1
|
Patiño P, Gallego C, Martínez N, Rey A, Iregui C. Intranasal instillation of Pasteurella multocida lipopolysaccharide in rabbits causes interstitial lung damage. Res Vet Sci 2022; 152:115-126. [DOI: 10.1016/j.rvsc.2022.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/22/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023]
|
2
|
Zhang MM, An LY, Hu WX, Li ZY, Qiang YY, Zhao BY, Han TS, Wu CC. Mechanism of endometrial MUC2 in reproductive performance in mice through PI3K/AKT signaling pathway after lipopolysaccharide treatment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113177. [PMID: 35030527 DOI: 10.1016/j.ecoenv.2022.113177] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
The objective of this study was to investigate the effects of exposure to endotoxin on the reproductive performance of humans and animals in pregnancy and delivery period. Mucin is considered to play a critical role in protecting the tissue epithelium. At pregnancy period, the MUC2 expression of uterus in the High LPS group was significantly higher than that in the Control group. The glycosaminoglycans of gland cells were secreted into the uterine cavity to protect the uterus. Then, the MUC2 layer became thinner, and LPS entered the lamina propria of the uterus. The mRNA expression of tight junction proteins showed a marked drop, and morphological damage of the uterus occurred. Subsequently, the glycosaminoglycans of gland cells in the High LPS and Low LPS groups increased with the increasing LPS dose, and the damage to the endometrial epithelium was repaired in female mice at Day 5 postdelivery. A low dose of LPS activated the PI3K/AKT signaling pathways to increase the glycosaminoglycans particles, while a high dose of LPS inhibited the PI3K/AKT signaling pathway to decrease the glycosaminoglycans particles. Taken together, our results suggest that gland cells secreted glycosaminoglycans particles into the uterine cavity by exocytosis to increase the thickness of the mucus layer to protect the uterus and that this process was regulated by PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Ming-Ming Zhang
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Li-Yan An
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Wen-Xiang Hu
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Zhong-Yang Li
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Yu-Yun Qiang
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Bao-Yu Zhao
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Tie-Suo Han
- Lanzhou Chia Tai Food CO. LTD, Lanzhou 730000, Gansu, People's Republic of China
| | - Chen-Chen Wu
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China.
| |
Collapse
|
3
|
Lv X, Yao T, He R, He Y, Li M, Han Y, Zhang Y, Long L, Jiang G, Cheng X, Xie Y, Huang L, Peng Z, Hu G, Li Q, Tao L, Meng J. Protective Effect of Fluorofenidone Against Acute Lung Injury Through Suppressing the MAPK/NF-κB Pathway. Front Pharmacol 2022; 12:772031. [PMID: 34987397 PMCID: PMC8721041 DOI: 10.3389/fphar.2021.772031] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a severe disease that presents serious damage and excessive inflammation in lungs with high mortality without effective pharmacological therapy. Fluorofenidone (AKFPD) is a novel pyridone agent that has anti-fibrosis, anti-inflammation, and other pharmacological activities, while the effect of fluorofenidone on ALI is unclarified. Here, we elucidated the protective effects and underlying mechanism of fluorofenidone on lipopolysaccharide (LPS)-induced ALI. In this study, fluorofenidone alleviated lung tissue structure injury and reduced mortality, decreased the pulmonary inflammatory cell accumulation and level of inflammatory cytokines IL-1β, IL-6, and TNF-α in the bronchoalveolar lavage fluid, and attenuated pulmonary apoptosis in LPS-induced ALI mice. Moreover, fluorofenidone could block LPS-activated phosphorylation of ERK, JNK, and P38 and further inhibited the phosphorylation of IκB and P65. These results suggested that fluorofenidone can significantly contrast LPS-induced ALI through suppressing the activation of the MAPK/NF-κB signaling pathway, which indicates that fluorofenidone could be considered as a novel therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Xin Lv
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Yao
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Rongling He
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yijun He
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Mengyu Li
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Han
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhang
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lingzhi Long
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guoliang Jiang
- Department of Respirology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun Cheng
- Department of Respirology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China
| | - Ling Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Gaoyun Hu
- Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,Faculty of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qianbin Li
- Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,Faculty of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Jie Meng
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| |
Collapse
|
4
|
Huang J, Liu J, Chang G, Wang Y, Ma N, Roy AC, Shen X. Glutamine Supplementation Attenuates the Inflammation Caused by LPS-Induced Acute Lung Injury in Mice by Regulating the TLR4/MAPK Signaling Pathway. Inflammation 2021; 44:2180-2192. [PMID: 34160729 DOI: 10.1007/s10753-021-01491-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022]
Abstract
Bacterial infection is one of the main causes of bovine respiratory disease (BRD), which can cause tremendous losses for the herd farming industry worldwide. L-Glutamine (GLN), a neutral amino acid, has been reported to have anti-inflammatory properties. This study aims to explore the potential protective effects and mechanisms of GLN on acute lung injury (ALI) induced by lipopolysaccharide (LPS) in mice. Forty ICR mice were randomly divided into four groups (n = 10): a PBS intratracheal instillation group, a LPS intratracheal instillation group, a GLN gavage group, and a LPS+GLN group (GLN was given 1 h before the LPS stimulation). Twelve hours after LPS administration, the lung tissue and blood were collected. The results showed that the concentrations of IL-6, IL-8, and IL-1β; the protein abundance of the toll-like receptor 4 (TLR4), phosphorylated p38 (p-p38), phosphorylated ERK1/2 (p-ERK1/2), and phosphorylated JNK (p-JNK); and the expression level of genes associated with inflammation, such as IL-1β, IL-8, TNF-α, IL-6, TLR4, p38, ERK1/2, and JNK, were significantly increased in the LPS group compared with those in the PBS group. However, these increases were attenuated by GLN pretreatment in the LPS+GLN group. Furthermore, the pathological change of the structure of lung tissue from the LPS group was obvious compared to that from the PBS group; however, with GLN administration, these pathological changes were alleviated. Additionally, the secretion level of mucus and the percentage of positive MUC5AC staining on the epithelial surface area of the airway increased dramatically in the LPS group; however, GLN pretreatment in the LPS+GLN group markedly decreased these phenomena compared with that of the LPS group. These results indicate that GLN supplementation ameliorates LPS-induced ALI in mice and this effect may be mediated by the TLR4/MAPK signaling pathway.
Collapse
Affiliation(s)
- Jie Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Jing Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yan Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Nana Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Animesh Chadra Roy
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
5
|
Abstract
The lungs are constantly exposed to the external environment and are therefore vulnerable to insults that can cause infection and injury. Maintaining the integrity and barrier function of the lung epithelium requires complex interactions of multiple cell lineages. Elucidating the cellular players and their regulation mechanisms provides fundamental information to deepen understanding about the responses and contributions of lung stem cells. This Review focuses on advances in our understanding of mammalian alveolar epithelial stem cell subpopulations and discusses insights about the regeneration-specific cell status of alveolar epithelial stem cells. We also consider how these advances can inform our understanding of post-injury lung repair processes and lung diseases.
Collapse
Affiliation(s)
- Huijuan Wu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Nan Tang
- National Institute of Biological Sciences, Beijing 102206, China .,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
6
|
An L, Zhao J, Sun X, Zhou Y, Zhao Z. S-allylmercaptocysteine inhibits mucin overexpression and inflammation via MAPKs and PI3K-Akt signaling pathways in acute respiratory distress syndrome. Pharmacol Res 2020; 159:105032. [PMID: 32574825 PMCID: PMC7305891 DOI: 10.1016/j.phrs.2020.105032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 11/17/2022]
Abstract
Cytokine storm is an important cause of acute respiratory distress syndrome and multiple organ failure. Excessive secretion and accumulation of mucins on the surface of airway cause airway obstruction and exacerbate lung infections. MUC5AC and MUC5B are the main secreted mucins and overexpressed in various inflammatory responses. S-allylmercaptocysteine, a water-soluble organic sulfur compound extracted from garlic, has anti-inflammatory and anti-oxidative effects for various pulmonary diseases. The aim of this work was to investigate the therapeutic effects of SAMC on mucin overproduction and inflammation in 16HBE cells and LPS-induced ARDS mice. Results show that SAMC treatment ameliorated inflammatory cell infiltration and lung histopathological changes in the LPS-induced ARDS mice. SAMC also inhibited the expressions of MUC5AC and MUC5B, decreased the production of pro-inflammatory markers (IL-6, TNF-α, CD86 and IL-12) and increased the production of anti-inflammatory markers (IL-10, CD206 and TGF-β). These results confirm that SAMC had potential beneficial effects on suppressed hyperinflammation and mucin overexpression. Furthermore, SAMC exerted the therapeutic effects through the inhibition of phosphorylation of MAPKs and PI3K-Akt signaling pathways in the 16HBE cells and mice. Overall, our results demonstrate the effects of SAMC on the LPS-induced mucin overproduction and inflammation both in the 16HBE cells and mice.
Collapse
Affiliation(s)
- Lulu An
- School of Pharmaceutical Sciences, Cheelloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Jianxiong Zhao
- School of Basic Medical Sciences, Cheelloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China
| | - Xiao Sun
- School of Pharmaceutical Sciences, Cheelloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Yingying Zhou
- School of Pharmaceutical Sciences, Cheelloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhongxi Zhao
- School of Pharmaceutical Sciences, Cheelloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Shandong Key University Laboratory of Pharmaceutics & Drug Delivery Systems, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Shandong Engineering & Technology Research Center for Jujube Food and Drug, 44 West Wenhua Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
7
|
Ta BTT, Nguyen DL, Jala I, Dontumprai R, Plumworasawat S, Aighewi O, Ong E, Shawley A, Potriquet J, Saichua P, van Diepen A, Sripa B, Hokke CH, Suttiprapa S. Identification, recombinant protein production, and functional analysis of a M60-like metallopeptidase, secreted by the liver fluke Opisthorchis viverrini. Parasitol Int 2019; 75:102050. [PMID: 31901435 DOI: 10.1016/j.parint.2019.102050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/30/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
The carcinogenic liver fluke Opisthorchis viverrini (O. viverrini) is endemic in Thailand and neighboring countries including Laos PDR, Vietnam and Cambodia. Infections with O. viverrini lead to hepatobiliary abnormalities including bile duct cancer-cholangiocarcinoma (CCA). Despite decades of extensive studies, the underlying mechanisms of how this parasite survives in the bile duct and causes disease are still unclear. Therefore, this study aims to identify and characterize the most abundant protein secreted by the parasite. Proteomics and bioinformatics analysis revealed that the most abundant secretory protein is a metallopeptidase, named Ov-M60-like-1. This protein contains an N-terminal carbohydrate-binding domain and a C-terminal M60-like domain with a zinc metallopeptidase HEXXH motif. Further analysis by mass spectrometry revealed that Ov-M60-like-1 is N-glycosylated. Recombinant Ov-M60-like-1 (rOv-M60-like-1) expressed in Escherichia coli (E. coli) was able to digest bovine submaxillary mucin (BSM). The mucinase activity was inhibited by the ion chelating agent EDTA, confirming its metallopeptidase identity. The enzyme was active at temperatures ranging 25-37 °C in a broad pH range (pH 2-10). The identification of Ov-M60-like-1 mucinase as the major secretory protein of O. viverrini worms warrants further research into the role of this glycoprotein in the pathology induced by this carcinogenic worm.
Collapse
Affiliation(s)
- Binh T T Ta
- Tropical Medicine Graduate Program, Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - D Linh Nguyen
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Isabelle Jala
- Tropical Medicine Graduate Program, Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Rieofarng Dontumprai
- Department of Microbiology, Faculty of Science, Mahidol University - RAMA VI, Bangkok 10400, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Omorose Aighewi
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Emily Ong
- Occidental College, 1600 Campus Road, Los Angeles, CA 90041, USA
| | - Audrey Shawley
- Occidental College, 1600 Campus Road, Los Angeles, CA 90041, USA
| | - Jeremy Potriquet
- Australian Institute of Tropical Health & Medicine, James Cook University, Douglas, QLD 4814, Australia
| | - Prasert Saichua
- Tropical Medicine Graduate Program, Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Angela van Diepen
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Banchob Sripa
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Sutas Suttiprapa
- Tropical Medicine Graduate Program, Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
8
|
Garić D, De Sanctis JB, Dumut DC, Shah J, Peña MJ, Youssef M, Petrof BJ, Kopriva F, Hanrahan JW, Hajduch M, Radzioch D. Fenretinide favorably affects mucins (MUC5AC/MUC5B) and fatty acid imbalance in a manner mimicking CFTR-induced correction. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158538. [PMID: 31678518 DOI: 10.1016/j.bbalip.2019.158538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/28/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Cystic fibrosis (CF) is the most common genetic disease in Caucasians. CF is manifested by abnormal accumulation of mucus in the lungs, which serves as fertile ground for the growth of microorganisms leading to recurrent infections and ultimately, lung failure. Mucus in CF patients consists of DNA from dead neutrophils as well as mucins produced by goblet cells. MUC5AC mucin leads to pathological plugging of the airways whereas MUC5B has a protective role against bacterial infection. Therefore, decreasing the level of MUC5AC while maintaining MUC5B intact would in principle be a desirable mucoregulatory treatment outcome. Fenretinide prevented the lipopolysaccharide-induced increase of MUC5AC gene expression, without affecting the level of MUC5B, in a lung goblet cell line. Additionally, fenretinide treatment reversed the pro-inflammatory imbalance of fatty acids by increasing docosahexaenoic acid and decreasing the levels of arachidonic acid in a lung epithelial cell line and primary leukocytes derived from CF patients. Furthermore, for the first time we also demonstrate the effect of fenretinide on multiple unsaturated fatty acids, as well as differential effects on the levels of long- compared to very-long-chain saturated fatty acids which are important substrates of complex phospholipids. Finally, we demonstrate that pre-treating mice with fenretinide in a chronic model of P. aeruginosa lung infection efficiently decreases the accumulation of mucus. These findings suggest that fenretinide may offer a new approach to therapeutic modulation of pathological mucus production in CF.
Collapse
Affiliation(s)
- Dušan Garić
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Program in Infectious Diseases and Immunity in Global Health, McGill University Health Center, Montreal, Quebec, Canada
| | - Juan B De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic; Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Bolivarian Republic of Venezuela
| | - Daciana Catalina Dumut
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Program in Infectious Diseases and Immunity in Global Health, McGill University Health Center, Montreal, Quebec, Canada
| | - Juhi Shah
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Program in Infectious Diseases and Immunity in Global Health, McGill University Health Center, Montreal, Quebec, Canada
| | - Maria Johanna Peña
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Bolivarian Republic of Venezuela
| | - Mina Youssef
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Program in Infectious Diseases and Immunity in Global Health, McGill University Health Center, Montreal, Quebec, Canada
| | - Basil J Petrof
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Francisek Kopriva
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - John W Hanrahan
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Danuta Radzioch
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Program in Infectious Diseases and Immunity in Global Health, McGill University Health Center, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Kaku N, Morinaga Y, Takeda K, Kosai K, Uno N, Hasegawa H, Miyazaki T, Izumikawa K, Mukae H, Yanagihara K. Efficacy and pharmacokinetics of ME1100, a novel optimized formulation of arbekacin for inhalation, compared with amikacin in a murine model of ventilator-associated pneumonia caused by Pseudomonas aeruginosa. J Antimicrob Chemother 2017; 72:1123-1128. [PMID: 27999047 DOI: 10.1093/jac/dkw517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/02/2016] [Indexed: 01/01/2023] Open
Abstract
Background Arbekacin is an aminoglycoside that shows strong antimicrobial activity against Gram-positive bacteria, including MRSA, as well as Pseudomonas aeruginosa . The therapeutic effectiveness of arbekacin is directly related to C max at the infection site. To maximize drug delivery to the respiratory tract and minimize the systemic toxicity, arbekacin optimized for inhalation, ME1100, is under development. In this study, we investigated the efficacy and pharmacokinetics of ME1100 in a murine model of ventilator-associated pneumonia caused by P. aeruginosa by using a customized investigational nebulizer system. Methods The mice were treated for 5 min, once daily, with placebo, 3, 10 or 30 mg/mL ME1100 or 30 mg/mL amikacin. Results In the survival study, the survival rate was significantly improved in the 10 and 30 mg/mL ME1100 treatment groups compared with that in the placebo group. The number of bacteria in the lungs was significantly lower in the 30 mg/mL ME1100 treatment group at 6 h after the initial treatment, compared with all other groups. In the pharmacokinetic study, the C max in the 30 mg/mL ME1100 treatment group in the epithelial lining fluid (ELF) and plasma was 31.1 and 1.2 mg/L, respectively. Furthermore, we compared the efficacy of ME1100 with that of amikacin. Although there were no significant differences in ELF and plasma concentrations between 30 mg/mL of ME1100 and 30 mg/mL of amikacin, ME1100 significantly improved the survival rate compared with amikacin. Conclusions The results of our study demonstrated the in vivo effectiveness of ME1100 and its superiority to amikacin.
Collapse
Affiliation(s)
- Norihito Kaku
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Yoshitomo Morinaga
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Kazuaki Takeda
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan.,Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Kosuke Kosai
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Naoki Uno
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Hiroo Hasegawa
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Taiga Miyazaki
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Koichi Izumikawa
- Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Hiroshi Mukae
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki city, Nagasaki, Japan
| |
Collapse
|
10
|
Efficacy of High-Dose Meropenem (Six Grams per Day) in Treatment of Experimental Murine Pneumonia Induced by Meropenem-Resistant Pseudomonas aeruginosa. Antimicrob Agents Chemother 2016; 61:AAC.02056-16. [PMID: 27799201 DOI: 10.1128/aac.02056-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/22/2016] [Indexed: 02/06/2023] Open
Abstract
High-dose meropenem (MEPM; 6 g/day) has been approved as a treatment for purulent meningitis; however, little is known regarding its in vivo efficacy in refractory lower respiratory tract infections. The purpose of this study was to evaluate the efficacy of MEPM at 6 g/day in a murine model of severe pneumonia caused by MEPM-resistant Pseudomonas aeruginosa Experimental pneumonia induced by MEPM-resistant P. aeruginosa was treated with normal-dose MEPM (150 mg/kg of body weight, simulating a 3-g/day regimen in humans) or high-dose MEPM (500 mg/kg, simulating a 6-g/day regimen in humans). Mice treated with high-dose MEPM showed significantly restored survival relative to that of untreated mice and tended to show a survival rate higher than that of mice treated with normal-dose MEPM. The viable bacterial counts (of two clinical isolates) in the lungs decreased significantly in mice treated with high-dose MEPM from those for untreated mice (P < 0.001) or mice treated with normal-dose MEPM (P, <0.01 and <0.05). The number of inflammatory cells in the bronchoalveolar lavage fluid (BALF) was also significantly lower in mice treated with high-dose MEPM than in untreated mice. The free MEPM concentration in the epithelial lining fluid (ELF) exceeded 16 μg/ml for 85 min in mice treated with high-dose MEPM, but not for mice treated with normal-dose MEPM. Our results demonstrate that high-dose MEPM (6 g/day) might provide better protection against pneumonia caused by MEPM-resistant strains of P. aeruginosa than the dose normally administered (less than 3 g/day).
Collapse
|
11
|
Moncla BJ, Chappell CA, Debo BM, Meyn LA. The Effects of Hormones and Vaginal Microflora on the Glycome of the Female Genital Tract: Cervical-Vaginal Fluid. PLoS One 2016; 11:e0158687. [PMID: 27437931 PMCID: PMC4954690 DOI: 10.1371/journal.pone.0158687] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022] Open
Abstract
In this study, we characterized the glycome of cervical-vaginal fluid, collected with a Catamenial cup. We quantified: glycosidase levels; sialic acid and high mannose specific lectin binding; mucins, MUC1, MUC4, MUC5AC, MUC7; and albumin in the samples collected. These data were analyzed in the context of hormonal status (day of menstrual cycle, hormonal contraception use) and role, if any, of the type of the vaginal microflora present. When the Nugent score was used to stratify the subjects by microflora as normal, intermediate, or bacterial vaginosis, several important differences were observed. The activities of four of six glycosidases in the samples from women with bacterial vaginosis were significantly increased when compared to normal or intermediate women: sialidase, P = <0.001; α-galactosidase, P = 0.006; β-galactosidase, P = 0.005; α-glucosidase, P = 0.056. Sialic acid binding sites as measured by two lectins, Maackia amurensis and Sambucus nigra binding, were significantly lower in women with BV compared to women with normal and intermediate scores (P = <0.0001 and 0.008 respectively). High mannose binding sites, a measure of innate immunity were also significantly lower in women with BV (P = <0.001). Additionally, we observed significant increases in MUC1, MUC4, MUC5AC, and MUC7 concentrations in women with BV (P = <0.001, 0.001, <0.001, 0.02 respectively). Among normal women we found that the membrane bound mucin MUC4 and the secreted MUC5AC were decreased in postmenopausal women (P = 0.02 and 0.07 respectively), while MUC7 (secreted) was decreased in women using levonorgestrel-containing IUDs (P = 0.02). The number of sialic acid binding sites was lower in the postmenopausal group (P = 0.04), but the number of high mannose binding sites, measured with Griffithsin, was not significantly different among the 6 hormonal groups. The glycosidase levels in the cervical-vaginal mucus were rather low in the groups, with exception of α-glucosidase activity that was much lower in the postmenopausal group (P<0.001). These studies present compelling evidence that the vaginal ecosystem responds to the presence of different vaginal microorganisms. These effects were so influential that it required us to remove subjects with BV for data interpretation of the impact of hormones. We also suggest that certain changes occurring in vaginal/cervical proteins are due to bacteria or their products. Therefore, the quantitation of vaginal mucins and lectin binding offers a new method to monitor bacteria-host interactions in the female reproductive tract. The data suggest that some of the changes in these components are the result of host processing, such as the increases in mucin content, while the microflora is responsible for the increases in glycosidases and the decreases in lectin binding. The methods should be considered a valid marker for insult to the female genital tract.
Collapse
Affiliation(s)
- Bernard J. Moncla
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Catherine A. Chappell
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Brian M. Debo
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Leslie A. Meyn
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
12
|
Toll-Like Receptor 4 Agonistic Antibody Promotes Host Defense against Chronic Pseudomonas aeruginosa Lung Infection in Mice. Infect Immun 2016; 84:1986-1993. [PMID: 27091927 DOI: 10.1128/iai.01384-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 04/11/2016] [Indexed: 12/22/2022] Open
Abstract
Chronic lower respiratory tract infection with Pseudomonas aeruginosa is difficult to treat due to enhanced antibiotic resistance and decreased efficacy of drug delivery to destroyed lung tissue. To determine the potential for restorative immunomodulation therapies, we evaluated the effect of Toll-like receptor 4 (TLR4) stimulation on the host immune response to Pseudomonas infection in mice. We implanted sterile plastic tubes precoated with P. aeruginosa in the bronchi of mice, administered the TLR4/MD2 agonistic monoclonal antibody UT12 intraperitoneally every week, and subsequently analyzed the numbers of viable bacteria and inflammatory cells and the levels of cytokines. We also performed flow cytometry-based phagocytosis and opsonophagocytic killing assays in vitro using UT12-treated murine peritoneal neutrophils. UT12-treated mice showed significantly enhanced bacterial clearance, increased numbers of Ly6G(+) neutrophils, and increased concentrations of macrophage inflammatory protein 2 (MIP-2) in the lungs (P < 0.05). Depletion of CD4(+) T cells eliminated the ability of the UT12 treatment to improve bacterial clearance and promote neutrophil recruitment and MIP-2 production. Additionally, UT12-pretreated peritoneal neutrophils exhibited increased opsonophagocytic killing activity via activation of the serine protease pathway, specifically neutrophil elastase activity, in a TLR4-dependent manner. These data indicated that UT12 administration significantly augmented the innate immune response against chronic bacterial infection, in part by promoting neutrophil recruitment and bactericidal function.
Collapse
|
13
|
Tulbah AS, Ong HX, Colombo P, Young PM, Traini D. Could simvastatin be considered as a potential therapy for chronic lung diseases? A debate on the pros and cons. Expert Opin Drug Deliv 2016; 13:1407-20. [PMID: 27212150 DOI: 10.1080/17425247.2016.1193150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Simvastatin (SV) is a drug from the statin class, currently used orally as an anti-cholesterolemic drug. It inhibits the 3-hydroxy-3-methyl-glutaryl-Coenzyme A (HMG-CoA) reductase to reduce cholesterol synthesis. Recently, it has been found that SV also has several other protective pharmacological actions unrelated to its anti-cholesterol effects that might be beneficial in the treatment of chronic airway diseases. AREAS COVERED This review summarizes the evidence relating to SV as a potential anti-inflammatory, anti-oxidant and muco-inhibitory agent, administered both orally and via pulmonary inhalation, and discusses its pro and cons. Evidence could potentially be used to support the delivery of SV as inhaled formulation for the treatment of chronic respiratory diseases. EXPERT OPINION The use of SV as anti-inflammatory, anti-oxidant and muco-inhibitory agent for drug delivery to the lung is promising. Inhaled SV formulations could allow the delivery profile to be customized and optimized to take advantage of the rapid onset of action, low systemic side effect and improved physico-chemical stability. This treatment could potentially to be used clinically for the localized treatment of lung diseases where inflammation and oxidative stress production is present.
Collapse
Affiliation(s)
- Alaa S Tulbah
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia.,b Faculty of Pharmacy , Umm Al Qura University , Makkah , Saudi Arabia
| | - Hui Xin Ong
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia
| | - Paolo Colombo
- c Department of Pharmacy , University of Parma , Parma , Italy
| | - Paul M Young
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia
| | - Daniela Traini
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School , Sydney University , Australia
| |
Collapse
|
14
|
Seki M, Sakata T, Toyokawa M, Nishi I, Tomono K. A Chronic Respiratory Pasteurella multocida Infection Is Well-Controlled by Long-Term Macrolide Therapy. Intern Med 2016; 55:307-10. [PMID: 26831030 DOI: 10.2169/internalmedicine.55.4929] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 57-year-old woman with severe bronchiectasis frequently received antibiotics, including penicillin, for acute exacerbations due to Pasteurella multocida. Although the bacteria showed a decrease in antibiotic susceptibility, her symptoms and X-ray findings became stable, and severe exacerbations were not observed for the last few years after a low-dose erythromycin treatment was started. The development of a respiratory infection with Pasteurella multocida is relatively uncommon, but it can be controlled by immunomodulation which is associated with long-term macrolide therapy.
Collapse
Affiliation(s)
- Masafumi Seki
- Division of Infection Control and Prevention, Osaka University Hospital, Japan
| | | | | | | | | |
Collapse
|
15
|
Adam D, Roux-Delrieu J, Luczka E, Bonnomet A, Lesage J, Mérol JC, Polette M, Abély M, Coraux C. Cystic fibrosis airway epithelium remodelling: involvement of inflammation. J Pathol 2014; 235:408-19. [DOI: 10.1002/path.4471] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/02/2014] [Accepted: 10/21/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Damien Adam
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
| | - Jacqueline Roux-Delrieu
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
| | - Emilie Luczka
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
| | - Arnaud Bonnomet
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
| | - Julien Lesage
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
| | | | - Myriam Polette
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
- Laboratory of Histology; University Hospital Centre; Reims France
| | - Michel Abély
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
- Pediatric Unit A, American Memorial Hospital; University Hospital Centre; Reims France
| | - Christelle Coraux
- INSERM UMR-S 903, SFR CAP-SANTE (FED 4231); University of Reims Champagne-Ardenne; Reims France
| |
Collapse
|
16
|
Yamada K, Morinaga Y, Yanagihara K, Kaku N, Harada Y, Uno N, Nakamura S, Imamura Y, Hasegawa H, Miyazaki T, Izumikawa K, Kakeya H, Mikamo H, Kohno S. Azithromycin inhibits MUC5AC induction via multidrug-resistant Acinetobacter baumannii in human airway epithelial cells. Pulm Pharmacol Ther 2014; 28:165-70. [DOI: 10.1016/j.pupt.2014.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 05/21/2014] [Accepted: 05/24/2014] [Indexed: 10/25/2022]
|
17
|
Harada Y, Morinaga Y, Kaku N, Nakamura S, Uno N, Hasegawa H, Izumikawa K, Kohno S, Yanagihara K. In vitro and in vivo activities of piperacillin-tazobactam and meropenem at different inoculum sizes of ESBL-producing Klebsiella pneumoniae. Clin Microbiol Infect 2014; 20:O831-9. [PMID: 24813594 DOI: 10.1111/1469-0691.12677] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 01/21/2023]
Abstract
The inoculum effect is a laboratory phenomenon in which the minimal inhibitory concentration (MIC) of an antibiotic is increased when a large number of organisms are exposed. Due to the emergence of extended-spectrum β-lactamase-producing Klebsiella pneumoniae (ESBL-Kpn) infections, the inoculum effect of ESBL-Kpn on β-lactams was studied in vitro and in vivo using an experimental model of pneumonia. The in vitro inoculum effect of 45 clinical ESBL-Kpn isolates on β-lactams was evaluated at standard (10(5) CFU/mL) and high (10(7) CFU/mL) organism concentrations. The MIC50 of piperacillin-tazobactam, cefotaxime and cefepime was increased eight-fold or more and that of meropenem was increased two-fold. The in vivo inoculum effect was evaluated in an ESBL-Kpn pneumonia mouse model treated with bacteriostatic effect-adjusted doses of piperacillin-tazobactam (1000 mg/kg four times daily, %T>MIC; 32.60%) or meropenem (100 mg/kg twice daily, %T>MIC; 28.65%) at low/standard (10(4) CFU/mouse) and high (10(6) CFU/mouse) inocula. In mice administered a low inoculum, no mice died after treatment with piperacillin-tazobactam or meropenem, whereas all the control mice died. In contrast, in the high inoculum model, all mice in the piperacillin-tazobactam-treated group died, whereas all meropenem-treated mice survived and had a decreased bacterial load in the lungs and no invasion into the blood. In conclusion, meropenem was more resistant to the inoculum effect of ESBL-Kpn than piperacillin-tazobactam both in vitro and in vivo. In the management of severe pneumonia caused by ESBL-Kpn, carbapenems may be the drugs of choice to achieve a successful outcome.
Collapse
Affiliation(s)
- Y Harada
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dong S, Zhong Y, Yang K, Xiong X, Mao B. Intervention effect and dose-dependent response of tanreqing injection on airway inflammation in lipopolysaccharide-induced rats. J TRADIT CHIN MED 2014; 33:505-12. [PMID: 24187873 DOI: 10.1016/s0254-6272(13)60156-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To assess the effect of Tanreqing injection on airway inflammation in rats. METHODS A rat model of airway inflammation was generated with lipopolysaccharide (LPS). Tanreqing injection was given by intratracheal instillation, and bronchoalveolar lavage fluid (BALF) from the right lung was collected. BALF total cell and neutrophil counts were then determined. In addition, BALF levels of inflammatory cytokines interleukin-13, cytokine-induced neutrophil chemoat-tractant-1, and tumor necrosis factor-alpha were measured using enzyme linked immunosorbent assay. The middle lobe of the right lung was stained with hematoxylin-eosin and histological changes examined. RESULTS LPS increased airway inflammation, decreased BALF inflammatory cell count, inflammatory cytokine levels, and suppressed leukocyte influx of the lung. The LPS-induced airway inflammation peaked at 24 h, decreased beginning at 48 h, and had decreased markedly by 96 h. CONCLUSION Tanreqing injection contains anti-inflammatory properties, and inhibits airway inflammation in a dose-dependent manner.
Collapse
Affiliation(s)
- Shoujin Dong
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu 610041, China.
| | | | | | | | | |
Collapse
|
19
|
Yamada K, Yanagihara K, Kaku N, Harada Y, Migiyama Y, Nagaoka K, Morinaga Y, Nakamura S, Imamura Y, Miyazaki T, Izumikawa K, Kakeya H, Hasegawa H, Yasuoka A, Kohno S. In vivo efficacy of biapenem with ME1071, a novel metallo-β-lactamase (MBL) inhibitor, in a murine model mimicking ventilator-associated pneumonia caused by MBL-producing Pseudomonas aeruginosa. Int J Antimicrob Agents 2013; 42:238-43. [PMID: 23891525 DOI: 10.1016/j.ijantimicag.2013.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/27/2013] [Accepted: 05/29/2013] [Indexed: 10/26/2022]
Abstract
ME1071, a maleic acid derivative, is a novel, specific inhibitor of metallo-β-lactamases (MBLs). In vitro, ME1071 can potentiate the activity of carbapenems against MBL-producing Pseudomonas aeruginosa. To confirm the clinical efficacy of ME1071 in ventilator-associated pneumonia (VAP) caused by MBL-producing P. aeruginosa, a mouse model that mimics VAP by placement of a plastic tube in the bronchus was used. Biapenem (100 mg/kg) or ME1071 plus biapenem (each 100 mg/kg) was administered intraperitoneally every 12 h beginning at 12 h after inoculation. Survival was evaluated over 7 days. At 30 h post infection, mice were sacrificed and the numbers of viable bacteria in the lungs and bronchoalveolar lavage fluid (BALF) were compared. Histopathological analysis of lung specimens was also performed. The pharmacokinetics of ME1071 was analysed after initial treatment. The ME1071 plus biapenem combination group displayed significantly longer survival compared with the control and biapenem monotherapy groups (P<0.05). Furthermore, the number of viable bacteria in the lungs was significantly lower in the combination group (P<0.05). Histopathological examination of lung specimens indicated that progression of lung inflammation was prevented in the combination group. Furthermore, total cell and neutrophil counts, as well as cytokine levels, in BALF were significantly decreased (P<0.05) in the combination group. The percentage time above the MIC (%T>MIC) for biapenem without ME1071 was 0% in plasma; however, this value was elevated to 10.8% with ME1071. These results suggest that ME1071 is potent and effective for treatment of VAP caused by MBL-producing P. aeruginosa.
Collapse
Affiliation(s)
- Koichi Yamada
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wu H, Liu Z, Ling G, Lawrence D, Ding X. Transcriptional suppression of CYP2A13 expression by lipopolysaccharide in cultured human lung cells and the lungs of a CYP2A13-humanized mouse model. Toxicol Sci 2013; 135:476-85. [PMID: 23884085 DOI: 10.1093/toxsci/kft165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
CYP2A13, a human P450 enzyme preferentially expressed in the respiratory tract, is highly efficient in the metabolic activation of tobacco-specific nitrosamines. The aim of this study was to test the hypothesis that inflammation suppresses CYP2A13 expression in the lung, thus explaining the large interindividual differences in CYP2A13 levels previously found in human lung biopsy samples. We first demonstrated that the bacterial endotoxin lipopolysaccharide (LPS) and the proinflammatory cytokine IL-6 can suppress CYP2A13 messenger RNA (mRNA) expression in the NCI-H441 human lung cell line. We then report that an ip injection of LPS (1mg/kg), which induces systemic and lung inflammation, caused substantial reductions in CYP2A13 mRNA (~50%) and protein levels (~80%) in the lungs of a newly generated CYP2A13-humanized mouse model. We further identified two critical CYP2A13 promoter regions, one (major) between -484 and -1008bp and the other (minor) between -134 and -216bp, for the response to LPS, through reporter gene assays in H441 cells. The potential involvement of the nuclear factor NF-κB in LPS-induced CYP2A13 downregulation was suggested by identification of putative NF-κB binding sites within the LPS response regions and effects of an NF-κB inhibitor (pyrrolidine dithiocarbamate) on CYP2A13 expression in H441 cells. Results from gel shift assays further confirmed binding of NF-κB-like nuclear proteins of H441 cells to the major LPS response region of the CYP2A13 promoter. Thus, our findings strongly support the hypothesis that CYP2A13 levels in human lung can be suppressed by inflammation associated with disease status in tissue donors, causing underestimation of CYP2A13 levels in healthy lung.
Collapse
Affiliation(s)
- Hong Wu
- * Wadsworth Center, New York State Department of Health, Albany, New York 12201
| | | | | | | | | |
Collapse
|
21
|
Azithromycin attenuates lung inflammation in a mouse model of ventilator-associated pneumonia by multidrug-resistant Acinetobacter baumannii. Antimicrob Agents Chemother 2013; 57:3883-8. [PMID: 23733468 DOI: 10.1128/aac.00457-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Acinetobacter baumannii is one of the main pathogens that cause ventilator-associated pneumonia (VAP) and is associated with a high rate of mortality. Little is known about the efficacy of macrolides against A. baumannii. In order to confirm the efficacy of azithromycin (AZM) against VAP caused by multidrug-resistant A. baumannii (MDRAB), we used a mouse model that mimics VAP by placement of a plastic tube in the bronchus. AZM (10 and 100 mg/kg of body weight) was administered subcutaneously every 24 h beginning at 3 h after inoculation. Phosphate-buffered saline was administered as the control. Survival was evaluated over 7 days. At 48 h postinfection, mice were sacrificed and the numbers of viable bacteria in lungs and bronchoalveolar lavage fluid were compared. Histopathological analysis of lung specimens was also performed. The treatment groups displayed significantly longer survival than the control group (P < 0.05). AZM did not have an antimicrobial effect. Histopathological examination of lung specimens indicated that the progression of lung inflammation was prevented in the AZM-treated groups. Furthermore, total cell and neutrophil counts, as well as cytokine levels, in bronchoalveolar lavage fluid were significantly decreased (P < 0.05) in the AZM-treated groups. AZM may have a role for the treatment of VAP with MDRAB because of its anti-inflammatory effects.
Collapse
|
22
|
Toll-like receptor 4 agonistic antibody promotes innate immunity against severe pneumonia induced by coinfection with influenza virus and Streptococcus pneumoniae. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:977-85. [PMID: 23637040 DOI: 10.1128/cvi.00010-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Coinfection with bacteria is a major cause of mortality during influenza epidemics. Recently, Toll-like receptor (TLR) agonists were shown to have immunomodulatory functions. In the present study, we investigated the effectiveness and mechanisms of the new TLR4 agonistic monoclonal antibody UT12 against secondary pneumococcal pneumonia induced by coinfection with influenza virus in a mouse model. Mice were intranasally inoculated with Streptococcus pneumoniae 2 days after influenza virus inoculation. UT12 was intraperitoneally administered 2 h before each inoculation. Survival rates were significantly increased and body weight loss was significantly decreased by UT12 administration. Additionally, the production of inflammatory mediators was significantly suppressed by the administration of UT12. In a histopathological study, pneumonia in UT12-treated mice was very mild compared to that in control mice. UT12 increased antimicrobial defense through the acceleration of macrophage recruitment into the lower respiratory tract induced by c-Jun N-terminal kinase (JNK) and nuclear factor kappaB (NF-κB) pathway-dependent monocyte chemoattractant protein 1 (MCP-1) production. Collectively, these findings indicate that UT12 promoted pulmonary innate immunity and may reduce the severity of severe pneumonia induced by coinfection with influenza virus and S. pneumoniae. This immunomodulatory effect of UT12 improves the prognosis of secondary pneumococcal pneumonia and makes UT12 an attractive candidate for treating severe infectious diseases.
Collapse
|
23
|
Macrophages are related to goblet cell hyperplasia and induce MUC5B but not MUC5AC in human bronchus epithelial cells. J Transl Med 2012; 92:937-48. [PMID: 22391959 DOI: 10.1038/labinvest.2012.15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Airway goblet cell hyperplasia (GCH)--detectable by mucin staining--and abnormal macrophage infiltrate are pathological features present in many chronic respiratory disorders. However, it is unknown if both factors are associated. Using in-vivo and in-vitro models, we investigated whether macrophages are related with GCH and changes in mucin immunophenotypes. Lung sections from Sprague-Dawley rats treated for 48 h with one intra-tracheal dose of PBS or LPS (n=4-6 per group) were immunophenotyped for rat-goblet cells, immune, and proliferation markers. Human monocyte-derived macrophages (MDM) were pre-treated with or without LPS, immunophenotyped, and their supernatant, as well as cytokines at levels equivalent to supernatant were used to challenge primary culture of normal human bronchus epithelial cells (HBEC) in air-liquid interface, followed by MUC5B and MUC5AC mucin immunostaining. An association between increased bronchiolar goblet cells and terminal-bronchiolar proliferative epithelial cells confirmed the presence of GCH in our LPS rat model, which was related with augmented bronchiolar CD68 macrophage infiltration. The in-vitro experiments have shown that MUC5AC phenotype was inhibited when HBEC were challenged with supernatant from MDM pre-treated with or without LPS. In contrast, TNF-α and interleukin-1β at levels equivalent to supernatant from LPS-treated MDM increased MUC5AC. MUC5B was induced by LPS, supernatant from LPS-treated MDM, a mix of cytokines including TNF-α and TNF-α alone at levels present in supernatant from LPS-treated MDM. We demonstrated that macrophages are related with bronchiolar GCH, and that they induced MUC5B and inhibited MUC5AC in HBEC, suggesting a role for them in the pathogenesis of airway MUC5B-related GCH.
Collapse
|
24
|
Meyer ML, Potts-Kant EN, Ghio AJ, Fischer BM, Foster WM, Voynow JA. NAD(P)H quinone oxidoreductase 1 regulates neutrophil elastase-induced mucous cell metaplasia. Am J Physiol Lung Cell Mol Physiol 2012; 303:L181-8. [PMID: 22659878 PMCID: PMC3423858 DOI: 10.1152/ajplung.00084.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mucous cell metaplasia (MCM) and neutrophil-predominant airway inflammation are pathological features of chronic inflammatory airway diseases. A signature feature of MCM is increased expression of a major respiratory tract mucin, MUC5AC. Neutrophil elastase (NE) upregulates MUC5AC in primary airway epithelial cells by generating reactive oxygen species, and this response is due in part to upregulation of NADPH quinone oxidoreductase 1 (NQO1) activity. Delivery of NE directly to the airway triggers inflammation and MCM and increases synthesis and secretion of MUC5AC protein from airway epithelial cells. We hypothesized that NE-induced MCM is mediated in vivo by NQO1. Male wild-type and Nqo1-null mice (C57BL/6 background) were exposed to human NE (50 μg) or vehicle via oropharyngeal aspiration on days 1, 4, and 7. On days 8 and 11, lung tissues and bronchoalveolar lavage (BAL) samples were obtained and evaluated for MCM, inflammation, and oxidative stress. MCM, inflammation, and production of specific cytokines, granulocyte-macrophage colony-stimulating factor, macrophage inflammatory protein-2, interleukin-4, and interleukin-5 were diminished in NE-treated Nqo1-null mice compared with NE-treated wild-type mice. However, in contrast to the role of NQO1 in vitro, we demonstrate that NE-treated Nqo1-null mice had greater levels of BAL and lung tissue lipid carbonyls and greater BAL iron on day 11, all consistent with increased oxidative stress. NQO1 is required for NE-induced inflammation and MCM. This model system demonstrates that NE-induced MCM directly correlates with inflammation, but not with oxidative stress.
Collapse
Affiliation(s)
- Marisa L Meyer
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | |
Collapse
|
25
|
Solatycka A, Owczarek T, Piller F, Piller V, Pula B, Wojciech L, Podhorska-Okolow M, Dziegiel P, Ugorski M. MUC1 in human and murine mammary carcinoma cells decreases the expression of core 2 β1,6-N-acetylglucosaminyltransferase and β-galactoside α2,3-sialyltransferase. Glycobiology 2012; 22:1042-54. [PMID: 22534569 DOI: 10.1093/glycob/cws075] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A good correlation between the expression of mucin1 (MUC1) and T antigen was found in breast cancer tumors and breast cancer cell lines, especially after treatment with neuraminidase. The association between the appearance of T antigen and the overexpression of MUC1 was further confirmed by transfecting MDA-MB-231 cells and murine 4T1 mammary carcinoma cells with cDNA for MUC1 and using an RNAi approach to inhibit the expression of MUC1 gene in T47D cells. Furthermore, we discovered that in 4T1 cells which express the sialyl Le(X) antigen, overexpression of MUC1 caused not only appearance of T antigen, but also loss of the sialyl Le(X) structure. As the observed changes in O-glycan synthesis can be associated with changes in the expression of specific glycosyltransferases, core 1 β1,3-galactosyltransferase, core 2 β1,6-N-acetylglucosaminyltransferase (C2GnT1) and β-galactoside α2,3-sialyltransferase (ST3Gal I), we studied their expression in parental, vector-transfected and MUC1-transfected MDA-MB-231 and 4T1 cells as well as T47D cells transduced with small hairpin RNA targeted MUC1 mRNA. It was found that the expression of C2GnT1 and ST3Gal I is highly decreased in MUC1-expressing MDA-MB-231 and 4T1 cells and increased in T47D cells with suppressed expression of MUC1. Therefore, we found that changes in the structure of O-linked oligosaccharides, resulting in the occurrence of T antigen, are at least partially associated with MUC1 overexpression which down-regulates the expression of C2GnT1 and ST3Gal I. We showed also that the overexpression of MUC1 in 4T1 cells changes their adhesive properties, as MUC1-expressing cells do not adhere to E-selectin, but bind galectin-3.
Collapse
Affiliation(s)
- Alicja Solatycka
- Laboratory of Glycobiology and Cell Interactions, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Morinaga Y, Yanagihara K, Araki N, Migiyama Y, Nagaoka K, Harada Y, Yamada K, Hasegawa H, Nishino T, Izumikawa K, Kakeya H, Yamamoto Y, Kohno S, Kamihira S. LiveLegionella pneumophilainduces MUC5AC production by airway epithelial cells independently of intracellular invasion. Can J Microbiol 2012; 58:151-7. [DOI: 10.1139/w11-123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshitomo Morinaga
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Nobuko Araki
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Yohei Migiyama
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kentaro Nagaoka
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yosuke Harada
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koichi Yamada
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroo Hasegawa
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Tomoya Nishino
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koichi Izumikawa
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Kakeya
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshihiro Yamamoto
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shigeru Kohno
- Second Department of Internal Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Global COE Program, Nagasaki University, Nagasaki, Japan
| | - Shimeru Kamihira
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
27
|
Yamada K, Yanagihara K, Araki N, Harada Y, Morinaga Y, Izumikawa K, Kakeya H, Yamamoto Y, Hasegawa H, Kohno S, Kamihira S. In vivo efficacy of KRP-109, a novel elastase inhibitor, in a murine model of severe pneumococcal pneumonia. Pulm Pharmacol Ther 2011; 24:660-5. [PMID: 21864700 DOI: 10.1016/j.pupt.2011.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Revised: 07/27/2011] [Accepted: 08/01/2011] [Indexed: 11/24/2022]
Abstract
KRP-109 is a novel specific inhibitor of neutrophil elastase (NE). Various studies suggest that NE inhibitors reduce lung injury associated with systemic inflammatory response syndrome (SIRS). In this study, the efficacy of KRP-109 was examined using a murine model of severe pneumonia induced by Streptococcus pneumoniae (S. pneumoniae). Female mice (CBA/J, aged 5 weeks) were inoculated intranasally with penicillin-susceptible S. pneumoniae (ATCC49619 strain, 2.5 × 10(8) CFU/mouse). KRP-109 (30 or 50 mg/kg) or physiological saline as a control was administered intraperitoneally every 8 h beginning at 8 h after inoculation, and survival rate was evaluated over 7 days. Histopathological and bacteriological analyses of the lung, and bronchoalveolar lavage were performed at 48 h post-infection. The mice treated with KRP-109 (KRP-109 mice) tended to have higher survival rate than those given saline. The lung tissues of the KRP-109 mice had few neutrophils in the alveolar walls and less inflammation. Furthermore, KRP-109 decreased significantly total cell and neutrophil counts, and cytokine levels (interleukin 1β and macrophage inflammatory protein 2) in bronchoalveolar lavage fluid. Viable bacterial numbers in lung were not influenced by treatment of KRP-109. The present results indicate that KRP-109 reduces lung inflammation in a murine model, and that KRP-109 may be useful for the treatment of patients with severe pneumonia.
Collapse
Affiliation(s)
- Koichi Yamada
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
N-acetylcysteine enhances cystic fibrosis sputum penetration and airway gene transfer by highly compacted DNA nanoparticles. Mol Ther 2011; 19:1981-9. [PMID: 21829177 DOI: 10.1038/mt.2011.160] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
For effective airway gene therapy of cystic fibrosis (CF), inhaled gene carriers must first penetrate the hyperviscoelastic sputum covering the epithelium. Whether clinically studied gene carriers can penetrate CF sputum remains unknown. Here, we measured the diffusion of a clinically tested nonviral gene carrier, composed of poly-l-lysine conjugated with a 10 kDa polyethylene glycol segment (CK(30)PEG(10k)). We found that CK(30)PEG(10k)/DNA nanoparticles were trapped in CF sputum. To improve gene carrier diffusion across sputum, we tested adjuvant regimens consisting of N-acetylcysteine (NAC), recombinant human DNase (rhDNase) or NAC together with rhDNase. While rhDNase alone did not enhance gene carrier diffusion, NAC and NAC + rhDNase increased average effective diffusivities by 6-fold and 13-fold, respectively, leading to markedly greater fractions of gene carriers that may penetrate sputum layers. We further tested the adjuvant effects of NAC in the airways of mice with Pseudomonas aeruginosa lipopolysaccharide (LPS)-induced mucus hypersecretion. Intranasal dosing of NAC prior to CK(30)PEG(10k)/DNA nanoparticles enhanced gene expression by up to ~12-fold compared to saline control, reaching levels observed in the lungs of mice without LPS challenge. Our findings suggest that a promising synthetic nanoparticle gene carrier may transfer genes substantially more effectively to lungs of CF patients if administered following adjuvant mucolytic therapy with NAC or NAC + rhDNase.
Collapse
|
29
|
McGuckin MA, Lindén SK, Sutton P, Florin TH. Mucin dynamics and enteric pathogens. Nat Rev Microbiol 2011. [PMID: 21407243 DOI: 10.1038/nrm] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The extracellular secreted mucus and the cell surface glycocalyx prevent infection by the vast numbers of microorganisms that live in the healthy gut. Mucin glycoproteins are the major component of these barriers. In this Review, we describe the components of the secreted and cell surface mucosal barriers and the evidence that they form an effective barricade against potential pathogens. However, successful enteric pathogens have evolved strategies to circumvent these barriers. We discuss the interactions between enteric pathogens and mucins, and the mechanisms that these pathogens use to disrupt and avoid mucosal barriers. In addition, we describe dynamic alterations in the mucin barrier that are driven by host innate and adaptive immune responses to infection.
Collapse
Affiliation(s)
- Michael A McGuckin
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute and The University of Queensland School of Medicine, South Brisbane, Queensland 4101, Australia.
| | | | | | | |
Collapse
|
30
|
Nakamura S, Yanagihara K, Araki N, Yamada K, Morinaga Y, Izumikawa K, Seki M, Kakeya H, Yamamoto Y, Kamihira S, Kohno S. High-dose tobramycin inhibits lipopolysaccharide-induced MUC5AC production in human lung epithelial cells. Eur J Pharmacol 2011; 659:67-71. [PMID: 21414310 DOI: 10.1016/j.ejphar.2011.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 02/11/2011] [Accepted: 03/01/2011] [Indexed: 12/29/2022]
Abstract
Tobramycin inhalation therapy is an effective therapy for cystic fibrosis as well as severe bronchiectasis that is colonized with Pseudomonas aeruginosa. The mechanism responsible for the efficacy of tobramycin in the treatment of severe chronic infectious diseases has not been elucidated. We demonstrate that high-dose tobramycin decreases MUC5AC gene expression and protein production in NCI-H292 cell stimulated with lipopolysaccharide of P. aeruginosa. MUC5AC protein of NCI-H292 cell stimulated by lipopolysaccharide was analyzed by an enzyme-linked immunosorbent assay and MUC5AC expression at the mRNA level was analyzed by RT-PCR. Western blot was performed to examine a potential role for the signaling molecules upstream of NFκB. High-dose tobramycin (500μg/ml) decreased the level of MUC5AC protein released into the supernatant of the NCI-H292 cell line at 24h after lipopolysaccharide stimulation (P<0.001). The tobramycin treatment also inhibited MUC5AC mRNA expression at 12h after lipopolysaccharide stimulation (P<0.05) and suppressed NFκB activation 60min after lipopolysaccharide stimulation (P<0.001). Tobramycin suppressed the phosphorylation of extracellular signal-regulated protein kinase, p38 MAP kinase. These results suggest that high-dose tobramycin, such as inhalation therapy, can inhibit MUC5AC gene expression and MUC5AC protein production in NCI-H292 cells, in part through the mitogen-activated protein kinase pathway. Thus, the activation of TLR4 and the subsequent immune/inflammatory responses induced by chronic infections such as P. aeruginosa might be modulated by tobramycin. Our data may reveal one of the mechanisms responsible for the clinical effect of tobramycin inhalation therapy against severe chronic respiratory diseases due to P. aeruginosa.
Collapse
Affiliation(s)
- Shigeki Nakamura
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Katsunori Yanagihara
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan; Department of Laboratory Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Nobuko Araki
- Department of Laboratory Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Koichi Yamada
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan; Department of Laboratory Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Yoshitomo Morinaga
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan; Department of Laboratory Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Koichi Izumikawa
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Masafumi Seki
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Hiroshi Kakeya
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Yoshihiro Yamamoto
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Shimeru Kamihira
- Department of Laboratory Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | - Shigeru Kohno
- Second Department of Internal Medicine, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan; Division of Molecular & Clinical Microbiology, Department of Molecular Microbiology & Immunology, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| |
Collapse
|
31
|
|
32
|
Cheng PW, Radhakrishnan P. Mucin O-glycan branching enzymes: structure, function, and gene regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 705:465-92. [PMID: 21618125 DOI: 10.1007/978-1-4419-7877-6_25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Pi-Wan Cheng
- Department of Biochemistry and Molecular Biology, College of Medicine and Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | | |
Collapse
|
33
|
Bandi N, Ayalasomayajula SP, Dhanda DS, Iwakawa J, Cheng PW, Kompella UB. Intratracheal budesonide-poly (lactide-co-glycolide) microparticles reduce oxidative stress, VEGF expression, and vascular leakage in a benzo(a)pyrene-fed mouse model. J Pharm Pharmacol 2010; 57:851-60. [PMID: 15969944 DOI: 10.1211/0022357056334] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Abstract
The purpose of this study was to determine whether intratracheally instilled polymeric budesonide microparticles could sustain lung budesonide levels for one week and inhibit early biochemical changes associated with benzo(a)pyrene (B[a]P) feeding in a mouse model for lung tumours. Polymeric microparticles of budesonide-poly (dl-lactide-co-glycolide) (PLGA 50:50) were prepared using a solvent evaporation technique and characterized for their size, morphology, encapsulation efficiency, and in-vitro release. The microparticles were administered intratracheally (i.t.) to B[a]P-fed A/J mice. At the end of one week drug levels in the lung tissue and bronchoalveolar lavage (BAL) were estimated using HPLC and compared with systemic (intramuscular) administration. In addition, in-vivo end points including malondialdehyde (MDA), glutathione (GSH), total protein levels and vascular endothelial growth factor (VEGF) in BAL, and VEGF and c-myc mRNA levels in the lung tissue were assessed at the end of one week following intratracheal administration of budesonide microparticles. Budesonide-PLGA microparticles (1–2 μm), with a budesonide loading efficiency of 69–94%, sustained in-vitro budesonide release for over 21 days. Compared with the intramuscular route, intratracheally administered budesonide-PLGA microparticles resulted in higher budesonide levels in the BAL and lung tissue. In-vivo, B[a]P-feeding increased BAL MDA, lung VEGF mRNA, lung c-myc mRNA, BAL total protein, and BAL VEGF levels by 60, 112, 71, 154, and 78%, respectively, and decreased BAL GSH by 62%. Interestingly, intratracheally administered budesonide-PLGA particles inhibited these biochemical changes. Thus, biodegradable budesonide microparticles sustained budesonide release and reduced MDA accumulation, GSH depletion, vascular leakage, and VEGF and c-myc expression in B[a]P-fed mice, indicating the potential of locally delivered sustained-release particles for inhibiting angiogenic factors in lung cancer.
Collapse
Affiliation(s)
- Nagesh Bandi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | | | | | | | | | | |
Collapse
|
34
|
Nishimoto Y, Hisatsune A, Katsuki H, Miyata T, Yokomizo K, Isohama Y. Glycyrrhizin Attenuates Mucus Production by Inhibition of MUC5AC mRNA Expression In Vivo and In Vitro. J Pharmacol Sci 2010; 113:76-83. [DOI: 10.1254/jphs.09344fp] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
35
|
Efficacy of clarithromycin against experimentally induced pneumonia caused by clarithromycin-resistant Haemophilus influenzae in mice. Antimicrob Agents Chemother 2009; 54:757-62. [PMID: 19949056 DOI: 10.1128/aac.00524-09] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clarithromycin is a 14-member lactone ring macrolide with potent activity against Haemophilus influenzae, including ampicillin-resistant strains. We evaluated the in vivo efficacy of clarithromycin at 40 mg/day and 100 mg/day for 3 days in the treatment of a murine model of pneumonia using a macrolide-resistant H. influenzae strain, which was also ampicillin resistant. The MIC of clarithromycin was 64 microg/ml. The viable bacterial counts in infected tissues after treatment with 100 mg clarithromycin/kg of body weight were lower than the counts obtained in control and 40-mg/kg clarithromycin-treated mice. The concentrations of macrophage inflammatory protein 2 (MIP-2) and interleukin 1beta (IL-1beta) in bronchoalveolar lavage fluid (BALF) samples from mice treated at both concentrations were lower than in the control group. Pathologically, following infection, clarithromycin-treated mice, particularly at a dose of 100 mg/kg, showed lower numbers of neutrophils in alveolar walls, and inflammatory changes had apparently improved, whereas large aggregates of inflammatory cells were observed within the alveoli of control mice. In addition, we demonstrated that clarithromycin has bacteriological effects against intracellular bacteria at levels below the MIC. Our results indicate that clarithromycin may be useful in vivo for macrolide-resistant H. influenzae, and this phenomenon may be related to the good penetration of clarithromycin into bronchoepithelial cells. We also believe that conventional drug susceptibility tests may not reflect the in vivo effects of clarithromycin.
Collapse
|
36
|
Azithromycin, clarithromycin and telithromycin inhibit MUC5AC induction by Chlamydophila pneumoniae in airway epithelial cells. Pulm Pharmacol Ther 2009; 22:580-6. [PMID: 19716898 DOI: 10.1016/j.pupt.2009.08.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 08/18/2009] [Accepted: 08/22/2009] [Indexed: 11/23/2022]
Abstract
BACKGROUND Airway mucus hypersecretion is an important problem in chronic respiratory diseases including bronchial asthma. Chlamydophila pneumoniae is recently confirmed to be a pathogen in bronchial asthma, but the relationship between C. pneumoniae and mucus hypersecretion is uncertain. In this study, we examined whether C. pneumoniae induces MUC5AC mucin in airway epithelial cells. We also examined the effects of macrolide and ketolide antibiotics on the C. pneumoniae-induced mucus production. METHODS MUC5AC production in bronchial epithelial cells after stimulation with C. pneumoniae was analyzed by ELISA and quantitative RT-PCR. NF-kappaB and phosphorylated ERK were also analyzed. For inhibition study, cells were pretreated with azithromycin, clarithromycin and telithromycin before stimulation. RESULTS C. pneumoniae dose-dependently induced MUC5AC production and gene expression. The ERK-NF-kappaB pathway was involved in C. pneumoniae-induced MUC5AC production. Macrolides and ketolides dose-dependently reduced C. pneumoniae-induced MUC5AC production. However, azithromycin was apparently less effective than the other antibiotics. Clarithromycin and telithromycin, but not azithromycin, reduced NF-kappaB activation. CONCLUSIONS Clarithromycin and telithromycin were thought to interfere with the signal pathways between ERK and NF-kappaB. These results suggest that airway mucus hypersecretion is one of the mechanisms of C. pneumoniae-induced bronchial asthma, and that macrolide and ketolide antibiotics represent a novel therapeutic intervention in these patients.
Collapse
|
37
|
Li RW, Li C, Elsasser TH, Liu G, Garrett WM, Gasbarre LC. Mucin biosynthesis in the bovine goblet cell induced by Cooperia oncophora infection. Vet Parasitol 2009; 165:281-9. [PMID: 19647371 DOI: 10.1016/j.vetpar.2009.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 07/06/2009] [Accepted: 07/07/2009] [Indexed: 10/20/2022]
Abstract
Mucin hypersecretion is considered to be one of the most common components of the immune response to gastrointestinal nematode infection. However, investigations have not been conducted in the Cattle-Cooperia oncophora system to verify the findings largely derived from murine models. In this study, we examined the expression of seven mucins and seven enzymes in the mucin biosynthesis pathway involved in O-linked glycosylation in the bovine small intestine including goblet cells enriched using laser capture microdissection during a primary C. oncophora infection. At the mRNA level, MUC2 expression was significantly higher in both lamina propria and goblet cells at 28 days post-infection compared to the naive control. MUC5B expression at the mRNA level was also higher in lamina propria at 28dpi. Expression of MUC1, MUC4, MUC5AC, and MUC6 was extremely low or not detectable in goblet cells, columnar epithelial cells, and lamina propria from both naive control and infected animals. Among the seven enzymes involved in post-translational O-linked glycosylation of mucins, GCNT3, which may represent one of the key rate-limiting steps in mucin biosynthesis, was up-regulated in goblet cells, columnar epithelial cells, lamina propria, and gross small intestine tissue during the course of infection. Western blot analysis revealed that MUC2 glycoprotein was strongly induced by infection in both gross small intestine tissue and its mucosal layer. In contrast, the higher MUC5B protein expression was observed only in the mucosal layer. Immunohistochemistry provided further evidence of the mucin glycoprotein production and localization. Our results provided insight into regulation of mucin biosynthesis in various cell types in the bovine small intestine during gastrointestinal nematode infection and will facilitate our understanding of mucins and their role in immune response against parasitic nematodes.
Collapse
Affiliation(s)
- Robert W Li
- Bovine Functional Genomics Laboratory, Animal and Natural Resources Institute, USDA-ARS, Beltsville, MD 20705, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Yanagihara K, Fukuda Y, Seki M, Izumikawa K, Miyazaki Y, Hirakata Y, Tsukamoto K, Yamada Y, Kamhira S, Kohno S. EFFECTS OF SPECIFIC NEUTROPHIL ELASTASE INHIBITOR, SIVELESTAT SODIUM HYDRATE, IN MURINE MODEL OF SEVERE PNEUMOCOCCAL PNEUMONIA. Exp Lung Res 2009; 33:71-80. [PMID: 17454103 DOI: 10.1080/01902140701198500] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
An excessive amount of neutrophil elastase (NE) released from neutrophils accumulated in the lung can cause tissue damage, despite its importance to host defense against microbial pathogens in severe pneumonia. Therefore, NE inhibitors may reduce tissue damage in lungs with severe pneumonia. In this study, the efficacy of a specific NE inhibitor, sivelestat sodium hydrate (sivelestat), was examined using a murine model of severe pneumonia with Streptococcus pneumoniae. Male mice (CBA/JNCrj, aged 5 weeks) were inoculated intranasally with penicillin-susceptible S. pneumonia (1.0 x 10(5) CFU/mouse). Sivelestat (3 mg/kg) or physiological saline was administered every 12 hours beginning at 12 hours after inoculation. Survival was primarily evaluated. Bronchoalveolar lavage fluid (BALF) and blood were collected at 30 hours after inoculation. Thus, cell counts in BALF and numbers of viable bacteria in blood were determined. Histopathological analysis was also performed. Sivelestat significantly prolonged survival when compared with the control group (P < .05), although all animals died within 4 days. Cell count and histopathological analysis indicated that sivelestat prevented the progression of lung inflammation, such as alveolar neutrophil infiltration and hemorrhage. Furthermore, the number of viable bacteria in blood was significantly lower in the sivelestat group than in the control group (5.69 +/- 0.27 and 6.75 +/- 0.32 log CFU/mL, respectively; mean +/- SEM, P < .01). Sivelestat prolonged survival in this model. A possible explanation for the improved survival is that sivelestat prevents tissue damage by inhibiting NE activity in the lung. Therefore, NE inhibitors may be useful for treating with patients with severe pneumonia.
Collapse
Affiliation(s)
- Katsunori Yanagihara
- Second Department of Internal Medicine, University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Atzori L, Lucattelli M, Scotton CJ, Laurent GJ, Bartalesi B, De Cunto G, Lunghi B, Chambers RC, Lungarella G. Absence of proteinase-activated receptor-1 signaling in mice confers protection from fMLP-induced goblet cell metaplasia. Am J Respir Cell Mol Biol 2009; 41:680-7. [PMID: 19307611 DOI: 10.1165/rcmb.2007-0386oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The morphological features of chronic obstructive pulmonary disease in man include emphysema and chronic bronchitis associated with mucus hypersecretion. These alterations can be induced in mice by a single intratracheal instillation of N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP), a chemoattractant and degranulating agent for neutrophils. The mechanisms underlying excessive mucus production and, in particular, goblet cell hyperplasia/metaplasia in chronic obstructive pulmonary disease remain poorly understood. The proteinase-activated receptors (PARs) are widely recognized for their modulatory properties during inflammation. In this study, we examined whether PAR-1 contributes to inflammation and lung damage induced by fMLP by comparing the response of PAR-1-deficient (PAR-1(-/-)) mice with that of wild-type (WT) mice. Mice were killed at various time points after fMLP instillation (200 microg/50 microl). WT mice developed emphysema and goblet cell metaplasia. The onset of pulmonary lesions was preceded by an increase in thrombin immunoreactivity in bronchial airways and alveolar tissue. This was followed by a decrease in PAR-1 immunoreactivity, and by an increase in IL-13 immunostaining on the luminal surface of airway epithelial cells. In PAR-1(-/-) mice, fMLP administration induced similar responses in terms of inflammation and emphysema, but these mice were protected from the development of goblet cell metaplasia. The involvement of PAR-1 in airway epithelial cell transdifferentiation was confirmed by demonstrating that intratracheal instillation of the selective PAR-1 agonist (TFLLR) induced goblet cell metaplasia in the airways of WT mice only. These data suggest that emphysema and goblet cell metaplasia occur independently, and that PAR-1 signaling through IL-13 stimulation may play an important role in inducing goblet cell metaplasia.
Collapse
Affiliation(s)
- Luigi Atzori
- Department of Toxicology, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Evans CM, Koo JS. Airway mucus: the good, the bad, the sticky. Pharmacol Ther 2008; 121:332-48. [PMID: 19059283 PMCID: PMC10079267 DOI: 10.1016/j.pharmthera.2008.11.001] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Indexed: 01/21/2023]
Abstract
Mucus production is a primary defense mechanism for maintaining lung health. However, the overproduction of mucin (the chief glycoprotein component of mucus) is a common pathological feature in asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), and lung cancer. Although it is associated with disease progression, effective therapies that directly target mucin overproduction and hypersecretion are lacking. Recent advances in our understanding of the control of mucin gene expression in the lungs, the cells that produce airway mucins, and the mechanisms used for releasing them into the airways have provided new potentials for the development of efficacious interventions that will be discussed in this review.
Collapse
Affiliation(s)
- Christopher M Evans
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.
| | | |
Collapse
|
41
|
|
42
|
OU XM, FENG YL, WEN FQ, WANG K, YANG J, DENG ZP, LIU DS, LI YP. Macrolides attenuate mucus hypersecretion in rat airways through inactivation of NF-κB. Respirology 2008; 13:63-72. [DOI: 10.1111/j.1440-1843.2007.01213.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Linden SK, Sutton P, Karlsson NG, Korolik V, McGuckin MA. Mucins in the mucosal barrier to infection. Mucosal Immunol 2008; 1:183-97. [PMID: 19079178 PMCID: PMC7100821 DOI: 10.1038/mi.2008.5] [Citation(s) in RCA: 818] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The mucosal tissues of the gastrointestinal, respiratory, reproductive, and urinary tracts, and the surface of the eye present an enormous surface area to the exterior environment. All of these tissues are covered with resident microbial flora, which vary considerably in composition and complexity. Mucosal tissues represent the site of infection or route of access for the majority of viruses, bacteria, yeast, protozoa, and multicellular parasites that cause human disease. Mucin glycoproteins are secreted in large quantities by mucosal epithelia, and cell surface mucins are a prominent feature of the apical glycocalyx of all mucosal epithelia. In this review, we highlight the central role played by mucins in accommodating the resident commensal flora and limiting infectious disease, interplay between underlying innate and adaptive immunity and mucins, and the strategies used by successful mucosal pathogens to subvert or avoid the mucin barrier, with a particular focus on bacteria.
Collapse
Affiliation(s)
- S K Linden
- grid.1003.20000 0000 9320 7537Mucosal Diseases Program, Mater Medical Research Institute and The University of Queensland, Level 3 Aubigny Place, Mater Hospitals, South Brisbane, Queensland Australia
| | - P Sutton
- grid.1008.90000 0001 2179 088XCentre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Melbourne, Victoria Australia
| | - N G Karlsson
- grid.6142.10000 0004 0488 0789Department of Chemistry, Centre for BioAnalytical Sciences, National University of Ireland, Galway, Ireland
| | - V Korolik
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Gold Coast, Queensland Australia
| | - M A McGuckin
- grid.1003.20000 0000 9320 7537Mucosal Diseases Program, Mater Medical Research Institute and The University of Queensland, Level 3 Aubigny Place, Mater Hospitals, South Brisbane, Queensland Australia
| |
Collapse
|
44
|
Abstract
The pathogenesis of chronic obstructive pulmonary disease (COPD) encompasses a number of injurious processes, including an abnormal inflammatory response in the lungs to inhaled particles and gases. Other processes, such as failure to resolve inflammation, abnormal cell repair, apoptosis, abnormal cellular maintenance programs, extracellular matrix destruction (protease/antiprotease imbalance), and oxidative stress (oxidant/antioxidant imbalance) also have a role. The inflammatory responses to the inhalation of active and passive tobacco smoke and urban and rural air pollution are modified by genetic and epigenetic factors. The subsequent chronic inflammatory responses lead to mucus hypersecretion, airway remodeling, and alveolar destruction. This article provides an update on the cellular and molecular mechanisms of these processes in the pathogenesis of COPD.
Collapse
Affiliation(s)
- William Macnee
- ELEGI Colt Research Laboratories, MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Avenue, Edinburgh EH16 4TJ, Scotland, UK.
| |
Collapse
|
45
|
Hauber HP, Goldmann T, Vollmer E, Wollenberg B, Zabel P. Effect of Dexamethasone and ACC on Bacteria-Induced Mucin Expression in Human Airway Mucosa. Am J Respir Cell Mol Biol 2007; 37:606-16. [PMID: 17600317 DOI: 10.1165/rcmb.2006-0404oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Gram-negative bacteria can stimulate mucin production, but excessive mucus supports bacterial infection and consequently leads to airway obstruction. Therefore, the effect of dexamethasone (DEX) and the antioxidant acetyl-cysteine (ACC) on bacteria-induced mucus expression was investigated. Explanted human airway mucosa and mucoepidermoid cells (Calu-3) were stimulated with lipopolysaccharide (LPS) or PAM3 (a synthetic lipoprotein). DEX or ACC were added to either LPS- or PAM3-stimulated airway mucosa or Calu-3 cells. Mucin mRNA expression (MUC5AC) and total mucus glycoconjugates (mucin protein) were quantified using real-time PCR and periodic acid Schiff staining. LPS and PAM3 significantly increased mucin expression in airway mucosa and Calu-3 cells (P < 0.05). DEX alone had no significant effect on mucin expression in airway mucosa or Calu-3 cells (P > 0.05). In contrast, DEX significantly reduced LPS- and PAM3-induced mucin expression in explanted mucosal tissue and mucin expression in Calu-3 cells (P < 0.05). In explanted human airway mucosa ACC alone significantly increased mucin expression (P < 0.05). In contrast, ACC significantly decreased LPS- and PAM3-induced mucin expression (P < 0.05). In Calu-3 cells ACC alone had no significant effect on mucin expression (P > 0.05). ACC decreased LPS- and PAM3-induced mucin expression, but this effect was not significant (P > 0.05). These data suggest that DEX can effectively reduce bacteria-induced mucin expression in the airways. ACC alone may increase mucin expression in noninfected mucosa, but it decreased bacteria-induced mucin expression. Further studies are warranted to evaluate whether the effect of DEX or ACC is clinically relevant.
Collapse
Affiliation(s)
- Hans-Peter Hauber
- Medical Clinic, Research Center Borstel, Research Center Borstel, Borstel, Germany.
| | | | | | | | | |
Collapse
|
46
|
Hauber HP, Goldmann T, Vollmer E, Wollenberg B, Hung HL, Levitt RC, Zabel P. LPS-induced mucin expression in human sinus mucosa can be attenuated by hCLCA inhibitors. ACTA ACUST UNITED AC 2007; 13:109-16. [PMID: 17621552 DOI: 10.1177/0968051907079168] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND hCLCA1 is a member of the calcium-activated chloride channel family and is associated with disease-inducible mucus expression. Niflumic acid (NFA) and a closely related chemical structure are reported inhibitors of calcium-activated chloride channels and endotoxin-inducible mucus expression in the mouse. Therefore, we tested the hypothesis that hCLCA1 may be involved in lipopolysaccharide (LPS) induced mucin up-regulation in human airways. We also investigated the effect of NFA and MSI-2216 on LPS-induced mucin up-regulation. MATERIALS AND METHODS Explanted human airways and the muco-epidermoid cell line Calu-3 were stimulated with LPS. Different concentrations of NFA or MSI-2216 were added to LPS-stimulated airway mucosa and Calu-3 cells. Expression of hCLCA1 and MUC5AC mRNA and protein was quantified in human airways using real-time PCR and PAS staining. In addition, immunohistochemistry was performed for quantification of inflammatory cells (lymphocytes, monocytes, eosinophils, and neutrophils) in the submucosa of the airways. Expression of hCLCA1 protein in Calu-3 cells was analysed by FACS. RESULTS LPS significantly induced hCLCA1 and MUC5AC mRNA and protein expression in human airway mucosa (P < 0.05). NFA and MSI-2216 significantly decreased LPS-induced mucus expression in explanted airway mucosa in a dose-dependent manner (P < 0.05). In Calu-3 cells, LPS significantly increased hCLCA1 surface expression whereas intracellular expression was significantly decreased (P < 0.05). In Calu-3 cells, NFA and MSI-2216 also significantly reduced MUC5AC mRNA expression (P < 0.05). CONCLUSIONS These data suggest that hCLCA1 may play a role in LPS-induced mucin expression in human airway mucosa. Calcium-activated chloride channel inhibitors significantly decreased LPS-induced mucus expression both ex vivo and in vitro . Therefore, blocking of hCLCA1 may offer a therapeutic approach to reduce bacterial-induced mucus hypersecretion.
Collapse
|
47
|
Yoshida T, Tuder RM. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol Rev 2007; 87:1047-82. [PMID: 17615396 DOI: 10.1152/physrev.00048.2006] [Citation(s) in RCA: 375] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary diseases (COPD), comprised of pulmonary emphysema, chronic bronchitis, and structural and inflammatory changes of small airways, is a leading cause of morbidity and mortality in the world. A better understanding of the pathobiology of COPD is critical for the developing of novel therapies, as the majority of patients with the disease have little therapeutic options at the present time. The pathobiology of COPD encompasses multiple injurious processes including inflammation (excessive or inappropriate innate and adaptive immunity), cellular apoptosis, altered cellular and molecular alveolar maintenance program, abnormal cell repair, extracellular matrix destruction (protease and anti-protease imbalance), and oxidative stress (oxidant and antioxidant imbalance). These processes are triggered by urban and rural air pollutants and active and/or passive cigarette smoke and modified by cellular senescence and infection. A series of receptor-mediated signal transduction pathways are activated by reactive oxygen species and tobacco components, resulting in impairment of a variety of cell signaling and cytokine networks, subsequently leading to chronic airway responses with mucus production, airway remodeling, and alveolar destruction. The authors provide an updated insight into the molecular and cellular pathobiology of COPD based on human and/or animal data.
Collapse
Affiliation(s)
- Toshinori Yoshida
- Division of Cardiopulmonary Pathology, Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
48
|
Tetaert D, Pierre M, Demeyer D, Husson MO, Béghin L, Galabert C, Gottrand F, Beermann C, Guery B, Desseyn JL. Dietary n-3 fatty acids have suppressive effects on mucin upregulation in mice infected with Pseudomonas aeruginosa. Respir Res 2007; 8:39. [PMID: 17550583 PMCID: PMC1899493 DOI: 10.1186/1465-9921-8-39] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Accepted: 06/05/2007] [Indexed: 12/02/2022] Open
Abstract
Background Mucin hypersecretion and mucus plugging in the airways are characteristic features of chronic respiratory diseases like cystic fibrosis (CF) and contribute to morbidity and mortality. In CF, Pseudomonas aeruginosa superinfections in the lung exacerbate inflammation and alter mucus properties. There is increasing evidence that n-3 polyunsaturated fatty acids (PUFAs) exhibit anti-inflammatory properties in many inflammatory diseases while n-6 PUFA arachidonic acid (AA) favors inflammatory mediators such as eicosanoids prostaglandin E2 (PGE2) and leukotriene B4 (LTB4) that may enhance inflammatory reactions. This suggests that n-3 PUFAs may have a protective effect against mucus over-production in airway diseases. Therefore, we hypothesized that n-3 PUFAs may downregulate mucins expression. Methods We designed an absolute real-time PCR assay to assess the effect of a 5-week diet enriched either with n-3 or n-6 PUFAs on the expression of large mucins in the lungs of mice infected by P. aeruginosa. Results Dietary fatty acids did not influence mucin gene expression in healthy mice. Lung infection induced an increase of the secreted gel-forming mucin Muc5b and a decrease of the membrane bound mucin Muc4. These deregulations are modulated by dietary fatty acids with a suppressive effect of n-3 PUFAs on mucin (increase of Muc5b from 19-fold up to 3.6 × 105-fold for the n-3 PUFAs treated group and the control groups, respectively, 4 days post-infection and decrease of Muc4 from 15-fold up to 3.2 × 104-fold for the control and the n-3 PUFAs treated groups, respectively, 4 days post-infection). Conclusion Our data suggest that n-3 PUFAs enriched diet represents an inexpensive strategy to prevent or treat mucin overproduction in pulmonary bacterial colonization.
Collapse
Affiliation(s)
- Daniel Tetaert
- INSERM, U837, JPARC Research Centre, IMPRT, place de Verdun, Lille, France
| | - Maud Pierre
- EA 3925, IMPRT, University of Lille 2 and CHRU of Lille, France
| | - Dominique Demeyer
- INSERM, U837, JPARC Research Centre, IMPRT, place de Verdun, Lille, France
| | | | - Laurent Béghin
- EA 3925, IMPRT, University of Lille 2 and CHRU of Lille, France
| | | | | | | | - Benoit Guery
- EA 2689, IMPRT, University of Lille 2 and CHRU of Lille, France
| | - Jean-Luc Desseyn
- INSERM, U837, JPARC Research Centre, IMPRT, place de Verdun, Lille, France
| |
Collapse
|
49
|
Kouznetsova I, Chwieralski CE, Bälder R, Hinz M, Braun A, Krug N, Hoffmann W. Induced trefoil factor family 1 expression by trans-differentiating Clara cells in a murine asthma model. Am J Respir Cell Mol Biol 2006; 36:286-95. [PMID: 16990615 DOI: 10.1165/rcmb.2006-0008oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Asthma is a chronic inflammatory disease of the airways that is accompanied by goblet cell metaplasia and mucus hypersecretion. Trefoil factor family (TFF) peptides represent major secretory products of the respiratory tract and are synthesized together with mucins. In the murine lung, TFF2 is mainly expressed, whereas TFF1 transcripts represent only a minor species. TFF peptides are well known for their motogenic and anti-apoptotic effects, and they modulate the inflammatory response of bronchial epithelial cells. Here, an established mouse model of asthma was investigated (i.e., exposure to Aspergillus fumigatus [AF] antigens). RT-PCR analysis of lung tissue showed elevated levels particularly of TFF1 transcripts in AF-sensitized/challenged animals. In contrast, transcripts encoding Clara cell secretory protein (CCSP/CC10) were strongly diminished in these animals. For comparison, the expression of the goblet cell secretory granule marker mCLCA3/Gob-5, the mucins Muc1-Muc6 and Muc19, and the secretoglobins ScgB3A1 and ScgB3A2, as well as the mammalian ependymin-related gene MERP2, were monitored. Immunohistochemistry localized TFF1 mainly in cells with a mixed phenotype (e.g., TFF1-positive cells stain with the lectin wheat germ agglutinin (WGA), which recognizes mucins characteristic of goblet cells). In addition, these cells express CCSP/CC10, a Clara cell marker. When compared with mucins or CCSP/CC10, TFF1 was stored in a different population of secretory granules localized at the more basolateral portion of these cells. Thus, the results presented indicate for the first time that allergen exposure leads to the trans-differentiation of Clara cells toward a TFF1-expressing mucous phenotype.
Collapse
Affiliation(s)
- Irina Kouznetsova
- Institut für Molekularbiologie und Medizinische Chemie, Otto-von-Guericke-Universität, Magdeburg; and Fraunhofer-Institut für Toxikologie und Experimentelle Medizin, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
50
|
Kuan YH, Lin RH, Chen YL, Tsao LT, Tzeng CC, Wang JP. Effective attenuation of acute lung injury in vivo and the formyl peptide-induced neutrophil activation in vitro by CYL-26z through the phosphoinositide 3-kinase γ pathway. Biochem Pharmacol 2006; 72:749-60. [PMID: 16887102 DOI: 10.1016/j.bcp.2006.06.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 06/06/2006] [Accepted: 06/09/2006] [Indexed: 10/24/2022]
Abstract
5-[4-Acridin-9-ylamino]phenyl]-5-methyl-3-methylenedihydrofuran-2-one (CYL-26z) inhibited the polymorphonuclear leukocyte (PMNL) infiltration and protein leakage into the lungs in lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice as determined on the basis of PMNL and protein contents in bronchoalveolar lavage (BAL) fluid and myeloperoxidase (MPO) content in whole lung extracts. CYL-26z also attenuated the formyl-Met-Leu-Phe (fMLP)-induced neutrophil chemotaxis and respiratory burst in vitro (IC(50) 8.4+/-0.9microM and 2.0+/-0.6microM, respectively). CYL-26z had no effect on superoxide anion (O(2)(-)) generation during dihydroxyfumaric acid autoxidation or on the NADPH oxidase activity in two cell-free systems (the arachidonic acid-induced assembly of NADPH oxidase and the preassembled oxidase caused by phorbol ester treatment), whereas it inhibited NaF-induced respiratory burst. Inhibition of respiratory burst by CYL-26z was readily reversible by washing. Only slight, but significant, inhibition of extracellular signal regulated kinase (ERK) phosphorylation and p38 mitogen-activated protein kinase (MAPK) activation in response to fMLP by CYL-26z up to 30microM was obtained. CYL-26z effectively blocked the formation of phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) as determined by immunofluorescence microscopy and flow cytometry assays and the dual phosphorylation of protein kinase B (PKB/Akt) on S473 and T308 residues in fMLP-stimulated neutrophils. The membrane recruitment of p110gamma and Ras, the Ras activation, and the association between p110gamma and Ras were also attenuated by CYL-26z. These results indicate that the blockade of Ras activation by CYL-26z attenuated the downstream phosphoinositide 3-kinase (PI3K) gamma signaling, which is involved in chemoattractant-induced neutrophil chemotaxis and respiratory burst, and may have a beneficial anti-inflammatory effect on ALI.
Collapse
Affiliation(s)
- Yu-Hsiang Kuan
- Department of Education and Research, Taichung Veterans General Hospital, Taichung 407, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|