, and neutrophil maturation through by IL-4 and OVA-specific IgE production. SIGNIFICANCE Rh1 protects against OVA/LPS-induced allergic asthma by suppressing immune cell infiltration by blocking the activation of MAPK, Akt, and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yujin Jin
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, South Korea
| | - Warisraporn Tangchang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, South Korea
| | - Oh Seong Kwon
- College of Pharmacy, Chung-Ang University, Seoul 06974, South Korea
| | - Ji-Yun Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, South Korea.
| | - Kyung-Sun Heo
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, South Korea.
| | - Hwa-Young Son
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, South Korea.
| |
Collapse
|
8
|
Liu J, Yang N, Yi X, Wang G, Wang C, Lin H, Sun L, Wang F, Zhu D. Integration of transcriptomics and metabolomics to reveal the effect of ginsenoside Rg3 on allergic rhinitis in mice. Food Funct 2023; 14:2416-2431. [PMID: 36786409 DOI: 10.1039/d2fo03885d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Increasing studies have demonstrated that ginsenoside Rg3 (Rg3) plays an important role in the prevention and treatment of various diseases, including allergic lower airway inflammation such as asthma. To investigate the role of Rg3 in allergic upper airway disease, the effect and therapeutic mechanism of Rg3 in allergic rhinitis (AR) were studied. Ovalbumin-induced AR model mice were intragastrically administered with Rg3. Nasal symptoms, levels of IgE, IL-4, IL-5, IL-13, SOD and MDA in serum, and histopathological analysis of nasal mucosa were used to evaluate the effect of Rg3 on ameliorating AR in mice. Moreover, nasal mucosa samples from the normal control group, AR model group and high dosage of Rg3 were collected to perform omics analysis. The differentially expressed genes and significantly changed metabolites were screened based on transcriptomics and metabolomics analyses, respectively. Integrative analysis was further performed to confirm the hub genes, metabolites and pathways. After Rg3 intervention, the nasal symptoms and inflammatory infiltration were effectively improved, the levels of IgE, IL-4, IL-5, IL-13 and MDA were significantly reduced, and the level of SOD was obviously increased. The results of the qRT-PCR assay complemented the transcriptomic findings. Integrated analysis showed that Rg3 played an anti-AR role mainly by regulating the interaction network, which was constructed by 12 genes, 8 metabolites and 4 pathways. Our findings suggested that Rg3 had a therapeutic effect on ovalbumin-induced AR in mice by inhibiting inflammation development and reducing oxidative stress. The present study could provide a potential natural agent for the treatment of AR.
Collapse
Affiliation(s)
- Jianming Liu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun 130021, China.
| | - Na Yang
- Clinical Pharmacy Department, First Hospital of Jilin University, Changchun 130021, China
| | - Xingcheng Yi
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun 130061, China
| | - Guoqiang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hongqiang Lin
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Liwei Sun
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun 130021, China.
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Dongdong Zhu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun 130021, China. .,Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases, Changchun 130021, China
| |
Collapse
|
9
|
DEC-205 receptor targeted poly(lactic-co-glycolic acid) nanoparticles containing Eucommia ulmoides polysaccharide enhances the immune response of foot-and-mouth disease vaccine in mice. Int J Biol Macromol 2023; 227:576-589. [PMID: 36549613 DOI: 10.1016/j.ijbiomac.2022.12.158] [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: 06/08/2022] [Revised: 11/24/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Nanoparticles targeting the DEC-205 receptor were found to induce antigen-specific protective immune response. When the delivery system carries both antigens and immunomodulators, it can maximize the expected therapeutic effect of the drug and induce effective humoral and cellular immune responses to antigens.In this study, we encapsulated the Eucommia ulmoides Oliv. polysaccharides (EUPS) into PLGA nanoparticles (NPs) and conjugated it with anti-CD205 monoclonal Ab (MAb) to produce a DEC-205 receptor targeted PLGA nanoparticles (anti-DEC-205-EUPS-PLGA NPs). The physicochemical characteristics and adjuvant activity of the above NPs were evaluated in vitro and in vivo. In the in vitro setting, 200 μg·mL-1 anti-DEC-205-EUPS-PLGA could improve the proliferation of DCs and promote their antigen up-take activity. In the in vivo setting, anti-DEC-205-EUPS-PLGA NPs remarkably controlled the release of drug and antigen to induce sustained immune responses and up-regulated the levels of FMDV-specific IgG antibodies, promoted the cytotoxic activity of CTLs and NK cells, and improved the proliferation of splenocytes. Moreover, the anti-DEC-205-EUPS-PLGA NPs facilitated the maturation of DCs. The above data indicated that anti-DEC-205-EUPS-PLGA NPs employed as an targeted adjuvant induced the humoral and cellular immune activity by promoting the maturation of DCs. These findings may provide a new insight onto the development of vaccine adjuvants.
Collapse
|
10
|
Recent advances in ginsenosides against respiratory diseases: Therapeutic targets and potential mechanisms. Biomed Pharmacother 2023; 158:114096. [PMID: 36502752 DOI: 10.1016/j.biopha.2022.114096] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Respiratory diseases mainly include asthma, influenza, pneumonia, chronic obstructive pulmonary disease, pulmonary hypertension, lung fibrosis, and lung cancer. Given their high prevalence and poor prognosis, the prevention and treatment of respiratory diseases are increasingly essential. In particular, the development for the novel strategies of drug treatment has been a hot topic in the research field. Ginsenosides are the major component of Panax ginseng C. A. Meyer (ginseng), a food homology and well-known medicinal herb. In this review, we summarize the current therapeutic effects and molecular mechanisms of ginsenosides in respiratory diseases. METHODS The reviewed studies were retrieved via a thorough analysis of numerous articles using electronic search tools including Sci-Finder, ScienceDirect, PubMed, and Web of Science. The following keywords were used for the online search: ginsenosides, asthma, influenza, pneumonia, chronic obstructive pulmonary disease (COPD), pulmonary hypertension (PH), lung fibrosis, lung cancer, and clinical trials. We summarized the findings and the conclusions from 176 manuscripts on ginsenosides, including research articles and reviews. RESULTS Ginsenosides Rb1, Rg1, Rg3, Rh2, and CK, which are the most commonly reported ginsenosides for treating of respiratory diseases, and other ginsenosides such as Rh1, Rk1, Rg5, Rd and Re, all primarily reduce pneumonia, fibrosis, and inhibit tumor progression by targeting NF-κB, TGF-β/Smad, PI3K/AKT/mTOR, and JNK pathways, thereby ameliorating respiratory diseases. CONCLUSION This review provides novel ideas and important aspects for the future research of ginsenosides for treating respiratory diseases.
Collapse
|
11
|
Zhou X, Zhang K, Liu L, Zhao Q, Huang M, Shao R, Wang Y, Qu B, Wang Y. Anti-fatigue effect from Ginseng Radix et Rhizoma: a suggestive and promising treatment for long COVID. ACUPUNCTURE AND HERBAL MEDICINE 2022; 2:69-77. [PMID: 37808250 PMCID: PMC9407182 DOI: 10.1097/hm9.0000000000000033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/05/2022] [Indexed: 11/26/2022]
Abstract
Two years after the coronavirus disease 2019 (COVID-19) outbreak, an increasing number of patients continue to suffer from long COVID (LC), persistent symptoms, and/or delayed or long-term complications beyond the initial 4 weeks from the onset of symptoms. Constant fatigue is one of the most common LC symptoms, leading to severely reduced quality of life among patients. Ginseng Radix et Rhizoma-known as the King of Herbs in traditional Chinese medicine-has shown clinical anti-fatigue effects. In this review, we summarize the underlying anti-fatigue mechanisms of Ginseng Radix et Rhizoma extracts and their bioactive compounds, with a special focus on anti-viral, immune remodeling, endocrine system regulation, and metabolism, suggesting that Ginseng Radix et Rhizoma is a potentially promising treatment for LC, especially in regard to targeting fatigue.
Collapse
Affiliation(s)
- Xiangda Zhou
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Keying Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lanbo Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qianru Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ming Huang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Shao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanyan Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Qu
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Yu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
12
|
PM2.5 Exposure and Asthma Development: The Key Role of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3618806. [PMID: 35419163 PMCID: PMC9001082 DOI: 10.1155/2022/3618806] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/24/2022] [Indexed: 12/21/2022]
Abstract
Oxidative stress is defined as the imbalance between reactive oxygen species (ROS) production and the endogenous antioxidant defense system, leading to cellular damage. Asthma is a common chronic inflammatory airway disease. The presence of asthma tends to increase the production of reactive oxygen species (ROS), and the antioxidant system in the lungs is insufficient to mitigate it. Therefore, asthma can lead to an exacerbation of airway hyperresponsiveness and airway inflammation. PM2.5 exposure increases ROS levels. Meanwhile, the accumulation of ROS will further enhance the oxidative stress response, resulting in DNA, protein, lipid, and other cellular and molecular damage, leading to respiratory diseases. An in-depth study on the relationship between oxidative stress and PM2.5-related asthma is helpful to understand the pathogenesis and progression of the disease and provides a new direction for the treatment of the disease. This paper reviews the research progress of oxidative stress in PM2.5-induced asthma as well as highlights the therapeutic potentials of antioxidant approaches in treatment of asthma.
Collapse
|
13
|
Choi JH, Lee YH, Kwon TW, Ko SG, Nah SY, Cho IH. Can Panax ginseng help control cytokine storm in COVID-19? J Ginseng Res 2022; 46:337-347. [PMID: 35233163 PMCID: PMC8876050 DOI: 10.1016/j.jgr.2022.02.006] [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: 02/03/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/05/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is currently a pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 are directly associated with hyper-activation of innate immune response that excessively produce pro-inflammatory cytokines and induce cytokine storm, leading to multi-organ-failure and significant morbidity/mortality. Currently, several antiviral drugs such as Paxlovid (nirmatrelvir and ritonavir) and molnupiravir are authorized to treat mild to moderate COVID-19, however, there are still no drugs that can specifically fight against challenges of SARS-CoV-2 variants. Panax ginseng, a medicinal plant widely used for treating various conditions, might be appropriate for this need due to its anti-inflammatory/cytokine/viral activities, fewer side effects, and cost efficiency. To review Panax ginseng and its pharmacologically active-ingredients as potential phytopharmaceuticals for treating cytokine storm of COVID-19, articles that reporting its positive effects on the cytokine production were searched from academic databases. Experimental/clinical evidences for the effectiveness of Panax ginseng and its active-ingredients in preventing or mitigating cytokine storm, especially for the cascade of cytokine storm, suggest that they might be beneficial as an adjunct treatment for cytokine storm of COVID-19. This review may provide a new approach to discover specific medications using Panax ginseng to control cytokine storm of COVID-19.
Collapse
|