1
|
Sadeghloo Z, Nabavi-Rad A, Zali MR, Klionsky DJ, Yadegar A. The interplay between probiotics and host autophagy: mechanisms of action and emerging insights. Autophagy 2024:1-23. [PMID: 39291740 DOI: 10.1080/15548627.2024.2403277] [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: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024] Open
Abstract
Autophagy, a lysosome-dependent protein degradation mechanism, is a highly conserved catabolic process seen in all eukaryotes. This cell protection system, which is present in all tissues and functions at a basic level, can be up- or downregulated in response to various stresses. A disruption in the natural route of the autophagy process is frequently followed by an interruption in the inherent operation of the body's cells and organs. Probiotics are live bacteria that protect the host through various mechanisms. One of the processes through which probiotics exert their beneficial effects on various cells and tissues is autophagy. Autophagy can assist in maintaining host homeostasis by stimulating the immune system and affecting numerous physiological and pathological responses. In this review, we particularly focus on autophagy impairments occurring in several human illnesses and investigate how probiotics affect the autophagy process under various circumstances.Abbreviation: AD: Alzheimer disease; AKT: AKT serine/threonine kinase; AMPK: 5'AMP-activated protein kinase; ATG: autophagy related; CCl4: carbon tetrachloride; CFS: cell-free supernatant; CMA: chaperone-mediated autophagy; CRC: colorectal cancer; EPS: L. plantarum H31 exopolysaccharide; HD: Huntington disease; HFD: high-fat diet; HPV: human papillomavirus; IFNG/IFN-γ: interferon gamma; IL6: interleukin 6; LGG: L. rhamnosus GG; LPS: lipopolysaccharide; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; PD: Parkinson disease; Pg3G: pelargonidin-3-O-glucoside; PI3K: phosphoinositide 3-kinase; PolyQ: polyglutamine; ROS: reactive oxygen species; SCFAs: short-chain fatty acids; SLAB51: a novel formulation of lactic acid bacteria and bifidobacteria; Slp: surface layer protein (of acidophilus NCFM); SNCA: synuclein alpha; ULK1: unc-51 like autophagy-activating kinase 1; YB: B. longum subsp. infantis YB0411; YFP: yeast fermentate prebiotic.
Collapse
Affiliation(s)
- Zahra Sadeghloo
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Pessino G, Scotti C, Maggi M, Immuno-Hub Consortium. Hepatocellular Carcinoma: Old and Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:901. [PMID: 38473265 DOI: 10.3390/cancers16050901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Liver cancer, predominantly hepatocellular carcinoma (HCC), globally ranks sixth in incidence and third in cancer-related deaths. HCC risk factors include non-viral hepatitis, alcohol abuse, environmental exposures, and genetic factors. No specific genetic alterations are unequivocally linked to HCC tumorigenesis. Current standard therapies include surgical options, systemic chemotherapy, and kinase inhibitors, like sorafenib and regorafenib. Immunotherapy, targeting immune checkpoints, represents a promising avenue. FDA-approved checkpoint inhibitors, such as atezolizumab and pembrolizumab, show efficacy, and combination therapies enhance clinical responses. Despite this, the treatment of hepatocellular carcinoma (HCC) remains a challenge, as the complex tumor ecosystem and the immunosuppressive microenvironment associated with it hamper the efficacy of the available therapeutic approaches. This review explores current and advanced approaches to treat HCC, considering both known and new potential targets, especially derived from proteomic analysis, which is today considered as the most promising approach. Exploring novel strategies, this review discusses antibody drug conjugates (ADCs), chimeric antigen receptor T-cell therapy (CAR-T), and engineered antibodies. It then reports a systematic analysis of the main ligand/receptor pairs and molecular pathways reported to be overexpressed in tumor cells, highlighting their potential and limitations. Finally, it discusses TGFβ, one of the most promising targets of the HCC microenvironment.
Collapse
Affiliation(s)
- Greta Pessino
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Claudia Scotti
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maristella Maggi
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Immuno-Hub Consortium
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
3
|
Lee IT, Yang CC, Yang CM. Harnessing peroxisome proliferator-activated receptor γ agonists to induce Heme Oxygenase-1: a promising approach for pulmonary inflammatory disorders. Cell Commun Signal 2024; 22:125. [PMID: 38360670 PMCID: PMC10868008 DOI: 10.1186/s12964-024-01501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/27/2024] [Indexed: 02/17/2024] Open
Abstract
The activation of peroxisome proliferator-activated receptor (PPAR)-γ has been extensively shown to attenuate inflammatory responses in conditions such as asthma, acute lung injury, and acute respiratory distress syndrome, as demonstrated in animal studies. However, the precise molecular mechanisms underlying these inhibitory effects remain largely unknown. The upregulation of heme oxygenase-1 (HO-1) has been shown to confer protective effects, including antioxidant, antiapoptotic, and immunomodulatory effects in vitro and in vivo. PPARγ is highly expressed not only in adipose tissues but also in various other tissues, including the pulmonary system. Thiazolidinediones (TZDs) are highly selective agonists for PPARγ and are used as antihyperglycemic medications. These observations suggest that PPARγ agonists could modulate metabolism and inflammation. Several studies have indicated that PPARγ agonists may serve as potential therapeutic candidates in inflammation-related diseases by upregulating HO-1, which in turn modulates inflammatory responses. In the respiratory system, exposure to external insults triggers the expression of inflammatory molecules, such as cytokines, chemokines, adhesion molecules, matrix metalloproteinases, and reactive oxygen species, leading to the development of pulmonary inflammatory diseases. Previous studies have demonstrated that the upregulation of HO-1 protects tissues and cells from external insults, indicating that the induction of HO-1 by PPARγ agonists could exert protective effects by inhibiting inflammatory signaling pathways and attenuating the development of pulmonary inflammatory diseases. However, the mechanisms underlying TZD-induced HO-1 expression are not well understood. This review aimed to elucidate the molecular mechanisms through which PPARγ agonists induce the expression of HO-1 and explore how they protect against inflammatory and oxidative responses.
Collapse
Affiliation(s)
- I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, 333008, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, 242062, Taiwan.
| |
Collapse
|
4
|
Yang CC, Lee IT, Lin YJ, Wu WB, Hsiao LD, Yang CM. Thrombin-Induced COX-2 Expression and PGE 2 Synthesis in Human Tracheal Smooth Muscle Cells: Role of PKCδ/Pyk2-Dependent AP-1 Pathway Modulation. Int J Mol Sci 2023; 24:15130. [PMID: 37894811 PMCID: PMC10606820 DOI: 10.3390/ijms242015130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
In this study, we confirmed that thrombin significantly increases the production of COX-2 and PGE2 in human tracheal smooth muscle cells (HTSMCs), leading to inflammation in the airways and lungs. These molecules are well-known contributors to various inflammatory diseases. Here, we investigated in detail the involved signaling pathways using specific inhibitors and small interfering RNAs (siRNAs). Our results demonstrated that inhibitors targeting proteins such as protein kinase C (PKC)δ, proline-rich tyrosine kinase 2 (Pyk2), c-Src, epidermal growth factor receptor (EGFR), phosphatidylinositol 3-kinase (PI3K), or activator protein-1 (AP-1) effectively reduced thrombin-induced COX-2 and PGE2 production. Additionally, transfection with siRNAs against PKCδ, Pyk2, c-Src, EGFR, protein kinase B (Akt), or c-Jun mitigated these responses. Furthermore, our observations revealed that thrombin stimulated the phosphorylation of key components of the signaling cascade, including PKCδ, Pyk2, c-Src, EGFR, Akt, and c-Jun. Thrombin activated COX-2 promoter activity through AP-1 activation, a process that was disrupted by a point-mutated AP-1 site within the COX-2 promoter. Finally, resveratrol (one of the most researched natural polyphenols) was found to effectively inhibit thrombin-induced COX-2 expression and PGE2 release in HTSMCs through blocking the activation of Pyk2, c-Src, EGFR, Akt, and c-Jun. In summary, our findings demonstrate that thrombin-induced COX-2 and PGE2 generation involves a PKCδ/Pyk2/c-Src/EGFR/PI3K/Akt-dependent AP-1 activation pathway. This study also suggests the potential use of resveratrol as an intervention for managing airway inflammation.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Taoyuan, Taoyuan 333008, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan;
| | - Yan-Jyun Lin
- Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung 406040, Taiwan;
| | - Wen-Bin Wu
- School of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| | - Li-Der Hsiao
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| |
Collapse
|
5
|
Gojani EG, Wang B, Li DP, Kovalchuk O, Kovalchuk I. Anti-Inflammatory Effects of Minor Cannabinoids CBC, THCV, and CBN in Human Macrophages. Molecules 2023; 28:6487. [PMID: 37764262 PMCID: PMC10534668 DOI: 10.3390/molecules28186487] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Inflammation is a natural response of the body to signals of tissue damage or infection caused by pathogens. However, when it becomes imbalanced, it can lead to various disorders such as cancer, obesity, cardiovascular problems, neurological conditions, and diabetes. The endocannabinoid system, which is present throughout the body, plays a regulatory role in different organs and influences functions such as food intake, pain perception, stress response, glucose tolerance, inflammation, cell growth and specialization, and metabolism. Phytocannabinoids derived from Cannabis sativa can interact with this system and affect its functioning. In this study, we investigate the mechanisms underlying the anti-inflammatory effects of three minor phytocannabinoids including tetrahydrocannabivarin (THCV), cannabichromene (CBC), and cannabinol (CBN) using an in vitro system. We pre-treated THP-1 macrophages with different doses of phytocannabinoids or vehicle for one hour, followed by treating the cells with 500 ng/mL of LPS or leaving them untreated for three hours. To induce the second phase of NLRP3 inflammasome activation, LPS-treated cells were further treated with 5 mM ATP for 30 min. Our findings suggest that the mitigation of the PANX1/P2X7 axis plays a significant role in the anti-inflammatory effects of THCV and CBC on NLRP3 inflammasome activation. Additionally, we observed that CBC and THCV could also downregulate the IL-6/TYK-2/STAT-3 pathway. Furthermore, we discovered that CBN may exert its inhibitory impact on the assembly of the NLRP3 inflammasome by reducing PANX1 cleavage. Interestingly, we also found that the elevated ADAR1 transcript responded negatively to THCV and CBC in LPS-macrophages, indicating a potential involvement of ADAR1 in the anti-inflammatory effects of these two phytocannabinoids. THCV and CBN inhibit P-NF-κB, downregulating proinflammatory gene transcription. In summary, THCV, CBC, and CBN exert anti-inflammatory effects by influencing different stages of gene expression: transcription, post-transcriptional regulation, translation, and post-translational regulation.
Collapse
Affiliation(s)
| | | | | | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.-P.L.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.-P.L.)
| |
Collapse
|
6
|
Chi MC, Lin ZC, Lee CW, Huang CCY, Peng KT, Lin CM, Lee HC, Fang ML, Chiang YC. Tanshinone IIA suppresses burning incense-induced oxidative stress and inflammatory pathways in astrocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 258:114987. [PMID: 37172407 DOI: 10.1016/j.ecoenv.2023.114987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
The burning incense (BI) behavior could be widely observed in Asia families. Incense sticks are often believed to be made from natural herbs and powders, and to have minimal impact on human health; however, there is limited research to support this claim. The current study aimed to identify the components of BI within the particulate matter 2.5 µm (PM2.5) range and explore if BI has bio-toxicity effects on rat astrocytes (CTX-TNA2). The study also examined the protective effects and underlying molecular mechanisms of tanshinone IIA, a primary lipid-soluble compound found in the herb danshen (Salvia miltiorrhiza Bunge), which has been shown to benefit the central nervous system. Results showed that despite the differences in BI components compared to the atmospheric particulate matter (PM) standards, BI still had a bio-toxicity on astrocytes. BI exposure caused early and late apoptosis, reactive oxygen species (ROS) production, MAPKs (JNK, p38, and ERK), and Akt signaling activation, and inflammation-related proteins (cPLA2, COX-2, HO-1, and MMP-9) increases. Our results further exhibit that the tanshinone IIA pre-treatment could significantly avoid the BI-induced apoptosis and inflammatory signals on rat astrocytes. These findings suggest that BI exposure may cause oxidative stress in rat astrocytes and increase inflammation-related proteins and support the potential of tanshinone IIA as a candidate for preventing BI-related adverse health effects.
Collapse
Affiliation(s)
- Miao-Ching Chi
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City 243, Taiwan; Division of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan
| | - Zih-Chan Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan
| | - Chiang-Wen Lee
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 61363, Taiwan
| | | | - Kuo-Ti Peng
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 61363, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chieh-Mo Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Division of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hui-Chun Lee
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan
| | - Mei-Ling Fang
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan; Super Micro Research and Technology Center, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Yao-Chang Chiang
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan; Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan.
| |
Collapse
|
7
|
Lu Q, Guo Y, Yang G, Cui L, Wu Z, Zeng X, Pan D, Cai Z. Structure and Anti-Inflammation Potential of Lipoteichoic Acids Isolated from Lactobacillus Strains. Foods 2022; 11:foods11111610. [PMID: 35681360 PMCID: PMC9180668 DOI: 10.3390/foods11111610] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 12/31/2022] Open
Abstract
Lactobacillus are normal inhabitants of the gastrointestinal tract and confer a variety of health effects. Lipoteichoic acid (LTA), an amphiphilic substance located in the cell membrane, is a key molecule in probiotic–host crosstalk. Through the characterization of structural characteristics of LTA molecules derived from Lactobacillus plantarum A3, Lactobacillus reuteri DMSZ 8533, and Lactobacillus acidophilus CICC 6074, there exists some heterogeneity in LTA molecules, which perhaps contributes to the distinguishable adhesion properties of Lactobacillus strains based on fluorescence microscopy observations. In LPS-induced RAW 264.7 cells, LTAs derived from three Lactobacillus strains obviously alleviated inflammatory responses as evidenced by the altered inflammatory cytokine levels of TNF-α, IL-6, and IL-10. Western blotting demonstrated that L. reuteri LTA blocked LPS-triggered expression of the MAPK and NF-κB pathways. The findings further validated that LTA is an important effector molecule and deserves further consideration as an alternative therapeutic for ulcerative colitis treatment.
Collapse
Affiliation(s)
- Qianqian Lu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
| | - Yingqi Guo
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
| | - Guo Yang
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
| | - Lei Cui
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
| | - Zhen Wu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China
| | - Xiaoqun Zeng
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China
| | - Daodong Pan
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China
| | - Zhendong Cai
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China; (Q.L.); (Y.G.); (G.Y.); (L.C.); (Z.W.); (X.Z.); (D.P.)
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China
- Correspondence:
| |
Collapse
|
8
|
Cheng CY, Chen YH, Thuy Tien Vo T, Chui Hong Y, Wang CS, Canh Vo Q, Chou HC, Huang TW, Lee IT. CORM-2 prevents human gingival fibroblasts from lipoteichoic acid-induced VCAM-1 and ICAM-1 expression by inhibiting TLR2/MyD88/TRAF6/PI3K/Akt/ROS/NF-κB signaling pathway. Biochem Pharmacol 2022; 201:115099. [PMID: 35617999 DOI: 10.1016/j.bcp.2022.115099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/02/2022]
Abstract
Periodontal diseases are prevalent worldwide. Lipoteichoic acid (LTA), a major component of gram-positive bacteria, may play a key role in periodontally inflammatory diseases. Carbon monoxide (CO) is a critical messenger in many biological processes. It can elicit various biological properties, especially anti-inflammatory effects. As the straight administration of CO remains difficult, CO-releasing molecules (CO-RMs) are emerging as promising alternatives. To explore the pharmacological actions and signaling pathways of CO battling LTA-induced periodontal inflammation, this study investigated the cytoprotective effects of CORM-2 against the adhesion of THP-1 monocytes to human gingival fibroblasts (HGFs) and the underlying molecular mechanism. After exposing HGFs to LTA with or without CORM-2 pretreatment, monocyte adhesion was determined. VCAM-1 and ICAM-1 expression in HGFs was measured by real-time PCR. To identify the signaling pathways of CO involved in the cytoprotective effects of CORM-2, HGFs underwent pharmacological or genetical interventions before LTA incubation. The expression and/or activity of possible regulatory molecules were determined. The release of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, were measured using ELISA. The results showed that LTA increased cytokine production and upregulated VCAM-1 and ICAM-1 expression in HGFs, promoting monocyte adhesion. These events were dependent on TLR2/MyD88/TRAF6- and PI3K/Akt/NADPH oxidase/ROS-regulated NF-κB activation. CORM-2 inhibited LTA-induced inflammatory cascades in HGFs, in which CO seemed to be the hitman. To conclude, CO released from CORM-2 can prevent the LTA-stimulated HGFs from increasing VCAM-1 and ICAM-1 expression and promoting monocyte adhesion by inhibiting TLR2/MyD88/TRAF6 association and PI3K/Akt/NADPH oxidase/ROS signaling, both converge on the canonical NF-κB activation.
Collapse
Affiliation(s)
- Ching-Yi Cheng
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, No. 261, Wenhua 1st Rd., Guishan Dist., Taoyuan City 333, Taiwan; Department of Pulmonary Infection and Immunology, Chang Gung Memorial Hospital at Linkou, No. 5, Fuxing St., Guishan Dist., Taoyuan City 333, Taiwan
| | - Yu-Hsu Chen
- Department of Orthopedic surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan; Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Thi Thuy Tien Vo
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying Chui Hong
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Quang Canh Vo
- Department of Dental Biomaterials Science, Dental Research Institute and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Han-Chin Chou
- Department of Chinese Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Ting-Wei Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Network Pharmacological Analysis and Experimental Study of the Antipharyngitis Mechanism of the Chaiqin Qingning Capsule. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5616942. [PMID: 35528163 PMCID: PMC9071881 DOI: 10.1155/2022/5616942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
Objective The study aimed to explore the active composition and mechanism of the Chaiqin Qingning capsule (CQQN) against pharyngitis based on the network pharmacology and through using a pharyngitis rat model. Methods The active ingredients and targets of CQQN were queried using the TCMSP database. Disease-related target genes were queried in the DrugBank, GeneCards, OMIM, and DisGeNEt databases using “pharyngitis” as the search term. The STRING database was used to establish a protein-protein interaction (PPI) network. GO function enrichment and KEGG pathway enrichment analyses were performed to identify active components and key targets. Cytoscape software (version 3.7.2) was used to construct an active component/target gene/enrichment pathway network. AutoDock software was used to select the best binding target for molecular docking. The effect of CQQN was verified in the pharyngitis rats. Results Network pharmacology showed 30 active compounds in CQQN with 240 targets, including 54 for the treatment of pharyngitis. Potential active ingredients included quercetin, kaempferol, stigmasterol, saikosaponin D, and isorhamnetin. The key targets involved were AKT1, TNF, IL-6, and IL-1β. Signaling pathways included virus infection, TNF, IL-17, and cancer pathways. The molecular docking results showed that the critical components in CQQN had good potential for binding to key target genes. Animal experiments showed that CQQN could significantly reduce the expression of TNF-α, IL-1β, IL-6, and IL-17 in the serum of rats with pharyngitis (P < 0.05). Hematoxylin and eosin staining showed that the inflammatory state of pharyngeal tissue in rats was significantly reduced compared to that in the model group. Conclusion CQQN can improve pharyngitis by regulating the TNF and IL-17 signaling pathways. The study makes a positive exploration and provides a new idea for a more comprehensive and in-depth excavation of CQQN with an intervention effect on pharyngitis and other upper respiratory diseases in the future.
Collapse
|
10
|
Thrombin Induces COX-2 and PGE2 Expression via PAR1/PKCalpha/MAPK-Dependent NF-kappaB Activation in Human Tracheal Smooth Muscle Cells. Mediators Inflamm 2022; 2022:4600029. [PMID: 35497094 PMCID: PMC9042634 DOI: 10.1155/2022/4600029] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/03/2022] [Indexed: 12/21/2022] Open
Abstract
The inflammation of the airway and lung could be triggered by upregulation cyclooxygenase (COX)-2 and prostaglandin E2 (PGE2) induced by various proinflammatory factors. COX-2 induction by thrombin has been shown to play a vital role in various inflammatory diseases. However, in human tracheal smooth muscle cells (HTSMCs), how thrombin enhanced the levels of COX-2/PGE2 is not completely characterized. Thus, in this study, the levels of COX-2 expression and PGE2 synthesis induced by thrombin were determined by Western blot, promoter-reporter assay, real-time PCR, and ELISA kit. The various signaling components involved in the thrombin-mediated responses were differentiated by transfection with siRNAs and selective pharmacological inhibitors. The role of NF-κB was assessed by a chromatin immunoprecipitation (ChIP) assay, immunofluorescent staining, as well as Western blot. Our results verified that thrombin markedly triggered PGE2 secretion via COX-2 upregulation which were diminished by the inhibitor of thrombin (PPACK), PAR1 (SCH79797), Gi/o protein (GPA2), Gq protein (GPA2A), PKCα (Gö6976), p38 MAPK (SB202190), JNK1/2 (SP600125), MEK1/2 (U0126), or NF-κB (helenalin) and transfection with siRNA of PAR1, Gqα, Giα, PKCα, JNK2, p38, p42, or p65. Moreover, thrombin induced PAR1-dependent PKCα phosphorylation in HTSMCs. We also observed that thrombin induced p38 MAPK, JNK1/2, and p42/p44 MAPK activation through a PAR1/PKCα pathway. Thrombin promoted phosphorylation of NF-κB p65, leading to nuclear translocation and binding to the COX-2 promoter element to enhance promoter activity, which was reduced by Gö6976, SP600125, SB202190, or U0126. These findings supported that COX-2/PGE2 expression triggered by thrombin was engaged in PAR1/Gq or Gi/o/PKCα/MAPK-dependent NF-κB activation in HTSMCs.
Collapse
|
11
|
Kiarely Souza E, Pereira-Dutra FS, Rajão MA, Ferraro-Moreira F, Goltara-Gomes TC, Cunha-Fernandes T, Santos JDC, Prestes EB, Andrade WA, Zamboni DS, Bozza MT, Bozza PT. Lipid droplet accumulation occurs early following Salmonella infection and contributes to intracellular bacterial survival and replication. Mol Microbiol 2021; 117:293-306. [PMID: 34783412 DOI: 10.1111/mmi.14844] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 01/20/2023]
Abstract
Salmonellosis is a public health problem caused by Salmonella sp., a highly adapted facultative intracellular pathogen. After internalization, Salmonella sp. Manipulates several host processes, mainly through the activation of the type III secretion system (T3SS), including modification of host lipid metabolism and lipid droplet (LD) accumulation. LDs are dynamic and complex lipid-rich organelles involved in several cellular processes. The present study investigated the mechanism involved in LD biogenesis in Salmonella-infected macrophages and its role in bacterial pathogenicity. Here, we reported that S. Typhimurium induced a rapid time-dependent increase of LD formation in macrophages. The LD biogenesis was demonstrated to depend on Salmonella's viability and SPI1-related T3SS activity, with the participation of Toll-Like Receptor (TLR) signaling. We also observed that LD accumulation occurs through TLR2-dependent signaling and is counter-regulated by TLR4. Last, the pharmacologic modulation of LD formation by inhibiting diacylglycerol O-acyltransferase 1 (DGAT1) and cytosolic phospholipase A2 (cPLA2) significantly reduced the intracellular bacterial proliferation and impaired the prostaglandin E2 (PGE2 ) synthesis. Collectively, our data suggest the role of LDs on S. typhimurium intracellular survival and replication in macrophages. This data set provides new perspectives for future investigations about LDs in host-pathogen interaction.
Collapse
Affiliation(s)
- Ellen Kiarely Souza
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Program of Immunology and Inflammation, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Filipe S Pereira-Dutra
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Matheus A Rajão
- Program of Immunology and Tumor Biology, Instituto Nacional do Câncer, INCA, Rio de Janeiro, Brazil
| | - Felipe Ferraro-Moreira
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Taynná C Goltara-Gomes
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Tamires Cunha-Fernandes
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Julia da Cunha Santos
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Elisa B Prestes
- Laboratory of Inflammation and Immunity, Department of Immunity, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Warrison A Andrade
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Dario S Zamboni
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo T Bozza
- Laboratory of Inflammation and Immunity, Department of Immunity, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Vo TTT, Wee Y, Chen YL, Cheng HC, Tuan VP, Lee IT. Surfactin attenuates particulate matter-induced COX-2-dependent PGE 2 production in human gingival fibroblasts by inhibiting TLR2 and TLR4/MyD88/NADPH oxidase/ROS/PI3K/Akt/NF-κB signaling pathway. J Periodontal Res 2021; 56:1185-1199. [PMID: 34486757 DOI: 10.1111/jre.12932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To evaluate the anti-inflammatory effects of surfactin and underlying mechanisms against particulate matter (PM)-induced inflammatory responses in human gingival fibroblasts (HGFs). BACKGROUND PM, a major air pollutant, may associate with certain oral diseases possibly by inducing inflammation and oxidative stress. Surfactin, a potent biosurfactant, possesses various biological properties including anti-inflammatory activity. However, the underlying mechanisms are unclear. Also, there is no study investigating the effects of surfactin on PM-induced oral inflammatory responses. As an essential constituent of human periodontal connective tissues which involves immune-inflammatory responses, HGFs serve as useful study models. METHODS HGFs were pretreated with surfactin prior to PM incubation. The PGE2 production was determined by ELISA, while the protein expression and mRNA levels of COX-2 and upstream regulators were measured using Western blot and real-time PCR, respectively. The transcriptional activity of COX-2 and NF-κB were determined using promoter assay. ROS generation and NADPH oxidase activity were identified by specific assays. Co-immunoprecipitation assay, pharmacologic inhibitors, and siRNA transfection were applied to explore the interplay of molecules. Mice were given one dose of surfactin or different pharmacologic inhibitors, then PM was delivered into the gingiva for three consecutive days. Gingival tissues were obtained for analyzing COX-2 expression. RESULTS PM-treated HGFs released significantly higher COX-2-dependent PGE2 , which were regulated by TLR2 and TLR4/MyD88/NADPH oxidase/ROS/PI3K/Akt/NF-κB pathway. PM-induced COX-2/PGE2 increase was effectively reversed by surfactin through the disruption of regulatory pathway. Similar inhibitory effects of surfactin was observed in mice. CONCLUSION Surfactin may elicit anti-inflammatory effects against PM-induced oral inflammatory responses.
Collapse
Affiliation(s)
- Thi Thuy Tien Vo
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yinshen Wee
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Chung Cheng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Vo Phuoc Tuan
- Endoscopy Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Yang CC, Yang CM. Chinese Herbs and Repurposing Old Drugs as Therapeutic Agents in the Regulation of Oxidative Stress and Inflammation in Pulmonary Diseases. J Inflamm Res 2021; 14:657-687. [PMID: 33707963 PMCID: PMC7940992 DOI: 10.2147/jir.s293135] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Several pro-inflammatory factors and proteins have been characterized that are involved in the pathogenesis of inflammatory diseases, including acute respiratory distress syndrome, chronic obstructive pulmonary disease, and asthma, induced by oxidative stress, cytokines, bacterial toxins, and viruses. Reactive oxygen species (ROS) act as secondary messengers and are products of normal cellular metabolism. Under physiological conditions, ROS protect cells against oxidative stress through the maintenance of cellular redox homeostasis, which is important for proliferation, viability, cell activation, and organ function. However, overproduction of ROS is most frequently due to excessive stimulation of either the mitochondrial electron transport chain and xanthine oxidase or reduced nicotinamide adenine dinucleotide phosphate (NADPH) by pro-inflammatory cytokines, such as interleukin-1β and tumor necrosis factor α. NADPH oxidase activation and ROS overproduction could further induce numerous inflammatory target proteins that are potentially mediated via Nox/ROS-related transcription factors triggered by various intracellular signaling pathways. Thus, oxidative stress is considered important in pulmonary inflammatory processes. Previous studies have demonstrated that redox signals can induce pulmonary inflammatory diseases. Thus, therapeutic strategies directly targeting oxidative stress may be effective for pulmonary inflammatory diseases. Therefore, drugs with anti-inflammatory and anti-oxidative properties may be beneficial to these diseases. Recent studies have suggested that traditional Chinese medicines, statins, and peroxisome proliferation-activated receptor agonists could modulate inflammation-related signaling processes and may be beneficial for pulmonary inflammatory diseases. In particular, several herbal medicines have attracted attention for the management of pulmonary inflammatory diseases. Therefore, we reviewed the pharmacological effects of these drugs to dissect how they induce host defense mechanisms against oxidative injury to combat pulmonary inflammation. Moreover, the cytotoxicity of oxidative stress and apoptotic cell death can be protected via the induction of HO-1 by these drugs. The main objective of this review is to focus on Chinese herbs and old drugs to develop anti-inflammatory drugs able to induce HO-1 expression for the management of pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Kwei-San, Tao-Yuan, 33302, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan.,Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, 40402, Taiwan.,Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Taichung, 41354, Taiwan
| |
Collapse
|
14
|
Antipseudomonal and Immunomodulatory Properties of Esc Peptides: Promising Features for Treatment of Chronic Infectious Diseases and Inflammation. Int J Mol Sci 2021; 22:ijms22020557. [PMID: 33429882 PMCID: PMC7826692 DOI: 10.3390/ijms22020557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 12/04/2022] Open
Abstract
Persistent infections, such as those provoked by the Gram-negative bacterium Pseudomonas aeruginosa in the lungs of cystic fibrosis (CF) patients, can induce inflammation with lung tissue damage and progressive alteration of respiratory function. Therefore, compounds having both antimicrobial and immunomodulatory activities are certainly of great advantage in fighting infectious diseases and chronic inflammation. We recently demonstrated the potent antipseudomonal efficacy of the antimicrobial peptide (AMP) Esc(1-21) and its diastereomer Esc(1-21)-1c, namely Esc peptides. Here, we confirmed this antimicrobial activity by reporting on the peptides’ ability to kill P. aeruginosa once internalized into alveolar epithelial cells. Furthermore, by means of enzyme-linked immunosorbent assay and Western blot analyses, we investigated the peptides’ ability to detoxify the bacterial lipopolysaccharide (LPS) by studying their effects on the secretion of the pro-inflammatory cytokine IL-6 as well as on the expression of cyclooxygenase-2 from macrophages activated by P. aeruginosa LPS. In addition, by a modified scratch assay we showed that both AMPs are able to stimulate the closure of a gap produced in alveolar epithelial cells when cell migration is inhibited by concentrations of Pseudomonas LPS that mimic lung infection conditions, suggesting a peptide-induced airway wound repair. Overall, these results have highlighted the two Esc peptides as valuable candidates for the development of new multifunctional therapeutics for treatment of chronic infectious disease and inflammation, as found in CF patients.
Collapse
|
15
|
Jia G, Liu X, Che N, Xia Y, Wang G, Xiong Z, Zhang H, Ai L. Human-origin Lactobacillus salivarius AR809 protects against immunosuppression in S. aureus-induced pharyngitis via Akt-mediated NF-κB and autophagy signaling pathways. Food Funct 2020; 11:270-284. [PMID: 31957758 DOI: 10.1039/c9fo02476j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lactobacillus salivarius AR809 is a newly discovered probiotic strain from a healthy human pharynx and has potential ability to adhere to the pharyngeal epithelium and inhibit Staphylococcus aureus (S. aureus)-induced inflammatory response. Pharyngeal spray administration of AR809 exhibited protective effects in a S. aureus-induced mouse model of pharyngitis. The inhibitory effect and underlying molecular mechanism of AR809 on S. aureus-stimulated pharyngitis were further investigated. AR809 significantly increased phagocytosis and bactericidal activity, reduced the production of inflammatory mediators (intracellular reactive oxygen species (ROS), prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), nitric oxide (NO), inducible NOS (iNOS)) and the expression of inflammatory cytokines (tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β)), and induced macrophages to adopt the M2 phenotype. AR809 also attenuated S. aureus-induced phosphorylations of protein kinase B (Akt) and rapamycin (mTOR), and elevated the autophagic protein (light chain 3 from II (LC3-II) and Beclin-1) level. Furthermore, AR809 inhibited nuclear transcription factor kappa-B (NF-κB) activation by suppressing the nuclear translocation of NF-κB p65. Likewise, 740Y-P (a PI3K activator) decreased the anti-inflammatory effect of AR809 against S. aureus-induced inflammatory response, while AR809 treatments with wortmannin (a PI3K inhibitor) markedly reversed this inflammatory response. AR809 prevents S. aureus-induced pharyngeal inflammatory response, possibly by regulating TLR/PI3K/Akt/mTOR signalling pathway-related autophagy and TLR/PI3K/Akt/IκB/NF-κB pathway activity, and therefore has potential for use in preventing pharyngitis and other inflammatory diseases.
Collapse
Affiliation(s)
- Guochao Jia
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Zha C, Xiao H, Song B, Zheng C, Yang X, Wang W, Wang L. Resveratrol promotes mammary cell proliferation and antioxidation capacity during pregnancy and lactation in mice. J Appl Microbiol 2020; 130:450-463. [PMID: 32544275 DOI: 10.1111/jam.14747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022]
Abstract
AIMS Reproduction can induce maternal oxidative stress, and resveratrol is able to alleviate oxidative stress. This study aimed to investigate the protective effects of resveratrol supplementation in mice during pregnancy and lactation. METHODS AND RESULTS Pregnant mice were randomly divided into three groups: (i) Pure water (Con); (ii) Cellulose sodium dissolves with 20 mg kg-1 resveratrol (R1) and (iii) Cellulose sodium dissolves with 40 mg kg-1 resveratrol (R2). The experiment starts at pregnancy and ends at weaning. The results showed that resveratrol increased mammary cell proliferation and the mRNA expressions of UDP glucuronosyltransferase family 1 member A1 (UGT1A1) and cytochrome P450 proteins (CYP1A1), while decreased superoxide dismutase, extracellular (SOD3) in the mammary gland. Further study showed that resveratrol promoted the mRNA expressions of genes involved in mitophagy, such as transcription factor Eβ (Tfeβ), Bcl1lc3β, homosapiens microtubule-associated protein 1 light-chain 3 beta (Map1lc3β), Parkin, sequestosome1 (p62), autophagy-related protein (Atg5) and Beclin-1 (Becn1) in the mammary gland. Moreover, resveratrol increased the abundances of some intestinal microbial species. 40 mg kg-1 resveratrol significantly increased the contents of Acidobacteri, unidentified acidbacteria, Bacilales, Staphylococcaceae and Staphylococcus at phylum, class, order, family and genus level respectively. CONCLUSION Our results indicate that resveratrol supplementation may promote mammary cell proliferation and antioxidant ability through mitophagy and regulating gut microbiota in pregnant mice. SIGNIFICANCE AND IMPACT OF THE STUDY Current study proved that resveratrol could affect mammary cell proliferation and antioxidation capacity during pregnancy and lactation in mice for the first time. The underlying mechanisms may be related to mitophagy and gut microbiota.
Collapse
Affiliation(s)
- C Zha
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - H Xiao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - B Song
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - C Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - X Yang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - W Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - L Wang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
17
|
Su BC, Chen JY. Epinecidin-1: An orange-spotted grouper antimicrobial peptide that modulates Staphylococcus aureus lipoteichoic acid-induced inflammation in macrophage cells. FISH & SHELLFISH IMMUNOLOGY 2020; 99:362-367. [PMID: 32084537 DOI: 10.1016/j.fsi.2020.02.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/30/2020] [Accepted: 02/17/2020] [Indexed: 06/10/2023]
Abstract
Orange-spotted grouper (Epinephelus coioides) is among the most economically important of all fish species farmed in Asia. This species expresses an antimicrobial peptide called epinecidin-1 (EPI), which is considered to be a host defense factor due to its strong bacterial killing activity. Antimicrobial peptides usually possess both bacterial killing and immunomodulatory activity, however, the modulatory activity of EPI on Gram-positive bacterial lipoteichoic acids (LTA)-induced inflammation has not been previously reported. In this study, we found that EPI effectively suppressed LTA-induced production of proinflammatory factors in macrophages. Mechanistically, EPI attenuated LTA-induced inflammation by inhibiting Toll-like receptor (TLR) 2 internalization and subsequent downstream signaling (reactive oxygen species, Akt, p38 and Nuclear factor κB). However, protein abundance of TLR2 was not altered by EPI or LTA. Taken together, our findings reveal for the first time that EPI possesses inhibitory activity toward LTA-induced inflammation in macrophages.
Collapse
Affiliation(s)
- Bor-Chyuan Su
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan.
| |
Collapse
|
18
|
Lima BHF, Marques PE, Gomides LF, Mattos MS, Kraemer L, Queiroz-Junior CM, Lennon M, Hirsch E, Russo RC, Menezes GB, Hessel EM, Amour A, Teixeira MM. Converging TLR9 and PI3Kgamma signaling induces sterile inflammation and organ damage. Sci Rep 2019; 9:19085. [PMID: 31836766 PMCID: PMC6910931 DOI: 10.1038/s41598-019-55504-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/24/2019] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptor 9 (TLR9) and Phosphatidylinositol-3-kinase gamma (PI3Kγ) are very important effectors of the immune response, however, the importance of such crosstalk for disease development is still a matter of discussion. Here we show that PI3Kγ is required for immune responses in which TLR9 is a relevant trigger. We demonstrate the requirement of PI3Kγ for TLR9-induced inflammation in a model of CpG-induced pleurisy. Such requirement was further observed in inflammatory models where DNA sensing via TLR9 contributes to disease, such as silicosis and drug-induced liver injury. Using adoptive transfer, we demonstrate that PI3Kγ is important not only in leukocytes but also in parenchymal cells for the progression of inflammation. We demonstrate this crosstalk between TLR9 and PI3Kγ in vitro using human PBMCs. The inhibition of PI3Kγ in CpG-stimulated PBMCs resulted in reduction of both cytokine production and phosphorylated Akt. Therefore, drugs that target PI3Kγ have the potential to treat diseases mediated by excessive TLR9 signalling.
Collapse
Affiliation(s)
- Braulio Henrique Freire Lima
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Elias Marques
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lindisley Ferreira Gomides
- Center for Gastrointestinal Biology, Instituto de Ciências Biológicas, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Matheus Silvério Mattos
- Physiology and Biophysics/Instituto de Ciencias Biologicas, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lucas Kraemer
- Physiology and Biophysics/Instituto de Ciencias Biologicas, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso M Queiroz-Junior
- Departament of Morphology, Institute of Biological Sciences, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mark Lennon
- Target Sciences, GlaxoSmithKline, Stevenage, Hertfordshire, Stevenage, United Kingdom
| | - Emilio Hirsch
- Department ot Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Remo Castro Russo
- Physiology and Biophysics/Instituto de Ciencias Biologicas, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Instituto de Ciências Biológicas, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edith M Hessel
- Refractory Respiratory Inflammation DPU, GlaxoSmithKline, Hertfordshire, Stevenage, United Kingdom
| | - Augustin Amour
- Refractory Respiratory Inflammation DPU, GlaxoSmithKline, Hertfordshire, Stevenage, United Kingdom
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Feredal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
19
|
Kumar NG, Contaifer D, Madurantakam P, Carbone S, Price ET, Van Tassell B, Brophy DF, Wijesinghe DS. Dietary Bioactive Fatty Acids as Modulators of Immune Function: Implications on Human Health. Nutrients 2019; 11:E2974. [PMID: 31817430 PMCID: PMC6950193 DOI: 10.3390/nu11122974] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/05/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Diet is major modifiable risk factor for cardiovascular disease that can influence the immune status of the individual and contribute to persistent low-grade inflammation. In recent years, there has been an increased appreciation of the role of polyunsaturated fatty acids (PUFA) in improving immune function and reduction of systemic inflammation via the modulation of pattern recognition receptors (PRR) on immune cells. Extensive research on the use of bioactive lipids such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and their metabolites have illustrated the importance of these pro-resolving lipid mediators in modulating signaling through PRRs. While their mechanism of action, bioavailability in the blood, and their efficacy for clinical use forms an active area of research, they are found widely administered as marine animal-based supplements like fish oil and krill oil to promote health. The focus of this review will be to discuss the effect of these bioactive fatty acids and their metabolites on immune cells and the resulting inflammatory response, with a brief discussion about modern methods for their analysis using mass spectrometry-based methods.
Collapse
Affiliation(s)
- Naren Gajenthra Kumar
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Daniel Contaifer
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Parthasarathy Madurantakam
- Department of General Practice, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA 23220, USA;
- VCU Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Elvin T. Price
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Benjamin Van Tassell
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Donald F. Brophy
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
| | - Dayanjan S. Wijesinghe
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (D.C.); (E.T.P.); (B.V.T.); (D.F.B.)
- da Vinci Center, Virginia Commonwealth University, Richmond, VA 23220, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University School of Pharmacy, Richmond, VA 23298, USA
| |
Collapse
|
20
|
Ethanol Extract of Sesamum indicum Linn. Inhibits Fc εRI-Mediated Allergic Reaction via Regulation of Lyn/Syk and Fyn Signaling Pathways in Rat Basophilic Leukemic RBL-2H3 Mast Cells. Mediators Inflamm 2019; 2019:5914396. [PMID: 31686984 PMCID: PMC6811790 DOI: 10.1155/2019/5914396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/12/2019] [Accepted: 08/07/2019] [Indexed: 01/26/2023] Open
Abstract
This study is aimed at determining whether Sesamum indicum Linn. beneficially influences FcεRI-mediated allergic reactions in RBL-2H3 mast cells; it is also aimed at further investigating Lyn/Fyn and Syk signaling pathways. To examine the antiallergic effect of Sesamum indicum Linn. extract (SIE), we treated antigen/immunoglobulin E- (IgE-) sensitized mast cells with extracts of various concentrations. We examined the degranulation release and concentrations of inflammatory mediators. Additionally, the expressions of genes involved in the FcεRI and arachidonate signaling pathways were examined. SIE inhibited the degranulation and secretion of inflammatory mediators in antigen/IgE-sensitized mast cells. SIE reduced the expressions of FcεRI signaling-related genes, such as Syk, Lyn, and Fyn, and the phosphorylation of extracellular signal-regulated kinase in antigen/IgE-sensitized mast cells. Additionally, in late allergic responses, SIE reduced PGD2 release and COX-2 and cPLA2 phosphorylation expression in FcεRI-mediated mast cell activation. Lastly, 250–500 mg/kg SIE significantly attenuated the Ag/IgE-induced passive cutaneous anaphylaxis (PCA) reaction in mice. The potent effect of SIE on RBL-2H3 mast cell activation indicates that the extract could potentially be used as a novel inhibitor against allergic reactions.
Collapse
|
21
|
Zhang L, Dong L, Tang Y, Li M, Zhang M. MiR-146b protects against the inflammation injury in pediatric pneumonia through MyD88/NF-κB signaling pathway. Infect Dis (Lond) 2019; 52:23-32. [PMID: 31583932 DOI: 10.1080/23744235.2019.1671987] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Pneumonia is a common respiratory disease worldwide that can be prevented and treated. However, it is considered to be the leading cause of children death. The present study was aimed to explore the functional role and molecular mechanism of miR-146b in the inflammation injury in pediatric pneumonia.Materials and methods: The lipopolysaccharide (LPS)-induced pulmonary injury cell model was established in WI-38 human lung fibroblasts cells. QRT-PCR and Western blot was applied to detect miR-146b and MyD88 expression. ELISA assay was used to analyze the production of pro-inflammatory factors. Cell viability was evaluated by CCK-8 assay. The apoptosis proteins and the downstream genes of NF-κB pathway were detected by Western blot.Results: we displayed that miR-146b was down-regulated, whereas MyD88 was up-regulated in the serum of children patients with pneumonia and in WI-38 cells treated with LPS. Moreover, re-expression of miR-146b suppressed the production of inflammatory factors in the serum of pneumonia patients and WI-38 cells treated with LPS. In addition, elevating miR-146b expression increased WI-38 cell viability and reduced cell apoptosis. More importantly, bioinformatics analysis revealed that MyD88 was a target of miR-146b and could overturn the protective effect of miR-146b on the inflammation injury in LPS-injured WI-38 cells. Furthermore, miR-146b over-expression inhibited the activation of NF-κB signaling pathway by suppressing MyD88.Conclusion: miR-146b attenuated the inflammation injury in pediatric pneumonia through inhibiting MyD88/NF-κB signaling pathway. These preliminarily findings further deepened our understanding of this mechanism and identified new potential therapeutic targets for pediatric pneumonia.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Lili Dong
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Yu Tang
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Min Li
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Mingming Zhang
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang City, China
| |
Collapse
|
22
|
Lee CW, Chi MC, Peng KT, Chiang YC, Hsu LF, Yan YL, Li HY, Chen MC, Lee IT, Lai CH. Water-Soluble Fullerenol C 60(OH) 36 toward Effective Anti-Air Pollution Induced by Urban Particulate Matter in HaCaT Cell. Int J Mol Sci 2019; 20:ijms20174259. [PMID: 31480310 PMCID: PMC6747515 DOI: 10.3390/ijms20174259] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Particulate matter (PM), a widespread air pollutant, consists of a complex mixture of solid and liquid particles suspended in air. Many diseases have been linked to PM exposure, which induces an imbalance in reactive oxygen species (ROS) generated in cells, and might result in skin diseases (such as aging and atopic dermatitis). New techniques involving nanomedicine and nano-delivery systems are being rapidly developed in the medicinal field. Fullerene, a kind of nanomaterial, acts as a super radical scavenger. Lower water solubility levels limit the bio-applications of fullerene. Hence, to improve the water solubility of fullerene, while retaining its radical scavenger functions, a fullerene derivative, fullerenol C60(OH)36, was synthesized, to examine its biofunctions in PM-exposed human keratinocyte (HaCaT) cells. The PM-induced increase in ROS levels and expression of phosphorylated mitogen-activated protein kinase and Akt could be inhibited via fullerenol pre-treatment. Furthermore, the expression of inflammation-related proteins, cyclooxygenase-2, heme oxygenase-1, and prostaglandin E2 was also suppressed. Fullerenol could preserve the impaired state of skin barrier proteins (filaggrin, involucrin, repetin, and loricrin), which was attributable to PM exposure. These results suggest that fullerenol could act against PM-induced cytotoxicity via ROS scavenging and anti-inflammatory mechanisms, and the maintenance of expression of barrier proteins, and is a potential candidate compound for the treatment of skin diseases.
Collapse
Affiliation(s)
- Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Guishan District, Taoyuan City 333, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 613, Taiwan
| | - Miao-Ching Chi
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Division of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Kuo-Ti Peng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 613, Taiwan
- College of Medicine, Chang Gung University, Guishan District, Taoyuan City 333, Taiwan
| | - Yao-Chang Chiang
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 613, Taiwan
| | - Lee-Fen Hsu
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Division of Neurosurgery, Department of Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 613, Taiwan
| | - Yi-Ling Yan
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 613, Taiwan
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Hsing-Yen Li
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Chun Chen
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chian-Hui Lai
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
23
|
Dong P, Ji X, Han W, Han H. Oxymatrine exhibits anti-neuroinflammatory effects on Aβ 1-42-induced primary microglia cells by inhibiting NF-κB and MAPK signaling pathways. Int Immunopharmacol 2019; 74:105686. [PMID: 31207405 DOI: 10.1016/j.intimp.2019.105686] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 06/01/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
Oxymatrine (OMT), isolated from Sophora flavescens or Sophora alopecuroides, possesses various pharmacological and biological activities, including anti-inflammatory, anti-oxidant, and anti-diabetic properties. Microglia cells, the resident immune cells in the central nervous system (CNS), play a key role in neurodegenerative diseases. In this study, the neuroinflammatory effects of OMT and its mechanisms were investigated by Aβ1-42-induced rat brain tissue model and primary microglia cells model. The hematoxylin-eosin (HE) staining and immunohistochemistry results showed that OMT could reduce neuronal damage and inhibit microglia activation in the model tissue. The in vitro experiments revealed that OMT could decrease the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and nitric oxide (NO), and down-regulate the expression of iNOS and COX-2 in a dose-dependent manner. Furthermore, OMT inhibited phosphorylation of JNK, ERK 1/2, P-p38 and NF-κB in Aβ1-42-induced microglia cells. In summary, OMT exhibits anti-neuroinflammatory effects and the anti-inflammatory activity of OMT is related to the regulation of MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Peiliang Dong
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiaomeng Ji
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Wei Han
- Guiyang College of Traditional Chinese Medicine, China
| | - Hua Han
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
24
|
Li B, Xi P, Wang Z, Han X, Xu Y, Zhang Y, Miao J. PI3K/Akt/mTOR signaling pathway participates in Streptococcus uberis-induced inflammation in mammary epithelial cells in concert with the classical TLRs/NF-ĸB pathway. Vet Microbiol 2018; 227:103-111. [PMID: 30473339 DOI: 10.1016/j.vetmic.2018.10.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022]
Abstract
Mammary epithelial cells (MECs) play an important role in debating Streptococcus uberis (S. uberis) infection. Toll like receptor (TLR) engagement leads to the recruitment of phosphatidylinositol 3 kinases (PI3K). In order to investigate the relationship of TLRs/NF-κB and PI3K/Akt/mTOR signaling pathways in S. uberis infection in MECs, we challenged MECs (EpH4-Ev) with S. uberis 0140 J and quantified the adaptor molecules in these two signaling pathways, as-well-as proinflammatory cytokines and cell damage. The results indicate that the host's responses to virulent S. uberis infection are complex. In MECs, both TLR2 and TLR4 are detecting S. uberis infection and TLR2 is the principal receptor. The role of the PI3K/Akt/mTOR pathway in inflammatory regulation is independent of the activation of TLRs/NF-κB. Cross-talk between PI3K/Akt/mTOR and TLRs/NF-κB signaling pathways promote inflammation. This study increases our understanding of the molecular defense mechanisms of MECs in S. uberis mastitis, and provides theoretical support for the prevention of this disease.
Collapse
Affiliation(s)
- Bin Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Panpan Xi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhenglei Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuanshu Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
25
|
Tsai MH, Wu CH, Lin WN, Cheng CY, Chuang CC, Chang KT, Jiang RS, Hsu JF, Lee IT. Infection with Staphylococcus aureus elicits COX-2/PGE2/IL-6/MMP-9-dependent aorta inflammation via the inhibition of intracellular ROS production. Biomed Pharmacother 2018; 107:889-900. [DOI: 10.1016/j.biopha.2018.08.096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 12/21/2022] Open
|
26
|
Gajenthra Kumar N, Contaifer D, Baker PRS, Ekroos K, Jefferson KK, Wijesinghe DS. Untargeted lipidomic analysis to broadly characterize the effects of pathogenic and non-pathogenic staphylococci on mammalian lipids. PLoS One 2018; 13:e0206606. [PMID: 30379915 PMCID: PMC6209338 DOI: 10.1371/journal.pone.0206606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 10/16/2018] [Indexed: 11/19/2022] Open
Abstract
Modification of the host lipidome via secreted enzymes is an integral, but often overlooked aspect of bacterial pathogenesis. In the current era of prevalent antibiotic resistance, knowledge regarding critical host pathogen lipid interactions has the potential for use in developing novel antibacterial agents. While most studies to date on this matter have focused on specific lipids, or select lipid classes, this provides an incomplete picture. Modern methods of untargeted lipidomics have the capacity to overcome these gaps in knowledge and provide a comprehensive understanding of the role of lipid metabolism in the pathogenesis of infections. In an attempt to determine the role of lipid modifying enzymes produced by staphylococci, we exposed bovine heart lipids, a standardized model for the mammalian lipidome, to spent medium from staphylococcal cultures, and analyzed lipid molecular changes by MS/MSALL shotgun lipidomics. We elucidate distinct effects of different staphylococcal isolates, including 4 clinical isolates of the pathogenic species Staphylococcus aureus, a clinical isolate of the normally commensal species S. epidermidis, and the non-pathogenic species S. carnosus. Two highly virulent strains of S. aureus had a more profound effect on mammalian lipids and modified more lipid classes than the other staphylococcal strains. Our studies demonstrate the utility of the applied untargeted lipidomics methodology to profile lipid changes induced by different bacterial secretomes. Finally, we demonstrate the promise of this lipidomics approach in assessing the specificity of bacterial enzymes for mammalian lipid classes. Our data suggests that there may be a correlation between the bacterial expression of lipid-modifying enzymes and virulence, and could facilitate the guided discovery of lipid pathways required for bacterial infections caused by S. aureus and thereby provide insights into the generation of novel antibacterial agents.
Collapse
Affiliation(s)
- Naren Gajenthra Kumar
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Daniel Contaifer
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | | | - Kim Ekroos
- Lipidomics Consulting Ltd., Esbo, Finland
| | - Kimberly K. Jefferson
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Dayanjan S. Wijesinghe
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
27
|
Resveratrol Attenuates Staphylococcus Aureus-Induced Monocyte Adhesion through Downregulating PDGFR/AP-1 Activation in Human Lung Epithelial Cells. Int J Mol Sci 2018; 19:ijms19103058. [PMID: 30301269 PMCID: PMC6213130 DOI: 10.3390/ijms19103058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a very common Gram-positive bacterium. It is widely distributed in air, soil, and water. S. aureus often causes septicemia and pneumonia in patients. In addition, it is considered to play a key role in mediating cell adhesion molecules upregulation. Resveratrol is a natural antioxidant with diverse biological effects, including the modulation of immune function, anti-inflammation, and cancer chemoprevention. In this study, we proved that S. aureus-upregulated vascular cell adhesion molecule-1 (VCAM-1) expression in human lung epithelial cells (HPAEpiCs) was inhibited by resveratrol. We also observed that resveratrol downregulated S. aureus-enhanced leukocyte count in bronchoalveolar lavage (BAL) fluid in mice. In HPAEpiCs, S. aureus stimulated c-Src, PDGFR, p38 MAPK, or JNK1/2 phosphorylation, which was inhibited by resveratrol. S. aureus induced the adhesion of THP-1 cells (a human monocytic cell line) to HPAEpiCs, which was also reduced by resveratrol. Finally, we found that S. aureus induced c-Src/PDGFR/p38 MAPK and JNK1/2-dependent c-Jun and ATF2 activation and in vivo binding of c-Jun and ATF2 to the VCAM-1 promoter, which were inhibited by resveratrol. Thus, resveratrol functions as a suppressor of S. aureus-induced inflammatory signaling, not only by inhibiting VCAM-1 expression but also by diminishing c-Src, PDGFR, JNK1/2, p38 MAPK, and AP-1 activation in HPAEpiCs.
Collapse
|
28
|
Tsai MH, Yang CM, Chang KT, Chuang CC, Lin WN, Jiang RS, Wu CH, Lee IT. Carbon monoxide ameliorates Staphylococcus aureus-elicited COX-2/IL-6/MMP-9-dependent human aortic endothelial cell migration and inflammatory responses. Immunol Lett 2018; 203:40-49. [PMID: 30236480 DOI: 10.1016/j.imlet.2018.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/28/2018] [Accepted: 09/14/2018] [Indexed: 12/14/2022]
Abstract
Staphylococcus aureus (S. aureus) can often lead to many life-threatening diseases. It has the ability to invade normal endovascular tissue. Acute inflammation and its resolution are important to ensure bacterial clearance and limit tissue injury. Carbon monoxide (CO) has been shown to exert anti-inflammatory effects in various tissues and organ systems. In our study, we investigated the effects and the mechanisms of carbon monoxide releasing molecule-2 (CORM-2) on S. aureus-induced inflammatory responses in human aortic endothelial cells (HAECs). We proved that S. aureus induced cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2)/interleukin-6 (IL-6)/matrix metallopeptidase-9 (MMP-9) expression and cell migration, which were decreased by CORM-2. Moreover, CORM-2 had no effects on TLR2 mRNA levels in response to S. aureus. Interestingly, we proved that S. aureus decreased intracellular ROS generation, suggesting that the inhibition of ROS further promoted inflammatory responses. However, CORM-2 significantly inhibited S. aureus-induced inflammation by increasing intracellular ROS generation. S. aureus-induced NF-κB activation was also inhibited by CORM-2. Finally, we proved that S. aureus induced levels of the biomarkers of inflammation in cardiovascular diseases, which were inhibited by CORM-2. Taken together, these results suggest that CORM-2 inhibits S. aureus-induced COX-2/PGE2/IL-6/MMP-9 expression and aorta inflammatory responses by increasing the ROS generation and reducing the inflammatory molecules levels.
Collapse
Affiliation(s)
- Ming-Horng Tsai
- Department of Pediatrics, Division of Neonatology and Pediatric Hematology/Oncology, Chang Gung Memorial Hospital, Yunlin, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| | - Kuo-Ting Chang
- Translational Medicine Center, Taoyuan General Hospital, Ministry of Healthy and Welfare, Taoyuan, Taiwan
| | - Chu-Chun Chuang
- Department of Physical Therapy, China Medical University, Taichung, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Rong-San Jiang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Cheng-Hsun Wu
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| | - I-Ta Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Nursing, College of Nursing, Hungkuang University, Taichung, Taiwan.
| |
Collapse
|
29
|
Zhang B, Wu H, Fang L, Ding P, Xu K, Yang Q, Liu R. MerTK Does Not Mediate Phagocytosis of Staphylococcus aureus but Attenuates Inflammation Induced by Staphylococcal Lipoteichoic Acid Through Blocking NF-κB Activation. Inflammation 2018; 40:1543-1552. [PMID: 28528507 DOI: 10.1007/s10753-017-0595-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Mer receptor tyrosine kinase (MerTK) expressed in macrophages is essential for phagocytosis of apoptotic cells. Here, we investigate whether MerTK is involved in the phagocytosis of Staphylococcus aureus (S. aureus) and regulation of staphylococcal lipoteichoic acid (LTA)-induced inflammatory response in macrophages. We found that stimulating RAW264.7 macrophages with S. aureus activated multiple signaling pathways including toll-like receptor 2 (TLR2), scavenger receptor A (SR-A), and MerTK. Meanwhile, S. aureus stimulation also induced activation of proteins focal adhesion kinase (FAK) and Rac1, which are related to phagocytosis. Pretreatment with a specific Mer-blocking antibody significantly inhibited S. aureus-induced phosphorylation of MerTK, while it had no effect on S. aureus-induced activation of FAK and Rac1. Moreover, by confocal laser microscope, we observed that the antibody blockade of MerTK had little impact on the phagocytosis of S. aureus by RAW264.7 macrophages. Additionally, pretreatment with this antibody further promoted LTA-induced phosphorylation of nuclear factor κB (NF-κB) p65 subunit and production of pro-inflammatory cytokines, such as TNF-α, IL-6, IL-1β, and macrophage inflammatory protein-2 (MIP-2). Collectively, these results suggest that MerTK does not play an essential role in the phagocytosis of S. aureus but attenuates inflammation induced by staphylococcal LTA through blocking NF-κB activation.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Respiration, Hefei Second People's Hospital, Anhui Medical University, Heping Road 246, Hefei, Anhui, 230022, People's Republic of China
| | - Huimei Wu
- Department of Respiration, Anhui Geriatric Institute, First Affiliated Hospital, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, People's Republic of China
| | - Lei Fang
- Department of Respiration, Anhui Geriatric Institute, First Affiliated Hospital, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, People's Republic of China
| | - Peishan Ding
- Department of Respiration, Anhui Geriatric Institute, First Affiliated Hospital, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, People's Republic of China
| | - Ke Xu
- Department of Respiration, Anhui Geriatric Institute, First Affiliated Hospital, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, People's Republic of China
| | - Qingbin Yang
- Department of Respiration, Hefei Second People's Hospital, Anhui Medical University, Heping Road 246, Hefei, Anhui, 230022, People's Republic of China
| | - Rongyu Liu
- Department of Respiration, Anhui Geriatric Institute, First Affiliated Hospital, Anhui Medical University, Jixi Road 218, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
30
|
Lee CW, Wu CH, Chiang YC, Chen YL, Chang KT, Chuang CC, Lee IT. Carbon monoxide releasing molecule-2 attenuates Pseudomonas aeruginosa-induced ROS-dependent ICAM-1 expression in human pulmonary alveolar epithelial cells. Redox Biol 2018; 18:93-103. [PMID: 30007888 PMCID: PMC6039312 DOI: 10.1016/j.redox.2018.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) infection in the lung is common in patients with cystic fibrosis (CF). Intercellular adhesion molecule-1 (ICAM-1) is known to play a key role in lung inflammation. Acute inflammation and its timely resolution are important to ensure bacterial clearance and limit tissue damage. Carbon monoxide (CO) has been shown to exert anti-inflammatory effects in various tissues and organ systems. Here, we explored the protective effects and mechanisms of carbon monoxide releasing molecule-2 (CORM-2) on P. aeruginosa-induced inflammatory responses in human pulmonary alveolar epithelial cells (HPAEpiCs). We showed that P. aeruginosa induced prostaglandin E2 (PGE2)/interleukin-6 (IL-6)/ICAM-1 expression and monocyte adherence to HPAEpiCs. Moreover, P. aeruginosa-induced inflammatory responses were inhibited by transfection with siRNA of Toll-like receptor 4 (TLR4), PKCα, p47phox, JNK2, p42, p50, or p65. P. aeruginosa also induced PKCα, JNK, ERK1/2, and NF-κB activation. We further demonstrated that P. aeruginosa increased intracellular ROS generation via NADPH oxidase activation. On the other hand, P. aeruginosa-induced inflammation was inhibited by pretreatment with CORM-2. Preincubation with CORM-2 had no effects on TLR4 mRNA levels in response to P. aeruginosa. However, CORM-2 inhibits P. aeruginosa-induced inflammation by decreasing intracellular ROS generation. P. aeruginosa-induced PKCα, JNK, ERK1/2, and NF-κB activation was inhibited by CORM-2. Finally, we showed that P. aeruginosa induced levels of the biomarkers of inflammation in respiratory diseases, which were inhibited by pretreatment with CORM-2. Taken together, these data suggest that CORM-2 inhibits P. aeruginosa-induced PGE2/IL-6/ICAM-1 expression and lung inflammatory responses by reducing the ROS generation and the inflammatory pathways. CORM-2 inhibits P. aeruginosa-induced PGE2/IL-6/ICAM-1 expression. CORM-2 reduced PKCα phosphorylation in response to P. aeruginosa. We provide molecular mechanisms for antibacterial effects of CORM-2.
Collapse
Affiliation(s)
- Chiang-Wen Lee
- Division of Basic Medical Sciences, Department of Nursing and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi, Taiwan, ROC; Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan, ROC; Department of Rehabilitation, Chang Gung Memorial Hospital, Chia-Yi, Taiwan, ROC
| | - Cheng-Hsun Wu
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Yao-Chang Chiang
- Division of Basic Medical Sciences, Department of Nursing and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi, Taiwan, ROC; Center for Drug Abuse and Addiction, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Kuo-Ting Chang
- Translational Medicine Center, Taoyuan General Hospital, Ministry of Healthy and Welfare, Taoyuan, Taiwan, ROC
| | - Chu-Chun Chuang
- Department of Physical Therapy, China Medical University, Taichung, Taiwan, ROC
| | - I-Ta Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, ROC; Department of Nursing, College of Nursing, Hungkuang University, Taichung, Taiwan, ROC.
| |
Collapse
|
31
|
Chen YF, You MM, Liu YC, Shi YZ, Wang K, Lu YY, Hu FL. Potential protective effect of Trans-10-hydroxy-2-decenoic acid on the inflammation induced by Lipoteichoic acid. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
32
|
Tsai MH, Liu JF, Chiang YC, Hu SCS, Hsu LF, Lin YC, Lin ZC, Lee HC, Chen MC, Huang CL, Lee CW. Artocarpin, an isoprenyl flavonoid, induces p53-dependent or independent apoptosis via ROS-mediated MAPKs and Akt activation in non-small cell lung cancer cells. Oncotarget 2018; 8:28342-28358. [PMID: 28423703 PMCID: PMC5438654 DOI: 10.18632/oncotarget.16058] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 02/27/2017] [Indexed: 12/13/2022] Open
Abstract
Artocarpin has been shown to exhibit cytotoxic effects on different cancer cells, including non-small cell lung carcinoma (NSCLC, A549). However, the underlying mechanisms remain unclear. Here, we explore both p53-dependent and independent apoptosis pathways in artocarpin-treated NSCLC cells. Our results showed that artocarpin rapidly induced activation of cellular protein kinases including Erk1/2, p38 and AktS473. Inhibition of these protein kinases prevented artocarpin-induced cell death. Moreover, artocarpin-induced phosphorylation of these protein kinases and apoptosis were mediated by induction of reactive oxygen species (ROS), as pretreatment with NAC (a ROS scavenger) and Apocynin (a Nox-2 inhibitor) blocked these events. Similarly, transient transfection of p47Phox or p91Phox siRNA attenuated artocarpin-induced NADPH oxidase activity and cell death. In addition, p53 dependent apoptotic proteins including PUMA, cytochrome c, Apaf-1 and caspase 3 were activated by artocarpin, and these effects can be abolished by antioxidants, MAPK inhibitors (U0126 and SB202190), but not by PI3K inhibitor (LY294002). Furthermore, we found that artocarpin-induced Akt phosphorylation led to increased NF-κB activity, which may act as an upstream regulator in the c-Myc and Noxa pathway. Therefore, we propose that enhancement of both ERK/ p38/ p53-dependent or independent AktS473/NF-κB/c-Myc/Noxa cascade by Nox-derived ROS generation plays an important role in artocarpin-induced apoptosis in NSCLC cells.
Collapse
Affiliation(s)
- Ming-Horng Tsai
- Department of Pediatrics, Division of Neonatology and Pediatric Hematology/Oncology, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Ju-Fang Liu
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yao-Chang Chiang
- Center for Drug Abuse and Addiction, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Chia-Yi, Taiwan
| | - Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Lee-Fen Hsu
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Yu-Ching Lin
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan.,Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Department of Respiratory Care, Chang Gung University, Taoyuan, Taiwan
| | - Zih-Chan Lin
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Chun Lee
- Department of Pediatrics, Division of Neonatology and Pediatric Hematology/Oncology, Chang Gung Memorial Hospital, Yunlin, Taiwan
| | - Mei-Chuan Chen
- Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chieh-Liang Huang
- Center for Drug Abuse and Addiction, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Chia-Yi, Taiwan.,Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chia-Yi, Taiwan.,Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| |
Collapse
|
33
|
Zhang J, Zhang Y, Huang H, Zhang H, Lu W, Fu G, Zhu Y. Forsythoside A inhibited S. aureus stimulated inflammatory response in primary bovine mammary epithelial cells. Microb Pathog 2018; 116:158-163. [DOI: 10.1016/j.micpath.2018.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/06/2018] [Accepted: 01/07/2018] [Indexed: 01/20/2023]
|
34
|
Hardman CS, Chen YL, Salimi M, Jarrett R, Johnson D, Järvinen VJ, Owens RJ, Repapi E, Cousins DJ, Barlow JL, McKenzie ANJ, Ogg G. CD1a presentation of endogenous antigens by group 2 innate lymphoid cells. Sci Immunol 2017; 2:eaan5918. [PMID: 29273672 PMCID: PMC5826589 DOI: 10.1126/sciimmunol.aan5918] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023]
Abstract
Group 2 innate lymphoid cells (ILC2) are effectors of barrier immunity, with roles in infection, wound healing, and allergy. A proportion of ILC2 express MHCII (major histocompatibility complex II) and are capable of presenting peptide antigens to T cells and amplifying the subsequent adaptive immune response. Recent studies have highlighted the importance of CD1a-reactive T cells in allergy and infection, activated by the presentation of endogenous neolipid antigens and bacterial components. Using a human skin challenge model, we unexpectedly show that human skin-derived ILC2 can express CD1a and are capable of presenting endogenous antigens to T cells. CD1a expression is up-regulated by TSLP (thymic stromal lymphopoietin) at levels observed in the skin of patients with atopic dermatitis, and the response is dependent on PLA2G4A. Furthermore, this pathway is used to sense Staphylococcus aureus by promoting Toll-like receptor-dependent CD1a-reactive T cell responses to endogenous ligands. These findings define a previously unrecognized role for ILC2 in lipid surveillance and identify shared pathways of CD1a- and PLA2G4A-dependent ILC2 inflammation amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Clare S Hardman
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yi-Ling Chen
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Maryam Salimi
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rachael Jarrett
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - David Johnson
- Department of Plastic and Reconstructive Surgery, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Valtteri J Järvinen
- Oxford Protein Production Facility-UK, Harwell and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Raymond J Owens
- Oxford Protein Production Facility-UK, Harwell and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Emmanouela Repapi
- Computational Biology Research Group, Weatherall Institute of Molecular Medicine, Oxford, UK
| | - David J Cousins
- Department of Infection, Immunity and Inflammation, NIHR Leicester Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | | | | | - Graham Ogg
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Su BC, Chen JY. Antimicrobial Peptide Epinecidin-1 Modulates MyD88 Protein Levels via the Proteasome Degradation Pathway. Mar Drugs 2017; 15:md15110362. [PMID: 29144391 PMCID: PMC5706051 DOI: 10.3390/md15110362] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 12/18/2022] Open
Abstract
The cationic antimicrobial peptide epinecidin-1 was identified from Epinephelus coioides and possesses multiple biological functions, including antibacterial, antifungal, anti-tumor, and immunomodulatory effects. In addition, epinecidin-1 suppresses lipopolysaccharide (LPS)-induced inflammation by neutralizing LPS and ameliorating LPS/Toll-like receptor (TLR)-4 internalization. However, it is unclear whether the actions of epinecidin-1 depend on the regulation of TLR adaptor protein MyD88 or endogenous TLR signaling antagonists, which include A20, interleukin-1 receptor associated kinase (IRAK)-M, and suppressor of cytokine signaling (SOCS)-1. Our results demonstrate that epinecidin-1 alone does not affect A20, IRAK-M, or SOCS-1 protein levels. However, pre-incubation of epinecidin-1 significantly inhibits LPS-induced upregulation of A20, IRAK-M, and SOCS-1. In addition, epinecidin-1 significantly reduces the abundance of MyD88 protein. Both MG132 (a specific proteasome inhibitor) and Heclin (a specific Smurf E3 ligase inhibitor) are able to abolish epinecidin-1-mediated MyD88 degradation. Thus, our data suggest that epinecidin-1 directly inhibits MyD88 via induction of the Smurf E3 ligase proteasome pathway.
Collapse
Affiliation(s)
- Bor-Chyuan Su
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd., Jiaushi, Ilan 262, Taiwan.
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd., Jiaushi, Ilan 262, Taiwan.
| |
Collapse
|
36
|
K ATP channel block inhibits the Toll-like receptor 2-mediated stimulation of NF-κB by suppressing the activation of Akt, mTOR, JNK and p38-MAPK. Eur J Pharmacol 2017; 815:190-201. [PMID: 28923349 DOI: 10.1016/j.ejphar.2017.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 08/23/2017] [Accepted: 09/12/2017] [Indexed: 11/22/2022]
Abstract
Changes in the KATP channel activity have been shown to regulate inflammation and immune responses. Using human keratinocytes, we investigated the effect of KATP channel inhibition on inflammatory mediator production in relation to the Toll like receptor-2-mediated-Akt, mTOR and NF-κB pathways, as well as JNK and p38-MAPK, which regulate the transcription genes involved in immune and inflammatory responses. 5-Hydroxydecanoate (a selective KATP channel blocker), glibenclamide (a cell surface and mitochondrial KATP channel inhibitor), the Akt inhibitor, rapamycin, Bay 11-7085 and N-acetylcysteine reduced the lipoteichoic acid- or peptidoglycan-induced production of cytokines and chemokines, and production of reactive oxygen species and increased the levels and activities of Kir 6.2, NF-κB, phosphorylated-Akt and mTOR, and the activation of JNK and p38-MAPK in keratinocytes. Inhibitors of c-JNK (SP600125) and p38-MAPK (SB203580) attenuated the lipoteichoic acid- or peptidoglycan-induced production of inflammatory mediators, the activation of the JNK and p38-MAPK, and the production of reactive oxygen species in keratinocytes. The results show that KATP channel blockers may reduce the bacterial component-stimulated production of inflammatory mediators in keratinocytes by suppressing the Toll-like receptor-2-mediated activation of the Akt, mTOR and NF-κB pathways, as well as JNK and p38-MAPK. The suppressive effect of KATP channel blockers appears to be achieved by the inhibition of reactive oxygen species production.
Collapse
|
37
|
Ribas JLC, Sherry JP, Zampronio AR, Silva de Assis HC, Simmons DBD. Inhibition of immune responses and related proteins in Rhamdia quelen exposed to diclofenac. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2017; 36:2092-2107. [PMID: 28106285 DOI: 10.1002/etc.3742] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/22/2016] [Accepted: 01/16/2017] [Indexed: 06/06/2023]
Abstract
Nonsteroidal anti-inflammatory drugs are among the most widely detected pharmaceuticals in surface water worldwide. The nonsteroidal anti-inflammatory drug diclofenac is used to treat many types of pain and inflammation. Diclofenac's potential to cause adverse effects in exposed wildlife is a growing concern. To evaluate the effects of waterborne diclofenac on the immune response in Rhamdia quelen (South American catfish), fish were exposed to 3 concentrations of diclofenac (0.2, 2.0, and 20.0 μg/L) for 14 d. Some of the exposed fish were also given an intraperitoneal injection on day 14 of 1 mg/kg of carrageenan to evaluate cell migration to the peritoneum. Total blood leukocyte count and carrageenan-induced leukocyte migration to the peritoneal cavity, particularly of polymorphonuclear cells, were significantly affected for all diclofenac exposure groups. Nitric oxide production was significantly reduced in the diclofenac-treated fish. Plasma and kidney proteins were analyzed by means of liquid chromatography-tandem mass spectrometry in a shotgun proteomic approach. In both plasma and kidney of diclofenac-exposed R. quelen, the expression of 20 proteins related to the inflammatory process, nitric oxide production, leukocyte migration, and the complement cascade was significantly altered. In addition, class I major histocompatibility complex was significantly decreased in plasma of diclofenac-treated fish. Thus, waterborne exposure to diclofenac could lead to suppression of the innate immune system in R. quelen. Environ Toxicol Chem 2017;36:2092-2107. © 2017 SETAC.
Collapse
Affiliation(s)
- João L C Ribas
- Department of Pharmacology, Federal University of Parana, Curitiba-Paraná, Brazil
- Department of Biomedicine, Positivo University, Curitiba-Paraná, Brazil
| | - James P Sherry
- Water Science and Technology, Aquatic Contaminants Research Division, Environment and Climate Change Canada, Burlington, Ontario, Canada
| | | | | | - Denina B D Simmons
- Water Science and Technology, Aquatic Contaminants Research Division, Environment and Climate Change Canada, Burlington, Ontario, Canada
| |
Collapse
|
38
|
Resveratrol inhibits urban particulate matter-induced COX-2/PGE 2 release in human fibroblast-like synoviocytes via the inhibition of activation of NADPH oxidase/ROS/NF-κB. Int J Biochem Cell Biol 2017; 88:113-123. [PMID: 28495310 DOI: 10.1016/j.biocel.2017.05.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 03/17/2017] [Accepted: 05/08/2017] [Indexed: 01/09/2023]
Abstract
Human fibroblast-like synoviocytes (FLSs) play a role in joint synovial inflammation in rheumatoid arthritis (RA). Some evidence indicates that particulate matter (PM) in air pollution could contribute to the progression of RA. However, more research is needed to clarify this relationship. Up-regulation of cyclooxygenase (COX)-2 and its metabolite prostaglandin E2 (PGE2) are implicated in various inflammatory diseases. Resveratrol, a polyphenol found mainly in grapes and red wine, has antioxidant and anti-inflammatory activities. In the present study, we demonstrated that resveratrol reduced PM-induced COX-2/PGE2 expression in human FLSs, and attenuated PM-enhanced NADPH oxidase activity and ROS generation. In addition, PM induced Akt, ERK1/2, or p38 MAPK activation, which was inhibited by resveratrol. Finally, we demonstrated that PM enhanced NF-κB p65 phosphorylation and the NF-κB promoter activity, which were reduced by pretreatment with a ROS inhibitor or resveratrol. Thus, we concluded that resveratrol functions as a suppressor of PM-induced inflammatory signaling pathways by inhibiting COX-2/PGE2 expression.
Collapse
|
39
|
Staphylococcus aureus Infection Reduces Nutrition Uptake and Nucleotide Biosynthesis in a Human Airway Epithelial Cell Line. Metabolites 2016; 6:metabo6040041. [PMID: 27834866 PMCID: PMC5192447 DOI: 10.3390/metabo6040041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022] Open
Abstract
The Gram positive opportunistic human pathogen Staphylococcus aureus induces a variety of diseases including pneumonia. S. aureus is the second most isolated pathogen in cystic fibrosis patients and accounts for a large proportion of nosocomial pneumonia. Inside the lung, the human airway epithelium is the first line in defence with regard to microbial recognition and clearance as well as regulation of the immune response. The metabolic host response is, however, yet unknown. To address the question of whether the infection alters the metabolome and metabolic activity of airway epithelial cells, we used a metabolomics approach. The nutrition uptake by the human airway epithelial cell line A549 was monitored over time by proton magnetic resonance spectroscopy (1H-NMR) and the intracellular metabolic fingerprints were investigated by gas chromatography and high performance liquid chromatography (GC-MS) and (HPLC-MS). To test the metabolic activity of the host cells, glutamine analogues and labelled precursors were applied after the infection. We found that A549 cells restrict uptake of essential nutrients from the medium after S. aureus infection. Moreover, the infection led to a shutdown of the purine and pyrimidine synthesis in the A549 host cell, whereas other metabolic routes such as the hexosamine biosynthesis pathway remained active. In summary, our data show that the infection with S. aureus negatively affects growth, alters the metabolic composition and specifically impacts the de novo nucleotide biosynthesis in this human airway epithelial cell model.
Collapse
|
40
|
Chen HM, Yang CM, Chang JF, Wu CS, Sia KC, Lin WN. AdipoR-increased intracellular ROS promotes cPLA2 and COX-2 expressions via activation of PKC and p300 in adiponectin-stimulated human alveolar type II cells. Am J Physiol Lung Cell Mol Physiol 2016; 311:L255-69. [PMID: 27288489 DOI: 10.1152/ajplung.00218.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 03/28/2016] [Indexed: 01/21/2023] Open
Abstract
Adiponectin, an adipokine, accumulated in lung system via T-cadherin after allergens/ozone challenge. However, the roles of adiponectin on lung pathologies were controversial. Here we reported that adiponectin stimulated expression of inflammatory proteins, cytosolic phospholipase A2 (cPLA2), cyclooxygenase-2 (COX-2), and production of reactive oxygen species (ROS) in human alveolar type II A549 cells. AdipoR1/2 involved in adiponectin-activated NADPH oxidase and mitochondria, which further promoted intracellular ROS accumulation. Protein kinase C (PKC) may involve an adiponectin-activated NADPH oxidase. Similarly, p300 phosphorylation and histone H4 acetylation occurred in adiponectin-challenged A549 cells. Moreover, adiponectin-upregulated cPLA2 and COX-2 expression was significantly abrogated by ROS scavenger (N-acetylcysteine) or the inhibitors of NADPH oxidase (apocynin), mitochondrial complex I (rotenone), PKC (Ro31-8220, Gö-6976, and rottlerin), and p300 (garcinol). Briefly, we reported that adiponectin stimulated cPLA2 and COX-2 expression via AdipoR1/2-dependent activation of PKC/NADPH oxidase/mitochondria resulting in ROS accumulation, p300 phosphorylation, and histone H4 acetylation. These results suggested that adiponectin promoted lung inflammation, resulting in exacerbation of pulmonary diseases via upregulating cPLA2 and COX-2 expression together with intracellular ROS production. Understanding the adiponectin signaling pathways on regulating cPLA2 and COX-2 may help develop therapeutic strategies on pulmonary diseases.
Collapse
Affiliation(s)
- Hsiao-Mei Chen
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Xinzhuang, New Taipei City, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo, Kwei-San, Tao-Yuan, Taiwan; Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| | - Jia-Feng Chang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, Xinzhuang, New Taipei City, Taiwan; Department of Internal Medicine, En-Chu-Kong Hospital, Sanxia, New Taipei City, Taiwan
| | - Chi-Sheng Wu
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Xinzhuang, New Taipei City, Taiwan
| | - Kee-Chin Sia
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Xinzhuang, New Taipei City, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Xinzhuang, New Taipei City, Taiwan
| |
Collapse
|
41
|
Fang P, Shi HY, Wu XM, Zhang YH, Zhong YJ, Deng WJ, Zhang YP, Xie M. Targeted inhibition of GATA-6 attenuates airway inflammation and remodeling by regulating caveolin-1 through TLR2/MyD88/NF-κB in murine model of asthma. Mol Immunol 2016; 75:144-50. [PMID: 27289030 DOI: 10.1016/j.molimm.2016.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 12/11/2022]
Abstract
The purpose of this study was to evaluate the effects of GATA-6 on airway inflammation and remodeling and the underlying mechanisms in a murine model of chronic asthma. Female BALB/c mice were randomly divided into four groups: phosphate-buffered saline control (PBS), ovalbumin (OVA)-induced asthma group (OVA), OVA+ siNC and OVA+ siGATA-6. In this mice model, GATA-6 expression level was significantly elevated and the expression in Caveolin-1 (Cav-1) inversely correlated with the abundance of GATA-6 in OVA-induced asthma of mice. Silencing of GATA-6 gene expression upregulated Cav-1 expression. Additionally, downregulation of GATA-6 dramatically decreased OVA-challenged inflammation, infiltration, and mucus production. Moreover, silencing of GATA-6 resulted in decreased levels of immunoglobulin E (IgE) and inflammatory mediators and reduced inflammatory cell accumulation, as well as inhibiting the expression of important mediators including matrix metalloproteinase (MMP)-2 and MMP-9, TGF-β1, and a disintegrin and metalloproteinase 8 (ADAM8) and ADAM33, which is related to airway remodeling. Further analysis confirmed that silencing of GATA-6 attenuated OVA-induced airway inflammation and remodeling through the TLR2/MyD88 and NF-κB pathway. In conclusion, these findings indicated that the downregulation of GATA-6 effectively inhibited airway inflammation and reversed airway remodeling via Cav-1, at least in part through downregulation of TLR2/MyD88/NF-κB, which suggests that GATA-6 represents a promising therapeutic strategy for human allergic asthma.
Collapse
Affiliation(s)
- Ping Fang
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China.
| | - Hong-Yang Shi
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China
| | - Xiao-Ming Wu
- Key Laboratory, Biochemical Information Engineering of Ministry of Education Xi'an Jiaotong University School of Life Science and Technology, Xi'an 710049, Shaanxi, PR China
| | - Yong-Hong Zhang
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China
| | - Yu-Jie Zhong
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China
| | - Wen-Jing Deng
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China
| | - Yu-Ping Zhang
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China
| | - Mei Xie
- Respiratory Department, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710004, Shaanxi, PR China
| |
Collapse
|
42
|
Hsu PS, Wu CS, Chang JF, Lin WN. Leptin Promotes cPLA₂ Gene Expression through Activation of the MAPK/NF-κB/p300 Cascade. Int J Mol Sci 2015; 16:27640-58. [PMID: 26593914 PMCID: PMC4661903 DOI: 10.3390/ijms161126045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/26/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
Hyperplasia or hypertrophy of adipose tissues plays a crucial role in obesity, which is accompanied by the release of leptin. Recently, obesity was determined to be associated with various pulmonary diseases including asthma, acute lung injury, and chronic obstructive pulmonary disease. However, how obesity contributes to pulmonary diseases and whether leptin directly regulates lung inflammation remains unclear. We used cell and animal models to study the mechanisms of leptin mediation of pulmonary inflammation. We found that leptin activated de novo synthesis of cytosolic phospholipase A2-α (cPLA2-α) in vitro in the lung alveolar type II cells, A549, and in vivo in ICR mice. Upregulated cPLA2-α protein was attenuated by pretreatment with an OB-R blocking antibody, U0126, SB202190, SP600125, Bay11-7086, garcinol, and p300 siRNA, suggesting roles of p42/p44 MAPK, p38 MAPK, JNK1/2, NF-κB, and p300 in leptin effects. Leptin enhanced the activities of p42/p44 MAPK, p38 MAPK, JNK1/2, and p65 NF-κB in a time-dependent manner. Additional studies have suggested the participation of OB-R, p42/p44 MAPK, and JNK1/2 in leptin-increased p65 phosphorylation. Furthermore, p300 phosphorylation and histone H4 acetylation were reduced by blockage of OB-R, p42/p44 MAPK, p38 MAPK, JNK1/2, and NF-κB in leptin-stimulated cells. Similarly, blockage of the MAPKs/NF-κB/p300 cascade significantly inhibited leptin-mediated cPLA2-α mRNA expression. Our data as a whole showed that leptin contributed to lung cPLA2-α expression through OB-R-dependent activation of the MAPKs/NF-κB/p300 cascade.
Collapse
Affiliation(s)
- Pei-Sung Hsu
- Division of Chest Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan.
| | - Chi-Sheng Wu
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, Xinzhuang 24205, Taiwan.
| | - Jia-Feng Chang
- Department of Internal Medicine, En-Chu-Kong Hospital, Sanxia 23702, Taiwan.
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, Xinzhuang 24205, Taiwan.
| | - Wei-Ning Lin
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, Xinzhuang 24205, Taiwan.
| |
Collapse
|
43
|
Hsu CK, Lin CC, Hsiao LD, Yang CM. Mevastatin ameliorates sphingosine 1-phosphate-induced COX-2/PGE2-dependent cell migration via FoxO1 and CREB phosphorylation and translocation. Br J Pharmacol 2015; 172:5360-76. [PMID: 26359950 DOI: 10.1111/bph.13326] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 08/19/2015] [Accepted: 09/03/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Sphingosine 1-phosphate (S1P), an important inflammatory mediator, has been shown to regulate COX-2 production and promote various cellular responses such as cell migration. Mevastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoA), effectively inhibits inflammatory responses. However, the mechanisms underlying S1P-evoked COX-2-dependent cell migration, which is modulated by mevastatin in human tracheal smooth muscle cells (HTSMCs) remain unclear. EXPERIMENTAL APPROACH The expression of COX-2 was determined by Western blotting, real time-PCR and promoter analyses. The signalling molecules were investigated by pretreatment with respective pharmacological inhibitors or transfection with siRNAs. The interaction between COX-2 promoter and transcription factors was determined by chromatin immunoprecipitation assay. Finally, the effect of mevastatin on HTSMC migration and leukocyte counts in BAL fluid and COX-2 expression induced by S1P was determined by a cell migration assay, cell counting and Western blot. KEY RESULTS S1P stimulated mTOR activation through the Nox2/ROS and PI3K/Akt pathways, which can further stimulate FoxO1 phosphorylation and translocation to the cytosol. We also found that S1P induced CREB activation and translocation via an mTOR-independent signalling pathway. Finally, we showed that pretreatment with mevastatin markedly reduced S1P-induced cell migration and COX-2/PGE2 production via a PPARγ-dependent signalling pathway. CONCLUSIONS AND IMPLICATIONS Mevastatin attenuates the S1P-induced increased expression of COX-2 and cell migration via the regulation of FoxO1 and CREB phosphorylation and translocation by PPARγ in HTSMCs. Mevastatin could be beneficial for prevention of airway inflammation in the future.
Collapse
Affiliation(s)
- Chih-Kai Hsu
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anaesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anaesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.,Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| |
Collapse
|
44
|
Yang CM, Lin CC, Lee IT, Hsu CK, Tai YC, Hsieh HL, Chi PL, Hsiao LD. c-Src-dependent transactivation of EGFR mediates CORM-2-induced HO-1 expression in human tracheal smooth muscle cells. J Cell Physiol 2015; 230:2351-61. [PMID: 25921464 DOI: 10.1002/jcp.24912] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 12/18/2014] [Indexed: 12/28/2022]
Abstract
Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, displays an anti-inflammatory effect in various cellular injuries, but the precise mechanisms of HO-1 expression remain unknown. We used the transition metal carbonyl compound carbon monoxide-releasing molecule-2 (CORM-2) that acts as carbon monoxide donor. The effects of CORM-2 on expression of HO-1 in human tracheal smooth muscle cells (HTSMCs) were determined by Western blot, real-time PCR, and promoter activity assay. In HTSMCs, CORM-2 activated Nrf2 through the activation of a c-Src/EGFR/PI3K/Akt-dependent pathway, resulting in HO-1 expression. We showed that CORM-2-induced HO-1 protein and mRNA levels were inhibited by the inhibitor of c-Src (PP1 or SU6656), EGFR (AG1478), PI3K (LY294002), Akt (SH-5), JNK1/2 (SP600125), or p38 MAPK (SB202190) and transfection with siRNA of c-Src, EGFR, Akt, p38, JNK2, or Nrf2 in HTSMCs. We also showed that CORM-2 stimulated c-Src, EGFR, Akt, p38 MAPK, and JNK1/2 phosphorylation. CORM-2 also enhanced Nrf2 translocation from the cytosol to the nucleus and antioxidant response element (ARE) promoter activity. Moreover, CORM-2 mediated p38 MAPK and JNK1/2 activation via a c-Src/EGFR/PI3K/Akt pathway, which further enhanced Nrf2 activation and translocation. Finally, we observed that CORM-2 induced in vivo binding of Nrf2 to the HO-1 promoter. CORM-2 activates the c-Src/EGFR/PI3K/Akt/JNK1/2 and p38 MAPK pathways, which in turn trigger Nrf2 activation and ultimately induces HO-1 expression in HTSMCs. Thus, the HO-1/CO system might be potential therapeutics in airway diseases.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - I-Ta Lee
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Kai Hsu
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Yu-Chen Tai
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Hsi-Lung Hsieh
- Department of Nursing, Division of Basic Medical Sciences, Chang Gung University of Science and Technology, Kwei-San, Tao-Yuan, Taiwan
| | - Pei-Ling Chi
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkuo and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| |
Collapse
|
45
|
Choi SJ, Kim MH, Jeon J, Kim OY, Choi Y, Seo J, Hong SW, Lee WH, Jeon SG, Gho YS, Jee YK, Kim YK. Active Immunization with Extracellular Vesicles Derived from Staphylococcus aureus Effectively Protects against Staphylococcal Lung Infections, Mainly via Th1 Cell-Mediated Immunity. PLoS One 2015; 10:e0136021. [PMID: 26333035 PMCID: PMC4558092 DOI: 10.1371/journal.pone.0136021] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 07/30/2015] [Indexed: 01/26/2023] Open
Abstract
Staphylococcus aureus is an important pathogenic bacterium that causes various infectious diseases. Extracellular vesicles (EVs) released from S. aureus contain bacterial proteins, nucleic acids, and lipids. These EVs can induce immune responses leading to similar symptoms as during staphylococcal infection condition and have the potential as vaccination agent. Here, we show that active immunization (vaccination) with S. aureus-derived EVs induce adaptive immunity of antibody and T cell responses. In addition, these EVs have the vaccine adjuvant ability to induce protective immunity such as the up-regulation of co-stimulatory molecules and the expression of T cell polarizing cytokines in antigen-presenting cells. Moreover, vaccination with S. aureus EVs conferred protection against lethality induced by airway challenge with lethal dose of S. aureus and also pneumonia induced by the administration of sub-lethal dose of S. aureus. These protective effects were also found in mice that were adoptively transferred with splenic T cells isolated from S. aureus EV-immunized mice, but not in serum transferred mice. Furthermore, this protective effect of S. aureus EVs was significantly reduced by the absence of interferon-gamma, but not by the absence of interleukin-17. Together, the study herein suggests that S. aureus EVs are a novel vaccine candidate against S. aureus infections, mainly via Th1 cellular response.
Collapse
Affiliation(s)
- Seng Jin Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Min-Hye Kim
- Department of Medicine, Ewha Womans University School of Medicine and Ewha Institute of Convergence Medicine, Ewha Womans Medical Center, Seoul, Republic of Korea
| | - Jinseong Jeon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Oh Youn Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Youngwoo Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jihye Seo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Sung-Wook Hong
- Academy of Immunology and Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Won-Hee Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Seong Gyu Jeon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- * E-mail: (YK); (SJ)
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Young-Koo Jee
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Yoon-Keun Kim
- Department of Medicine, Ewha Womans University School of Medicine and Ewha Institute of Convergence Medicine, Ewha Womans Medical Center, Seoul, Republic of Korea
- * E-mail: (YK); (SJ)
| |
Collapse
|
46
|
Cengiz M, Ozenirler S, Elbeg S. Role of serum toll-like receptors 2 and 4 in non-alcoholic steatohepatitis and liver fibrosis. J Gastroenterol Hepatol 2015; 30:1190-6. [PMID: 25684563 DOI: 10.1111/jgh.12924] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM Non-alcoholic fatty liver disease is a common cause of chronic liver disease, including non-alcoholic steatohepatitis (NASH). Our aim was to investigate whether serum toll-like receptors 2 and 4 (TLR2 and TLR4) levels are correlated with NASH and able to predict liver fibrosis, as well as to compare these markers with other non-invasive fibrosis scores (aspartate aminotransferase [AST] to alanine aminotransferase ratio, AST to platelet ratio index, fibrosis index, fibrosis 4, and fibrosis cirrhosis index). METHODS Serum samples were obtained from consecutive biopsy proven NASH patients and healthy controls. Serum TLR2 and TLR4 were measured using ELISA. Stage of fibrosis was evaluated using the Brunt Criteria. The different non-invasive fibrosis scores were compared using areas under the curve. RESULTS Fifty-seven patients with NASH and 57 healthy individuals were enrolled in the study. Serum TLR2 levels were not significantly different between the healthy controls and NASH patients. The medians were 3.88 ng/mL ± 0.29 versus 3.81 ng/mL ± 0.32, respectively (P = 0.587). In comparing the levels of TLR4 between groups, the medians were 1.05 ng/mL ± 0.13 versus 1.46 ng/mL ± 0.27, respectively (P < 0.001). In NASH patients, the levels of serum TLR4 increased with the stage of fibrosis: TLR4 medians were F0:1.01, F1:1.46, F2:2.14, F3:3.74, F4:5.83 (P < 0.001). TLR4 produced AUCs for ≥ F1, ≥ F2, and ≥ F3 of 0.862, 0.810, and 0.905, respectively (P < 0.001). TLR4 levels were more predictive than other non-invasive fibrosis scores in liver fibrosis. CONCLUSION Serum TLR4 levels but not TLR2 were elevated in NASH patients in comparison with healthy controls. And in NASH patients, serum TLR4 levels both correlated with and were able to predict liver fibrosis.
Collapse
Affiliation(s)
- Mustafa Cengiz
- Department of Gastroenterology, Dr. A.Y. Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | - Seren Ozenirler
- Department of Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Sehri Elbeg
- Department of Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
47
|
JANG JAEWOONG, KIM WONYONG, KIM KIJEONG, CHUNG SANGIN, SHIM YAEJIE, KIM SEOKMIN, YOON YOOSIK. Lipoteichoic acid upregulates NF-κB and proinflammatory cytokines by modulating β-catenin in bronchial epithelial cells. Mol Med Rep 2015; 12:4720-4726. [DOI: 10.3892/mmr.2015.3965] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 05/15/2015] [Indexed: 11/06/2022] Open
|
48
|
McGarry T, Veale DJ, Gao W, Orr C, Fearon U, Connolly M. Toll-like receptor 2 (TLR2) induces migration and invasive mechanisms in rheumatoid arthritis. Arthritis Res Ther 2015; 17:153. [PMID: 26055925 PMCID: PMC4495696 DOI: 10.1186/s13075-015-0664-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/27/2015] [Indexed: 01/09/2023] Open
Abstract
Introduction This study investigates the role of Toll-like receptor 2 (TLR2) in the regulation of migratory and invasive mechanisms in rheumatoid arthritis (RA). Methods Invasion, migration, matrix metalloproteinase (MMP)-1, -3 and tissue inhibitor of matrix metalloproteinase-3 (TIMP-3) expression, β-integrin binding, cytoskeletal rearrangement and Ras-related C3 botulinum toxin substrate 1 (Rac1) activation in response to a TLR2-ligand, Pam3CSK4 (1 μg/ml), in ex vivo RA synovial tissue explants, primary RA synovial fibroblasts (RASFC) and microvascular endothelial cells (HMVEC) were assessed by Transwell Matrigel™ invasion chambers, enzyme-linked immunosorbent assay (ELISA), multiplex adhesion binding assay, reverse transcription polymerase chain reaction (RT-PCR), F-actin immunofluorescent staining, matrigel synovial outgrowths, Rac1 pull-down assays/Western blot and zymography. β1-integrin expression in RA/control synovial tissue was assessed by immunohistology. The effect of Pam3CSK4 on cell migration, invasion, MMP-3 and Rac1 activation was examined in the presence or absence of anti-β1-integrin (10 μg/ml) or anti-IgG control (10 μg/ml). The effect of an anti-TLR-2 mAb (OPN301)(1 μg/ml) or immunoglobulin G (IgG) control (1 μg/ml) on RASFC migration and RA synovial tissue MMP activity was assessed by wound assays, ELISA and zymography. Results Pam3CSK4 significantly induced cell migration, invasion, MMP-1, MMP-3, MMP-2 and MMP-9 expression and induced the MMP-1/TIMP-3 and MMP-3/TIMP-3 ratio in RASFC and explants (p <0.05). β1-integrin expression was significantly higher in RA synovial tissue compared to controls (p <0.05). Pam3CSK4 specifically induced β1-integrin binding in RASFC (p <0.05), with no effect observed for β2-4, β6, αvβ5 or α5β1. Pam3CSK4 increased β1-integrin mRNA expression, Rac1 activation, RASFC outgrowths and altered cytoskeletal dynamic through induction of filopodia formation. Pam3CSK4-regulated cell migration and invasion processes, but not MMP-3, were inhibited in the presence of anti-β1-integrin (p <0.05), with no effect observed for anti-IgG control. Furthermore, anti-β1-integrin inhibited Pam3CSK4-induced Rac1 activation. Finally, blockade of TLR2 with OPN301 significantly decreased spontaneous release of MMP-3, MMP-2 and MMP-9 and increased TIMP-3 secretion from RA synovial explant cultures (p <0.05). Incubation of RASFC with OPN301 RA ex vivo conditioned media inhibited migration and invasion compared to IgG control. Conclusions TLR2 activation induces migrational and invasive mechanisms, which are critically involved in the pathogenesis of RA, suggesting TLR2 as a potential therapeutic target for the treatment of RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0664-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Trudy McGarry
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin Academic Health Care and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Douglas J Veale
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin Academic Health Care and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Wei Gao
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin Academic Health Care and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Carl Orr
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin Academic Health Care and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Ursula Fearon
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin Academic Health Care and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Mary Connolly
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin Academic Health Care and The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
49
|
Goy SD, Olling A, Neumann D, Pich A, Gerhard R. Human neutrophils are activated by a peptide fragment of Clostridium difficile toxin B presumably via formyl peptide receptor. Cell Microbiol 2015; 17:893-909. [PMID: 25529763 DOI: 10.1111/cmi.12410] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 01/23/2023]
Abstract
Clostridium difficile may induce antibiotic-associated diarrhoea and, in severe cases, pseudomembranous colitis characterized by tremendous neutrophil infiltration. All symptoms are caused by two exotoxins: TcdA and TcdB. We describe here the activation of isolated human blood neutrophils by TcdB and, moreover, by toxin fragments generated by limited proteolytical digestion. Kinetics and profiles of TcdB-induced rise in intracellular-free Ca(2+) and reactive oxygen species production were similar to that induced by fMLF, which activates the formyl peptide receptor (FPR) recognizing formylated bacterial peptide sequences. Transfection assays with the FPR-1 isoform hFPR26 in HEK293 cells, heterologous desensitization experiments and FPR inhibition via cyclosporine H strongly suggest activation of cells via FPR-1. Domain analyses revealed that the N-terminal glucosyltransferase domain of TcdB is a potent activator of FPR pointing towards an additional mechanism that might contribute to pathogenesis. This pro-inflammatory ligand effect can be triggered even by cleaved and, thus, non-cytotoxic toxin. In summary, we report (i) a ligand effect on neutrophils as completely new molecular mode of action, (ii) pathogenic potential of truncated or proteolytically cleaved 'non-cytotoxic' fragments and (iii) an interaction of the N-terminal glucosyltransferase domain instead of the C-terminal receptor binding domain of TcdB with target cells.
Collapse
Affiliation(s)
| | | | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical School, Germany
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Germany
| | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, Germany
| |
Collapse
|
50
|
Hsu CK, Lee IT, Lin CC, Hsiao LD, Yang CM. Sphingosine-1-Phosphate Mediates COX-2 Expression and PGE2/IL-6 Secretion via c-Src-Dependent AP-1 Activation. J Cell Physiol 2014; 230:702-15. [DOI: 10.1002/jcp.24795] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 09/05/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Chih-Kai Hsu
- Department of Physiology and Pharmacology and Health Aging Research Center; College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - I-Ta Lee
- Department of Physiology and Pharmacology and Health Aging Research Center; College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics; Chang Gung Memorial Hospital at Lin-Kou and College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics; Chang Gung Memorial Hospital at Lin-Kou and College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center; College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| |
Collapse
|