1
|
Zhang K, Wu Z, Zhao Y, Qiu X, Li F, Chen Q, Cui F. LC3 Accelerated Brain-Lung Axis Abscopal Effects after Fractionated Whole-Brain Radiation by Promoting Motoneurons to Secrete Periostin. Radiat Res 2023; 200:462-473. [PMID: 37796808 DOI: 10.1667/rade-23-00075.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/14/2023] [Indexed: 10/07/2023]
Abstract
The effect of autophagy on the radiation-induced bystander effect (RIBE) in vivo is unclear. Here, the whole brains of microtubule-associated protein 1A/1B-light chain 3 (LC3) and C57BL/6 (B6) mice were irradiated once (10 Gy)(IR1), given 3 fractions in three weeks (IR3), or 6 fractions in six weeks (IR6). The median survival of LC3 mice was 56.5 days, and that of B6 mice was 65 days after IR6. LC3 mice showed more congestion and fibrosis in the lung after the IR3 and IR6 irradiation protocols than B6 mice. Quantitative proteomics of serum samples and lung RNA sequencing of the LC3 group showed that the common most clustered pathway of the IR3 group was the elastic fiber formation pathway, which contained Periostin (POSTN). POSTN in the motoneurons increased with increasing number of radiation fractions in LC3 mice. A 1 μg/g POSTN neutralizing antibody reduced the lung fibrosis of LC3 mice exposed to IR3 by one-third, and significantly prolonged the survival time of LC3 mice exposed to IR6. LDN-214117 and LRRK2-in-1 were the best two of sixteen transforming growth factor-beta1 (TGF-β) receptor and autophagy mediators to decrease Postn mRNA. These data led us to conclude that LC3 accelerated motoneuron secretion of POSTN and aggravated the RIBE in the lung after brain irradiation.
Collapse
Affiliation(s)
- Ke Zhang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, P R China
- Qidong People's Hospital/Affiliated Qidong Hospital of Nantong University, Qidong 226200, P R China
| | - Zhuojun Wu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, P R China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education 215123, P R China
| | - Ying Zhao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, P R China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education 215123, P R China
| | - Xinyu Qiu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, P R China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education 215123, P R China
| | - Fang Li
- School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, P R China
| | - Qiu Chen
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, P R China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education 215123, P R China
| | - Fengmei Cui
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, P R China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education 215123, P R China
| |
Collapse
|
2
|
Santos L, Nascimento R, Duarte A, Railean V, Amaral MD, Harrison PT, Gama-Carvalho M, Farinha CM. Mutation-class dependent signatures outweigh disease-associated processes in cystic fibrosis cells. Cell Biosci 2023; 13:26. [PMID: 36759923 PMCID: PMC9912517 DOI: 10.1186/s13578-023-00975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/28/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND The phenotypic heterogeneity observed in Cystic Fibrosis (CF) patients suggests the involvement of other genes, besides CFTR. Here, we combined transcriptome and proteome analysis to understand the global gene expression patterns associated with five prototypical CFTR mutations. RESULTS Evaluation of differentially expressed genes and proteins unveiled common and mutation-specific changes revealing functional signatures that are much more associated with the specific molecular defects associated with each mutation than to the CFTR loss-of-function phenotype. The combination of both datasets revealed that mutation-specific detected translated-transcripts (Dtt) have a high level of consistency. CONCLUSIONS This is the first combined transcriptomic and proteomic study focusing on prototypical CFTR mutations. Analysis of Dtt provides novel insight into the pathophysiology of CF, and the mechanisms through which each mutation class causes disease and will likely contribute to the identification of new therapeutic targets and/or biomarkers for CF.
Collapse
Affiliation(s)
- Lúcia Santos
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal ,grid.7872.a0000000123318773Department of Physiology, University College Cork, Cork, T12 K8AF Ireland
| | - Rui Nascimento
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Aires Duarte
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Violeta Railean
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Margarida D. Amaral
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Patrick T. Harrison
- grid.7872.a0000000123318773Department of Physiology, University College Cork, Cork, T12 K8AF Ireland
| | - Margarida Gama-Carvalho
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Carlos M. Farinha
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| |
Collapse
|
3
|
Jaganathan D, Bruscia EM, Kopp BT. Emerging Concepts in Defective Macrophage Phagocytosis in Cystic Fibrosis. Int J Mol Sci 2022; 23:7750. [PMID: 35887098 PMCID: PMC9319215 DOI: 10.3390/ijms23147750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Chronic inflammation and decline in lung function are major reasons for morbidity in CF. Mutant CFTR expressed in phagocytic cells such as macrophages contributes to persistent infection, inflammation, and lung disease in CF. Macrophages play a central role in innate immunity by eliminating pathogenic microbes by a process called phagocytosis. Phagocytosis is required for tissue homeostasis, balancing inflammation, and crosstalk with the adaptive immune system for antigen presentation. This review focused on (1) current understandings of the signaling underlying phagocytic mechanisms; (2) existing evidence for phagocytic dysregulation in CF; and (3) the emerging role of CFTR modulators in influencing CF phagocytic function. Alterations in CF macrophages from receptor initiation to phagosome formation are linked to disease progression in CF. A deeper understanding of macrophages in the context of CFTR and phagocytosis proteins at each step of phagosome formation might contribute to the new therapeutic development of dysregulated innate immunity in CF. Therefore, the review also indicates future areas of research in the context of CFTR and macrophages.
Collapse
Affiliation(s)
- Devi Jaganathan
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Emanuela M. Bruscia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Benjamin T. Kopp
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Division of Pulmonary Medicine, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| |
Collapse
|
4
|
Gao Q, Chang X, Yang M, Zheng J, Gong X, Liu H, Li K, Wang X, Zhan H, Li S, Feng S, Sun X, Sun Y. LncRNA MEG3 restrained pulmonary fibrosis induced by NiO NPs via regulating hedgehog signaling pathway-mediated autophagy. ENVIRONMENTAL TOXICOLOGY 2022; 37:79-91. [PMID: 34608745 DOI: 10.1002/tox.23379] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Long noncoding RNA maternally expressed gene 3 (lncRNA MEG3) was down-regulated in pulmonary fibrosis of rats induced by Nickel oxide nanoparticles (NiO NPs), while the downstream regulatory mechanisms of MEG3 remain unclear. This study aimed to investigate the relationship among MEG3, Hedgehog (Hh) signaling pathway and autophagy in pulmonary fibrosis caused by NiO NPs. The pulmonary fibrosis model in rats was constructed by intratracheal instillation of 0.015, 0.06, and 0.24 mg/kg NiO NPs twice a week for 9 weeks. Collagen deposition model was established by treating A549 cells with 25, 50, and 100 μg/mL NiO NPs for 24 h. Our results indicated that NiO NPs activated Hh pathway, down-regulated the expression of MEG3, and reduced autophagy activity in vivo and in vitro. Meanwhile, the autophagy process was promoted by Hh pathway inhibitor (CDG-0449), while the collagen formation in A549 cells was reduced by autophagy activator (Rapamycin). Furthermore, the overexpressed MEG3 inhibited the activation of Hh pathway, resulting in autophagy activity enhancement along with collagen formation reduction. In summary, lncRNA MEG3 can restrain pulmonary fibrosis induced by NiO NPs via regulating hedgehog signaling pathway-mediated autophagy, which may serve as a potential therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Qing Gao
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Mengmeng Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Jinfa Zheng
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuefeng Gong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Han Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Kun Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoxia Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haibing Zhan
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Sheng Li
- Department of Public Health, The First People's Hospital of Lanzhou city, Lanzhou, China
| | - Sanwei Feng
- Institute of Occupational Diseases, Gansu Baoshihua Hospital, Lanzhou, China
| | - Xingchang Sun
- Institute of Occupational Diseases, Gansu Baoshihua Hospital, Lanzhou, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
5
|
Künzi L, Easter M, Hirsch MJ, Krick S. Cystic Fibrosis Lung Disease in the Aging Population. Front Pharmacol 2021; 12:601438. [PMID: 33935699 PMCID: PMC8082404 DOI: 10.3389/fphar.2021.601438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/15/2021] [Indexed: 01/02/2023] Open
Abstract
The demographics of the population with cystic fibrosis (CF) is continuously changing, with nowadays adults outnumbering children and a median predicted survival of over 40 years. This leads to the challenge of treating an aging CF population, while previous research has largely focused on pediatric and adolescent patients. Chronic inflammation is not only a hallmark of CF lung disease, but also of the aging process. However, very little is known about the effects of an accelerated aging pathology in CF lungs. Several chronic lung disease pathologies show signs of chronic inflammation with accelerated aging, also termed “inflammaging”; the most notable being chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). In these disease entities, accelerated aging has been implicated in the pathogenesis via interference with tissue repair mechanisms, alterations of the immune system leading to impaired defense against pulmonary infections and induction of a chronic pro-inflammatory state. In addition, CF lungs have been shown to exhibit increased expression of senescence markers. Sustained airway inflammation also leads to the degradation and increased turnover of cystic fibrosis transmembrane regulator (CFTR). This further reduces CFTR function and may prevent the novel CFTR modulator therapies from developing their full efficacy. Therefore, novel therapies targeting aging processes in CF lungs could be promising. This review summarizes the current research on CF in an aging population focusing on accelerated aging in the context of chronic airway inflammation and therapy implications.
Collapse
Affiliation(s)
- Lisa Künzi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Epidemiology, Biostatistics and Prevention Institute, Department of Public and Global Health, University of Zürich, Zürich, Switzerland
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Gregory Fleming Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
6
|
Abstract
Antimicrobial therapies against cystic fibrosis (CF) lung infections are largely aimed at the traditional, well-studied CF pathogens such as Pseudomonas aeruginosa and Burkholderia cepacia complex, despite the fact that the CF lung harbors a complex and dynamic polymicrobial community. A clinical focus on the dominant pathogens ignores potentially important community-level interactions in disease pathology, perhaps explaining why these treatments are often less effective than predicted based on in vitro testing. Antimicrobial therapies against cystic fibrosis (CF) lung infections are largely aimed at the traditional, well-studied CF pathogens such as Pseudomonas aeruginosa and Burkholderia cepacia complex, despite the fact that the CF lung harbors a complex and dynamic polymicrobial community. A clinical focus on the dominant pathogens ignores potentially important community-level interactions in disease pathology, perhaps explaining why these treatments are often less effective than predicted based on in vitro testing. A better understanding of the ecological dynamics of this ecosystem may enable clinicians to harness these interactions and thereby improve treatment outcomes. Like all ecosystems, the CF lung microbial community develops through a series of stages, each of which may present with distinct microbial communities that generate unique host-microbe and microbe-microbe interactions, metabolic profiles, and clinical phenotypes. While insightful models have been developed to explain some of these stages and interactions, there is no unifying model to describe how these infections develop and persist. Here, we review current perspectives on the ecology of the CF airway and present the CF Ecological Succession (CFES) model that aims to capture the spatial and temporal complexity of CF lung infection, address current challenges in disease management, and inform the development of ecologically driven therapeutic strategies.
Collapse
|
7
|
Abstract
Acute Respiratory Distress Syndrome is a severe disorder affecting thousands of individuals worldwide. The available medical countermeasures do not sufficiently suppress the unacceptable high mortality rates associated with those in need. Thus, intense efforts aim to delineate the function of the lung endothelium, so to deliver new therapeutic approaches against this disease. The present manuscript attempts to shed light on the interrelations between the unfolded protein response and autophagy towards lung disease, to deliver a new line of possible therapeutic approaches against the ferocious Acute Respiratory Distress Syndrome.
Collapse
Affiliation(s)
- Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA
| | - Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana 71201, USA
| |
Collapse
|
8
|
Recchiuti A, Isopi E, Romano M, Mattoscio D. Roles of Specialized Pro-Resolving Lipid Mediators in Autophagy and Inflammation. Int J Mol Sci 2020; 21:E6637. [PMID: 32927853 PMCID: PMC7555248 DOI: 10.3390/ijms21186637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a catabolic pathway that accounts for degradation and recycling of cellular components to extend cell survival under stress conditions. In addition to this prominent role, recent evidence indicates that autophagy is crucially involved in the regulation of the inflammatory response, a tightly controlled process aimed at clearing the inflammatory stimulus and restoring tissue homeostasis. To be efficient and beneficial to the host, inflammation should be controlled by a resolution program, since uncontrolled inflammation is the underlying cause of many pathologies. Resolution of inflammation is an active process mediated by a variety of mediators, including the so-called specialized pro-resolving lipid mediators (SPMs), a family of endogenous lipid autacoids known to regulate leukocyte infiltration and activities, and counterbalance cytokine production. Recently, regulation of autophagic mechanisms by these mediators has emerged, uncovering unappreciated connections between inflammation resolution and autophagy. Here, we summarize mechanisms of autophagy and resolution, focusing on the contribution of autophagy in sustaining paradigmatic examples of chronic inflammatory disorders. Then, we discuss the evidence that SPMs can restore dysregulated autophagy, hypothesizing that resolution of inflammation could represent an innovative approach to modulate autophagy and its impact on the inflammatory response.
Collapse
Affiliation(s)
| | | | | | - Domenico Mattoscio
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Sciences, University of Chieti—Pescara, 66100 Chieti, Italy; (A.R.); (E.I.); (M.R.)
| |
Collapse
|
9
|
Rezaei S, Mahjoubin-Tehran M, Aghaee-Bakhtiari SH, Jalili A, Movahedpour A, Khan H, Moghoofei M, Shojaei Z, R Hamblin M, Mirzaei H. Autophagy-related MicroRNAs in chronic lung diseases and lung cancer. Crit Rev Oncol Hematol 2020; 153:103063. [DOI: 10.1016/j.critrevonc.2020.103063] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 06/11/2020] [Accepted: 07/12/2020] [Indexed: 12/24/2022] Open
|
10
|
Pehote G, Vij N. Autophagy Augmentation to Alleviate Immune Response Dysfunction, and Resolve Respiratory and COVID-19 Exacerbations. Cells 2020; 9:cells9091952. [PMID: 32847034 PMCID: PMC7565665 DOI: 10.3390/cells9091952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of cellular homeostasis requires the synthesis of new proteins (proteostasis) and organelles, and the effective removal of misfolded or impaired proteins and cellular debris. This cellular homeostasis involves two key proteostasis mechanisms, the ubiquitin proteasome system and the autophagy–lysosome pathway. These catabolic pathways have been known to be involved in respiratory exacerbations and the pathogenesis of various lung diseases, such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and coronavirus disease-2019 (COVID-19). Briefly, proteostasis and autophagy processes are known to decline over time with age, cigarette or biomass smoke exposure, and/or influenced by underlying genetic factors, resulting in the accumulation of misfolded proteins and cellular debris, elevating apoptosis and cellular senescence, and initiating the pathogenesis of acute or chronic lung disease. Moreover, autophagic dysfunction results in an impaired microbial clearance, post-bacterial and/or viral infection(s) which contribute to the initiation of acute and recurrent respiratory exacerbations as well as the progression of chronic obstructive and restrictive lung diseases. In addition, the autophagic dysfunction-mediated cystic fibrosis transmembrane conductance regulator (CFTR) immune response impairment further exacerbates the lung disease. Recent studies demonstrate the therapeutic potential of novel autophagy augmentation strategies, in alleviating the pathogenesis of chronic obstructive or restrictive lung diseases and exacerbations such as those commonly seen in COPD, CF, ALI/ARDS and COVID-19.
Collapse
Affiliation(s)
- Garrett Pehote
- Michigan State University College of Osteopathic Medicine, East Lansing, MI 48823, USA;
| | - Neeraj Vij
- Department of Pediatrics and Pulmonary Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- PRECISION THERANOSTICS INC, Baltimore, MD 21202, USA
- VIJ BIOTECH, Baltimore, MD 21202, USA
- Correspondence: or ; Tel.: +1-240-623-0757
| |
Collapse
|
11
|
Costantini C, Puccetti M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Bellet MM, Cellini B, Giovagnoli S, Romani L. Selectively targeting key inflammatory pathways in cystic fibrosis. Eur J Med Chem 2020; 206:112717. [PMID: 32823008 DOI: 10.1016/j.ejmech.2020.112717] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/10/2020] [Accepted: 07/21/2020] [Indexed: 01/04/2023]
Abstract
Cystic fibrosis (CF) is a rare genetic disorder caused by a defect in the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR), resulting in ionic imbalance of surface fluid. Although affecting multiple organs, the progressive deterioration of respiratory function by recurrent infections and chronic inflammation represents the main cause of morbidity and mortality in CF patients. The development of modulators targeting the basic defect of CFTR has represented a major breakthrough in CF therapy, but the impact on inflammation has remained enigmatic. The emerging scenario taking hold in the field points to inflammation as a major, somehow missed, therapeutic target for prevention of lung decline. Not surprisingly, the development of anti-inflammatory drugs is taking its share in the drug development pipeline. But the path is not straightforward and targeting inflammation should be balanced with the increased risk of infection. The strategy to restore the homeostatic regulation of inflammation to efficiently respond to infection while preventing lung damage needs to be based on identifying and targeting endogenous immunoregulatory pathways that are defective in CF. We herein provide an overview of anti-inflammatory drugs currently approved or under investigation in CF patients, and present our recent studies on how the knowledge on defective immune pathways in CF may translate into innovative and selective anti-inflammatory therapeutics. Through the discovery of naturally occurring molecules or their synthetic mimics, this review emphasizes the critical importance of selectively targeting key inflammatory pathways to preserve immunocompetence in CF patients.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, 06132, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Claudia Stincardini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Barbara Cellini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, Perugia, 06132, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| |
Collapse
|
12
|
Dhillon BK, Smith M, Baghela A, Lee AHY, Hancock REW. Systems Biology Approaches to Understanding the Human Immune System. Front Immunol 2020; 11:1683. [PMID: 32849587 PMCID: PMC7406790 DOI: 10.3389/fimmu.2020.01683] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022] Open
Abstract
Systems biology is an approach to interrogate complex biological systems through large-scale quantification of numerous biomolecules. The immune system involves >1,500 genes/proteins in many interconnected pathways and processes, and a systems-level approach is critical in broadening our understanding of the immune response to vaccination. Changes in molecular pathways can be detected using high-throughput omics datasets (e.g., transcriptomics, proteomics, and metabolomics) by using methods such as pathway enrichment, network analysis, machine learning, etc. Importantly, integration of multiple omic datasets is becoming key to revealing novel biological insights. In this perspective article, we highlight the use of protein-protein interaction (PPI) networks as a multi-omics integration approach to unravel information flow and mechanisms during complex biological events, with a focus on the immune system. This involves a combination of tools, including: InnateDB, a database of curated interactions between genes and protein products involved in the innate immunity; NetworkAnalyst, a visualization and analysis platform for InnateDB interactions; and MetaBridge, a tool to integrate metabolite data into PPI networks. The application of these systems techniques is demonstrated for a variety of biological questions, including: the developmental trajectory of neonates during the first week of life, mechanisms in host-pathogen interaction, disease prognosis, biomarker discovery, and drug discovery and repurposing. Overall, systems biology analyses of omics data have been applied to a variety of immunology-related questions, and here we demonstrate the numerous ways in which PPI network analysis can be a powerful tool in contributing to our understanding of the immune system and the study of vaccines.
Collapse
Affiliation(s)
- Bhavjinder K. Dhillon
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Maren Smith
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Arjun Baghela
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy H. Y. Lee
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Molecular Biology & Biochemistry Department, Simon Fraser University, Burnaby, BC, Canada
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
13
|
Fang L, Shen Q, Wu H, He F, Ding P, Xu K, Yan X, Wang M, Li S, Liu R. TLR2 favors OVA-induced allergic airway inflammation in mice through JNK signaling pathway with activation of autophagy. Life Sci 2020; 256:117896. [PMID: 32504758 DOI: 10.1016/j.lfs.2020.117896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/21/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022]
Abstract
AIMS Numerous studies indicate that toll-like receptor 2 (TLR2) led to divergent effects in asthma. The occurrence of autophagy in asthma pathogenesis is still incompletely understood. Here, we aimed to investigate the role of TLR2 and the underlying mechanisms in allergic airway inflammation and autophagy activation. MAIN METHODS C57BL/6 and TLR2 knockout (TLR2-/-) mice were subjected to an ovalbumin (OVA)-immunized allergic airway model, and were treated with SP600125. Differential cell counts in bronchoalveolar lavage fluid were determined by Wright's staining. Histological analysis of airway inflammation was determined by haematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining. The levels of OVA-specific immunoglobulin E (IgE), tumor necrosis factor α (TNF-α) and interleukin 10 (IL-10) were detected by enzyme-linked immunosorbent assay (ELISA). Proteins expression in lung tissues was detected by western blot, expression of TLR2 was further observed by immunofluorescence. Autophagy activation was determined by western blot and transmission electron microscopy (TEM). KEY FINDINGS TLR2 expression was increased upon OVA challenge, and TLR2 deficiency was associated with decreased allergic airway inflammation. Meanwhile, TLR2 deficiency weakened autophagy activation. Moreover, inhibition of c-Jun N-terminal kinase (JNK) by SP600125 also suppressed OVA-induced allergic airway inflammation and autophagy activation. Interestingly, treating TLR2-/- mice with SP600125 showed similar OVA-induced allergic airway inflammation and autophagy activation compared to that in vehicle-treated TLR2-/- mice. SIGNIFICANCE TLR2 might contribute to the maintenance of allergic airway inflammation through JNK signaling pathway accompanying with autophagy activation. These findings may provide a novel signal target for prevention of allergic airway inflammation.
Collapse
Affiliation(s)
- Lei Fang
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Qiying Shen
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Huimei Wu
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Fang He
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; No.1 Department of Respiratory Medicine, Anhui Chest Hospital, Jixi Road 397, Hefei, Anhui 230022, China
| | - Peishan Ding
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Ke Xu
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230031, China
| | - Xuebo Yan
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Muzi Wang
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China
| | - Shuai Li
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China; Intensive Care Unit, The Fourth Affiliated Hospital of Anhui Medical University, Huaihai Avenue 100, Hefei, Anhui 230012, China
| | - Rongyu Liu
- Department of Geriatric Respiratory and Critical Care, Provincial Key Laboratory of Molecular Medicine for Geriatric Disease, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, China.
| |
Collapse
|
14
|
Wrigley R, Phipps-Green AJ, Topless RK, Major TJ, Cadzow M, Riches P, Tausche AK, Janssen M, Joosten LAB, Jansen TL, So A, Harré Hindmarsh J, Stamp LK, Dalbeth N, Merriman TR. Pleiotropic effect of the ABCG2 gene in gout: involvement in serum urate levels and progression from hyperuricemia to gout. Arthritis Res Ther 2020; 22:45. [PMID: 32164793 PMCID: PMC7069001 DOI: 10.1186/s13075-020-2136-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The ABCG2 Q141K (rs2231142) and rs10011796 variants associate with hyperuricaemia (HU). The effect size of ABCG2 rs2231142 on urate is ~ 60% that of SLC2A9, yet the effect size on gout is greater. We tested the hypothesis that ABCG2 plays a role in the progression from HU to gout by testing for association of ABCG2 rs2231142 and rs10011796 with gout using HU controls. METHODS We analysed 1699 European gout cases and 14,350 normouricemic (NU) and HU controls, and 912 New Zealand (NZ) Polynesian (divided into Eastern and Western Polynesian) gout cases and 696 controls. Association testing was performed using logistic and linear regression with multivariate adjusting for confounding variables. RESULTS In Europeans and Polynesians, the ABCG2 141K (T) allele was associated with gout using HU controls (OR = 1.85, P = 3.8E- 21 and ORmeta = 1.85, P = 1.3E- 03, respectively). There was evidence for an effect of 141K in determining HU in European (OR = 1.56, P = 1.7E- 18) but not in Polynesian (ORmeta = 1.49, P = 0.057). For SLC2A9 rs11942223, the T allele associated with gout in the presence of HU in European (OR = 1.37, P = 4.7E- 06), however significantly weaker than ABCG2 rs2231142 141K (PHet = 0.0023). In Western Polynesian and European, there was epistatic interaction between ABCG2 rs2231142 and rs10011796. Combining the presence of the 141K allele with the rs10011796 CC-genotype increased gout risk, in the presence of HU, 21.5-fold in Western Polynesian (P = 0.009) and 2.6-fold in European (P = 9.9E- 06). The 141K allele of ABCG2 associated with increased gout flare frequency in Polynesian (Pmeta = 2.5E- 03). CONCLUSION These data are consistent with a role for ABCG2 141K in gout in the presence of established HU.
Collapse
Affiliation(s)
- Rebekah Wrigley
- Department of Biochemistry, University of Otago, Box 56, Dunedin, New Zealand
| | | | - Ruth K Topless
- Department of Biochemistry, University of Otago, Box 56, Dunedin, New Zealand
| | - Tanya J Major
- Department of Biochemistry, University of Otago, Box 56, Dunedin, New Zealand
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, Box 56, Dunedin, New Zealand
| | - Philip Riches
- Rheumatic Diseases Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Anne-Kathrin Tausche
- Department of Rheumatology, University Clinic "Carl-Gustav-Carus", Dresden, Germany
| | - Matthijs Janssen
- Department of Rheumatology, VieCuri Medical Center, Venlo, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tim L Jansen
- Department of Rheumatology, VieCuri Medical Center, Venlo, The Netherlands
| | - Alexander So
- Laboratory of Rheumatology, University of Lausanne, CHUV, Nestlé 05-5029, 1011, Lausanne, Switzerland
| | | | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Box 56, Dunedin, New Zealand.
| |
Collapse
|
15
|
Zhao H, Wang Y, Qiu T, Liu W, Yao P. Autophagy, an important therapeutic target for pulmonary fibrosis diseases. Clin Chim Acta 2019; 502:139-147. [PMID: 31877297 DOI: 10.1016/j.cca.2019.12.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
As an evolutionarily conserved intracellular degradation pathway, autophagy is essential to cellular homeostasis. Several studies have demonstrated that autophagy showed an important effect on some pulmonary fibrosis diseases, including idiopathic pulmonary fibrosis (IPF), cystic fibrosis lung disease, silicosis and smoking-induced pulmonary fibrosis. For example, autophagy mitigates the pathological progression of IPF by regulating the apoptosis of fibroblasts and the senescence of alveolar epithelial cells. In addition, autophagy ameliorates cystic fibrosis lung disease via rescuing transmembrane conductance regulators (CFTRs) to the plasma membrane. Furthermore, autophagy alleviates the silica-induced pulmonary fibrosis by decreasing apoptosis of alveolar epithelial cells in silicosis. However, excessive macrophage autophagy aggravates the pathogenesis of silicosis fibrosis by promoting the proliferation and migration of lung fibroblasts in silicosis. Autophagy is also involved in smoking-induced pulmonary fibrosis, coal workers' pneumoconiosis, ionizing radiation-mediated pulmonary fibrosis and heavy metal nanoparticle-mediated pulmonary fibrosis. In this review, the role and signalling mechanisms of autophagy in the progression of pulmonary fibrosis diseases have been systematically analysed. It has provided a new insight into the therapeutic potential associated with autophagy in pulmonary fibrosis diseases. In conclusion, the targeting of autophagy might prove to be a prospective avenue for the therapeutic intervention of pulmonary fibrosis diseases.
Collapse
Affiliation(s)
- Hong Zhao
- Nursing College, University of South China, Hengyang, 421001, China
| | - Yiqun Wang
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China
| | - Tingting Qiu
- Nursing College, University of South China, Hengyang, 421001, China
| | - Wei Liu
- Department of Intensive Care Units, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China.
| | - Pingbo Yao
- Department of Clinical Technology, Changsha Health Vocational College, Changsha 410100, China.
| |
Collapse
|
16
|
Protein Misfolding and Endoplasmic Reticulum Stress in Chronic Lung Disease: Will Cell-Specific Targeting Be the Key to the Cure? Chest 2019; 157:1207-1220. [PMID: 31778676 DOI: 10.1016/j.chest.2019.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic lung disease accounts for a significant global burden with respect to death, disability, and health-care costs. Due to the heterogeneous nature and limited treatment options for these diseases, it is imperative that the cellular and molecular mechanisms underlying the disease pathophysiology are further understood. The lung is a complex organ with a diverse cell population, and each cell type will likely have different roles in disease initiation, progression, and resolution. The effectiveness of a given therapeutic agent may depend on the net effect on each of these cell types. Over the past decade, it has been established that endoplasmic reticulum stress and the unfolded protein response are involved in the development of several chronic lung diseases. These conserved cellular pathways are important for maintaining cellular proteostasis, but their aberrant activation can result in pathology. This review discusses the current understanding of endoplasmic reticulum stress and the unfolded protein response at the cellular level in the development and progression of various chronic lung diseases. We highlight the need for increased understanding of the specific cellular contributions of unfolded protein response activation to these pathologies and suggest that the development of cell-specific targeted therapies is likely required to further decrease disease progression and to promote resolution of chronic lung disease.
Collapse
|
17
|
Alencar-Silva T, Zonari A, Foyt D, Gang M, Pogue R, Saldanha-Araujo F, Dias SC, Franco OL, Carvalho JL. IDR-1018 induces cell proliferation, migration, and reparative gene expression in 2D culture and 3D human skin equivalents. J Tissue Eng Regen Med 2019; 13:2018-2030. [PMID: 31408919 DOI: 10.1002/term.2953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 01/12/2023]
Abstract
Skin lesions are associated with functional/cosmetic problems for those afflicted. Scarless regeneration is a challenge, not limited to the skin, and focus of active investigation. Recently, the host defense peptide innate defense regulatory peptide 1018 (IDR-1018) has shown exciting regenerative properties. Nevertheless, literature regarding IDR-1018 regenerative potential is scarce and limited to animal models. Here, we evaluated the regenerative potential of IDR-1018 using human 2D and 3D human skin equivalents. First, we investigated IDR-1018 using human cells found in skin-primary fibroblasts, primary keratinocytes, and the MeWo melanocytes cell line. IDR-1018 promoted cell proliferation and expression of marker of proliferation Ki-67, matrix metalloproteinase 1, and hyaluronan synthase 2 by fibroblasts. In keratinocytes, a drastic increase in expression was observed for Ki-67, matrix metalloproteinase 1, C-X-C motif chemokine receptor type 4, C-X-C motif chemokine receptor type 7, fibroblast growth factor 2, hyaluronan synthase 2, vascular endothelial growth factor, and elastin, reflecting an intense stimulation of these cells. In melanocytes, increased migration and proliferation were observed following IDR-1018 treatment. The capacity of IDR-1018 to promote dermal contraction was verified using a dermal model. Finally, using a 3D human skin equivalent lesion model, we revealed that the regenerative potential of IDR1018, previously tested in mice and pigs, is valid for human skin tissue. Lesions closed faster in IDR-1018-treated samples, and the gene expression signature observed in 2D was reproduced in the 3D human skin equivalents. Overall, the present data show the regenerative potential of IDR-1018 in an experimental system comprising human cells, underscoring the potential application for clinical investigation.
Collapse
Affiliation(s)
- Thuany Alencar-Silva
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brazil
| | | | - Daniel Foyt
- OneSkin Technologies, San Francisco, CA, USA
| | | | - Robert Pogue
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brazil
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia, Departamento de Ciências da Saúde, Universidade de Brasília, Brasilia, Brazil.,Programa de Pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
| | - Simoni Campos Dias
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brazil.,Pós-Graduação em Biologia Animal, Universidade de Brasília, Campus Darcy Ribeiro, Brasília, Brazil
| | - Octavio Luiz Franco
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brazil.,S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil.,Programa de Pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Juliana Lott Carvalho
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brazil.,OneSkin Technologies, San Francisco, CA, USA.,Faculdade de Medicina, Universidade de Brasília, Brasilia, Brazil
| |
Collapse
|
18
|
Rossi GA, Morelli P, Galietta LJ, Colin AA. Airway microenvironment alterations and pathogen growth in cystic fibrosis. Pediatr Pulmonol 2019; 54:497-506. [PMID: 30620146 DOI: 10.1002/ppul.24246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
Abstract
Cystic Fibrosis Transmembrane Regulator (CFTR) dysfunction is associated with epithelial cell vulnerability and with dysregulation of the local inflammatory responses resulting in excessive airway neutrophilic inflammation and pathogen growth. In combination with impaired mucociliary clearance, and dysregulation of defense function, bacterial infection follows with eventual airway damage and remodeling. Because of these inherent vulnerabilities, viral infections are also more severe and prolonged and appear to render the airway even more prone to bacterial infection. Airway acidity, deficient nitric oxide production and increased iron concentrations, further enhance the airway milieu's susceptibility to infection. Novel diagnostic techniques of the airway microbiome elucidate the coexistence of an array of non-virulent taxa beyond the recognized virulent organisms, predominantly Pseudomonas aeruginosa. The complex interplay between these two bacterial populations, including upregulation of virulence genes and utilization of mucin as a nutrient source, modulates the action of pathogens, modifies the CF airway milieu and contributes to the processes leading to airway derangement. The review provides an update on recent advances of the complex mechanisms that render the CF airway vulnerable to inflammation, infection and ultimately structural damage, the key pathogenetic elements of CF. The recent contributions on CF pathogenesis will hopefully help in identifying new prophylactic measures and therapeutic targets for this highly destructive disorder.
Collapse
Affiliation(s)
- Giovanni A Rossi
- Department of Pediatrics, Pulmonary and Allergy Disease Unit and Cystic Fibrosis Center, Genoa, Italy
| | - Patrizia Morelli
- Microbiology Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Luis J Galietta
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Andrew A Colin
- Division of Pediatric Pulmonology, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
19
|
Ideozu JE, Zhang X, McColley S, Levy H. Transcriptome Profiling and Molecular Therapeutic Advances in Cystic Fibrosis: Recent Insights. Genes (Basel) 2019; 10:genes10030180. [PMID: 30813620 PMCID: PMC6470978 DOI: 10.3390/genes10030180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
In cystic fibrosis (CF), mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene disrupt the capacity of the encoded protein to function as a channel to transport chloride ions and water across cell membranes. The consequences are deleterious, system-wide, and immensely variable, even among patients with the same CFTR genotype. This underscores the need to characterize the mechanisms contributing to CF pathophysiology. Gene replacement and gene editing therapies have been pursued intensively and are expected to provide a one-time treatment for CF. However, gene replacement therapy is limited by the lack of efficient vectors to deliver functional copies of CFTR to cells without immunological complications, while gene editing technologies such as CRISPR/Cas9 are still in their infancy, mainly useful in somatic cells and limited by off-target insertions. Small molecule treatments targeted at potentiating or correcting CFTR have shown clinical benefits, but they are limited to a few CFTR mutations and insufficient to overcome challenges related to clinical heterogeneity. Transcriptome profiling approaches have emerged as robust tools capable of characterizing phenotypic variability and revealing novel molecular targets with therapeutic potential for CF. We summarize current insights gained through transcriptome profiling approaches in CF studies and recent advances in molecular therapeutics.
Collapse
Affiliation(s)
- Justin E Ideozu
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Human Molecular Genetics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
- Feinberg School of Medicine at Northwestern University Chicago, Chicago, IL 60611, USA.
| | - Xi Zhang
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Human Molecular Genetics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
- Feinberg School of Medicine at Northwestern University Chicago, Chicago, IL 60611, USA.
| | - Susanna McColley
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Feinberg School of Medicine at Northwestern University Chicago, Chicago, IL 60611, USA.
| | - Hara Levy
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Human Molecular Genetics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
- Feinberg School of Medicine at Northwestern University Chicago, Chicago, IL 60611, USA.
| |
Collapse
|
20
|
Haney EF, Straus SK, Hancock REW. Reassessing the Host Defense Peptide Landscape. Front Chem 2019; 7:43. [PMID: 30778385 PMCID: PMC6369191 DOI: 10.3389/fchem.2019.00043] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/15/2019] [Indexed: 12/18/2022] Open
Abstract
Current research has demonstrated that small cationic amphipathic peptides have strong potential not only as antimicrobials, but also as antibiofilm agents, immune modulators, and anti-inflammatories. Although traditionally termed antimicrobial peptides (AMPs) these additional roles have prompted a shift in terminology to use the broader term host defense peptides (HDPs) to capture the multi-functional nature of these molecules. In this review, we critically examined the role of AMPs and HDPs in infectious diseases and inflammation. It is generally accepted that HDPs are multi-faceted mediators of a wide range of biological processes, with individual activities dependent on their polypeptide sequence. In this context, we explore the concept of chemical space as it applies to HDPs and hypothesize that the various functions and activities of this class of molecule exist on independent but overlapping activity landscapes. Finally, we outline several emerging functions and roles of HDPs and highlight how an improved understanding of these processes can potentially be leveraged to more fully realize the therapeutic promise of HDPs.
Collapse
Affiliation(s)
- Evan F Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Protective Features of Autophagy in Pulmonary Infection and Inflammatory Diseases. Cells 2019; 8:cells8020123. [PMID: 30717487 PMCID: PMC6406971 DOI: 10.3390/cells8020123] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process involving autolysosomal degradation of cellular components, including protein aggregates, damaged organelles (such as mitochondria, endoplasmic reticulum, and others), as well as various pathogens. Thus, the autophagy pathway represents a major adaptive response for the maintenance of cellular and tissue homeostasis in response to numerous cellular stressors. A growing body of evidence suggests that autophagy is closely associated with diverse human diseases. Specifically, acute lung injury (ALI) and inflammatory responses caused by bacterial infection or xenobiotic inhalation (e.g., chlorine and cigarette smoke) have been reported to involve a spectrum of alterations in autophagy phenotypes. The role of autophagy in pulmonary infection and inflammatory diseases could be protective or harmful dependent on the conditions. In this review, we describe recent advances regarding the protective features of autophagy in pulmonary diseases, with a focus on ALI, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), tuberculosis, pulmonary arterial hypertension (PAH) and cystic fibrosis.
Collapse
|
22
|
Ryter SW, Bhatia D, Choi ME. Autophagy: A Lysosome-Dependent Process with Implications in Cellular Redox Homeostasis and Human Disease. Antioxid Redox Signal 2019; 30:138-159. [PMID: 29463101 PMCID: PMC6251060 DOI: 10.1089/ars.2018.7518] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/20/2018] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Autophagy, a lysosome-dependent homeostatic process inherent to cells and tissues, has emerging significance in the pathogenesis of human disease. This process enables the degradation and turnover of cytoplasmic substrates via membrane-dependent sequestration in autophagic vesicles (autophagosomes) and subsequent lysosomal delivery of cargo. Recent Advances: Selective forms of autophagy can target specific substrates (e.g., organelles, protein aggregates, and lipids) for processing. Autophagy is highly regulated by oxidative stress, including exposure to altered oxygen tension, by direct and indirect mechanisms, and contributes to inducible defenses against oxidative stress. Mitochondrial autophagy (mitophagy) plays a critical role in the oxidative stress response, through maintenance of mitochondrial integrity. CRITICAL ISSUES Autophagy can impact a number of vital cellular processes including inflammation and adaptive immunity, host defense, lipid metabolism and storage, mitochondrial homeostasis, and clearance of aggregated proteins, all which may be of significance in human disease. Autophagy can exert both maladaptive and adaptive roles in disease pathogenesis, which may also be influenced by autophagy impairment. This review highlights the essential roles of autophagy in human diseases, with a focus on diseases in which oxidative stress or inflammation play key roles, including human lung, liver, kidney and heart diseases, metabolic diseases, and diseases of the cardiovascular and neural systems. FUTURE DIRECTIONS Investigations that further elucidate the complex role of autophagy in the pathogenesis of disease will facilitate targeting this pathway for therapies in specific diseases.
Collapse
Affiliation(s)
- Stefan W. Ryter
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York
| | - Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Mary E. Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
- NewYork-Presbyterian Hospital, Weill Cornell Medical Center, New York, New York
| |
Collapse
|
23
|
Assani K, Shrestha CL, Rinehardt H, Zhang S, Robledo-Avila F, Wellmerling J, Partida-Sanchez S, Cormet-Boyaka E, Reynolds SD, Schlesinger LS, Kopp BT. AR-13 reduces antibiotic-resistant bacterial burden in cystic fibrosis phagocytes and improves cystic fibrosis transmembrane conductance regulator function. J Cyst Fibros 2018; 18:622-629. [PMID: 30366849 DOI: 10.1016/j.jcf.2018.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 09/13/2018] [Accepted: 10/14/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND There are no effective treatments for Burkholderia cenocepacia in patients with cystic fibrosis (CF) due to bacterial multi-drug resistance and defective host killing. We demonstrated that decreased bacterial killing in CF is caused by reduced macrophage autophagy due to defective cystic fibrosis transmembrane conductance regulator (CFTR) function. AR-12 is a small molecule autophagy inducer that kills intracellular pathogens such as Francisella. We evaluated the efficacy of AR-12 and a new analogue AR-13 in reducing bacterial burden in CF phagocytes. METHODS Human CF and non-CF peripheral blood monocyte-derived macrophages, neutrophils, and nasal epithelial cells were exposed to CF bacterial strains in conjunction with treatment with antibiotics and/or AR compounds. RESULTS AR-13 and not AR-12 had growth inhibition on B. cenocepacia and methicillin-resistantStaphylococcus aureus (MRSA) in media alone. There was a 99% reduction in MRSA in CF macrophages, 71% reduction in Pseudomonas aeruginosa in CF neutrophils, and 70% reduction in non-CF neutrophils using AR-13. Conversely, there was no reduction in B. cenocepacia in infected CF and non-CF macrophages using AR-13 alone, but AR-13 and antibiotics synergistically reduced B. cenocepacia in CF macrophages. AR-13 improved autophagy in CF macrophages and CF patient-derived epithelial cells, and increased CFTR protein expression and channel function in CF epithelial cells. CONCLUSIONS The novel AR-12 analogue AR-13, in combination with antibiotics, reduced antibiotic-resistant bacterial burden in CF phagocytes, which correlated with increased autophagy and CFTR expression. AR-13 is a novel therapeutic for patients infected with B. cenocepacia and other resistant organisms that lack effective therapies.
Collapse
Affiliation(s)
- Kaivon Assani
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Chandra L Shrestha
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Hannah Rinehardt
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Shuzhong Zhang
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Frank Robledo-Avila
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Jack Wellmerling
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Susan D Reynolds
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | | | - Benjamin T Kopp
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, United States.
| |
Collapse
|
24
|
Massip-Copiz MM, Santa-Coloma TA. Extracellular pH and lung infections in cystic fibrosis. Eur J Cell Biol 2018; 97:402-410. [PMID: 29933921 DOI: 10.1016/j.ejcb.2018.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by CFTR mutations. It is characterized by high NaCl concentration in sweat and the production of a thick and sticky mucus, occluding secretory ducts, intestine and airways, accompanied by chronic inflammation and infections of the lungs. This causes a progressive and lethal decline in lung function. Therefore, finding the mechanisms driving the high susceptibility to lung infections has been a key issue. For decades the prevalent hypothesis was that a reduced airway surface liquid (ASL) volume and composition, and the consequent increased mucus concentration (dehydration), create an environment favoring infections. However, a few years ago, in a pig model of CF, the Na+/K+ concentrations and the ASL volume were found intact. Immediately a different hypothesis arose, postulating a reduced ASL pH as the cause for the increased susceptibility to infections, due to a diminished bicarbonate secretion through CFTR. Noteworthy, a recent report found normal ASL pH values in CF children and in cultured primary airway cells, challenging the ASL pH hypothesis. On the other hand, recent evidences revitalized the hypothesis of a reduced ASL secretion. Thus, the role of the ASL pH in the CF is still a controversial matter. In this review we discuss the basis that sustain the role of CFTR in modulating the extracellular pH, and the recent results sustaining the different points of view. Finding the mechanisms of CFTR signaling that determine the susceptibility to infections is crucial to understand the pathophysiology of CF and related lung diseases.
Collapse
Affiliation(s)
- María Macarena Massip-Copiz
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED UCA-CONICET), The National Scientific and Technical Research Council (CONICET), and School of Medical Sciences, The Pontifical Catholic University of Argentina (UCA), Buenos Aires, Argentina
| | - Tomás Antonio Santa-Coloma
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED UCA-CONICET), The National Scientific and Technical Research Council (CONICET), and School of Medical Sciences, The Pontifical Catholic University of Argentina (UCA), Buenos Aires, Argentina.
| |
Collapse
|
25
|
Tosco A, Villella VR, Castaldo A, Kroemer G, Maiuri L, Raia V. Repurposing therapies for the personalised treatment of cystic fibrosis. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1483231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Antonella Tosco
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Valeria R. Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Alice Castaldo
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Guido Kroemer
- Equipe11 labellisée Ligue Nationale Contrele Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Paris, Sorbonne Paris Cité, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, HôpitalEuropéen Georges Pompidou, AP-HP, Paris, France
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
26
|
Piyadasa H, Hemshekhar M, Altieri A, Basu S, van der Does AM, Halayko AJ, Hiemstra PS, Mookherjee N. Immunomodulatory innate defence regulator (IDR) peptide alleviates airway inflammation and hyper-responsiveness. Thorax 2018; 73:908-917. [PMID: 29853649 DOI: 10.1136/thoraxjnl-2017-210739] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 04/19/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Exacerbation in asthma is associated with decreased expression of specific host defence peptides (HDPs) in the lungs. We examined the effects of a synthetic derivative of HDP, innate defence regulator (IDR) peptide IDR-1002, in house dust mite (HDM)-challenged murine model of asthma, in interleukin (IL)-33-challenged mice and in human primary bronchial epithelial cells (PBECs). METHODS IDR-1002 (6 mg/kg per mouse) was administered (subcutaneously) in HDM-challenged and/or IL-33-challenged BALB/c mice. Lung function analysis was performed with increasing dose of methacholine by flexiVent small animal ventilator, cell differentials in bronchoalveolar lavage performed by modified Wright-Giemsa staining, and cytokines monitored by MesoScale Discovery assay and ELISA. PBECs stimulated with tumour necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ), with or without IDR-1002, were analysed by western blots. RESULTS IDR-1002 blunted HDM challenge-induced airway hyper-responsiveness (AHR), and lung leucocyte accumulation including that of eosinophils and neutrophils, in HDM-challenged mice. Concomitantly, IDR-1002 suppressed HDM-induced IL-33 in the lungs. IFN-γ/TNF-α-induced IL-33 production was abrogated by IDR-1002 in PBECs. Administration of IL-33 in HDM-challenged mice, or challenge with IL-33 alone, mitigated the ability of IDR-1002 to control leucocyte accumulation in the lungs, suggesting that the suppression of IL-33 is essential for the anti-inflammatory activity of IDR-1002. In contrast, the peptide significantly reduced either HDM, IL-33 or HDM+IL-33 co-challenge-induced AHR in vivo. CONCLUSION This study demonstrates that an immunomodulatory IDR peptide controls the pathophysiology of asthma in a murine model. As IL-33 is implicated in steroid-refractory severe asthma, our findings on the effects of IDR-1002 may contribute to the development of novel therapies for steroid-refractory severe asthma.
Collapse
Affiliation(s)
- Hadeesha Piyadasa
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anthony Altieri
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sujata Basu
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Anne M van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.,The Canadian Respiratory Research Network, Ottawa, Ontario, Canada
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.,The Canadian Respiratory Research Network, Ottawa, Ontario, Canada
| |
Collapse
|
27
|
Marin-Luevano P, Trujillo V, Rodriguez-Carlos A, González-Curiel I, Enciso-Moreno JA, Hancock REW, Rivas-Santiago B. Induction by innate defence regulator peptide 1018 of pro-angiogenic molecules and endothelial cell migration in a high glucose environment. Peptides 2018; 101:135-144. [PMID: 29353019 DOI: 10.1016/j.peptides.2018.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/03/2018] [Accepted: 01/14/2018] [Indexed: 02/06/2023]
Abstract
Synthetic innate defence regulator (IDR) peptides such as IDR-1018 modulate immunity to promote key protective functions including chemotaxis, wound healing, and anti-infective activity, while suppressing pro-inflammatory responses to non-pathological levels. Here we demonstrated that IDR-1018 induced, by up to 75-fold, pro-angiogenic VEGF-165 in keratinocytes but suppressed this isoform in endothelial cells. It also induced early angiogenin and prolonged anti-inflammatory TGFβ expression on endothelial cells, while suppressing early pro-inflammatory IL-1β expression levels. IDR-1018 also down-regulated the hypoxia induced transcription factor HIF-1α in both keratinocytes and endothelial cells. Consistent with these data, in an in vitro wound healing scratch assay, IDR-1018 induced migration of endothelial cells under conditions of hypoxia while in epithelial cells migration increased only under conditions of normoxia.
Collapse
Affiliation(s)
- Paulina Marin-Luevano
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico; Department of Immunology, School of Medicine, Universidad Autonoma de San Luis Potosi, Mexico
| | - Valentin Trujillo
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico; Department of Immunology, School of Medicine, Universidad Autonoma de San Luis Potosi, Mexico
| | - Adrian Rodriguez-Carlos
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | | | - Jose A Enciso-Moreno
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, 2259 Lower Mall Research Station, Vancouver, BC, Canada
| | - Bruno Rivas-Santiago
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico.
| |
Collapse
|
28
|
Racanelli AC, Kikkers SA, Choi AM, Cloonan SM. Autophagy and inflammation in chronic respiratory disease. Autophagy 2018; 14:221-232. [PMID: 29130366 PMCID: PMC5902194 DOI: 10.1080/15548627.2017.1389823] [Citation(s) in RCA: 345] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022] Open
Abstract
Persistent inflammation within the respiratory tract underlies the pathogenesis of numerous chronic pulmonary diseases including chronic obstructive pulmonary disease, asthma and pulmonary fibrosis. Chronic inflammation in the lung may arise from a combination of genetic susceptibility and environmental influences, including exposure to microbes, particles from the atmosphere, irritants, pollutants, allergens, and toxic molecules. To this end, an immediate, strong, and highly regulated inflammatory defense mechanism is needed for the successful maintenance of homeostasis within the respiratory system. Macroautophagy/autophagy plays an essential role in the inflammatory response of the lung to infection and stress. At baseline, autophagy may be critical for inhibiting spontaneous pulmonary inflammation and fundamental for the response of pulmonary leukocytes to infection; however, when not regulated, persistent or inefficient autophagy may be detrimental to lung epithelial cells, promoting lung injury. This perspective will discuss the role of autophagy in driving and regulating inflammatory responses of the lung in chronic lung diseases with a focus on potential avenues for therapeutic targeting. Abbreviations AR allergic rhinitis AM alveolar macrophage ATG autophagy-related CF cystic fibrosis CFTR cystic fibrosis transmembrane conductance regulator COPD chronic obstructive pulmonary disease CS cigarette smoke CSE cigarette smoke extract DC dendritic cell IH intermittent hypoxia IPF idiopathic pulmonary fibrosis ILD interstitial lung disease MAP1LC3B microtubule associated protein 1 light chain 3 beta MTB Mycobacterium tuberculosis MTOR mechanistic target of rapamycin kinase NET neutrophil extracellular traps OSA obstructive sleep apnea PAH pulmonary arterial hypertension PH pulmonary hypertension ROS reactive oxygen species TGFB1 transforming growth factor beta 1 TNF tumor necrosis factor.
Collapse
Affiliation(s)
- Alexandra C. Racanelli
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- New York-Presbyterian Hospital, New York, NY, USA
| | - Sarah Ann Kikkers
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- New York-Presbyterian Hospital, New York, NY, USA
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
29
|
Tazi MF, Dakhlallah DA, Caution K, Gerber MM, Chang SW, Khalil H, Kopp BT, Ahmed AE, Krause K, Davis I, Marsh C, Lovett-Racke AE, Schlesinger LS, Cormet-Boyaka E, Amer AO. Elevated Mirc1/Mir17-92 cluster expression negatively regulates autophagy and CFTR (cystic fibrosis transmembrane conductance regulator) function in CF macrophages. Autophagy 2017; 12:2026-2037. [PMID: 27541364 DOI: 10.1080/15548627.2016.1217370] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cystic fibrosis (CF) is a fatal, genetic disorder that critically affects the lungs and is directly caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR function. Macroautophagy/autophagy is a highly regulated biological process that provides energy during periods of stress and starvation. Autophagy clears pathogens and dysfunctional protein aggregates within macrophages. However, this process is impaired in CF patients and CF mice, as their macrophages exhibit limited autophagy activity. The study of microRNAs (Mirs), and other noncoding RNAs, continues to offer new therapeutic targets. The objective of this study was to elucidate the role of Mirs in dysregulated autophagy-related genes in CF macrophages, and then target them to restore this host-defense function and improve CFTR channel function. We identified the Mirc1/Mir17-92 cluster as a potential negative regulator of autophagy as CF macrophages exhibit decreased autophagy protein expression and increased cluster expression when compared to wild-type (WT) counterparts. The absence or reduced expression of the cluster increases autophagy protein expression, suggesting the canonical inverse relationship between Mirc1/Mir17-92 and autophagy gene expression. An in silico study for targets of Mirs that comprise the cluster suggested that the majority of the Mirs target autophagy mRNAs. Those targets were validated by luciferase assays. Notably, the ability of macrophages expressing mutant F508del CFTR to transport halide through their membranes is compromised and can be restored by downregulation of these inherently elevated Mirs, via restoration of autophagy. In vivo, downregulation of Mir17 and Mir20a partially restored autophagy expression and hence improved the clearance of Burkholderia cenocepacia. Thus, these data advance our understanding of mechanisms underlying the pathobiology of CF and provide a new therapeutic platform for restoring CFTR function and autophagy in patients with CF.
Collapse
Affiliation(s)
- Mia F Tazi
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Duaa A Dakhlallah
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Kyle Caution
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Madelyn M Gerber
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Sheng-Wei Chang
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA.,c Department of Veterinary Biosciences , The Ohio State University , Columbus , OH , USA
| | - Hany Khalil
- d Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute , University of Sadat City , Egypt
| | | | - Amr E Ahmed
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Kathrin Krause
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Ian Davis
- c Department of Veterinary Biosciences , The Ohio State University , Columbus , OH , USA
| | - Clay Marsh
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Amy E Lovett-Racke
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA
| | - Larry S Schlesinger
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Estelle Cormet-Boyaka
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA.,c Department of Veterinary Biosciences , The Ohio State University , Columbus , OH , USA
| | - Amal O Amer
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| |
Collapse
|
30
|
Strategies for the etiological therapy of cystic fibrosis. Cell Death Differ 2017; 24:1825-1844. [PMID: 28937684 PMCID: PMC5635223 DOI: 10.1038/cdd.2017.126] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022] Open
Abstract
Etiological therapies aim at repairing the underlying cause of cystic fibrosis (CF), which is the functional defect of the cystic fibrosis transmembrane conductance regulator (CFTR) protein owing to mutations in the CFTR gene. Among these, the F508del CFTR mutation accounts for more than two thirds of CF cases worldwide. Two somehow antinomic schools of thought conceive CFTR repair in a different manner. According to one vision, drugs should directly target the mutated CFTR protein to increase its plasma membrane expression (correctors) or improve its ion transport function (potentiators). An alternative strategy consists in modulating the cellular environment and proteostasis networks in which the mutated CFTR protein is synthesized, traffics to its final destination, the plasma membrane, and is turned over. We will analyze distinctive advantages and drawbacks of these strategies in terms of their scientific and clinical dimensions, and we will propose a global strategy for CF research and development based on a reconciliatory approach. Moreover, we will discuss the utility of preclinical biomarkers that may guide the personalized, patient-specific implementation of CF therapies.
Collapse
|
31
|
Brockman SM, Bodas M, Silverberg D, Sharma A, Vij N. Dendrimer-based selective autophagy-induction rescues ΔF508-CFTR and inhibits Pseudomonas aeruginosa infection in cystic fibrosis. PLoS One 2017; 12:e0184793. [PMID: 28902888 PMCID: PMC5597233 DOI: 10.1371/journal.pone.0184793] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Background Cystic Fibrosis (CF) is a genetic disorder caused by mutation(s) in the CF-transmembrane conductance regulator (Cftr) gene. The most common mutation, ΔF508, leads to accumulation of defective-CFTR protein in aggresome-bodies. Additionally, Pseudomonas aeruginosa (Pa), a common CF pathogen, exacerbates obstructive CF lung pathology. In the present study, we aimed to develop and test a novel strategy to improve the bioavailability and potentially achieve targeted drug delivery of cysteamine, a potent autophagy-inducing drug with anti-bacterial properties, by developing a dendrimer (PAMAM-DEN)-based cysteamine analogue. Results We first evaluated the effect of dendrimer-based cysteamine analogue (PAMAM-DENCYS) on the intrinsic autophagy response in IB3-1 cells and observed a significant reduction in Ub-RFP and LC3-GFP co-localization (aggresome-bodies) by PAMAM-DENCYS treatment as compared to plain dendrimer (PAMAM-DEN) control. Next, we observed that PAMAM-DENCYS treatment shows a modest rescue of ΔF508-CFTR as the C-form. Moreover, immunofluorescence microscopy of HEK-293 cells transfected with ΔF508-CFTR-GFP showed that PAMAM-DENCYS is able to rescue the misfolded-ΔF508-CFTR from aggresome-bodies by inducing its trafficking to the plasma membrane. We further verified these results by flow cytometry and observed significant (p<0.05; PAMAM-DEN vs. PAMAM-DENCYS) rescue of membrane-ΔF508-CFTR with PAMAM-DENCYS treatment using non-permeabilized IB3-1 cells immunostained for CFTR. Finally, we assessed the autophagy-mediated bacterial clearance potential of PAMAM-DENCYS by treating IB3-1 cells infected with PA01-GFP, and observed a significant (p<0.01; PAMAM-DEN vs. PAMAM-DENCYS) decrease in intracellular bacterial counts by immunofluorescence microscopy and flow cytometry. Also, PAMAM-DENCYS treatment significantly inhibits the growth of PA01-GFP bacteria and demonstrates potent mucolytic properties. Conclusions We demonstrate here the efficacy of dendrimer-based autophagy-induction in preventing sequestration of ΔF508-CFTR to aggresome-bodies while promoting its trafficking to the plasma membrane. Moreover, PAMAM-DENCYS decreases Pa infection and growth, while showing mucolytic properties, suggesting its potential in rescuing Pa-induced ΔF508-CF lung disease that warrants further investigation in CF murine model.
Collapse
Affiliation(s)
- Scott Mackenzie Brockman
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Manish Bodas
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - David Silverberg
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Ajit Sharma
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan, United States of America
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mount Pleasant, Michigan, United States of America
- Department of Pediatric Respiratory Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
32
|
de Araújo AA, Pereira ADSBF, de Medeiros CACX, Brito GADC, Leitão RFDC, Araújo LDS, Guedes PMM, Hiyari S, Pirih FQ, de Araújo Júnior RF. Effects of metformin on inflammation, oxidative stress, and bone loss in a rat model of periodontitis. PLoS One 2017; 12:e0183506. [PMID: 28847008 PMCID: PMC5573680 DOI: 10.1371/journal.pone.0183506] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/04/2017] [Indexed: 11/18/2022] Open
Abstract
Aim To evaluate the effects of metformin (Met) on inflammation, oxidative stress, and bone loss in a rat model of ligature-induced periodontitis. Materials & methods Male albino Wistar rats were divided randomly into five groups of twenty-one rats each, and given the following treatments for 10 days: (1) no ligature + water, (2) ligature + water, (3) ligature + 50 mg/kg Met, (4) ligature + 100 mg/kg Met, and (5) ligature + 200 mg/kg Met. Water or Met was administered orally. Maxillae were fixed and scanned using Micro-computed Tomography (μCT) to quantitate linear and bone volume/tissue volume (BV/TV) volumetric bone loss. Histopathological characteristics were assessed through immunohistochemical staining for MMP-9, COX-2, the RANKL/RANK/OPG pathway, SOD-1, and GPx-1. Additionally, confocal microscopy was used to analyze osteocalcin fluorescence. UV-VIS analysis was used to examine the levels of malondialdehyde, glutathione, IL-1β and TNF-α from gingival tissues. Quantitative RT-PCR reaction was used to gene expression of AMPK, NF-κB (p65), and Hmgb1 from gingival tissues. Significance among groups were analysed using a one-way ANOVA. A p-value of p<0.05 indicated a significant difference. Results Treatment with 50 mg/kg Met significantly reduced concentrations of malondialdehyde, IL-1β, and TNF-α (p < 0.05). Additionally, weak staining was observed for COX-2, MMP-9, RANK, RANKL, SOD-1, and GPx-1 after 50 mg/kg Met. OPG and Osteocalcin showed strong staining in the same group. Radiographically, linear measurements showed a statistically significant reduction in bone loss after 50 mg/kg Met compared to the ligature and Met 200 mg/kg groups. The same pattern was observed volumetrically in BV/TV and decreased osteoclast number (p<0.05). RT-PCR showed increased AMPK expression and decreased expression of NF-κB (p65) and HMGB1 after 50 mg/kg Met. Conclusions Metformin, at a concentration of 50 mg/kg, decreases the inflammatory response, oxidative stress and bone loss in ligature-induced periodontitis in rats.
Collapse
Affiliation(s)
- Aurigena Antunes de Araújo
- Department of Biophysics and Pharmacology, Post Graduation Program Public Health / Post Graduation Program in Pharmaceutical Science, UFRN, Natal, RN, Brazil
- * E-mail:
| | | | | | | | | | | | - Paulo Marcos Matta Guedes
- Department of Microbiology and Parasitology, Post Graduation Program in Parasitary Biology/Post Guaduation Biological Science, UFRN, Natal, RN, Brazil
| | - Sarah Hiyari
- Section of Periodontics, School of Dentistry, University of California, UCLA, Los Angeles, California, United States of America
| | - Flávia Q. Pirih
- Section of Periodontics, School of Dentistry, University of California, UCLA, Los Angeles, California, United States of America
| | - Raimundo Fernandes de Araújo Júnior
- Department of Morphology, Post Graduation Program in Functional and Structural Biology/ Post Graduation Program Health Science/Department of Morphology, UFRN, Natal, RN, Brazil
| |
Collapse
|
33
|
Silymarin attenuates cigarette smoke extract-induced inflammation via simultaneous inhibition of autophagy and ERK/p38 MAPK pathway in human bronchial epithelial cells. Sci Rep 2016; 6:37751. [PMID: 27874084 PMCID: PMC5118787 DOI: 10.1038/srep37751] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 10/28/2016] [Indexed: 02/05/2023] Open
Abstract
Cigarette smoke (CS) is a major risk of chronic obstructive pulmonary disease (COPD), contributing to airway inflammation. Our previous study revealed that silymarin had an anti-inflammatory effect in CS-exposed mice. In this study, we attempt to further elucidate the molecular mechanisms of silymarin in CS extract (CSE)-induced inflammation using human bronchial epithelial cells. Silymarin significantly suppressed autophagy activation and the activity of ERK/p38 mitogen-activated protein kinase (MAPK) pathway in Beas-2B cells. We also observed that inhibiting the activity of ERK with specific inhibitor U0126 led to reduced autophagic level, while knockdown of autophagic gene Beclin-1 and Atg5 decreased the levels of ERK and p38 phosphorylation. Moreover, silymarin attenuated CSE-induced upregulation of inflammatory cytokines TNF-α, IL-6 and IL-8 which could also be dampened by ERK/p38 MAPK inhibitors and siRNAs for Beclin-1 and Atg5. Finally, we validated decreased levels of both autophagy and inflammatory cytokines (TNF-α and KC) in CS-exposed mice after silymarin treatment. The present research has demonstrated that CSE-induced autophagy in bronchial epithelia, in synergism with ERK MAPK pathway, may initiate and exaggerate airway inflammation. Silymarin could attenuate inflammatory responses through intervening in the crosstalk between autophagy and ERK MAPK pathway, and might be an ideal agent treating inflammatory pulmonary diseases.
Collapse
|
34
|
Stick SM, Kicic A, Ranganathan S. Of Pigs, Mice, and Men: Understanding Early Triggers of Cystic Fibrosis Lung Disease. Am J Respir Crit Care Med 2016; 194:784-785. [PMID: 27689703 DOI: 10.1164/rccm.201605-1094ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Stephen M Stick
- 1 Telethon Kids Institute Perth, Australia.,2 School of Paediatrics and Child Health University of Western Australia Perth, Australia.,3 Department of Respiratory and Sleep Medicine Princess Margaret Hospital for Children Perth, Australia
| | - Anthony Kicic
- 1 Telethon Kids Institute Perth, Australia.,2 School of Paediatrics and Child Health University of Western Australia Perth, Australia
| | - Sarath Ranganathan
- 4 Department of Respiratory and Sleep Medicine Royal Children's Hospital Melbourne, Australia.,5 Department of Paediatrics University of Melbourne Melbourne, Australia and.,6 Infection and Immunology Murdoch Children's Research Institute Melbourne, Australia
| |
Collapse
|
35
|
c- Src and its role in cystic fibrosis. Eur J Cell Biol 2016; 95:401-413. [DOI: 10.1016/j.ejcb.2016.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 12/15/2022] Open
|
36
|
Huang C, Jiang D, Francisco D, Berman R, Wu Q, Ledford JG, Moore CM, Ito Y, Stevenson C, Munson D, Li L, Kraft M, Chu HW. Tollip SNP rs5743899 modulates human airway epithelial responses to rhinovirus infection. Clin Exp Allergy 2016; 46:1549-1563. [PMID: 27513438 DOI: 10.1111/cea.12793] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 07/15/2016] [Accepted: 07/19/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Rhinovirus (RV) infection in asthma induces varying degrees of airway inflammation (e.g. neutrophils), but the underlying mechanisms remain unclear. OBJECTIVE The major goal was to determine the role of genetic variation [e.g. single nucleotide polymorphisms (SNPs)] of Toll-interacting protein (Tollip) in airway epithelial responses to RV in a type 2 cytokine milieu. METHODS DNA from blood of asthmatic and normal subjects was genotyped for Tollip SNP rs5743899 AA, AG and GG genotypes. Human tracheobronchial epithelial (HTBE) cells from donors without lung disease were cultured to determine pro-inflammatory and antiviral responses to IL-13 and RV16. Tollip knockout and wild-type mice were challenged with house dust mite (HDM) and infected with RV1B to determine lung inflammation and antiviral response. RESULTS Asthmatic subjects carrying the AG or GG genotype (AG/GG) compared with the AA genotype demonstrated greater airflow limitation. HTBE cells with AG/GG expressed less Tollip. Upon IL-13 and RV16 treatment, cells with AG/GG (vs. AA) produced more IL-8 and expressed less antiviral genes, which was coupled with increased NF-κB activity and decreased expression of LC3, a hallmark of the autophagic pathway. Tollip co-localized and interacted with LC3. Inhibition of autophagy decreased antiviral genes in IL-13- and RV16-treated cells. Upon HDM and RV1B, Tollip knockout (vs. wild-type) mice demonstrated higher levels of lung neutrophilic inflammation and viral load, but lower levels of antiviral gene expression. CONCLUSIONS AND CLINICAL RELEVANCE Our data suggest that Tollip SNP rs5743899 may predict varying airway response to RV infection in asthma.
Collapse
Affiliation(s)
- C Huang
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - D Jiang
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - D Francisco
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| | - R Berman
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Q Wu
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - J G Ledford
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| | - C M Moore
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Y Ito
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - C Stevenson
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - D Munson
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - L Li
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - M Kraft
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| | - H W Chu
- Department of Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|
37
|
Considerations for the Conduct of Clinical Trials with Antiinflammatory Agents in Cystic Fibrosis. A Cystic Fibrosis Foundation Workshop Report. Ann Am Thorac Soc 2016; 12:1398-406. [PMID: 26146892 DOI: 10.1513/annalsats.201506-361ot] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inflammation leads to lung destruction and loss of pulmonary function in patients with cystic fibrosis (CF). Drugs that modulate the cystic fibrosis transmembrane conductance regulator (CFTR) have recently been approved. Although the impact of CFTR modulators on sweat chloride and lung function are exciting, they have not yet demonstrated an effect on inflammation. Therefore, CF antiinflammatory drug development must continue. Unfortunately, the lack of clarity with this process has left investigators and industry sponsors frustrated. The Cystic Fibrosis Foundation established a working group in early 2014 to address this issue. There are many inflammatory processes disrupted in CF, and, therefore, there are many potential targets amenable to antiinflammatory therapy. Regardless of a drug's specific mechanism of action, it must ultimately affect the neutrophil or its products to impact CF. The working group concluded that before bringing new antiinflammatory drugs to clinical trial, preclinical safety studies must be conducted in disease-relevant models to assuage safety concerns. Furthermore, although studies of antiinflammatory therapies must first establish safety in adults, subsequent studies must involve children, as they are most likely to reap the most benefit. The working group also recommended that pharmacokinetic-pharmacodynamic studies and early-phase safety studies be performed before proceeding to larger studies of longer duration. In addition, innovative study designs may improve the likelihood of adequately assessing treatment response and mitigating risk before conducting multiyear studies. Learning from past experiences and incorporating this knowledge into new drug development programs will be instrumental in bringing new antiinflammatory therapies to patients.
Collapse
|
38
|
|
39
|
Zeki AA, Yeganeh B, Kenyon NJ, Post M, Ghavami S. Autophagy in airway diseases: a new frontier in human asthma? Allergy 2016; 71:5-14. [PMID: 26335713 DOI: 10.1111/all.12761] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2015] [Indexed: 12/11/2022]
Abstract
The study of autophagy ('self-eating'), a fundamental cell fate pathway involved in physiological and pathological subcellular processes, opens a new frontier in the continuous search for novel therapies for human asthma. Asthma is a complex syndrome with different disease phenotypes. Autophagy plays a central role in cell physiology, energy and metabolism, and cell survival. Autophagy's hallmark is the formation of double-membrane autophagic autophagosomes, and this process is operational in airway epithelial and mesenchymal cells in asthma. Genetic associations between autophagy genes and asthma have been observed including single nucleotide polymorphisms in Atg5 which correlate with reduced lung function. Immune mechanisms important in asthma such as Th2 cells and eosinophils also manifest autophagy. Lastly, we address the role of autophagy in extracellular matrix deposition and fibrosis in asthmatic airways remodeling, a pathologic process still without effective therapy, and discuss potential pharmacologic inhibitors. We end by offering two opposing but plausible hypotheses as to how autophagy may be directly involved in airway fibrosis.
Collapse
Affiliation(s)
- A. A. Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine; Center for Comparative Respiratory Biology and Medicine; Davis CA USA
| | - B. Yeganeh
- Physiology and Experimental Medicine; Hospital for Sick Children (Sickkids); University of Toronto; Toronto ON Canada
| | - N. J. Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine; Center for Comparative Respiratory Biology and Medicine; Davis CA USA
| | - M. Post
- Physiology and Experimental Medicine; Hospital for Sick Children (Sickkids); University of Toronto; Toronto ON Canada
| | - S. Ghavami
- Department of Human Anatomy & Cell Science; Faculty of Health Sciences; College of Medicine; University of Manitoba; Winnipeg MB Canada
| |
Collapse
|
40
|
Abstract
BACKGROUND Mutation of cystic fibrosis transmembrane conductance regulator (CFTR) in the airway epithelial cells can lead to recurrent airway inflammation in cystic fibrosis (CF). Dysfunction of CFTR in neutrophils could contribute to LPS-induced acute lung inflammation. Deficiency of CFTR could also facilitate platelet aggregation and neutrophil-platelet interaction and promote inflammation. AIM To study whether inhibition or mutation of CFTR in alveolar macrophages (AMs) or peritoneal macrophages (PMs) would promote their proinflammatory responses and whether dysfunction of CFTR would deteriorate acute E. coli-induced lung or peritoneal inflammation. DESIGN Laboratory study. METHODS ELISA was used to determine production of proinflammatory cytokines in the CFTR inhibited or mutated macrophages under LPS challenge. Lung or peritoneum lavage was used to analyze proinflammatory parameters and cell differentiation. Excess lung water and lung vascular permeability were measured for evaluating severity of acute lung inflammation. RESULTS Escherichia coli LPS simulation in AMs increased CFTR expression. Inhibition or mutation of CFTR in both AMs and PMs enhanced production of tumor necrosis factor alpha (TNF-α) and macrophage inflammatory protein-2 (MIP-2). Mutation of CFTR in macrophages exaggerated production of cytokines through NF-kB and p38 MAPK. Inhibition of CFTR by MalH2 or CFTRinh-172 deteriorates E. coli-induced acute lung inflammation. Deficiency of CFTR promotes migration of monocytes and neutrophils in E. coli pneumonia and peritonitis mouse models. CONCLUSIONS CFTR expressed by alveolar or peritoneal macrophages regulates acute proinflammatory responses.
Collapse
Affiliation(s)
- Z Gao
- From the Institute of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - X Su
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China and Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0130, USA
| |
Collapse
|
41
|
Abstract
BACKGROUND Innate defense regulator peptide-1018 (IDR-1018) is a 12-amino acid, synthetic, immunomodulatory host defense peptide that can reduce soft tissue infections and is less likely to induce bacterial resistance than conventional antibiotics. However, IDRs have not been tested on orthopaedic infections and the immunomodulatory effects of IDR-1018 have only been characterized in response to lipopolysacharide, which is exclusively produced by Gram-negative bacteria. QUESTIONS/PURPOSES We sought (1) to more fully characterize the immunomodulatory effects of IDR-1018, especially in response to Staphylococcus aureus; and (2) to determine whether IDR-1018 decreases S aureus infection of orthopaedic implants in mice and thereby protects the implants from failure to osseointegrate. METHODS In vitro effects of IDR-1018 on S aureus were assessed by determining minimum inhibitory concentrations in bacterial broth without and with supplementation of physiologic ion levels. In vitro effects of IDR-1018 on macrophages were determined by measuring production of monocyte chemoattractant protein-1 (MCP-1) and proinflammatory cytokines by enzyme-linked immunosorbent assay. In vivo effects of IDR-1018 were determined in a murine model of S aureus implant infection by quantitating bacterial burden, macrophage recruitment, MCP-1, proinflammatory cytokines, and osseointegration in nine mice per group on Day 1 postimplantation and 20 mice per group on Day 15 postimplantation. RESULTS IDR-1018 demonstrated antimicrobial activity by directly killing S aureus even in the presence of physiologic ion levels, increasing recruitment of macrophages to the site of infections by 40% (p = 0.036) and accelerating S aureus clearance in vivo (p = 0.008) with a 2.6-fold decrease in bacterial bioburden on Day 7 postimplantation. In vitro immunomodulatory activity of IDR-1018 included inducing production of MCP-1 in the absence of other inflammatory stimuli and to potently blunt excess production of proinflammatory cytokines and MCP-1 induced by lipopolysaccharide. Higher concentrations of IDR-1018 were required to blunt production of proinflammatory cytokines and MCP-1 in the presence S aureus. The largest in vivo immunomodulatory effect of IDR-1018 was to reduce tumor necrosis factor-α levels induced by S aureus by 60% (p = 0.006). Most importantly, IDR-1018 reduced S aureus-induced failures of osseointegration by threefold (p = 0.022) and increased osseointegration as measured by ultimate force (5.4-fold, p = 0.033) and average stiffness (4.3-fold, p = 0.049). CONCLUSIONS IDR-1018 is potentially useful to reduce orthopaedic infections by directly killing bacteria and by recruiting macrophages to the infection site. CLINICAL RELEVANCE These findings make IDR-1018 an attractive candidate to explore in larger animal models to ascertain whether its effects in our in vitro and mouse experiments can be replicated in more clinically relevant settings.
Collapse
|
42
|
Kanayama M, He YW, Shinohara ML. The lung is protected from spontaneous inflammation by autophagy in myeloid cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:5465-71. [PMID: 25911758 DOI: 10.4049/jimmunol.1403249] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/24/2015] [Indexed: 11/19/2022]
Abstract
The lung is constantly exposed to the outer environment; thus, it must maintain a state of immune ignorance or tolerance not to overrespond to harmless environmental stimuli. How cells in the lung control immune responses under nonpathogenic condition is not fully understood. In this study, we found that autophagy plays a critical role in the lung-specific immune regulation that prevents spontaneous inflammation. Autophagy in pulmonary myeloid cells plays a role in maintaining low burdens of environmental microbes in the lung, as well as in lowering mitochondrial reactive oxygen species production and preventing overresponse to TLR4 ligands in alveolar macrophages. Based on these mechanisms, we also found that intranasal instillation of antibiotics or an inhibitor of reactive oxygen species was efficient in preventing spontaneous pulmonary inflammation. Thus, autophagy in myeloid cells, particularly alveolar macrophages, is critical for inhibiting spontaneous pulmonary inflammation, and pulmonary inflammation caused by dysfunctional autophagy is pharmacologically prevented.
Collapse
Affiliation(s)
- Masashi Kanayama
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical Center, Durham, NC 27710; and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
43
|
Schultz A, Stick S. Early pulmonary inflammation and lung damage in children with cystic fibrosis. Respirology 2015; 20:569-78. [PMID: 25823858 DOI: 10.1111/resp.12521] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/11/2014] [Accepted: 02/17/2015] [Indexed: 12/21/2022]
Abstract
Individuals with cystic fibrosis (CF) suffer progressive airway inflammation, infection and lung damage. Airway inflammation and infection are present from early in life, often before children are symptomatic. CF gene mutations cause changes in the CF transmembrane regulator protein that result in an aberrant airway microenvironment including airway surface liquid (ASL) dehydration, reduced ASL acidity, altered airway mucin and a dysregulated inflammatory response. This review discusses how an altered microenvironment drives CF lung disease before overt airway infection, the response of the CF airway to early infection, and methods to prevent inflammation and early lung disease.
Collapse
Affiliation(s)
- André Schultz
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; School of Paediatric and Child Health, University of Western Australia, Perth, Western Australia, Australia; Telethon Kids Institute, Perth, Western Australia, Australia
| | | |
Collapse
|
44
|
Hyaluronic Acid-Based Nanogels Produced by Microfluidics-Facilitated Self-Assembly Improves the Safety Profile of the Cationic Host Defense Peptide Novicidin. Pharm Res 2015; 32:2727-35. [PMID: 25813840 DOI: 10.1007/s11095-015-1658-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE Cationic host defence peptides constitute a promising class of therapeutic drug leads with a wide range of therapeutic applications, including anticancer therapy, immunomodulation, and antimicrobial activity. Although potent and efficacious, systemic toxicity and low chemical stability have hampered their commercial development. To overcome these challenges a novel nanogel-based drug delivery system was designed. METHOD The peptide novicidin was self-assembled with an octenyl succinic anhydride-modified analogue of hyaluronic acid, and this formulation was optimized using a microfluidics-based quality-by-design approach. RESULTS By applying design-of-experiment it was demonstrated that the encapsulation efficiency of novicidin (15% to 71%) and the zeta potential (-24 to -57 mV) of the nanogels could be tailored by changing the preparation process parameters, with a maximum peptide loading of 36 ± 4%. The nanogels exhibited good colloidal stability under different ionic strength conditions and allowed complete release of the peptide over 14 days. Furthermore, self-assembly of novicidin with hyaluronic acid into nanogels significantly improved the safety profile at least five-fold and six-fold when tested in HUVECs and NIH 3T3 cells, respectively, whilst showing no loss of antimicrobial activity against Escherichia coli and Staphylococcus aureus. CONCLUSION Formulation in nanogels could be a viable approach to improve the safety profile of host defence peptides.
Collapse
|
45
|
Baker B, Geng S, Chen K, Diao N, Yuan R, Xu X, Dougherty S, Stephenson C, Xiong H, Chu HW, Li L. Alteration of lysosome fusion and low-grade inflammation mediated by super-low-dose endotoxin. J Biol Chem 2015; 290:6670-8. [PMID: 25586187 PMCID: PMC4358298 DOI: 10.1074/jbc.m114.611442] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/02/2015] [Indexed: 11/06/2022] Open
Abstract
Subclinical super-low-dose endotoxin LPS is a risk factor for the establishment of low-grade inflammation during the pathogenesis and progression of chronic diseases. However, the underlying mechanisms are not well understood. At the cellular level, a disruption of lysosome fusion with endosomes or autophagosomes may contribute to the potentiation of low-grade inflammation. In this study, we identified that subclinical super-low-dose endotoxin LPS can potently inhibit the process of endosome acidification and lysosome fusion with endosomes or autophagosomes in primary macrophages. Super-low-dose LPS induced the inhibitory phosphorylation of VPS34, thus leading to the disruption of endosome-lysosome fusion. This effect may depend upon the clearance and relocation of Tollip in macrophages by super-low-dose LPS. Consistent with this notion, Tollip-deficient macrophages had constitutively elevated levels of VPS34 inhibitory phosphorylation and constitutive disruption of endosome-lysosome fusion. By employing a skin excision wound-healing model, we observed that Tollip-deficient mice had significantly elevated levels of cell stress and reduced wound repair. This study reveals a novel mechanism responsible for the modulation of endosome-lysosome fusion and low-grade inflammation in innate macrophages.
Collapse
Affiliation(s)
- Bianca Baker
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Shuo Geng
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Keqiang Chen
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Na Diao
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Ruoxi Yuan
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Xiguang Xu
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Sean Dougherty
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Caroline Stephenson
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910
| | - Huabao Xiong
- the Department of Medicine, Mt. Sinai School of Medicine, New York, New York 10029, and
| | - Hong Wei Chu
- the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Liwu Li
- From the Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061-0910,
| |
Collapse
|
46
|
Ryter SW, Choi AMK. Autophagy in lung disease pathogenesis and therapeutics. Redox Biol 2015; 4:215-25. [PMID: 25617802 PMCID: PMC4803789 DOI: 10.1016/j.redox.2014.12.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 12/18/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022] Open
Abstract
Autophagy, a cellular pathway for the degradation of damaged organelles and proteins, has gained increasing importance in human pulmonary diseases, both as a modulator of pathogenesis and as a potential therapeutic target. In this pathway, cytosolic cargos are sequestered into autophagosomes, which are delivered to the lysosomes where they are enzymatically degraded and then recycled as metabolic precursors. Autophagy exerts an important effector function in the regulation of inflammation, and immune system functions. Selective pathways for autophagic degradation of cargoes may have variable significance in disease pathogenesis. Among these, the autophagic clearance of bacteria (xenophagy) may represent a crucial host defense mechanism in the pathogenesis of sepsis and inflammatory diseases. Our recent studies indicate that the autophagic clearance of mitochondria, a potentially protective program, may aggravate the pathogenesis of chronic obstructive pulmonary disease by activating cell death programs. We report similar findings with respect to the autophagic clearance of cilia components, which can contribute to airways dysfunction in chronic lung disease. In certain diseases such as pulmonary hypertension, autophagy may confer protection by modulating proliferation and cell death. In other disorders, such as idiopathic pulmonary fibrosis and cystic fibrosis, impaired autophagy may contribute to pathogenesis. In lung cancer, autophagy has multiple consequences by limiting carcinogenesis, modulating therapeutic effectiveness, and promoting tumor cell survival. In this review we highlight the multiple functions of autophagy and its selective autophagy subtypes that may be of significance to the pathogenesis of human disease, with an emphasis on lung disease and therapeutics. Autophagy may impact the pathogenesis of pulmonary diseases. Mitophagy may exert deleterious effects in the pathogenesis of COPD. Autophagy can exert pleiotropic effects in lung cancer. Targeting autophagy may represent a promising therapeutic strategy in human diseases.
Collapse
Affiliation(s)
- Stefan W Ryter
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA.
| | - Augustine M K Choi
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
47
|
Host defense peptides: front-line immunomodulators. Trends Immunol 2014; 35:443-50. [DOI: 10.1016/j.it.2014.07.004] [Citation(s) in RCA: 349] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 12/30/2022]
|
48
|
Abstract
The term "Translational Genomics" reflects both title and mission of this new journal. "Translational" has traditionally been understood as "applied research" or "development", different from or even opposed to "basic research". Recent scientific and societal developments have triggered a re-assessment of the connotation that "translational" and "basic" are either/or activities: translational research nowadays aims at feeding the best science into applications and solutions for human society. We therefore argue here basic science to be challenged and leveraged for its relevance to human health and societal benefits. This more recent approach and attitude are catalyzed by four trends or developments: evidence-based solutions; large-scale, high dimensional data; consumer/patient empowerment; and systems-level understanding.
Collapse
Affiliation(s)
- Martin Kussmann
- Molecular Biomarkers Core, Nestlé Institute of Health Sciences (NIHS), Lausanne, Switzerland; Faculty of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland; Faculty of Science, Interdisciplinary NanoScience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Jim Kaput
- Systems Nutrition and Health Unit, Nestlé Institute of Health Sciences (NIHS), Lausanne, Switzerland; Service Endocrinol. Diabetes, Metabol. Univ. Hospital Lausanne (CHUV), Univ. Lausanne, Switzerland
| |
Collapse
|
49
|
Whiteson KL, Bailey B, Bergkessel M, Conrad D, Delhaes L, Felts B, Harris JK, Hunter R, Lim YW, Maughan H, Quinn R, Salamon P, Sullivan J, Wagner BD, Rainey PB. The upper respiratory tract as a microbial source for pulmonary infections in cystic fibrosis. Parallels from island biogeography. Am J Respir Crit Care Med 2014; 189:1309-15. [PMID: 24702670 DOI: 10.1164/rccm.201312-2129pp] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A continuously mixed series of microbial communities inhabits various points of the respiratory tract, with community composition determined by distance from colonization sources, colonization rates, and extinction rates. Ecology and evolution theory developed in the context of biogeography is relevant to clinical microbiology and could reframe the interpretation of recent studies comparing communities from lung explant samples, sputum samples, and oropharyngeal swabs. We propose an island biogeography model of the microbial communities inhabiting different niches in human airways. Island biogeography as applied to communities separated by time and space is a useful parallel for exploring microbial colonization of healthy and diseased lungs, with the potential to inform our understanding of microbial community dynamics and the relevance of microbes detected in different sample types. In this perspective, we focus on the intermixed microbial communities inhabiting different regions of the airways of patients with cystic fibrosis.
Collapse
|
50
|
Morgenthau A, Partha SK, Adamiak P, Schryvers AB. The specificity of protection against cationic antimicrobial peptides by lactoferrin binding protein B. Biometals 2014; 27:923-33. [PMID: 25038734 DOI: 10.1007/s10534-014-9767-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/22/2014] [Indexed: 10/25/2022]
Abstract
A variety of Gram-negative pathogens possess host-specific lactoferrin (Lf) receptors that mediate the acquisition of iron from host Lf. The integral membrane protein component of the receptor, lactoferrin binding protein A specifically binds host Lf and is required for acquisition of iron from Lf. In contrast, the role of the bi-lobed surface lipoprotein, lactoferrin binding protein B (LbpB), in Lf binding and iron acquisition is uncertain. A common feature of LbpBs from most species is the presence of clusters of negatively charged amino acids in the protein's C-terminal lobe. Recently it has been shown that the negatively charged regions from the Neisseria meningitidis LbpB are responsible for protecting against an 11 amino acid cationic antimicrobial peptide (CAP), lactoferricin (Lfcin), derived from human Lf. In this study we investigated whether the LbpB confers resistance to other CAPs since N. meningitidis is likely to encounter other CAPs from the host. LbpB provided protection against the cathelicidin derived peptide, cathelicidin related antimicrobial peptide (mCRAMP), but did not confer protection against Tritrp 1 or LL37 under our experimental conditions. When tested against a range of rationally designed synthetic peptides, LbpB was shown to protect against IDR-1002 and IDR-0018 but not against HH-2 or HHC10.
Collapse
Affiliation(s)
- Ari Morgenthau
- Departments of Microbiology, Immunology & Infectious Diseases and Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | | | | | | |
Collapse
|