1
|
Dong Q, Sun Y, Li J, Tian X, Liu S, Fu Y, Luo R, Guo L, Zong B, Lu Q, Ye C, Fu S, Qiu Y. LAMTOR1/mTORC1 promotes CD276 to induce immunosuppression via PI3K/Akt/MMP signaling pathway in Clostridium perfringens-induced necrotic enteritis of laying hens. Poult Sci 2024; 103:104216. [PMID: 39270482 PMCID: PMC11417168 DOI: 10.1016/j.psj.2024.104216] [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: 07/02/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Clostridium perfringens (C. perfringens) causes avian necrotic enteritis, leading to huge economic losses to the poultry industry. This pathogen induces host immunosuppression; however, the molecular mechanism is still unclear. Thus, we established a laying hen infection model to explore this mechanism. We randomly divided 20 one-old-day laying hens into the control and infection groups. The infection group was infected intragastrically with 1 × 109 colony-forming units of C. perfringens in 1 mL of sterile phosphate-buffered saline (PBS) once a day from d 17 to 20; the control group received the same volume of PBS without the bacterium. Twenty-four hours after the last challenge, we sacrificed the laying hens and collected the jejunum for analysis. The infection group presented alterations in blood biochemical parameters and necrotic lesion scores as well as damage to the jejunum. Proteomics revealed 427 upregulated and 291 downregulated proteins in the infection group. In the infection group, CD3, CD4, and CD8 messenger RNA expression (mRNA) expression was decreased; LAMTOR1 and mTORC1 mRNA expression was increased; CD276 protein expression was enhanced; and the PI3K/Akt/MMP pathway was activated in jejunum of laying hens. This is the first study to report CD276 expression in the jejunum related to immunosuppression in a laying hen model of necrotic enteritis. It provides some new key targets to potentially control avian necrotic enteritis.
Collapse
Affiliation(s)
- Qiaoli Dong
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yamin Sun
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jingyang Li
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xinyue Tian
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Siyu Liu
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yunjian Fu
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ronghui Luo
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ling Guo
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Bingbing Zong
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qirong Lu
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chun Ye
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shulin Fu
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Yinsheng Qiu
- School of Animal Science and Nutritional Engineering, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| |
Collapse
|
2
|
Yue C, Li J, Zhang S, Ma R, Suo M, Chen Y, Jin H, Zeng Y, Chen Y. Activation of the NLRP3-CASP-1 inflammasome is restrained by controlling autophagy during Glaesserella parasuis infection. Vet Microbiol 2024; 295:110160. [PMID: 38964034 DOI: 10.1016/j.vetmic.2024.110160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
Infection with Glaesserella parasuis, the primary pathogen behind Glässer's disease, is often associated with diverse clinical symptoms, including serofibrinous polyserositis, arthritis, and meningitis. Autophagy plays a dual role in bacterial infections, exerting either antagonistic or synergistic effects depending on the nature of the pathogen. Our previous studies have demonstrated that autophagy serves as a defense mechanism, combating inflammation and invasion caused by infection of highly virulent G. parasuis. However, the precise mechanisms remain to be elucidated. Pathogens exhibit distinct interactions with inflammasomes and autophagy processes. Herein, we explored the effect of autophagy on inflammasomes during G. parasuis infection. We found that G. parasuis infection triggers NLRP3-dependent pro-CASP-1-IL-18/IL-1β processing and maturation pathway, resulting in increased release of IL-1β and IL-18. Inhibition of autophagy enhances NLRP3 inflammasome activity, whereas stimulation of autophagy restricts it during G. parasuis infection. Furthermore, assembled NLRP3 inflammasomes undergo ubiquitination and recruit the autophagic adaptor, p62, facilitating their sequestration into autophagosomes during G. parasuis infection. These results suggest that the induction of autophagy mitigates inflammation by eliminating overactive NLRP3 inflammasomes during G. parasuis infection. Our research uncovers a mechanism whereby G. parasuis infection initiates inflammatory responses by promoting the assembly of the NLRP3 inflammasomes and activating NLRP3-CASP-1, both of which processes are downregulated by autophagy. This suggests that pharmacological manipulation of autophagy could be a promising approach to modulate G. parasuis-induced inflammatory responses.
Collapse
Affiliation(s)
- Chaoxiong Yue
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China; State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jinquan Li
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Siming Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Ruyi Ma
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingjiao Suo
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yiwen Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China.
| | - Yushan Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Wang J, Yan P, Jia Y, Guo Z, Guo Y, Yin R, Wang L, Fan Z, Zhou Y, Yuan J, Yin R. Expression profiles of miRNAs in the lung tissue of piglets infected with Glaesserella parasuis and the roles of ssc-miR-135 and ssc-miR-155-3p in the regulation of inflammation. Comp Immunol Microbiol Infect Dis 2024; 111:102214. [PMID: 39002176 DOI: 10.1016/j.cimid.2024.102214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
MicroRNAs (miRNAs) have been shown to play an important regulatory role in the process of pathogenic infection. However, the miRNAs that regulate the pathogenic process of G. parasuis and their functions are still unknown. Here, high-throughput sequencing was used to quantify the expression of miRNA in piglet lung tissue after G. parasuis XX0306 strain infection. A total of 25 differentially expressed microRNAs (DEmiRNAs) were identified. GO and KEGG pathway enrichment analysis showed that many of the functions of genes that may be regulated by DEmiRNA are related to inflammatory response and immune regulation. Further studies found that ssc-miR-135 may promote the expression of inflammatory factors through NF-κB signaling pathway. Whereas, ssc-miR-155-3p inhibited the inflammatory response induced by G. parasuis, and its regulatory mechanism remains to be further investigated. This study provides a valuable reference for revealing the regulatory effects of miRNAs on the pathogenesis of G. parasuis. DATA AVAILABILITY: The datasets generated during the current study are not publicly available due to this study is currently in the ongoing research stage, and some of the data cannot be made public sooner yet, but are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Jingyi Wang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China; College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China.
| | - Ping Yan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yongchao Jia
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Zhongbo Guo
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Ying Guo
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Ronglan Yin
- Research Academy of Animal Husbandry and Veterinary Medicine Sciences of Jilin Province, Changchun 130062, China.
| | - Linxi Wang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Zenglei Fan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yuanyuan Zhou
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Jing Yuan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Ronghuan Yin
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
4
|
He Y, Peng Y, Sun Y, Wan Y, Zhuo R, Hu S, Wang Y, Hu X, Jin H, Hua K. AMPK signaling pathway regulated the expression of the ApoA1 gene via the transcription factor Egr1 during G. parasuis stimulation. Vet Microbiol 2024; 294:110106. [PMID: 38776767 DOI: 10.1016/j.vetmic.2024.110106] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/21/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024]
Abstract
Glaesserella parasuis (G. parasuis) is the causative agent of porcine Glässer's disease, resulting in high mortality rates in pigs due to excessive inflammation-induced tissue damage. Previous studies investigating the protective effects of G. parasuis vaccination indicated a possible role of ApoA1 in reflecting disease progression following G. parasuis infection. However, the mechanisms of ApoA1 expression and its role in these infections are not well understood. In this investigation, newborn porcine tracheal (NPTr) epithelial cells infected with G. parasuis were used to elucidate the molecular mechanism and role of ApoA1. The study revealed that the AMPK pathway activation inhibited ApoA1 expression in NPTr cells infected with G. parasuis for the first time. Furthermore, Egr1 was identified as a core transcription factor regulating ApoA1 expression using a CRISPR/Cas9-based system. Importantly, it was discovered that APOA1 protein significantly reduced apoptosis, pyroptosis, necroptosis, and inflammatory factors induced by G. parasuis in vivo. These findings not only enhance our understanding of ApoA1 in response to bacterial infections but also highlight its potential in mitigating tissue damage caused by G. parasuis infection.
Collapse
Affiliation(s)
- Yanling He
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Yuna Peng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Yu Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Yanxi Wan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Ran Zhuo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Shuai Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Yi Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Xueying Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China.
| | - Kexin Hua
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, China; College of Veterinary Medicine, Huazhong Agricultural University, China; Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, China.
| |
Collapse
|
5
|
Ye C, Chen Y, Yu R, Zhao M, Yin R, Qiu Y, Fu S, Liu Y, Wu Z. Baicalin-aluminum complex on the regulation of IPEC-1 infected with enterotoxigenic Escherichia coli. Heliyon 2024; 10:e33038. [PMID: 39027442 PMCID: PMC11254522 DOI: 10.1016/j.heliyon.2024.e33038] [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: 02/20/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the main bacterial cause of diarrhea in weaned piglets. Baicalin-aluminum (BA) complex is the main active ingredient of Scutellaria baicalensis Georgi extracted-aluminum complex, which has been used to treat diarrhea in weaning piglets, however the underlying mechanism remains unclear. To investigate the effects of the BA complex on the regulation of porcine intestinal epithelial (IPEC-1) cells infected with ETEC, IPEC-1 cells were incubated with an ETEC bacterial strain at a multiplicity of infection of 1 for 6 h and then treated with different concentrations of the BA complex for 6 h. ETEC infection increased the levels of cAMP and cGMP, upregulated CFTR (cystic fibrosis transmembrane conductance regulator) mRNA, and downregulated NHE4 mRNA in IPEC-1 cells. Treatment with the BA complex inhibited ETEC adhesion and the production of cAMP and cGMP, reduced CFTR mRNA expression, and increased NHE4 mRNA expression. Overall, the BA complex weakened the adhesion of ETEC to IPEC-1 cells, and inhibited cAMP/cGMP-CFTR signaling in IPEC-1 cells.
Collapse
Affiliation(s)
- Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Yuqian Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Ruixue Yu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Ming Zhao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Ronghua Yin
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430000, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430000, PR China
| |
Collapse
|
6
|
Sun P, Yang Y, Yang L, Qian Y, Liang M, Chen H, Zhang J, Qiu Y, Guo L, Fu S. Quercetin Protects Blood-Brain Barrier Integrity via the PI3K/Akt/Erk Signaling Pathway in a Mouse Model of Meningitis Induced by Glaesserella parasuis. Biomolecules 2024; 14:696. [PMID: 38927100 PMCID: PMC11201931 DOI: 10.3390/biom14060696] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Glaesserella parasuis (G. parasuis) causes serious inflammation and meningitis in piglets. Quercetin has anti-inflammatory and anti-bacterial activities; however, whether quercetin can alleviate brain inflammation and provide protective effects during G. parasuis infection has not been studied. Here, we established a mouse model of G. parasuis infection in vivo and in vitro to investigate transcriptome changes in the mouse cerebrum and determine the protective effects of quercetin on brain inflammation and blood-brain barrier (BBB) integrity during G. parasuis infection. The results showed that G. parasuis induced brain inflammation, destroyed BBB integrity, and suppressed PI3K/Akt/Erk signaling-pathway activation in mice. Quercetin decreased the expression of inflammatory cytokines (Il-18, Il-6, Il-8, and Tnf-α) and BBB-permeability marker genes (Mmp9, Vegf, Ang-2, and Et-1), increased the expression of angiogenetic genes (Sema4D and PlexinB1), reduced G. parasuis-induced tight junction disruption, and reactivated G. parasuis-induced suppression of the PI3K/Akt/Erk signaling pathway in vitro. Thus, we concluded that quercetin may protect BBB integrity via the PI3K/Akt/Erk signaling pathway during G. parasuis infection. This was the first attempt to explore the protective effects of quercetin on brain inflammation and BBB integrity in a G. parasuis-infected mouse model. Our findings indicated that quercetin is a promising natural agent for the prevention and treatment of G. parasuis infection.
Collapse
Affiliation(s)
- Peiyan Sun
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yaqiong Yang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Linrong Yang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuanzhuo Qian
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mingxia Liang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Hongbo Chen
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan 430023, China
| | - Jing Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan 430023, China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Ling Guo
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| |
Collapse
|
7
|
Noor G, Badruddeen, Akhtar J, Singh B, Ahmad M, Khan MI. An outlook on the target-based molecular mechanism of phytoconstituents as immunomodulators. Phytother Res 2023; 37:5058-5079. [PMID: 37528656 DOI: 10.1002/ptr.7969] [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: 11/16/2022] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023]
Abstract
The immune system is one of the essential defense mechanisms. Immune system inadequacy increases the risk of infections and cancer diseases, whereas over-activation of the immune system causes allergies or autoimmune disorders. Immunomodulators have been used in the treatment of immune-related diseases. There is growing interest in using herbal medicines as multicomponent agents to modulate the complex immune system in immune-related diseases. Many therapeutic phytochemicals showed immunomodulatory effects by various mechanisms. This mechanism includes stimulation of lymphoid cell, phagocytosis, macrophage, and cellular immune function enhancement. In addition increased antigen-specific immunoglobulin production, total white cell count, and inhibition of TNF-α, IFN-γ, NF-kB, IL-2, IL-6, IL-1β, and other cytokines that influenced the immune system. This review aims to overview, widely investigated plant-derived phytoconstituents by targeting cells to modulate cellular and humoral immunity in in vivo and in vitro. However, further high-quality research is needed to confirm the clinical efficacy of plant-based immunomodulators.
Collapse
Affiliation(s)
- Gazala Noor
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Badruddeen
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Juber Akhtar
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Bhuwanendra Singh
- Department of Pharmacognosy, S.D. College of Pharmacy and Vocational Studies, Muzaffarnagar, India
| | - Mohammad Ahmad
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Mohammad Irfan Khan
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, India
| |
Collapse
|
8
|
Guan Z, Pang L, Ouyang Y, Jiang Y, Zhang J, Qiu Y, Li Z, Li B, Liu K, Shao D, Ma Z, Wei J. Secondary Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus (HP-PRRSV2) Infection Augments Inflammatory Responses, Clinical Outcomes, and Pathogen Load in Glaesserella-parasuis-Infected Piglets. Vet Sci 2023; 10:vetsci10050365. [PMID: 37235448 DOI: 10.3390/vetsci10050365] [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: 03/14/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Glaesserella parasuis (Gps), Gram-negative bacteria, are a universal respiratory-disease-causing pathogen in swine that colonize the upper respiratory tract. Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus (HP-PRRSV2HP-PRRSV2) and Gps coinfections are epidemics in China, but little is known about the influence of concurrent coinfection on disease severity and inflammatory responses. Herein, we studied the effects of secondary HP-PRRS infection on clinical symptoms, pathological changes, pathogen load, and inflammatory response of Gps coinfection in the upper respiratory tract of piglets. All coinfected piglets (HP-PRRSV2 + Gps) displayed fever and severe lesions in the lungs, while fever was present in only a few animals with a single infection (HP-PRRSV2 or Gps). Additionally, HP-PRRSV2 and Gps loading in nasal swabs and blood and lung tissue samples was significantly increased in the coinfected group. Necropsy data showed that coinfected piglets suffered from severe lung damage and had significantly higher antibody titers of HP-PRRSV2 or Gps than single-infected piglets. Moreover, the serum and lung concentrations of inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-8) were also significantly higher in coinfected piglets than in those infected with HP-PRRSV2 or Gps alone. In conclusion, our results show that HP-PRRSV2 promotes the shedding and replication of Gps, and their coinfection in the upper respiratory tract aggravates the clinical symptoms and inflammatory responses, causing lung damage. Therefore, in the unavoidable situation of Gps infection in piglets, necessary measures must be made to prevent and control secondary infection with HP-PRRSV2, which can save huge economic losses to the pork industry.
Collapse
Affiliation(s)
- Zhixin Guan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Linlin Pang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Yan Ouyang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
- College of Agriculture, Hubei Three Gorges Polytechnic, Yichang 443000, China
| | - Yifeng Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Junjie Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Zongjie Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai 200241, China
| |
Collapse
|
9
|
Xu X, Qin Z, Zhang C, Mi X, Zhang C, Zhou F, Wang J, Zhang L, Hua F. TRIM29 promotes podocyte pyroptosis in diabetic nephropathy through the NF-kB/NLRP3 inflammasome pathway. Cell Biol Int 2023; 47:1126-1135. [PMID: 36841942 DOI: 10.1002/cbin.12006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/09/2022] [Accepted: 02/20/2023] [Indexed: 02/27/2023]
Abstract
Diabetic nephropathy (DN) is one of the most common complications of diabetes. Gradual loss of podocytes is a sign of DN and pyroptosis mechanistically correlates with podocyte injury in DN; however, the mechanism(s) involved remain unknown. Here we reveal that TRIM29 is overexpressed in high glucose (HG)-treated murine podocytes cells and that TRIM29 silencing significantly inhibits podocyte damage due to HG treatment, as evidenced by lower desmin expression and greater nephrin expression. Additionally, flow cytometry analysis showed that TRIM29 silencing significantly inhibited HG treatment-induced pyroptosis, which was confirmed by immunoblotting for NLRP3, active Caspase-1, GSDMD-N, and phosphorylated NF-κB-p65. Conversely, overexpression of TRIM29 could trigger pyroptosis that was attenuated by NF-κB inhibition, indicating that TRIM29 promotes pyroptosis through the NF-κB pathway. Mechanistic studies revealed that TRIM29 interacts with IκBα to mediate its ubiquitination-dependent degradation, which in turn leads to NF-κB activation. Taken together, our data demonstrate that TRIM29 can promote podocyte pyroptosis by activating the NF-κB/NLRP3 pathway. Thus, TRIM29 represents a potentially novel therapeutic target that may also be clinically relevant in the management of DN.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China.,Department of Nephrology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Zihan Qin
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ce Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Xia Mi
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Chi Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Feihong Zhou
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Junsheng Wang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Liexiang Zhang
- Department of Neurosurgery, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China.,Department of Neurosurgery, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
10
|
Ding Y, Wang Y, Li J, Tang M, Chen H, Wang G, Guo J, Gui S. Microemulsion-thermosensitive gel composites as in situ-forming drug reservoir for periodontitis tissue repair through alveolar bone and collagen regeneration strategy. Pharm Dev Technol 2023; 28:30-39. [PMID: 36541732 DOI: 10.1080/10837450.2022.2161574] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A satisfactory clinical effect in treating periodontitis is often difficult to achieve by conventional non-surgical systemic drug delivery due to the narrow anatomical structure of the periodontal pocket and insufficient drug concentration at lesion sites. In addition, the feasibility of combating periodontal tissue lesions by restoring the alveolar bone and allowing collagen regeneration has not been fully explored. The objective of this study was to prepare a microemulsion integrating the anti-inflammatory and osteogenic active ingredients of baicalin and clove oil (BC-MEs). Then, the composite hydrogel obtained by mixing poloxamer 407 and 188 was used as the thermosensitive gel matrix to load BC-MEs and form a drug reservoir (Gel-BC-MEs) injectable in situ. Gel-BC-MEs exhibited a significant, sustained release of baicalin for 12 h, gelation temperature was 33.4 ± 0.36 °C, and pH was 5.45 ± 0.12. The experiment on a rat periodontitis model demonstrated that Gel-BC-MEs significantly improved periodontal tissue repair by collagen regeneration and osteogenesis by inhibiting osteoclast infiltration. This study proposes a novel strategy for periodontal tissue repair by enhancing the therapeutic potential of a microemulsion using an in situ nano-gel delivery system.
Collapse
Affiliation(s)
- Yang Ding
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, Anhui, China.,Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, China
| | - Yuxiao Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jiaxin Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Maomao Tang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hairong Chen
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guichun Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jian Guo
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, Anhui, China.,Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, China
| | - Shuangying Gui
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, China.,Anhui Province Key Laboratory of Pharmaceutical Technology and Application, Hefei, Anhui, China.,Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, China
| |
Collapse
|
11
|
Zeng Z, Zhang H, Gui G, Luo J, Liu S. Macleaya cordata extract modulates inflammation via inhibition of the NF-κB and MAPK signaling pathways in porcine alveolar macrophages induced by Glaesserella parasuis. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2022; 86:254-260. [PMID: 36211213 PMCID: PMC9536353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/15/2022] [Indexed: 04/03/2023]
Abstract
Glässer's disease in pigs is associated with infection by Glaesserella parasuis and is characterized by pneumonia-like symptoms, fibrinous polyserositis, polyarthritis, and meningitis. Macleaya cordata, a commonly used traditional Chinese medication, has been shown to have anti-inflammatory, antiviral, antioxidative, antimicrobial, insecticidal, and antitumor properties. However, the anti-inflammatory effects of M. cordata on G. parasuis stimulation are still poorly understood. This study explored the anti-inflammatory effects and mechanisms of M. cordata extract on G. parasuis-induced inflammatory responses, via the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, in porcine alveolar macrophages (PAMs). Porcine alveolar macrophages, when stimulated with G. parasuis, initiated transcription of interleukin (IL)-1α, IL-1β, IL-6, IL-8, and tumor necrosis factor alpha (TNF-α). Furthermore, p65, IκBα, p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) phosphorylation were upregulated via the NF-κB and MAPK signaling pathways. However, treatment with M. cordata extract inhibited transcription of IL-1α, IL-1β, IL-6, IL-8, and TNF-α and reduced p65, IκBα, p38, ERK, and JNK phosphorylation, by inhibiting activation of the NF-κB and MAPK signaling pathways in PAMs induced by G. parasuis. These findings reveal that M. cordata extract can reverse the inflammatory effect initiated by G. parasuis in vitro and that it possesses significant immunosuppression activity; thus, it may offer a novel strategy for controlling and treating G. parasuis infection.
Collapse
Affiliation(s)
- Ze Zeng
- Key Open Laboratory of Chinese Veterinary Medicine of State Ethnic Affairs Commission, Tongren Vocational and Technical College, Tongren, Guizhou 554300, China
| | - Huaqi Zhang
- Key Open Laboratory of Chinese Veterinary Medicine of State Ethnic Affairs Commission, Tongren Vocational and Technical College, Tongren, Guizhou 554300, China
| | - Ganbei Gui
- Key Open Laboratory of Chinese Veterinary Medicine of State Ethnic Affairs Commission, Tongren Vocational and Technical College, Tongren, Guizhou 554300, China
| | - Jie Luo
- Key Open Laboratory of Chinese Veterinary Medicine of State Ethnic Affairs Commission, Tongren Vocational and Technical College, Tongren, Guizhou 554300, China
| | - Shanshan Liu
- Key Open Laboratory of Chinese Veterinary Medicine of State Ethnic Affairs Commission, Tongren Vocational and Technical College, Tongren, Guizhou 554300, China
| |
Collapse
|
12
|
Wang K, Yang A, Peng X, Lv F, Wang Y, Cui Y, Wang Y, Zhou J, Si H. Linkages of Various Calcium Sources on Immune Performance, Diarrhea Rate, Intestinal Barrier, and Post-gut Microbial Structure and Function in Piglets. Front Nutr 2022; 9:921773. [PMID: 35782941 PMCID: PMC9248811 DOI: 10.3389/fnut.2022.921773] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/20/2022] [Indexed: 01/08/2023] Open
Abstract
The purpose of this experiment was to investigate the effects of different sources of calcium on immune performance, diarrhea rate, intestinal barrier, and post-intestinal flora structure and function in weaned piglets. A total of 1,000 weaned piglets were randomly assigned to five groups 10 replicate pens per treatment, 20 piglets per pen and fed calcium carbonate, calcium citrate, multiple calcium, and organic trace minerals of different concentrations of acidifier diets. The results of the study showed that the replacement of calcium carbonate with calcium citrate and multiple calcium had almost no significant effect on immune indexes (IL-1β, IL-6, IL-10, TNF-α) of piglets compared with the control group (p > 0.05). The five groups did not show a change in the diarrhea rate and diarrhea index (p > 0.05). The diet containing multiple calcium dramatically decreased the TP compared to the C and L diet (p < 0.05). No significant difference in HDL was noted in the five groups (p > 0.05). However, the concentration of LDL in blood in the multiple calcium group was significantly higher than that in groups L and D (p < 0.05). Moreover, the concentration of Glu in blood in the multiple calcium group was significantly higher than that in group C (p < 0.05). Compared with the control group, calcium citrate plus organic trace minerals diet markedly increased UCG-005 abundance in the colon (p < 0.05). In addition, the relative abundance of Prevotellaceae_NK3B31_group had an upward trend in the colon of the M group compared to the D group (p = 0.070). Meanwhile, calcium citrate plus organic trace minerals diet markedly increased Clostridium_sensu_stricto_1 abundance in the colon (p < 0.05). Metagenomic predictions by PICRUSt suggested that the colonic and fecal microbiota was mainly involved in carbohydrate metabolism, amino acid metabolism, energy metabolism, and metabolism of cofactors and vitamins.
Collapse
Affiliation(s)
- Kaijun Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Anqi Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiaomin Peng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Feifei Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ying Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yao Cui
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yuhan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | | | - Hongbin Si
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Hongbin Si,
| |
Collapse
|
13
|
Chen X, Shao Z, Wu L, He B, Yang W, Chen J, Jin E, Huang Q, Lei L, Xu J, Li H, Zhang H, Wan Y, Liu W, Zhou R. Involvement of the Actinobacillus pleuropneumoniae ompW Gene in Confrontation of Environmental Pressure. Front Vet Sci 2022; 9:846322. [PMID: 35664844 PMCID: PMC9161549 DOI: 10.3389/fvets.2022.846322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Actinobacillus pleuropneumoniae causes porcine pleuropneumonia. The function of the outer membrane protein W gene (ompW) of A. pleuropneumoniae has not been evaluated. Thus a deletion mutant of ompW, ΔompW, was constructed to explore the effect of ompW gene deletion on bacterial growth, biofilm formation, bacterial morphology, oxidative tolerance, susceptibility to antibiotics, and the expression of ribosome synthesis and ABC transporter related genes. Results showed that the ompW gene deletion did not affect biofilm formation and the growth of A. pleuropneumoniae but did affect bacterial morphology during steady growth, oxidative tolerance, and bacterial susceptibility to polymyxin B, kanamycin, and penicillin. The ompW gene deletion also affected the expression of ribosome synthesis and ABC transporter related genes. These results suggested that ompW may regulate the biological phenotype of A. pleuropneumoniae.
Collapse
Affiliation(s)
- Xiabing Chen
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
- *Correspondence: Xiabing Chen
| | - Zhiyong Shao
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Lijun Wu
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Bin He
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Wenhai Yang
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Jie Chen
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Erguang Jin
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liancheng Lei
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, China
| | - Jiajia Xu
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hui Zhang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yun Wan
- Wuhan Animal Disease Control Center, Wuhan, China
| | - Wu Liu
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Sciences, Wuhan, China
- Wu Liu
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Cooperative Innovation Center for Sustainable Pig Production, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Rui Zhou
| |
Collapse
|
14
|
Zaghloul RA, Zaghloul AM, El-Kashef DH. Hepatoprotective effect of Baicalin against thioacetamide-induced cirrhosis in rats: Targeting NOX4/NF-κB/NLRP3 inflammasome signaling pathways. Life Sci 2022; 295:120410. [PMID: 35182557 DOI: 10.1016/j.lfs.2022.120410] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022]
Abstract
Aim Liver cirrhosis is the result of a vicious cycle of both chronic oxidative stress and inflammation. NADPH oxidase-4 (NOX4) and its companion, NOD-like receptor protein 3 (NLRP3) inflammasome, are emerging as therapeutic targets of liver fibrosis. MAIN METHODS Baicalin (BA), a natural flavone, has been investigated for its therapeutic potential against cirrhosis induced by thioacetamide (TAA) (200 mg/kg, twice/week) for 12 weeks in Sprague-Dawley rats. Two doses of BA were administered (25 and 75 mg/kg/day, orally, a week after TAA was stopped and continued for 4 weeks). KEY FINDINGS BA was able to reduce fibrosis visualized by Masson trichrome and immunohistochemical staining of the hepatic α-smooth muscle actin (α-SMA) and transforming growth factor-β1. Moreover, BA was able to ameliorate inflammation by reducing hepatic NLRP3 inflammasome subunits, NLRP3 and caspase-1, both parts of the complex responsible for the activation of different interleukins (IL), measured as IL-1β. In addition, BA was able to reduce hepatic nuclear factor kappa B (NF-κB)-driven inflammation through IL-6. BA targeted inflammation through its anti-oxidant ability evidenced by the enhancement of the hepatic superoxide dismutase (SOD) and reduced glutathione (GSH) activity and level, respectively, and the reduction of both hepatic malondialdehyde (MDA) and nitric oxide (NOx) contents. Treatment with BA significantly decreased TAA-induced elevation in hepatic NOX4, a key enzyme for reactive oxygen species (ROS) generation, as well as, inducible nitric oxide synthase (iNOS). SIGNIFICANCE therefore, the study could conclude, the anti-fibrotic effect of BA through TGF- β1/NOX4/NF-κB/NLRP3 pathway, exerting both anti-inflammatory and anti-oxidant effects.
Collapse
Affiliation(s)
- Randa A Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Egypt.
| | - Ahmed M Zaghloul
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, 35516, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
15
|
Liu D, Wang Q, He W, Ge L, Huang K. Deoxynivalenol aggravates the immunosuppression in piglets and PAMs under the condition of PEDV infection through inhibiting TLR4/NLRP3 signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113209. [PMID: 35051765 DOI: 10.1016/j.ecoenv.2022.113209] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
Mycotoxins are toxic metabolites produced by fungi, which are ubiquitous in cereals and feed worldwide and threaten human and animal health. Deoxynivalenol (DON) is one of the most prevalent mycotoxins and causes a series of toxicities, especially enterotoxicity and immunotoxicity. Porcine epidemic diarrhea virus (PEDV) is a destructive enteropathogenic animal coronavirus, is often accompanied with DON contamination in the swine herd. Previous studies have shown that PEDV infection leads severe immunosuppression in pigs. However, whether DON exposure aggravates the PEDV-induced immunosuppression remains unclear. In this study, weaned piglet and porcine alveolar macrophage cell (PAM) models were established to explore the effects of DON on the PEDV-induced immunosuppression and to clarify its underlying mechanism. The in vivo results showed that 2.25 mg/kg feed DON significantly exacerbated the immunosuppressive effects on the PEDV-infected piglets, as demonstrated by the decreases in growth performance, the numbers of goblet cells and CD3+T cells, as well as the protein expressions of ZO-1, Claudin1 and Muc2, in addition to the increases in anti-inflammatory factors levels and the intestinal injury. Similarly, the in vitro results demonstrated that 3-4 μM DON markedly aggravated apoptosis, enhanced the expressions of anti-inflammatory factors, but reduced the migration and phagocytosis abilities of the PEDV-infected PAMs. Furthermore, DON significantly suppressed the expressions of TLR4/NLRP3 in vivo and in vitro. To contrast, lipopolysaccharide (LPS), the corresponding activator, obviously alleviated the DON-exacerbated immunosuppression. Our findings suggest that DON could aggravate host immunosuppression under the condition of PEDV infection through inhibiting TLR4/NLRP3 signaling pathway, and provide novel theoretical insights into the further studies on the immunotoxicity of DON contamination and PEDV-induced immunosuppression.
Collapse
Affiliation(s)
- Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Qing Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Wenmiao He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Lei Ge
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| |
Collapse
|
16
|
Skrajnowska D, Brumer M, Kankowska S, Matysek M, Miazio N, Bobrowska-Korczak B. Covid 19: Diet Composition and Health. Nutrients 2021; 13:2980. [PMID: 34578858 PMCID: PMC8472186 DOI: 10.3390/nu13092980] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022] Open
Abstract
The virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the disease coronavirus disease 2019 (COVID-19). The cumulative number of cases reported globally is now nearly 197 million and the number of cumulative deaths is 4.2 million (26 July to 1 August 2021). Currently we are focusing primarily on keeping a safe distance from others, washing our hands, and wearing masks, and the question of the effects of diet and diet-dependent risk factors remains outside the center of attention. Nevertheless, numerous studies indicate that diet can play an important role in the course of COVID-19. In this paper, based on select scientific reports, we discuss the structure and replication cycle of SARS-CoV-2, risk factors, dietary standards for sick patients, and the roles of the microbiome and dietary components supporting the immune system in preventing COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | - Barbara Bobrowska-Korczak
- Department of Bromatology, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (D.S.); (M.B.); (S.K.); (M.M.); (N.M.)
| |
Collapse
|
17
|
Kim CH. Anti-SARS-CoV-2 Natural Products as Potentially Therapeutic Agents. Front Pharmacol 2021; 12:590509. [PMID: 34122058 PMCID: PMC8194829 DOI: 10.3389/fphar.2021.590509] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 04/19/2021] [Indexed: 12/21/2022] Open
Abstract
Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2), a β-coronavirus, is the cause of the recently emerged pandemic and worldwide outbreak of respiratory disease. Researchers exchange information on COVID-19 to enable collaborative searches. Although there is as yet no effective antiviral agent, like tamiflu against influenza, to block SARS-CoV-2 infection to its host cells, various candidates to mitigate or treat the disease are currently being investigated. Several drugs are being screened for the ability to block virus entry on cell surfaces and/or block intracellular replication in host cells. Vaccine development is being pursued, invoking a better elucidation of the life cycle of the virus. SARS-CoV-2 recognizes O-acetylated neuraminic acids and also several membrane proteins, such as ACE2, as the result of evolutionary switches of O-Ac SA recognition specificities. To provide information related to the current development of possible anti-SARS-COV-2 viral agents, the current review deals with the known inhibitory compounds with low molecular weight. The molecules are mainly derived from natural products of plant sources by screening or chemical synthesis via molecular simulations. Artificial intelligence-based computational simulation for drug designation and large-scale inhibitor screening have recently been performed. Structure-activity relationship of the anti-SARS-CoV-2 natural compounds is discussed.
Collapse
Affiliation(s)
- Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkhwan University, Suwon, South Korea
| |
Collapse
|
18
|
Luo X, Chang X, Zhou H, Lin H, Fan H. Glaesserella parasuis induces inflammatory response in 3D4/21 cells through activation of NLRP3 inflammasome signaling pathway via ROS. Vet Microbiol 2021; 256:109057. [PMID: 33799227 DOI: 10.1016/j.vetmic.2021.109057] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/21/2021] [Indexed: 02/08/2023]
Abstract
Glaesserella parasuis (G. parasuis) is an important pathogenic bacterium that can cause Glässer's disease, and it has resulted in tremendous economic losses to the global swine industry. The intensive pulmonary inflammatory response caused by G. parasuis infection is the main cause of lung injury and death in pigs. However, the exact mechanism by which it causes severe pulmonary inflammation is not fully understood yet. In this study, severe pneumonia was observed in piglets infected with G. parasuis; and an infection cell model was established using porcine alveolar macrophages cell line 3D4/21, which was determined to be susceptible to G. parasuis infection in vitro. G. parasuis infection of 3D4/21 cells induced upregulation of proinflammatory cytokines TNF-α, IL-1β, IL-18 and production of intracellular reactive oxygen species (ROS). The expression of IL-1β related to activation of the NLRP3 inflammasome signaling pathway, which had not been shown before in G. parasuis infection. Furthermore, it was first found that release of intracellular ROS, which was mediated by NADPH oxidase in 3D4/21 cells, was found crucial for the activation of the NLRP3 signaling pathway and promoted the expression of proinflammatory cytokines, such as TNF-α and IL-1. In general, this study explored the specific mechanism of severe pulmonary inflammation caused by G. parasuis infection, and provides a foundation for further elucidating the pathogenic mechanism of G. parasuis.
Collapse
Affiliation(s)
- Xinran Luo
- MOE Joint International Reasearch Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiaojing Chang
- MOE Joint International Reasearch Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Hong Zhou
- MOE Joint International Reasearch Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Huixing Lin
- MOE Joint International Reasearch Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Hongjie Fan
- MOE Joint International Reasearch Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
19
|
Rizzo V, Ferlazzo N, Currò M, Isola G, Matarese M, Bertuccio MP, Caccamo D, Matarese G, Ientile R. Baicalin-Induced Autophagy Preserved LPS-Stimulated Intestinal Cells from Inflammation and Alterations of Paracellular Permeability. Int J Mol Sci 2021; 22:2315. [PMID: 33652555 PMCID: PMC7956379 DOI: 10.3390/ijms22052315] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Several studies have demonstrated a relevant role of intestinal epithelial cells in the immune response and in chronic inflammatory conditions, including ulcers, colitis, and Crohn's disease. Baicalin (BA), extracted from the root of Scutellaria baicalensis, has various beneficial healthy effects, including anti-inflammatory activity. However, few studies have evaluated BA effects on autophagic signaling in epithelial cell response to inflammatory stimuli. To explore possible beneficial effects of BA, HT-29 cells were exposed to lipopolysaccharide (LPS), in presence or absence of BA, for 4 h. We evaluated mRNA levels of autophagy-related genes and cytokines, triggering inflammatory response. Furthermore, the expression of claudin 1, involved in the regulation of paracellular permeability was analyzed. BA treatment repressed LPS-induced expression of TNF-α and IL-1β. The down-regulation of autophagy-related genes induced by LPS was counteracted by cell pretreatment with BA. Under these conditions, BA reduced the NF-κB activation caused by LPS. Also, BA restored mRNA and protein levels of claudin 1, which were reduced by LPS. In conclusion, in intestinal epithelial cells BA regulates the NF-κB activation and modulates both autophagic and inflammatory processes, leading to an improvement of paracellular permeability. These results suggest that the anti-inflammatory effects of BA can be associated to the regulation of autophagic flux.
Collapse
Affiliation(s)
| | | | - Monica Currò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Polyclinic Hospital University, 98158 Messina, Italy; (V.R.); (N.F.); (G.I.); (M.M.); (M.P.B.); (D.C.); (G.M.); (R.I.)
| | | | | | | | | | | | | |
Collapse
|
20
|
Liao H, Ye J, Gao L, Liu Y. The main bioactive compounds of Scutellaria baicalensis Georgi. for alleviation of inflammatory cytokines: A comprehensive review. Biomed Pharmacother 2020; 133:110917. [PMID: 33217688 DOI: 10.1016/j.biopha.2020.110917] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/11/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Scutellaria baicalensis Georgi., a plant used in traditional Chinese medicine, has multiple biological activities, including anti-inflammatory, antiviral, antitumor, antioxidant, and antibacterial effects, and can be used to treat respiratory tract infections, pneumonia, colitis, hepatitis, and allergic diseases. The main active substances of S. baicalensis, baicalein, baicalin, wogonin, wogonoside, and oroxylin A, can act directly on immune cells such as lymphocytes, macrophages, mast cells, dendritic cells, monocytes, and neutrophils, and inhibit the production of the inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α, and other inflammatory mediators such as nitric oxide, prostaglandins, leukotrienes, and reactive oxygen species. The molecular mechanisms underlying the immunomodulatory and anti-inflammatory effects of the active compounds of S. baicalensis include downregulation of toll-like receptors, activation of the Nrf2 and PPAR signaling pathways, and inhibition of the nuclear thioredoxin system and inflammation-associated pathways such as those of MAPK, Akt, NFκB, and JAK-STAT. Given that in addition to the downregulation of cytokine production, the active constituents of S. baicalensis also have antiviral and antibacterial effects, they may be more promising candidate therapeutics for the prevention of infection-related cytokine storms than are drugs having only antimicrobial or anti-inflammatory activities.
Collapse
Affiliation(s)
- Hengfeng Liao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lili Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
21
|
Xu C, Fang MY, Wang K, Liu J, Tai GP, Zhang ZT, Ruan BF. Discovery and Development of Inflammatory Inhibitors from 2-Phenylchromonone (Flavone) Scaffolds. Curr Top Med Chem 2020; 20:2578-2598. [PMID: 32972343 DOI: 10.2174/1568026620666200924115611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/01/2020] [Accepted: 06/14/2020] [Indexed: 12/19/2022]
Abstract
Flavonoids are compounds based on a 2-phenylchromonone scaffold. Flavonoids can be divided into flavonoids, flavonols, dihydroflavones, anthocyanins, chalcones and diflavones according to the oxidation degree of the central tricarbonyl chain, the connection position of B-ring (2-or 3-position), and whether the tricarbonyl chain forms a ring or not. There are a variety of biological activities about flavonoids, such as anti-inflammatory activity, anti-oxidation and anti-tumor activity, and the antiinflammatory activity is apparent. This paper reviews the anti-inflammatory activities and mechanisms of flavonoids and their derivatives reported in China and abroad from 2011 till date (2011-2020), in order to find a good drug scaffold for the study of anti-inflammatory activities.
Collapse
Affiliation(s)
- Chen Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Meng-Yuan Fang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Ke Wang
- Center of Tobacco Industry Development, Xuanzhou District, Xuancheng, 242000, China
| | - Jing Liu
- Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei 230601, China,Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Guang-Ping Tai
- Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei 230601, China
| | - Zhao-Ting Zhang
- Center of Tobacco Industry Development, Xuanzhou District, Xuancheng, 242000, China
| | - Ban-Feng Ruan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China,Key Lab of Biofabrication of Anhui Higher Education, Hefei University, Hefei 230601, China
| |
Collapse
|
22
|
Fu S, Liu J, Xu J, Zuo S, Zhang Y, Guo L, Qiu Y, Ye C, Liu Y, Wu Z, Hou Y, Hu CAA. The effect of baicalin on microRNA expression profiles in porcine aortic vascular endothelial cells infected by Haemophilus parasuis. Mol Cell Biochem 2020; 472:45-56. [PMID: 32519231 DOI: 10.1007/s11010-020-03782-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 06/04/2020] [Indexed: 01/10/2023]
Abstract
Glässer's disease, caused by Haemophilus parasuis (H. parasuis), is associated with vascular damage and vascular inflammation in pigs. Therefore, early assessment and treatment are essential to control the inflammatory disorder. MicroRNAs have been shown to be involved in the vascular pathology. Baicalin has important pharmacological functions, including anti-inflammatory, antimicrobial and antioxidant effects. In this study, we investigated the changes of microRNAs in porcine aortic vascular endothelial cells (PAVECs) induced by H. parasuis and the effect of baicalin in this model by utilizing high-throughput sequencing. The results showed that 155 novel microRNAs and 76 differentially expressed microRNAs were identified in all samples. Subsequently, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the target genes of the differentially expressed microRNAs demonstrated that regulation of actin cytoskeleton, focal adhesion, ECM-receptor interaction, bacterial invasion of epithelial cells, and adherens junction were the most interesting pathways after PAVECs were infected with H. parasuis. In addition, when the PAVECs were pretreated with baicalin, mismatch repair, peroxisome, oxidative phosphorylation, DNA replication, and ABC transporters were the most predominant signaling pathways. STRING analysis showed that most of the target genes of the differentially expressed microRNAs were associated with each other. The expression levels of the differentially expressed microRNAs were negatively co-regulated with their target genes' mRNA following pretreatment with baicalin in the H. parasuis-induced PAVECs using co-expression networks analysis. This is the first report that microRNAs might have key roles in inflammatory damage of vascular tissue during H. parasuis infection. Baicalin regulated the microRNAs changes in the PAVECs following H. parasuis infection, which may represent useful novel targets to prevent or treat H. parasuis infection.
Collapse
Affiliation(s)
- Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Jun Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Jianfeng Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Sanling Zuo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Yunfei Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China.
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Chien-An Andy Hu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
- Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| |
Collapse
|
23
|
The effects of baicalin on piglets challenged with Glaesserella parasuis. Vet Res 2020; 51:102. [PMID: 32795339 PMCID: PMC7427943 DOI: 10.1186/s13567-020-00826-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022] Open
Abstract
Glaesserella parasuis (G. parasuis) causes porcine vascular inflammation and damage. Baicalin is reported to have antioxidant and anti-inflammatory functions. However, whether baicalin protects piglets against G. parasuis challenge and the potential protective mechanism have not been investigated. Therefore, in this study, we comprehensively examined the protective efficacy of baicalin in piglets challenged with G. parasuis and the possible protective mechanism. Our results show that baicalin attenuated the release of the inflammation-related cytokines interleukin (IL) 1β, IL6, IL8, IL10, and tumour necrosis factor α (TNF-α) and reduced high mobility group box 1 (HMGB1) production and cell apoptosis in piglets infected with G. parasuis. Baicalin also inhibited the activation of the mitogen-activated protein kinase (MAPK) signalling pathway and protected piglets against G. parasuis challenge. Taken together, our data suggest that baicalin could protect piglets from G. parasuis by reducing HMGB1 release, attenuating cell apoptosis, and inhibiting MAPK signalling activation, thereby alleviating the inflammatory response induced by the bacteria. Our results suggest that baicalin has utility as a novel therapeutic drug to control G. parasuis infection.
Collapse
|
24
|
McKee DL, Sternberg A, Stange U, Laufer S, Naujokat C. Candidate drugs against SARS-CoV-2 and COVID-19. Pharmacol Res 2020; 157:104859. [PMID: 32360480 PMCID: PMC7189851 DOI: 10.1016/j.phrs.2020.104859] [Citation(s) in RCA: 347] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 12/20/2022]
Abstract
Outbreak and pandemic of coronavirus SARS-CoV-2 in 2019/2020 will challenge global health for the future. Because a vaccine against the virus will not be available in the near future, we herein try to offer a pharmacological strategy to combat the virus. There exists a number of candidate drugs that may inhibit infection with and replication of SARS-CoV-2. Such drugs comprise inhibitors of TMPRSS2 serine protease and inhibitors of angiotensin-converting enzyme 2 (ACE2). Blockade of ACE2, the host cell receptor for the S protein of SARS-CoV-2 and inhibition of TMPRSS2, which is required for S protein priming may prevent cell entry of SARS-CoV-2. Further, chloroquine and hydroxychloroquine, and off-label antiviral drugs, such as the nucleotide analogue remdesivir, HIV protease inhibitors lopinavir and ritonavir, broad-spectrum antiviral drugs arbidol and favipiravir as well as antiviral phytochemicals available to date may limit spread of SARS-CoV-2 and morbidity and mortality of COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Ariane Sternberg
- Center and Network for Targeted Oncology, Muehlackerweg 8, D-69239, Heidelberg, Germany
| | - Ulrike Stange
- Center and Network for Targeted Oncology, Muehlackerweg 8, D-69239, Heidelberg, Germany
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität, Tübingen and Tuebingen Center for Academic Drug Discovery, Auf Der Morgenstelle 8, 72076, Tuebingen, Germany
| | - Cord Naujokat
- Institute of Immunology, University of Heidelberg, Im Neuenheimer Feld 305, D-69120, Heidelberg, Germany.
| |
Collapse
|
25
|
Wu X, Deng X, Wang J, Li Q. Baicalin Inhibits Cell Proliferation and Inflammatory Cytokines Induced by Tumor Necrosis Factor α (TNF-α) in Human Immortalized Keratinocytes (HaCaT) Human Keratinocytes by Inhibiting the STAT3/Nuclear Factor kappa B (NF-κB) Signaling Pathway. Med Sci Monit 2020; 26:e919392. [PMID: 32321906 PMCID: PMC7193247 DOI: 10.12659/msm.919392] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background Baicalin is a flavone isolated from the root of Scutellaria baicalensis and is used in traditional Chinese medicine. Psoriasis is a persistent and recurrent chronic inflammatory skin disease that is characterized by inflammation and increased proliferation of keratinocytes. This study aimed to investigate the effects of baicalin on HaCaT immortalized human keratinocytes in vitro and the molecular mechanisms involved. Material/Methods HaCaT keratinocytes were cultured in increasing concentrations of baicalin at 6.25 μM, 12.5 μM, and 25 μM. The in vitro model of psoriasis was established using HaCaT cells treated with tumor necrosis factor-α (TNF-α). The MTT assay was used to asses cell viability and apoptosis. Western blot was used to measure the expression of Bcl-2, Bax, pro-caspase-3, and cleaved caspase-3, and enzyme-linked immunosorbent assay (ELISA) was performed to detect inflammatory cytokines. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used to detect the levels of STAT3 and p65 mRNA. Results Baicalin reduced cell viability and induced apoptosis of HaCaT human keratinocytes in a dose-dependent manner. Increased cell viability and the expression of inflammatory cytokines by HaCaT cells induced by TNF-α were significantly inhibited by baicalin. Baicalin significantly inhibited the activation of the STAT3/NF-κB pathway in HaCaT cells stimulated by TNF-α. Conclusions Baicalin inhibited the proliferation and expression of inflammatory cytokines in HaCaT immortalized human keratinocytes in vitro through the inhibition of the STAT3/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xianwei Wu
- Department of Dermatology, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| | - Xiue Deng
- Department of Dermatology, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| | - Jiandi Wang
- Department of Dermatology, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| | - Qin Li
- Department of Dermatology, Gansu Provincial Hospital, Lanzhou, Gansu, China (mainland)
| |
Collapse
|
26
|
Owona BA, Abia WA, Moundipa PF. Natural compounds flavonoids as modulators of inflammasomes in chronic diseases. Int Immunopharmacol 2020; 84:106498. [PMID: 32304996 DOI: 10.1016/j.intimp.2020.106498] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/12/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
The use of dietary or medicinal plant based natural compounds to disease treatment has become a unique trend in clinical research. Flavonoids, a group of polyphenolic compounds have drawn significant attention due to their modulatory effects on inflammasomes associated with the initiation and progression of chronic disorders including metabolic, neurodegenerative diseases and cancer. In this article, the role of most commonly studied natural flavonoids with their disease-specific impact via inflammasomes as a potential molecular target has been described. Since the role of inflammation is evident in multiple diseases, flavonoids may serve as a promising tool in drug discovery for the intervention of chronic diseases by manipulating the status of inflammation via inflammasome targeting.
Collapse
Affiliation(s)
- Brice Ayissi Owona
- Department of Biochemistry, Laboratory of Molecular Pharmacology and Toxicology, PO 812 AEFAS Faculty of Science, University of Yaounde I, Cameroon.
| | - Wilfred Angie Abia
- Department of Biochemistry, Laboratory of Molecular Pharmacology and Toxicology, PO 812 AEFAS Faculty of Science, University of Yaounde I, Cameroon
| | - Paul Fewou Moundipa
- Department of Biochemistry, Laboratory of Molecular Pharmacology and Toxicology, PO 812 AEFAS Faculty of Science, University of Yaounde I, Cameroon
| |
Collapse
|
27
|
Alschuler L, Weil A, Horwitz R, Stamets P, Chiasson AM, Crocker R, Maizes V. Integrative considerations during the COVID-19 pandemic. Explore (NY) 2020; 16:354-356. [PMID: 32229082 PMCID: PMC7270871 DOI: 10.1016/j.explore.2020.03.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Lise Alschuler
- University of Arizona College of Medicine, United States; Andrew Weil Center for Integrative Medicine, United States.
| | - Andrew Weil
- Andrew Weil Center for Integrative Medicine, United States; University of Arizona, United States
| | - Randy Horwitz
- University of Arizona College of Medicine, United States; Andrew Weil Center for Integrative Medicine, United States
| | | | - Ann Marie Chiasson
- University of Arizona College of Medicine, United States; Andrew Weil Center for Integrative Medicine, United States
| | - Robert Crocker
- University of Arizona College of Medicine, United States; Andrew Weil Center for Integrative Medicine, United States
| | - Victoria Maizes
- University of Arizona College of Medicine, United States; Andrew Weil Center for Integrative Medicine, United States
| |
Collapse
|
28
|
Shi H, Zhang Y, Xing J, Liu L, Qiao F, Li J, Chen Y. Baicalin attenuates hepatic injury in non-alcoholic steatohepatitis cell model by suppressing inflammasome-dependent GSDMD-mediated cell pyroptosis. Int Immunopharmacol 2020; 81:106195. [PMID: 32028242 DOI: 10.1016/j.intimp.2020.106195] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/21/2019] [Accepted: 01/05/2020] [Indexed: 12/12/2022]
Abstract
Baicalin (BA), a flavone glycoside, is the constituent of Scutellaria baicalensis, a Chinese herbal medicine used to treat non-alcoholic steatohepatitis (NASH). However, the mechanism of BA on NASH is still not clear. Here, the improving effect of BA on hepatocyte through inhibition of pyroprosis was investigated in vitro. With a cell model of NASH exposing HepG2 cells in free fatty acids (FFA), we revealed that BA could improve hepatocyte from FFA-induced morphological damage and death. And then through transcriptomes screening, a significant down-regulation of NLR pyrin domain containing 3 (Nlrp3), gasdermin D (Gsdmd), andinterleukin-1 beta (IL-1β) expression were found after BA treatment. Further analysis confirmed that BA could decrease the levels of NLRP3 and GSDMD, as well as the release of IL-1β and IL-18, resulting in the reduction of pyroptosis. Moreover, the improving effect of BA could be attenuated by Gsdmd knockdown. In conclusion, BA can reduce pyroptosis of hepatocyte by blocking NLRP3-GSDMD signaling in vitro.
Collapse
Affiliation(s)
- Huilian Shi
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Department of Infectious Diseases, Nanjing, Jiangsu, China; Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Infectious Diseases, Nanjing, Jiangsu, China
| | - Yanliang Zhang
- Nanjing Hospital of TCM, Department of Infectious Diseases, Nanjing, Jiangsu, China; Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Department of Infectious Diseases, Nanjing, Jiangsu, China
| | - Jing Xing
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Gastroenterology, Nanjing, Jiangsu, China
| | - Lina Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Infectious Diseases, Nanjing, Jiangsu, China
| | - Fei Qiao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Infectious Diseases, Nanjing, Jiangsu, China
| | - Jun Li
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Department of Infectious Diseases, Nanjing, Jiangsu, China.
| | - Yuanyuan Chen
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
29
|
Zhou Y, Feng S, He X, Zhou Q, Wang Y, Yue H, Tang C, Zhang B. Surface-exposed loops L7 and L8 of Haemophilus (Glaesserella) parasuis OmpP2 contribute to the expression of proinflammatory cytokines in porcine alveolar macrophages. Vet Res 2019; 50:105. [PMID: 31783919 PMCID: PMC6884870 DOI: 10.1186/s13567-019-0721-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Outer membrane protein P2 (OmpP2) of the virulent Haemophilus (Glaesserella) parasuis has been shown to induce the release of proinflammatory cytokines. The OmpP2 protein is composed of eight or nine surface-exposed loops, but it is unclear which of them participates in the OmpP2-induced inflammatory response. In this study, we synthesized linear peptides corresponding to surface-exposed loops L1–L8 of OmpP2 from the virulent H. parasuis SC096 strain to stimulate porcine alveolar macrophages (PAMs) in vitro. We found that both L7 and L8 significantly upregulated the mRNA expression of interleukin (IL)-1α, IL-1β, IL-6, IL-8, IL-17, and IL-23 and the chemokines CCL-4 and CCL-5 in a time- and dose-dependent manner. Additionally, we constructed ompP2ΔLoop7 and ompP2ΔLoop8 mutant SC096 strains and extracted their native OmpP2 proteins to stimulate PAMs. These mutant proteins induced significantly less mRNA expression of inflammatory cytokines than SC096 OmpP2. Next, the amino acid sequences of L7 and L8 from 15 serovars of H. parasuis OmpP2 were aligned. These sequences were relatively conserved among the most virulent reference strains, suggesting that L7 and L8 are the most active peptides of the OmpP2 protein. Furthermore, L7 and L8 significantly upregulated the NF-κB and AP-1 activity levels based on luciferase reporter assays in a dose-dependent manner. Therefore, our results demonstrated that both surface-exposed loops L7 and L8 of H. parasuis OmpP2 induced the expression of proinflammatory cytokines possibly by activating the NF-κB and MAPK signalling pathways in cells infected by H. parasuis.
Collapse
Affiliation(s)
- Ye Zhou
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China
| | - Saixiang Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xinyi He
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China
| | - Qun Zhou
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China
| | - Yuanwei Wang
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China
| | - Hua Yue
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China.,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China
| | - Cheng Tang
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China.,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China
| | - Bin Zhang
- College of Life Science and Technology, Southwest Minzu University, Chengdu, 610041, China. .,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu, 610041, China.
| |
Collapse
|
30
|
Liu Y, Zhao W, Xu J, Yu X, Ye C, Fu S, Qiu Y. Pharmacokinetics of sodium baicalin following intravenous and intramuscular administration to piglets. J Vet Pharmacol Ther 2019; 42:580-584. [PMID: 31373697 DOI: 10.1111/jvp.12797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/31/2019] [Accepted: 06/23/2019] [Indexed: 12/29/2022]
Abstract
The purpose of this study was to determine the pharmacokinetics of baicalin after intravenous and intramuscular administration of sodium baicalin at 50 mg/kg to piglets. Plasma baicalin levels were determined by high-performance liquid chromatography. The plasma concentration-time data of baicalin for both administration routes were best described by two-compartmental open model. The area under the plasma concentration-time curve and the elimination half-lives were 77.47 ± 6.14 µg/ml × h and 1.73 ± 0.16 hr for intravenous and 64.85 ± 5.67 µg/ml × h and 2.42 ± 0.15 hr for intramuscular administration, respectively. The apparent volume of distribution and body clearance were 1.63 ± 0.23 L/kg and 2.74 ± 0.30 L h-1 kg-1 for intravenous and 0.51 ± 0.10 L/kg and 0.78 ± 0.08 L h-1 kg-1 for intramuscular routes, respectively. An intramuscular injection of sodium baicalin in piglets resulted in rapid and complete absorption, with a mean maximal plasma concentration of 77.28 ± 7.40 µg/ml at 0.17 hr and a high absolute bioavailability of 83.73 ± 5.53%.
Collapse
Affiliation(s)
- Yu Liu
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Wenhua Zhao
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Jianfeng Xu
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Xiaolin Yu
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Chun Ye
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Shulin Fu
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Yinsheng Qiu
- Hubei key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
31
|
Fu S, Zhao W, Xiong C, Guo L, Guo J, Qiu Y, Hu CAA, Ye C, Liu Y, Wu Z, Hou Y. Baicalin modulates apoptosis via RAGE, MAPK, and AP-1 in vascular endothelial cells during Haemophilus parasuis invasion. Innate Immun 2019; 25:420-432. [PMID: 31271085 PMCID: PMC6900640 DOI: 10.1177/1753425919856078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Glässer’s disease, caused by Haemophilus parasuis, is a chronic
disease related to an inflammatory immune response. Baicalin exerts important
biological functions. In this study, we explored the protective efficacy of
treatment with baicalin and the potential mechanism of activation of the MAPK
signaling pathway in porcine aortic vascular endothelial cells (PAVECs) induced
by H. parasuis. H. parasuis stimulated
expression of receptor for advanced glycation end products, induced a
significant increase in the level of protein kinase-α and protein kinase-δ
phosphorylation, and significantly up-regulated ERK, c-Jun N-terminal kinase,
and p38 phosphorylation in PAVECs. H. parasuis also
up-regulated the levels of apoptotic genes (Bax,
C-myc, and Fasl) and the expression levels
of c-Jun and c-Fos, and induced S-phase arrest in PAVECs. However, treatment
with baicalin inhibited expression of RAGE, suppressed H.
parasuis-induced protein kinase-α and protein kinase-δ
phosphorylation, reduced ERK, c-Jun N-terminal kinase, and p38 phosphorylation,
down-regulated apoptotic genes (Bax, C-myc,
and Fasl), attenuated phospho-c-Jun production from the
extracellular to the nuclei, and reversed S-phase arrest in PAVECs. In
conclusion, baicalin treatment inhibited the MAPK signaling pathway, thereby
achieving its anti-inflammatory responses, which provides a new strategy to
control H. parasuis infection.
Collapse
Affiliation(s)
- Shulin Fu
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Wenhua Zhao
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China
| | - Chunhong Xiong
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China
| | - Ling Guo
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Jing Guo
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Yinsheng Qiu
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Chien-An Andy Hu
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,3 Biochemistry and Molecular Biology, University of New Mexico School of Medicine, USA
| | - Chun Ye
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Yu Liu
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Zhongyuan Wu
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| | - Yongqing Hou
- 1 Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, PR China.,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, PR China
| |
Collapse
|
32
|
Luo Z, Xu X, Sho T, Luo W, Zhang J, Xu W, Yao J, Xu J. Effects of n-acetyl-cysteine supplementation in late gestational diet on maternal-placental redox status, placental NLRP3 inflammasome, and fecal microbiota in sows1. J Anim Sci 2019; 97:1757-1771. [PMID: 30789643 DOI: 10.1093/jas/skz058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 12/14/2022] Open
Abstract
Although n-acetyl-cysteine (NAC) has been shown to efficiently alleviate oxidative stress, inflammatory response, and alter gut microbiota, little attention has been focused on their interactions with placental metabolic status of sows. The effects of NAC on the placental redox status, function, inflammasome, and fecal microbiota in sows were explored to clarify the correlation between the fecal microbiota and placenta. Sows were divided into either the control group or the NAC group which received dietary 0.5% NAC supplementation from day 85 of gestation to delivery. Plasma redox status, placental growth factors, nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome, fecal microbial metabolites, and communities were evaluated. Compared with the control group, although NAC did not ameliorate reproductive performance of sows (P > 0.05), it significantly improved maternal-placental health, which was accompanied by increased activities of glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD), decreased level of malondialdehyde (MDA), and lowered expression of interleukin (IL)-1β and IL-18 through inhibiting NLRP3 inflammasome (P < 0.05). Additionally, NAC significantly increased placental insulin-like growth factors (IGFs) and E-cadherin contents (P < 0.05), elevated the expression of genes involved in angiogenesis and amino acids transporters (P < 0.05), and decreased the microtubule-associated protein light chain 3B (LC3B) and Beclin-1 protein expression (P < 0.05). Furthermore, NAC increased the relative abundances of fecal Prevotella, Clostridium cluster XIVa, and Roseburial/Eubacterium rectale (P < 0.05), which were negatively correlated with placental NLRP3 and positively with solute carrier family 7, member 8 (Slc7a8; P < 0.05). In conclusion, NAC supplementation during late gestation alleviated maternal-placental oxidative stress and inflammatory response, improved placental function, and altered fecal microbial communities.
Collapse
Affiliation(s)
- Zhen Luo
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Xu
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Takami Sho
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Wenli Luo
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhang
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Weina Xu
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jianbo Yao
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV
| | - Jianxiong Xu
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
33
|
Enhanced efficacy of baicalin-loaded TPGS polymeric micelles against periodontitis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:387-395. [PMID: 31029332 DOI: 10.1016/j.msec.2019.03.103] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 05/14/2018] [Accepted: 03/27/2019] [Indexed: 02/03/2023]
Abstract
As a chronic infectious disease, periodontitis is the main cause of teeth exfoliation due to its severe inflammatory reaction and periodontal tissue destruction. Recent reports have shown that baicalin could inhibit the NF-κB signaling pathway in inflammatory activity of periodontitis, but the efficacy of baicalin is limited due to its poor water solubility. In this work, we report the fabrication and application of baicalin encapsulated D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) polymeric micelles (PMs) through thin-film hydration method. The monodispersed micelles showed a spherical shape in aqueous solution and a prolonged drug-release kinetic. After baicalin was loaded into PMs, cytotoxicity and apoptosis induction were both decreased. The expression of genes (including TNF-α, IL-1β, RANKL and NF-κB) and the phosphorylation level of NF-κB p65 protein in lipopolysaccharide (LPS)-induced rat gingival fibroblasts were also reduced. Further investigation of drug efficacy in a rat periodontal disease model confirmed that the use of baicalin-PMs could reduce the destruction of alveolar bone and gingival fiber. Moreover, the therapeutic effect of baicalin-PMs was significantly better than that of free baicalin. These results suggest that the direct injection of micelles containing water-insoluble drugs may become a simple but effective method for treating periodontitis.
Collapse
|
34
|
Ye C, Li R, Xu L, Qiu Y, Fu S, Liu Y, Wu Z, Hou Y, Hu CAA. Effects of Baicalin on piglet monocytes involving PKC-MAPK signaling pathways induced by Haemophilus parasuis. BMC Vet Res 2019; 15:98. [PMID: 30909903 PMCID: PMC6434632 DOI: 10.1186/s12917-019-1840-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/07/2019] [Indexed: 12/11/2022] Open
Abstract
Background Haemophilus parasuis (HPS) is the causative agent of Glässer’s disease, characterized by arthritis, fibrinous polyserositis and meningitis, and resulting in worldwide economic losses in the swine industry. Baicalin (BA), a commonly used traditional Chinese medication, has been shown to possess a series of activities, such as anti-bacterial, anti-viral, anti-tumor, anti-oxidant and anti-inflammatory activities. However, whether BA has anti-apoptotic effects following HPS infection is unclear. Here, we investigated the anti-apoptotic effects and mechanisms of BA in HPS-induced apoptosis via the protein kinase C (PKC)–mitogen-activated protein kinase (MAPK) pathway in piglet’s mononuclear phagocytes (PMNP). Results Our data demonstrated that HPS could induce reactive oxygen species (ROS) production, arrest the cell cycle and promote apoptosis via the PKC–MAPK signaling pathway in PMNP. Moreover, when BA was administered, we observed a reduction in ROS production, suppression of cleavage of caspase-3 in inducing apoptosis, and inhibition of activation of the PKC–MAPK signaling pathway for down-regulating p-JNK, p-p38, p-ERK, p-PKC-α and PKC-δ in PMNP triggered by HPS. Conclusions Our data strongly suggest that BA can reverse the apoptosis initiated by HPS through regulating the PKC–MAPK signaling pathway, which represents a promising therapeutic agent in the treatment of HPS infection.
Collapse
Affiliation(s)
- Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Ruizhi Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Lei Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China. .,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China.
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China.,Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, People's Republic of China
| | - Chien-An Andy Hu
- Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| |
Collapse
|
35
|
Wang S, Yuan YH, Chen NH, Wang HB. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson's disease. Int Immunopharmacol 2019; 67:458-464. [DOI: 10.1016/j.intimp.2018.12.019] [Citation(s) in RCA: 324] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 01/15/2023]
|
36
|
Lai YS, Putra RBDS, Aui SP, Chang KT. M2 C Polarization by Baicalin Enhances Efferocytosis via Upregulation of MERTK Receptor. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:1899-1914. [PMID: 30518232 DOI: 10.1142/s0192415x18500957] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Baicalin is the main active ingredient primary isolated from the Chinese herb, Scutellaria baicalensis Georgi. Although baicalin can induce M2 macrophage polarization, we still do not know the subtype of macrophages polarized by baicalin. In this study, we characterized that murine bone marrow derived macrophages induced by M-CSF can be further polarized into M2C phenotype by baicalin. The signatures of M2C macrophages for mRNA expression like interferon regulatory factor 4 (IRF4), interleukin-10 (IL-10), MERTK and PTX3 were up-regulated. Moreover, we observed the concomitantly decreasing of tumor necrosis factor alpha (TNF- α ), interferon regulatory factor 5 (IRF5), IL-6. In contrast, M2 macrophages polarized by IL-4 increased gene transcript of arginase-1 (Arg-1) and surface marker of CD206 indicates that their identity as M2A rather than M2C subtypes. Interestingly, the phagocytosis as well as efferocytosis activity were significantly enhanced in M2C macrophage polarized by baicalin and these capacities were associated with the expression of MERTK receptor. Finally, we conclude that baicalin induced M2C macrophages polarization with both elevations of efferocytosis and anti-inflammatory activity.
Collapse
Affiliation(s)
- Yin-Siew Lai
- * Department of Biological Science and Technology, National Pingtung University of Science and Technology, 1, Shuefu Rd., Neipu, Pingtung 91201, Taiwan.,† Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, 1, Shuefu Rd., Neipu, Pingtung 91201, Taiwan
| | | | - Shin-Peir Aui
- * Department of Biological Science and Technology, National Pingtung University of Science and Technology, 1, Shuefu Rd., Neipu, Pingtung 91201, Taiwan
| | - Ko-Tung Chang
- * Department of Biological Science and Technology, National Pingtung University of Science and Technology, 1, Shuefu Rd., Neipu, Pingtung 91201, Taiwan.,† Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, 1, Shuefu Rd., Neipu, Pingtung 91201, Taiwan
| |
Collapse
|
37
|
Luo Z, Xu X, Sho T, Zhang J, Xu W, Yao J, Xu J. ROS-induced autophagy regulates porcine trophectoderm cell apoptosis, proliferation, and differentiation. Am J Physiol Cell Physiol 2018; 316:C198-C209. [PMID: 30485137 DOI: 10.1152/ajpcell.00256.2018] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Significant embryo loss remains a serious problem in pig production. Reactive oxygen species (ROS) play a critical role in embryonic implantation and placentation. However, the potential mechanism of ROS on porcine trophectoderm (pTr) cell fate during the peri-implantation period has not been investigated. This study aimed to elucidate the effects of ROS on pTr cell phenotypes and the regulatory role in cell attachment and differentiation. Herein, results showed that exogenous H2O2 inhibited pTr cell viability, arrested the cell cycle at S and G2/M phases, and increased cell apoptosis and autophagy protein light chain 3B and Beclin-1, whereas these effects were reversed by different concentrations of N-acetyl-l-cysteine (NAC) posttreatment. In addition, NAC abolished H2O2-induced autophagic flux, inhibited intracellular and mitochondrial ROS, and restored expression of genes important for mitochondrial DNA and biogenesis, cell attachment, and differentiation. NAC reversed H2O2-activated MAPK and Akt/mammalian target of rapamycin pathways in dose-dependent manners. Furthermore, analyses with pharmacological and RNA interference approaches suggested that autophagy regulated cell apoptosis and gene expression of caudal-related homeobox 2 and IL-1β. Collectively, these results provide new insights into the role of the ROS-induced autophagy in pTr cell apoptosis, attachment, and differentiation, indicating a promising target for decreasing porcine conceptus loss during the peri-implantation period.
Collapse
Affiliation(s)
- Zhen Luo
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology , Shanghai , China
| | - Xue Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology , Shanghai , China
| | - Takami Sho
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology , Shanghai , China
| | - Jing Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology , Shanghai , China
| | - Weina Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology , Shanghai , China
| | - Jianbo Yao
- Division of Animal and Nutritional Sciences, West Virginia University , Morgantown, West Virginia
| | - Jianxiong Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Veterinary Biotechnology , Shanghai , China
| |
Collapse
|
38
|
Ma M, Pei Y, Wang X, Feng J, Zhang Y, Gao MQ. LncRNA XIST mediates bovine mammary epithelial cell inflammatory response via NF-κB/NLRP3 inflammasome pathway. Cell Prolif 2018; 52:e12525. [PMID: 30362186 PMCID: PMC6430464 DOI: 10.1111/cpr.12525] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/16/2018] [Accepted: 08/10/2018] [Indexed: 12/26/2022] Open
Abstract
Objectives The correlations between long non‐coding RNAs (lncRNAs) and diverse mammal diseases have been clarified by many researches, but the cognition about bovine mastitis‐related lncRNAs remains limited. This study aimed to investigate the potential role of lncRNA X‐inactive specific transcript (XIST) in the inflammatory response of bovine mammary epithelial cells. Materials and methods Two inflammatory bovine mammary alveolar cell‐T (MAC‐T) models were established by infecting the cells with Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). The expressions of pro‐inflammatory cytokines were measured, and the proliferation, viability and apoptosis of the inflammatory cells were evaluated after XIST was knocked down by an siRNA. The relationship among XIST, NF‐κB pathway and NOD‐like receptor protein 3 (NLRP3) inflammasome was investigated using an inhibitor of NF‐κB signal pathway. Results The expression of XIST was abnormally increased in bovine mastitic tissues and inflammatory MAC‐T cells. Silencing of XIST significantly increased the expression of E. coli or S. aureus‐induced pro‐inflammatory cytokines. Additionally, knockdown of XIST could inhibit cell proliferation, suppress cell viability and promote cell apoptosis under inflammatory conditions. Furthermore, XIST inhibited E. coli or S. aureus‐induced NF‐κB phosphorylation and the production of NLRP3 inflammasome. Conclusions The expression of XIST was promoted by activated NF‐κB pathway and, in turn, XIST generated a negative feedback loop to regulate NF‐κB/NLRP3 inflammasome pathway for mediating the process of inflammation.
Collapse
Affiliation(s)
- Mengru Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yifei Pei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xixi Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jiaxin Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Ming-Qing Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| |
Collapse
|
39
|
Baicalin alleviates IL-1β-induced inflammatory injury via down-regulating miR-126 in chondrocytes. Biomed Pharmacother 2018; 99:184-190. [PMID: 29331857 DOI: 10.1016/j.biopha.2018.01.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/03/2018] [Accepted: 01/03/2018] [Indexed: 12/28/2022] Open
Abstract
Baicalin is a flavonoid extracted from Scutellaria baicalensis Georgi, with anti-inflammatory and anti-apoptotic activities. The objective of this study was to explore the effect and mechanism of baicalin on chondrocyte inflammatory response in OA. Different concentrations of IL-1β (0, 0.1, 2, 5 and 10 ng/mL) were used to simulate inflammatory injury in CHON-001 cells. The expression of miR-126 was altered by transfection with miR-126 mimic. Thereafter, cells were treated with baicalin, and cell viability, apoptosis, the expressions of apoptosis-related protein and pro-inflammatory factors were respectively detected using CCK-8 assay, flow cytometry, qRT-PCR and western blot analysis. We found that IL-1β induced a significantly inflammatory injury in CHON-001 cells. Baicalin alleviated IL-1β-induced inflammatory injury, as it increased cell viability, decreased cell apoptosis and repressed the production of IL-6, IL-8 and TNF-α. miR-126 was up-regulated by IL-1β treatment while was down-regulated by baicalin. More interestingly, the protective actions of baicalin on IL-1β-injured CHON-001 cells were partially eliminated by miR-126 overexpression. Further, NF-κB signaling pathway was activated by IL-1β, and deactivated by addition of baicalin. The deactivation of NF-κB signaling pathway induced by baicalin upon IL-1β exposure was recovered by miR-126 overexpression. In conclusion, this study demonstrated that baicalin protected CHON-001 cells against IL-1β-induced inflammatory injury possibly via down-regulation of miR-126 and thereby deactivation of NF-κB signaling pathway.
Collapse
|
40
|
Zang Y, Zhou X, Wang Q, Li X, Huang H. LncRNA FIRRE/NF-kB feedback loop contributes to OGD/R injury of cerebral microglial cells. Biochem Biophys Res Commun 2018; 501:131-138. [DOI: 10.1016/j.bbrc.2018.04.194] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 04/25/2018] [Indexed: 11/16/2022]
|
41
|
Fu S, Liu H, Chen X, Qiu Y, Ye C, Liu Y, Wu Z, Guo L, Hou Y, Hu CAA. Baicalin Inhibits Haemophilus Parasuis-Induced High-Mobility Group Box 1 Release during Inflammation. Int J Mol Sci 2018; 19:ijms19051307. [PMID: 29702580 PMCID: PMC5983759 DOI: 10.3390/ijms19051307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/04/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023] Open
Abstract
Haemophilus parasuis (H. parasuis) can cause Glässer’s disease in pigs. However, the molecular mechanism of the inflammation response induced by H. parasuis remains unclear. The high-mobility group box 1 (HMGB1) protein is related to the pathogenesis of various infectious pathogens, but little is known about whether H. parasuis can induce the release of HMGB1 in piglet peripheral blood monocytes. Baicalin displays important anti-inflammatory and anti-microbial activities. In the present study, we investigated whether H. parasuis can trigger the secretion of HMGB1 in piglet peripheral blood monocytes and the anti-inflammatory effect of baicalin on the production of HMGB1 in peripheral blood monocytes induced by H. parasuis during the inflammation response. In addition, host cell responses stimulated by H. parasuis were determined with RNA-Seq. The RNA-Seq results showed that H. parasuis infection provokes the expression of cytokines and the activation of numerous pathways. In addition, baicalin significantly reduced the release of HMGB1 in peripheral blood monocytes induced by H. parasuis. Taken together, our study showed that H. parasuis can induce the release of HMGB1 and baicalin can inhibit HMGB1 secretion in an H. parasuis-induced peripheral blood monocytes model, which may provide a new strategy for preventing the inflammatory disorders induced by H. parasuis.
Collapse
Affiliation(s)
- Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Huashan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Xiao Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China.
| | - Chien-An Andy Hu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
- Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| |
Collapse
|
42
|
You J, Cheng J, Yu B, Duan C, Peng J. Baicalin, a Chinese Herbal Medicine, Inhibits the Proliferation and Migration of Human Non-Small Cell Lung Carcinoma (NSCLC) Cells, A549 and H1299, by Activating the SIRT1/AMPK Signaling Pathway. Med Sci Monit 2018; 24:2126-2133. [PMID: 29632297 PMCID: PMC5909419 DOI: 10.12659/msm.909627] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Baicalin is a flavonoid derived from Scutellaria baicalensis, used in Chinese herbal medicine. Activation of the sirtuin 1 gene (SIRT1) and adenosine monophosphate (AMP)-activated protein kinase gene (AMPK), the SIRT1/AMPK signaling pathway, is associated with human malignant tumors. The aim of this study was to investigate the effects of baicalin on the cell viability, apoptosis, proliferation, and migration of human non-small cell lung cancer (NSCLC) cells, A549 and H1299, in vitro. Material/Methods Human NSCLC cells, A549 and H1299, were treated with serial doses of baicalin. Small interfering RNA (siRNA) silencing of the SIRT1 and AMPK genes was performed using cell transfection. The MTT assay was used to determine cell viability, flow cytometry was used to measure cell apoptosis, wound healing and transwell assays were used to assess cell migration of A549 and H1299 cells. Western blotting was used to measure protein expression and phosphorylation levels in untreated A549 and H1299 cells, and cells treated with increasing doses of baicalin. Results Baicalin inhibited the viability, migration, and invasion of A549 and H1299 cells, and increased cell apoptosis in a dose-dependent manner. Baicalin activated the SIRT1/AMPK and mechanistic target of rapamycin (mTOR), and SIRT1/AMPK and matrix metalloproteinase (MMP) signaling in A549 and H1299 cells in a dose-dependent manner. siRNA silencing of SIRT1 and AMPK reduced the effects of baicalin on cell proliferation and migration. Conclusions Baicalin, a flavonoid used in Chinese herbal medicine, inhibited the proliferation and migration of human NSCLC cells, A549 and H1299, by activating the SIRT1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Jiawen You
- Department of Thoracic Surgery, Fenghua Peoples' Hospital of Ningbo City, Ningbo, Zhejiang, China (mainland)
| | - Jun Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Bing Yu
- Department of Thoracic Surgery, Fenghua Peoples' Hospital of Ningbo City, Ningbo, Zhejiang, China (mainland)
| | - Changhua Duan
- Department of Thoracic Surgery, Fenghua Peoples' Hospital of Ningbo City, Ningbo, Zhejiang, China (mainland)
| | - Jinghua Peng
- Department of Thoracic Surgery, Fenghua Peoples' Hospital of Ningbo City, Ningbo, Zhejiang, China (mainland)
| |
Collapse
|
43
|
Dai K, He L, Chang YF, Cao S, Zhao Q, Huang X, Wu R, Huang Y, Yan Q, Han X, Ma X, Wen X, Wen Y. Basic Characterization of Natural Transformation in a Highly Transformable Haemophilus parasuis Strain SC1401. Front Cell Infect Microbiol 2018; 8:32. [PMID: 29473023 PMCID: PMC5809987 DOI: 10.3389/fcimb.2018.00032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/25/2018] [Indexed: 11/13/2022] Open
Abstract
Haemophilus parasuis causes Glässer's disease and pneumonia, incurring serious economic losses in the porcine industry. In this study, natural competence was investigated in H. parasuis. We found competence genes in H. parasuis homologous to ones in Haemophilus influenzae and a high consensus battery of Sxy-dependent cyclic AMP (cAMP) receptor protein (CRP-S) regulons using bioinformatics. High rates of natural competence were found from the onset of stationary-phase growth condition to mid-stationary phase (OD600 from 0.29 to 1.735); this rapidly dropped off as cells reached mid-stationary phase (OD600 from 1.735 to 1.625). As a whole, bacteria cultured in liquid media were observed to have lower competence levels than those grown on solid media plates. We also revealed that natural transformation in this species is stable after 200 passages and is largely dependent on DNA concentration. Transformation competition experiments showed that heterogeneous DNA cannot outcompete intraspecific natural transformation, suggesting an endogenous uptake sequence or other molecular markers may be important in differentiating heterogeneous DNA. We performed qRT-PCR targeting multiple putative competence genes in an effort to compare bacteria pre-cultured in TSB++ vs. TSA++ and SC1401 vs. SH0165 to determine expression profiles of the homologs of competence-genes in H. influenzae. Taken together, this study is the first to investigate natural transformation in H. parasuis based on a highly naturally transformable strain SC1401.
Collapse
Affiliation(s)
- Ke Dai
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lvqin He
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Sanjie Cao
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
| | - Qin Zhao
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaobo Huang
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rui Wu
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qigui Yan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoping Ma
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xintian Wen
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center of Swine Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
44
|
Baicalin modulates NF-κB and NLRP3 inflammasome signaling in porcine aortic vascular endothelial cells Infected by Haemophilus parasuis Causing Glässer's disease. Sci Rep 2018; 8:807. [PMID: 29339754 PMCID: PMC5770393 DOI: 10.1038/s41598-018-19293-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022] Open
Abstract
Haemophilus parasuis (H. parasuis) can cause vascular inflammatory injury, but the molecular basis of this effect remains unclear. In this study,we investigated the effect of the anti-inflammatory, anti-microbial and anti-oxidant agent, baicalin, on the nuclear factor (NF)-κB and NLRP3 inflammasome signaling pathway in pig primary aortic vascular endothelial cells. Activation of the NF-κB and NLRP3 inflammasome signaling pathway was induced in H. parasuis-infected cells. However, baicalin reduced the production of reactive oxygen species, apoptosis, and activation of the NF-κB and NLRP3 inflammasome signaling pathway in infected cells. These results revealed that baicalin can inhibit H. parasuis-induced inflammatory responses in porcine aortic vascular endothelial cells, and may thus offer a novel strategy for controlling and treating H. parasuis infection. Furthermore, the results suggest that piglet primary aortic vascular endothelial cells may provide an experimental model for future studies of H. parasuis infection.
Collapse
|
45
|
Zhang J, Zhang H, Deng X, Zhang Y, Xu K. Baicalin protects AML-12 cells from lipotoxicity via the suppression of ER stress and TXNIP/NLRP3 inflammasome activation. Chem Biol Interact 2017; 278:189-196. [DOI: 10.1016/j.cbi.2017.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/28/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023]
|
46
|
Liu S, Li W, Wang Y, Gu C, Liu X, Charreyre C, Fan S, He Q. Coinfection with Haemophilus parasuis serovar 4 increases the virulence of porcine circovirus type 2 in piglets. Virol J 2017; 14:227. [PMID: 29157279 PMCID: PMC5696968 DOI: 10.1186/s12985-017-0890-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 11/02/2017] [Indexed: 11/26/2022] Open
Abstract
Background Postweaning multisystemic wasting syndrome (PMWS) is an emerging disease in swine. Pigs with PMWS are often infected with a variety of other pathogens, including bacteria, viruses and mycoplasm, in addition to porcine circovirus type 2 (PCV2). PCV2 and Haemophilus parasuis serovar 4 (HPS4) coinfection remain epidemic in China. Methods Here we report construction of a three-week-old naturally farrowed, colostrum-deprived (NFCD) piglet’s infection model and demonstrate that PCV2-infected piglets with the HPS4 coinfection increased the virulence of PCV2 and these pathogens interact acquired PMWS. Results All the single infected piglets were transiently bacteremic or viremic. All the PCV2/HPS4 coinfected piglets developed PMWS, characterized by dyspnea, anorexia, prostration and lose weight severely. Co-infection with PCV2 and HPS4 resulted in an increased amount of virus in serum and tissues, presented a slower generation and lower levels of antibodies against PCV2. Co-infection with PCV2 and HPS4 resulted in further reductions in total and differential peripheral blood leukocyte counts. Meantime, PCV2/ HPS4 coinfection potentiated the severity of lung and lymphoid lesions by PCV2-associated, increased the virulence of PCV2-antigen and enhanced the incidence of PMWS in piglets. Conclusion Co-infection with PCV2 and HPS4 induce the exacerbation of system injuries and enhance the pathogenicity of PCV2 in piglets.
Collapse
Affiliation(s)
- Shuqing Liu
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.,Key Laboratory of Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yang Wang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Changqin Gu
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Xiaoli Liu
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | | | - Shenxian Fan
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China. .,Department of Animal Infectious Disease, State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
47
|
Bai C, Li T, Sun Q, Xin Q, Xu T, Yu J, Wang Y, Wei L. Protective effect of baicalin against severe burn‑induced remote acute lung injury in rats. Mol Med Rep 2017; 17:2689-2694. [PMID: 29207058 DOI: 10.3892/mmr.2017.8120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 10/03/2017] [Indexed: 11/05/2022] Open
Abstract
Baicalin exhibits antibacterial, anti‑viral, anti‑oxidative, antipyretic, analgesic, anti‑inflammatory and anti‑tumor properties. The chemical scavenges oxygen free radicals, protects the cardiovascular system and neurons, protects the liver, and has been used for the prevention and treatment of diabetes‑associated complications. The present study investigated the effect of baicalin on severe burn‑induced remote acute lung injury (ALI). The present study demonstrated that baicalin significantly decreased the lung wet‑to‑dry weight ratio, improved pulmonary histological alterations and reduced the expression of high mobility group protein B1 in the rat model of ALI. In addition, treatment with baicalin decreased tumor necrosis factor‑α, interleukin (IL)‑8, IL‑1β and IL‑18 concentrations in the serum, reduced myeloperoxidase activity and malondialdehyde content, and increased the level of superoxide dismutase in the serum in treated model rats with ALI. As a result, baicalin significantly suppressed nucleotide‑binding oligomerization, NACHT, LRR and PYD domains‑containing protein 3 (NLRP3), caspase‑1, nuclear factor‑κB and matrix metalloproteinase‑9 protein expression in the rat model of ALI. The results of the present study suggested that baicalin may serve a protective role against ALI in rats through the NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Chongfeng Bai
- Postdoctoral Research Station of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Tao Li
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Qing Sun
- Department of Hyperbaric Medicine, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Qun Xin
- Department of General Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Tongyi Xu
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Jinjian Yu
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Yun Wang
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Li Wei
- Postdoctoral Research Station of Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
48
|
Zhang Q, Sun J, Wang Y, He W, Wang L, Zheng Y, Wu J, Zhang Y, Jiang X. Antimycobacterial and Anti-inflammatory Mechanisms of Baicalin via Induced Autophagy in Macrophages Infected with Mycobacterium tuberculosis. Front Microbiol 2017; 8:2142. [PMID: 29163427 PMCID: PMC5673628 DOI: 10.3389/fmicb.2017.02142] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis (TB) remains a leading killer worldwide among infectious diseases and the effective control of TB is still challenging. Autophagy is an intracellular self-digestion process which has been increasingly recognized as a major host immune defense mechanism against intracellular microorganisms like Mycobacterium tuberculosis (Mtb) and serves as a key negative regulator of inflammation. Clinically, chronic inflammation surrounding Mtb can persist for decades leading to lung injury that can remain even after successful treatment. Adjunct host-directed therapy (HDT) based on both antimycobacterial and anti-inflammatory interventions could be exploited to improve treatment efficacy and outcome. Autophagy occurring in the host macrophages represents a logical host target. Here, we show that herbal medicine, baicalin, could induce autophagy in macrophage cell line Raw264.7 and caused increased killing of intracellular Mtb. Further, baicalin inhibited Mtb-induced NLRP3 inflammasome activation and subsequent inflammasome-derived IL-1β. To investigate the molecular mechanisms of baicalin, the signaling pathways associated with autophagy were examined. Results indicated that baicalin decreased the levels of phosphorylated protein kinase B (p-Akt) and phosphorylated mammalian target of rapamycin (p-mTOR) at Ser473 and Ser2448, respectively, but did not alter the phosphorylation of p38, JNK, or ERK both in Raw264.7 and primary peritoneal macrophages. Moreover, baicalin exerted an obvious inhibitory effect on nuclear factor-kappa B (NF-κB) activity. Finally, immunofluorescence studies demonstrated that baicalin promoted the co-localization of inflammasome with autophagosome may serve as the underlying mechanism of autophagic degradative effect on reducing inflammasome activation. Together, baicalin definitely induces the activation of autophagy on the Mtb-infected macrophages through PI3K/Akt/mTOR pathway instead of MAPK pathway. Furthermore, baicalin inhibited the PI3K/Akt/NF-κB signal pathway, and both autophagy induction and NF-κB inhibition contribute to limiting the NLRP3 inflammasome as well as subsequent production of pro-inflammatory cytokine IL-1β. Based on these results, we conclude that baicalin is a promising antimycobacterial and anti-inflammatory agent which can be a novel candidate for the development of new adjunct drugs targeting HDT for possible improved treatment.
Collapse
Affiliation(s)
- Qingwen Zhang
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinxia Sun
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuli Wang
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weigang He
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lixin Wang
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuejuan Zheng
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Wu
- Department of Infectious Diseases, Institute of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Xin Jiang
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
49
|
Li CG, Yan L, Mai FY, Shi ZJ, Xu LH, Jing YY, Zha QB, Ouyang DY, He XH. Baicalin Inhibits NOD-Like Receptor Family, Pyrin Containing Domain 3 Inflammasome Activation in Murine Macrophages by Augmenting Protein Kinase A Signaling. Front Immunol 2017; 8:1409. [PMID: 29163487 PMCID: PMC5674921 DOI: 10.3389/fimmu.2017.01409] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/11/2017] [Indexed: 01/24/2023] Open
Abstract
The flavonoid baicalin has been reported to possess potent anti-inflammatory activities by suppressing inflammatory signaling pathways. However, whether baicalin can suppress the activation of NOD-like receptor (NLR) family, pyrin containing domain 3 (NLRP3) inflammasome in macrophages is largely unknown. Here, we showed that baicalin treatment dose-dependently inhibited adenosine triphosphate (ATP) or nigericin-induced NLRP3 inflammasome activation, as revealed by the decreased release of mature interleukin (IL)-1β, active caspase-1p10, and high-mobility group box-1 protein from lipopolysaccharide (LPS)-primed bone marrow-derived macrophages. The formation of ASC specks, a critical marker of NLRP3 inflammasome assembly, was robustly inhibited by baicalin in the macrophages upon ATP or nigericin stimulation. All these inhibitory effects of baicalin could be partly reversed by MDL12330A or H89, both of which are inhibitors of the protein kinase A (PKA) signaling pathway. Consistent with this, baicalin strongly enhanced PKA-mediated phosphorylation of NLRP3, which has been suggested to prevent ASC recruitment into the inflammasome. Of note, the PKA inhibitor H89 could block baicalin-induced NLRP3 phosphorylation on PKA-specific sites, further supporting PKA's role in this process. In addition, we showed that when administered pre and post exposure to Escherichia coli infection baicalin treatment significantly improved mouse survival in bacterial sepsis. Baicalin administration also significantly reduced IL-1β levels in the sera of bacterial infected mice. Altogether, our results revealed that baicalin inhibited NLRP3 inflammasome activation at least partly through augmenting PKA signaling, highlighting its therapeutic potential for the treatment of NLRP3-related inflammatory diseases.
Collapse
Affiliation(s)
- Chen-Guang Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Liang Yan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Feng-Yi Mai
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zi-Jian Shi
- Department of Fetal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li-Hui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yan-Yun Jing
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qing-Bing Zha
- Department of Fetal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dong-Yun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
50
|
Fu S, Liu L, Han L, Yu Y. Leptin promotes IL-18 secretion by activating the NLRP3 inflammasome in RAW 264.7 cells. Mol Med Rep 2017; 16:9770-9776. [DOI: 10.3892/mmr.2017.7797] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 08/31/2017] [Indexed: 11/05/2022] Open
|