1
|
Sun Y, Wang Y, Yang Z, Han X, Zhang Y, Chen L, Huo J, Wu R, Wang W, Wang N. Neutral Polysaccharide from Platycodonis Radix-Ameliorated PM 2.5-Induced Lung Injury by Inhibiting the TLR4/NF-κB p65 Pathway and Regulating the Lung and Gut Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27923-27938. [PMID: 39626068 DOI: 10.1021/acs.jafc.4c07319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Platycodonis radix (PR) has been reported to play a protective role in lung injury. However, much less is known about the protective effect and mechanism of its main component PR polysaccharides (PRPs) in particulate matter (PM2.5)-induced lung injury. Here, a neutral polysaccharide (MW: 244.56 kDa) was isolated from PR, mainly composed of Rha, Ara, Gal, Glc, Xyl, and Man. PRPs significantly improved PM2.5-induced pulmonary edema, oxidative damage, and cell apoptosis and downregulated inflammatory factor levels in bronchoalveolar lavage fluid. Mechanistically, PRPs reduced intestinal mucosal barrier damage, thereby lowering serum lipopolysaccharide levels and inhibiting the overactivation of the TLR4/NF-κB signaling pathway in the lung tissue. Notably, PRPs could optimize the composition of pulmonary and intestinal microbiota. Oral administration of PRPs resulted in enrichment of short-chain fatty acid (SCFA)-producing bacteria, thereby upregulating the levels of acetate, butyrate, and isovalerate. Taken together, PRPs have great potential in preventing and repairing the lung injury caused by PM2.5.
Collapse
Affiliation(s)
- Yang Sun
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yanchun Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zaiming Yang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xianlei Han
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yue Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Liyan Chen
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, China
| | - Jinhai Huo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Weiming Wang
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
2
|
Ren Y, Sun Y, Liao YY, Wang S, Liu Q, Duan CY, Sun L, Li XY, Yuan JL. Mechanisms of action and applications of Polygonatum sibiricum polysaccharide at the intestinal mucosa barrier: a review. Front Pharmacol 2024; 15:1421607. [PMID: 39224782 PMCID: PMC11366640 DOI: 10.3389/fphar.2024.1421607] [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: 04/22/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
As a medicinal and edible homologous Chinese herb, Polygonatum sibiricum has been used as a primary ingredient in various functional and medicinal products. Damage to the intestinal mucosal barrier can lead to or worsen conditions such as type 2 diabetes and Alzheimer's disease. Traditional Chinese medicine and its bioactive components can help prevent and manage these conditions by restoring the integrity of the intestinal mucosal barrier. This review delves into the mode of action of P. sibiricum polysaccharide in disease prevention and management through the restoration of the intestinal barrier. Polysaccharide from P. sibiricum effectively treats conditions by repairing the intestinal mucosal barrier, offering insights for treating complex diseases and supporting the application of P. sibiricum in clinical settings.
Collapse
Affiliation(s)
- Yu Ren
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Sun
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yu-Ying Liao
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Si Wang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qian Liu
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Chun-Yan Duan
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Lan Sun
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Xiao-Ya Li
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jia-Li Yuan
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Diseasein Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
3
|
Park JY, Kim MJ, Choi YA, Kim YY, Lee S, Chung JM, Kim SY, Jeong GS, Kim SH. Anti-Inflammatory Effects of Clematis terniflora Leaf on Lipopolysaccharide-Induced Acute Lung Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2024; 2024:6653893. [PMID: 38230250 PMCID: PMC10791263 DOI: 10.1155/2024/6653893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/03/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024]
Abstract
For centuries, natural products are regarded as vital medicines for human survival. Clematis terniflora var. mandshurica (Rupr.) Ohwi is an ingredient of the herbal medicine, Wei Ling Xian, which has been used in Chinese medicine to alleviate pain, fever, and inflammation. In particular, C. terniflora leaves have been used to cure various inflammatory diseases, including tonsillitis, cholelithiasis, and conjunctivitis. Based on these properties, this study aimed to scientifically investigate the anti-inflammatory effect of an ethanol extract of leaves of C. terniflora (EELCT) using activated macrophages that play central roles in inflammatory response. In this study, EELCT inhibited the essential inflammatory mediators, such as nitric oxide, cyclooxygenase-2, tumor necrosis factor-α, interleukin- (IL-) 6, IL-1β, and inducible nitric oxide synthase, by suppressing the nuclear factor-κB and mitogen-activated protein kinase activation in macrophages. Acute lung injury (ALI) is a fatal respiratory disease accompanied by serious inflammation. With high mortality rate, the disease has no effective treatments. Therefore, new therapeutic agents must be developed for ALI. We expected that EELCT can be a promising therapeutic agent for ALI by reducing inflammatory responses and evaluated its action in a lipopolysaccharide- (LPS-) induced ALI model. EELCT alleviated histological changes, immune cell infiltration, inflammatory mediator production, and protein-rich pulmonary edema during ALI. Collectively, our results may explain the traditional usage of C. terniflora in inflammatory diseases and suggest the promising potential of EELCT as therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Ji-Yeong Park
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Min-Jong Kim
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Young-Ae Choi
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Yeon-Yong Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Soyoung Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Jae-Min Chung
- Department of Gardens and Education, Korea National Arboretum, Pocheon 11186, Republic of Korea
| | - Sang-Yong Kim
- DMZ Botanic Garden, Korea National Arboretum, Yanggu 24564, Republic of Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sang-Hyun Kim
- Cell and Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
4
|
Hua F, Cui E, Lv L, Wang B, Li L, Lu H, Chen N, Chen W. Fecal microbiota transplantation from HUC-MSC-treated mice alleviates acute lung injury in mice through anti-inflammation and gut microbiota modulation. Front Microbiol 2023; 14:1243102. [PMID: 37840733 PMCID: PMC10569429 DOI: 10.3389/fmicb.2023.1243102] [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: 06/20/2023] [Accepted: 08/29/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Acute lung injury (ALI) is a severe respiratory tract disorder facilitated by dysregulated inflammation, oxidative stress and intestinal ecosystem. Fecal microbiota transplantation (FMT) is a rapid method for gut microbiota (GM) reconstruction. Furthermore, our previous studies have confirmed that human umbilical cord mesenchymal stromal cells (HUC-MSCs) can alleviate ALI by improving GM composition. Therefore, we aimed to explore the efficacy and mechanism of FMT from HUC-MSCs-treated mice on ALI. Methods In brief, fresh feces from HUC-MSCs-treated mice were collected for FMT, and the mice were randomly assigned into NC, FMT, LPS, ABX-LPS, and ABX-LPS-FMT groups (n = 12/group). Subsequently, the mice were administrated with antibiotic mixtures to deplete GM, and given lipopolysaccharide and FMT to induce ALI and rebuild GM. Next, the therapeutic effect was evaluated by bronchoalveolar lavage fluid (BALF) and histopathology. Immune cells in peripheral blood and apoptosis in lung tissues were measured. Furthermore, oxidative stress- and inflammation-related parameter levels were tested in BALF, serum, lung and ileal tissues. The expressions of apoptosis-associated, TLR4/NF-κB pathway-associated, Nrf2/HO-1 pathway related and tightly linked proteins in the lung and ileal tissues were assessed. Moreover, 16S rRNA was conducted to assess GM composition and distribution. Results Our results revealed that FMT obviously improved the pathological damage of lung and ileum, recovered the immune system of peripheral blood, decreased the cell apoptosis of lung, and inhibited inflammation and oxidative stress in BALF, serum, lung and ileum tissues. Moreover, FMT also elevated ZO-1, claudin-1, and occludin protein expressions, activating the Nrf2/HO-1 pathway but hindering the TLR4/NF-κB pathway. Of note, the relative abundances of Bacteroides, Christensenella, Coprococcus, and Roseburia were decreased, while the relative abundances of Xenorhabdus, Sutterella, and Acinetobacter were increased in the ABX-LPS-FMT group. Conclusion FMT from HUC-MSCs-treated mice may alleviate ALI by inhibiting inflammation and reconstructing GM, additionally, we also found that the TLR4/NF-κB and Nrf2/HO-1 pathways may involve in the improvement of FMT on ALI, which offers novel insights for the functions and mechanisms of FMT from HUC-MSCs-treated mice on ALI.
Collapse
Affiliation(s)
- Feng Hua
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Enhai Cui
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Lu Lv
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Bin Wang
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Liqin Li
- Traditional Chinese Medicine Key Laboratory Cultivation Base of Zhejiang Province for the Development and Clinical Transformation of Immunomodulatory Drugs, Huzhou, China
| | - Huadong Lu
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Na Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Wenyan Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| |
Collapse
|
5
|
Hou JY, Wu JR, Chen YB, Xu D, Liu S, Shang DD, Fan GW, Cui YL. Systematic identification of the interventional mechanism of Qingfei Xiaoyan Wan (QFXYW) in treatment of the cytokine storm in acute lung injury using transcriptomics-based system pharmacological analyses. PHARMACEUTICAL BIOLOGY 2022; 60:743-754. [PMID: 35357989 PMCID: PMC8979529 DOI: 10.1080/13880209.2022.2055090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
CONTEXT Acute lung injury (ALI) is a complex, severe inflammation disease with high mortality, and there is no specific and effective treatment for ALI. Qingfei Xiaoyan Wan (QFXYW) has been widely used to treat lung-related diseases for centuries. OBJECTIVE This study evaluates the potential effects and elucidates the therapeutic mechanism of QFXYW against LPS induced ALI in mice. MATERIALS AND METHODS BALB/c Mice in each group were first orally administered medicines (0.9% saline solution for the control group, 0.5 mg/kg Dexamethasone, or 1.3, 2.6, 5.2 g/kg QFXYW), after 4 h, the groups were injected LPS (1.0 mg/kg) to induce ALI, then the same medicines were administered repeatedly. The transcriptomics-based system pharmacological analyses were applied to screen the hub genes, RT-PCR, ELISA, and protein array assay was applied to verify the predicted hub genes and key pathways. RESULTS QFXYW significantly decreased the number of leukocytes from (6.34 ± 0.51) × 105/mL to (4.01 ± 0.11) × 105/mL, accompanied by the neutrophil from (1.41 ± 0.19) × 105/mL to (0.77 ± 0.10) × 105/mL in bronchoalveolar lavage fluid (BALF). Based on Degree of node connection (Degree) and BottleNeck (BN), important parameters of network topology, the protein-protein interaction (PPI) network screened hub genes, including IL-6, TNF-α, CCL2, TLR2, CXCL1, and MMP-9. The results of RT-PCR, ELISA, and protein chip assay revealed that QFXYW could effectively inhibit ALI via multiple key targets and the cytokine-cytokine signalling pathway. CONCLUSIONS This study showed that QFXYW decreased the number of leukocytes and neutrophils by attenuating inflammatory response, which provides an important basis for the use of QFXYW in the treatment of ALI.
Collapse
Affiliation(s)
- Jing-Yi Hou
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Jia-Rong Wu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Yi-Bing Chen
- Tianjin Key Laboratory of Transformation of Traditional Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dong Xu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Shu Liu
- Tianjin Zhongxin Pharmaceutical Group Corporation Limited Darentang Pharmaceutical Factory, Tianjin, China
| | - Dan-dan Shang
- Tianjin Zhongxin Pharmaceutical Group Corporation Limited Darentang Pharmaceutical Factory, Tianjin, China
| | - Guan-Wei Fan
- Tianjin Key Laboratory of Transformation of Traditional Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Guan-Wei Fan Tianjin Key Laboratory of Transformation of Traditional Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- CONTACT Yuan-Lu Cui State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| |
Collapse
|
6
|
Liu Z, Yang Y. Ginkgolide A Participates in LPS-Induced PMVEC Injury by Regulating miR-224 and Inhibiting p21 in a Targeted Manner. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6384334. [PMID: 36134118 PMCID: PMC9482518 DOI: 10.1155/2022/6384334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022]
Abstract
Most studies have focused on the protective effects of ginkgolide A against ischemia/reperfusion-induced cardiomyopathy and injury of the brain, liver, and other organs, but there are few reports about the protection of lung tissues. This study was designed to clarify the protection of ginkgolide A against lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cell (PMVEC) injury. PMVECs were extracted and fell into control, LPS, and ginkgolide A groups. Next, we delved into the growth activity and apoptosis rate of cells via the CCK-8 assay and Hoechst staining, independently. Beyond that, western blotting (WB) was implemented for measurement of the expressions of cyclin D1, cyclin-dependent kinase 4 (CDK4), and CDK inhibitor (p21) that pertained to the cell cycle. The target sites of ginkgolide A were confirmed by miRNA array and real-time quantitative PCR. The relationship between miR-224 and p21 was analyzed using dual-luciferase reporter gene assay. Compared with the control group, the LPS group and ginkgolide A group had significantly decreased cell growth activity and relative expressions of cyclin D1 and CDK4 and elevated apoptosis rate and p21 expression. Pronounced elevations were observable in the cell growth activity and expressions of cyclin D1, CDK4, and p21, while the ginkgolide A group presented with a reduced apoptosis rate in comparison with the LPS group (P < 0.05). MiR-224 was the target of ginkgolide A, which had targeted regulatory effects on p21. Ginkgolide A can modulate miR-224 expression and regulate p21 expression in a targeted manner to enhance the resistance of PMVECs to LPS-induced cell apoptosis.
Collapse
Affiliation(s)
- Zhonglin Liu
- Department of Traditional Chinese Medicine, Affiliated Nanhua Hospital University of South China, Hengyang 421000, Hunan, China
| | - Yan Yang
- Department of Pain Medicine, Affiliated Nanhua Hospital University of South China, Hengyang 421000, Hunan, China
| |
Collapse
|
7
|
Polygonatum Polysaccharide Regulates Macrophage Polarization and Improves LPS-Induced Acute Lung Injury through TLR4-MAPK/NF-κB Pathway. Can Respir J 2022; 2022:2686992. [PMID: 35874106 PMCID: PMC9303503 DOI: 10.1155/2022/2686992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 12/02/2022] Open
Abstract
Objective To investigate the effects of polygonatum sibiricum polysaccharides (PSPs) on the polarization of macrophages to M1 and M2 phenotypes and their potential mechanism. Methods PSPs samples were prepared through water extraction and alcohol precipitation assay. The properties of PSPs were identified and analyzed by high-performance liquid chromatography, FT-IR, and NMR assay. Then, the effects of PSPs on mouse macrophage RAW264.7 viability were measured by CCK-8 assay. The cells were randomly divided into the control group, PSPs group, LPS group, and LPS + PSPs group. M1 phenotype polarization of RAW264.7 cells was induced by LPS treatment. The effects of various treatments on expression of M2 phenotype CD206, activation of TLR4-MAPK/NF-κB signal pathway, and translocation of NF-κB into the nucleus were determined by ELISA, western blot, and immunofluorescence assay, respectively. TLR4 inhibitor, TAK-242, and MAPK inhibitor, BIRB 796, were used to verify the effects of PSPs on the TLR4-MAPK/NF-κB pathway. The mice model of acute lung injury (ALI) was established and randomly divided into control group, PSPs group, LPS group, and LPS + PSPs group. Bronchoalveolar lavage fluid (BALF) and lung tissue were collected to measure protein, inflammatory cells, neutrophil and macrophage cells number, and the levels of IL-6 and TNF-α in BALF. Flow cytometry and western blot assay measured the phenotypic changes of macrophages and the activation of the TLR4-MAPK/NF-κB signaling pathway. Results The concentrations of PSPs lower than 100 μg/mL showed no toxicity to RAW264.7 cells. PSPs treatment could significantly reverse the reduction of CD206 protein expression (P < 0.05) and the increase of the expression of inflammatory factor TNF-α, IL-1β, and IL-6 (all P < 0.05), TLR4-MAPK/NF-κB signaling pathway activation (all P < 0.05), and NF-κB translocation into the nucleus induced by LPS. The effect of inhibitors TAK-242 and BIRB 796 was consistent with that of PSPs. In the mice model of ALI, PSPs treatment could reduce the total protein levels of BALF and the number of inflammatory cells level, reverse the number changes of neutrophils and macrophages, and downregulate the proinflammatory factors IL-6 and TNF-α caused by LPS (all P < 0.05). In addition, PSPs treatment could also significantly reverse the increase in the number of iNOS expressing macrophages in alveolar lavage fluid induced by LPS (P < 0.05). In contrast, CD206-expressed cells decreased (P < 0.05). PSPs could also reverse LPS-induced TLR4-MAPK/NF-κB signal pathway protein activation (all P < 0.05). Conclusion PSPs could suppress TLR4-MAPK/NF-κB activation induced by LPS, inhibit M1 phenotypic polarization of macrophages, and promote M2 phenotypic polarization, thus playing an anti-inflammatory role.
Collapse
|
8
|
PTX3 Protects Intestinal Mucosal Barrier Damage in Sepsis Through Toll-Like Receptor Signaling Pathway. Inflammation 2022; 45:2339-2351. [PMID: 35687213 DOI: 10.1007/s10753-022-01696-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/18/2022] [Accepted: 05/31/2022] [Indexed: 01/16/2023]
Abstract
This study aims to confirm the protective effect of Pentraxin 3 (PTX3) on intestinal mucosal barrier damage in sepsis in animal and cell models and explore its mechanism. Analysis of the GSE147775 gene set revealed that the level of PTX3 was upregulated in the lipopolysaccharide (LPS)-induced rat sepsis model. The mice sepsis model was established by cecal ligation perforation (CLP), and the cell inflammation model was induced by LPS. Cell apoptosis and the expression of apoptosis-related protein were detected by flow cytometry and Western blotting. The PTX3 level was significantly upregulated in the mice sepsis model. Intestinal mucosal barrier damage was aggravated and inflammatory factor expression was upregulated after PTX3 downregulation in sepsis mice. After upregulation of PTX3, intestinal mucosal barrier damage was alleviated and inflammatory factor expression was decreased in sepsis mice. Further data mining suggested that the anti-inflammatory effect of PTX3 might be realized through inhibition of the toll-like receptor (TLR) signaling pathway. Moreover, compared with the LPS group, downregulation of PTX3 increased cell apoptosis and the levels of BCL2-associated X (Bax), myeloperoxidase (MPO), tumor necrosis factor-alfa (TNF-α), interleukin 1 beta (IL-1β), and interferon-gamma (IFN-γ), and decreased the levels of B-cell lymphoma-2 (Bcl-2), zona occludens (ZO)-1, and occludin. On the contrary, overexpression of PTX3 reduced cell apoptosis and the levels of Bax, MPO, TNF-α, IL-1β, and IFN-γ. Moreover, downregulation of PTX3 reversed the inhibitive effects on cell apoptosis and inflammation and promotive effects on the levels of Zo-1 and occludin induced by CLI-095 (a TLR signaling pathway inhibitor). In the CLP-induced mice sepsis model and LPS-induced cell inflammation model, PTX3 inhibits inflammatory response and reduces intestinal mucosal barrier damage through the TLR signaling pathway.
Collapse
|
9
|
Wang J, Wang F, Yuan L, Ruan H, Zhu Z, Fan X, Zhu L, Peng X. Blood-Enriching Effects and Immune-Regulation Mechanism of Steam-Processed Polygonatum Sibiricum Polysaccharide in Blood Deficiency Syndrome Mice. Front Immunol 2022; 13:813676. [PMID: 35250989 PMCID: PMC8892585 DOI: 10.3389/fimmu.2022.813676] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Polygonatum sibiricum Red. has been used as a medicinal herb and nutritional food in traditional Chinese medicine for a long time. It must be processed prior to clinical use for safe and effective applications. However, the present studies mainly focused on crude Polygonatum sibiricum (PS). This study aimed to investigate the chemical properties, blood-enriching effects and mechanism of polysaccharide from the steam-processed Polygonatum sibiricum (SPS), which is a common form of PS in clinical applications. Instrumentation analyses and chemistry analyses revealed the structure of SPS polysaccharide (SPSP). A mice model of blood deficiency syndrome (BDS) was induced by acetylphenylhydrazine (APH) and cyclophosphamide (CTX). Blood routine test, spleen histopathological changes, serum cytokines, etc. were measured. The spleen transcriptome changes of BDS mice were detected by RNA sequencing (RNA-seq). The results showed that SPSP consists predominantly of Gal and GalA together with fewer amounts of Man, Glc, Ara, Rha and GlcN. It could significantly increase peripheral blood cells, restore the splenic trabecular structure, and reverse hematopoietic cytokines to normal levels. RNA-seq analysis showed that 122 differentially expressed genes (DEGs) were obtained after SPSP treatment. GO and KEGG analysis revealed that SPSP-regulated DEGs were mainly involved in hematopoiesis, immune regulation signaling pathways. The reliability of transcriptome profiling was validated by quantitative real-time PCR and Western blot, and the results indicated that the potential molecular mechanisms of the blood-enriching effects of SPSP might be associated with the regulating of JAK1-STAT1 pathway, and elevated the hematopoietic cytokines (EPO, G-CSF, TNF-α and IL-6). This work provides important information on the potential mechanisms of SPSP against BDS.
Collapse
Affiliation(s)
- Juan Wang
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Furong Wang
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Lixia Yuan
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Hongsheng Ruan
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Zhibiao Zhu
- Department of Quality Control, Zhejiang Sanxitang Chinese Medicine Co., LTD, Yiwu, China
| | - Xiaoling Fan
- Department of Quality Control, Zhejiang Sanxitang Chinese Medicine Co., LTD, Yiwu, China
| | - Lingyan Zhu
- Department of Quality Control, Zhejiang Sanxitang Chinese Medicine Co., LTD, Yiwu, China
| | - Xin Peng
- Ningbo Research Institute of Zhejiang University, Ningbo, China
| |
Collapse
|
10
|
Fu HY, Hu ZS, Dong XT, Zhou RB, Du HY. Gelsolin Attenuates Lipopolysaccharide-Induced Acute Lung Injury in Rats by Modulating TLR4/Myd88/NF-κB Signaling Pathway. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.511.521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
11
|
Untargeted Metabolomics Analysis Revealed the Major Metabolites in the Seeds of four Polygonatum Species. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041445. [PMID: 35209244 PMCID: PMC8874640 DOI: 10.3390/molecules27041445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/29/2022]
Abstract
Most Polygonatum species are widely used in China as a source of medicine and food. In this study, a UPLC-QTOF-MS/MS system was used to conduct an untargeted metabolomics analysis and compare the classes and relative contents of metabolites in the seeds of four Polygonatum species: P. sibiricum (Ps), P. cyrtonema (Pc), P. kingianum (Pk), and P. macropodium (Pm). The objectives of this study were to clarify the metabolic profiles of these seeds and to verify their medicinal and nutritional value via comparative analyses. A total of 873 metabolites were identified, including 185 flavonoids, 127 lipids, 105 phenolic acids, and 36 steroids. The comparative analysis of metabolites among Polygonatum seed samples indicated that flavonoids, steroids, and terpenoids were the main differentially abundant compounds. The results of principal component analysis and hierarchical clustering were consistent indicating that the metabolites in Ps and Pm are similar, but differ greatly from Pc and Pk. The data generated in this study provide additional evidence of the utility of Polygonatum seeds for producing food and medicine.
Collapse
|
12
|
Li S, Lei Y, Lei J, Li H. All‑trans retinoic acid promotes macrophage phagocytosis and decreases inflammation via inhibiting CD14/TLR4 in acute lung injury. Mol Med Rep 2021; 24:868. [PMID: 34676874 PMCID: PMC8554390 DOI: 10.3892/mmr.2021.12508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical emergency and all-trans retinoic acid (ATRA) can alleviate organ injury. Therefore, the present study investigated the role of ATRA in ALI. Lipopolysaccharide (LPS)-induced ALI rats were treated with ATRA and the arterial partial pressure of oxygen (PaO2), lung wet/dry weight (W/D) ratio and protein content in the bronchial alveolar lavage fluid (BALF) were measured to evaluate the effect of ATRA on ALI rats. Alveolar macrophages were isolated from the BALF. The phagocytic function of macrophages was detected using the chicken erythrocyte phagocytosis method and flow cytometry. The viability of macrophages was measured using a Cell Counting Kit-8 assay, and apoptosis was analyzed using a TUNEL assay and flow cytometry. The expression levels of Toll-like receptor 4 (TLR4) and cluster of differentiation (CD)14 on the macrophage membrane were detected by immunofluorescence staining. The protein levels of TLR4, CD14, phosphorylated (p)-65, p65, p-IκBα and IκBα were analyzed using western blotting. The concentrations of IL-6, IL-1β and macrophage inflammatory protein-2 in the plasma of rats were detected by ELISA. Macrophages were treated with IAXO-102 (TLR4 inhibitor) to verify the involvement of CD14/TLR4 in the effect of ATRA on ALI. ATRA provided protection against LPS-induced ALI, as evidenced by the increased PaO2 and reduced lung W/D ratio and protein content in the BALF. ATRA enhanced macrophage phagocytosis and viability and reduced apoptosis and inflammation in ALI rats. Mechanically, ATRA inhibited CD14 and TLR4 expression and NF-κB pathway activation. ATRA enhanced macrophage phagocytosis and reduced inflammation by inhibiting the CD14/TLR4-NF-κB pathway in LPS-induced ALI. In summary, ATRA inactivated the NF-κB pathway by inhibiting the expression of CD14/TLR4 receptor in the alveolar macrophages of rats, thus enhancing the phagocytic function of macrophages in ALI rats, improving the activity of macrophages, inhibiting apoptosis, reducing the levels of inflammatory factors, and consequently playing a protective role in ALI model rats. This study may offer novel insights for the clinical management of ALI.
Collapse
Affiliation(s)
- Shuangxue Li
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yuansheng Lei
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jieyun Lei
- Department of Cardiology, Taiyuan Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Hui Li
- Department of Gynecology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
13
|
Zhao L, Mao Y, Yu H, Liu H, Wang C, Liu J, Han Y, Bi Y, Zhang D. The Preventive Effects of Lactobacillus casei on Acute Lung Injury Induced by Lipopolysaccharide. Indian J Microbiol 2021; 61:370-382. [PMID: 34092818 PMCID: PMC8169435 DOI: 10.1007/s12088-021-00949-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
Lactobacillus has been reported to inhibit acute lung injury (ALI). However, the molecular mechanism of Lactobacillus casei (L. casei) in preventing ALI has not been identified, so we investigated whether L. casei pretreatment could inhibit the activation of TLR4/MyD88/NF-κB signaling pathway following ALI. ALI model was established by intraperitoneal injection of 2 mg/kg lipopolysaccharide (LPS) to female BALB/c mice. In L. casei LC2W group, mice were intragastrically administrated L. casei LC2W for a week, before the ALI modeling. The serum of normal BALB/c mice after intragastric administration of L. casei LC2W was used for in vitro cell assays. The serum was pre-incubated with mouse macrophage cell line (RAW264.7) and human lung cell line (HLF-A), then LPS was added to co-incubate. Compared with ALI model group, L. casei LC2W pretreatment significantly reduced lung pathological damage, the number of neutrophils and total cells in bronchoalveolar lavage fluid. Besides, L. casei LC2W pretreatment could significantly reverse the abnormal expression of ICAM-1, IL-6, TNF-α and IL-10 in lung tissue and serum, plus, L. casei LC2W significantly reduced the phosphorylation levels of IRAK-1 and NF-κB p65. In vitro, the serum decreased the up-regulation of IL-6 and TNF-α in cell lines induced by LPS. In conclusion, L. casei LC2W intragastric administration pretreatment could significantly improve LPS-induced ALI in mice, probably through circulation to reach the lungs so as to inhibit the inflammatory response induced by activation of TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lihui Zhao
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Ying Mao
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Haiming Yu
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - He Liu
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Chao Wang
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Jianwei Liu
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Yutong Han
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Yang Bi
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Donghai Zhang
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| |
Collapse
|