1
|
Sharma R, Tiwari A, Kho AT, Wang AL, Srivastava U, Piparia S, Desai B, Wong R, Celedón JC, Peters SP, Smith LJ, Irvin CG, Castro M, Weiss ST, Tantisira KG, McGeachie MJ. Circulating microRNAs associated with bronchodilator response in childhood asthma. BMC Pulm Med 2024; 24:553. [PMID: 39497092 PMCID: PMC11536898 DOI: 10.1186/s12890-024-03372-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Bronchodilator response (BDR) is a measure of improvement in airway smooth muscle tone, inhibition of liquid accumulation and mucus section into the lumen in response to short-acting beta-2 agonists that varies among asthmatic patients. MicroRNAs (miRNAs) are well-known post-translational regulators. Identifying miRNAs associated with BDR could lead to a better understanding of the underlying complex pathophysiology. OBJECTIVE The purpose of this study is to identify circulating miRNAs associated with bronchodilator response in asthma and decipher possible mechanism of bronchodilator response variation. METHODS We used available small RNA sequencing on blood serum from 1,134 asthmatic children aged 6 to 14 years who participated in the Genetics of Asthma in Costa Rica Study (GACRS). We filtered the participants into the highest and lowest bronchodilator response (BDR) quartiles and used DeSeq2 to identify miRNAs with differential expression (DE) in high (N = 277) vs. low (N = 278) BDR group. Replication was carried out in the Leukotriene modifier Or Corticosteroids or Corticosteroid-Salmeterol trial (LOCCS), an adult asthma cohort. The putative target genes of DE miRNAs were identified, and pathway enrichment analysis was performed. RESULTS We identified 10 down-regulated miRNAs having odds ratios (OR) between 0.37 and 0.76 for a doubling of miRNA counts and one up-regulated miRNA (OR = 2.26) between high and low BDR group. These were assessed for replication in the LOCCS cohort, where two miRNAs (miR-200b-3p and miR-1246) were associated. Further, functional annotation of 11 DE miRNAs were performed as well as of two replicated miRs. Target genes of these miRs were enriched in regulation of cholesterol biosynthesis by SREBPs, ESR-mediated signaling, G1/S transition, RHO GTPase cycle, and signaling by TGFB family pathways. CONCLUSION MiRNAs miR-1246 and miR-200b-3p are associated with both childhood and adult asthma BDR. Our findings add to the growing body of evidence that miRNAs play a significant role in the difference of asthma treatment response among patients as it points to genomic regulatory machinery underlying difference in bronchodilator response among patients. TRIAL REGISTRATION LOCCS cohort [ClinicalTrials.gov number NCT00156819, Registration date 20050912], GACRS cohort [ClinicalTrials.gov number NCT00021840].
Collapse
Affiliation(s)
- Rinku Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alvin T Kho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Alberta L Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Upasna Srivastava
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
- Department of MEDCSC Neurodevelopment (Child Study Center), Yale University School of Medicine, New Haven, CT, USA
| | - Shraddha Piparia
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Brinda Desai
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Richard Wong
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen P Peters
- Department of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Lewis J Smith
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Charles G Irvin
- Pulmonary and Critical Care Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, KS, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kelan G Tantisira
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Parekh AB. House dust mite allergens, store-operated Ca 2+ channels and asthma. J Physiol 2024; 602:6021-6038. [PMID: 38054814 DOI: 10.1113/jp284931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
The house dust mite is the principal source of aero-allergen worldwide. Exposure to mite-derived allergens is associated with the development of asthma in susceptible individuals, and the majority of asthmatics are allergic to the mite. Mite-derived allergens are functionally diverse and activate multiple cell types within the lung that result in chronic inflammation. Allergens activate store-operated Ca2+ release-activated Ca2+ (CRAC) channels, which are widely expressed in multiple cell types within the lung that are associated with the pathogenesis of asthma. Opening of CRAC channels stimulates Ca2+-dependent transcription factors, including nuclear factor of activated T cells and nuclear factor-κB, which drive expression of a plethora of pro-inflammatory cytokines and chemokines that help to sustain chronic inflammation. Here, I describe drivers of asthma, properties of mite-derived allergens, how the allergens are recognized by cells, the signalling pathways used by the receptors and how these are transduced into functional effects, with a focus on CRAC channels. In vivo experiments that demonstrate the effectiveness of targeting CRAC channels as a potential new therapy for treating mite-induced asthma are also discussed, in tandem with other possible approaches.
Collapse
Affiliation(s)
- Anant B Parekh
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| |
Collapse
|
3
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
4
|
Dos Santos TM, Righetti RF, do Nascimento Camargo L, Leick EA, Fukuzaki S, de Campos EC, Galli TT, Saraiva-Romanholo BM, da Silva LLS, Barbosa JAS, João JMLG, Prado CM, de Rezende BG, Bourotte CLM, Dos Santos Lopes FDTQ, de Arruda Martins M, Bensenor IM, de Oliveira Cirillo JV, Bezerra SKM, Silva FJA, Paulo MSL, Lotufo PA, Lopes Calvo Tibério IDF. Effect of VAChT reduction on lung alterations induced by exposure to iron particles in an asthma model. J Inflamm (Lond) 2024; 21:24. [PMID: 38961398 PMCID: PMC11223391 DOI: 10.1186/s12950-024-00399-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
INTRODUCTION Pollution harms the health of people with asthma. The effect of the anti-inflammatory cholinergic pathway in chronic allergic inflammation associated to pollution is poorly understood. METHODS One hundred eight animals were divided into 18 groups (6 animals). Groups included: wild type mice (WT), genetically modified with reduced VAChT (VAChTKD), and those sensitized with ovalbumin (VAChTKDA), exposed to metal powder due to iron pelletizing in mining company (Local1) or 3.21 miles away from a mining company (Local2) in their locations for 2 weeks during summer and winter seasons. It was analyzed for hyperresponsivity, inflammation, remodeling, oxidative stress responses and the cholinergic system. RESULTS During summer, animals without changes in the cholinergic system revealed that Local1 exposure increased the hyperresponsiveness (%Rrs, %Raw), and inflammation (IL-17) relative to vivarium animals, while animals exposed to Local2 also exhibited elevated IL-17. During winter, animals without changes in the cholinergic system revealed that Local2 exposure increased the hyperresponsiveness (%Rrs) relative to vivarium animals. Comparing the exposure local of these animals during summer, animals exposed to Local1 showed elevated %Rrs, Raw, and IL-5 compared to Local 2, while in winter, Local2 exposure led to more IL-17 than Local1. Animals with VAChT attenuation displayed increased %Rrs, NFkappaB, IL-5, and IL-13 but reduced alpha-7 compared to animals without changes in the cholinergic system WT. Animals with VAChT attenuation and asthma showed increased the hyperresponsiveness, all inflammatory markers, remodeling and oxidative stress compared to animals without chronic lung inflammation. Exposure to Local1 exacerbated the hyperresponsiveness, oxidative stressand inflammation in animals with VAChT attenuation associated asthma, while Local2 exposure led to increased inflammation, remodeling and oxidative stress. CONCLUSIONS Reduced cholinergic signaling amplifies lung inflammation in a model of chronic allergic lung inflammation. Furthermore, when associated with pollution, it can aggravate specific responses related to inflammation, oxidative stress, and remodeling.
Collapse
Affiliation(s)
- Tabata Maruyama Dos Santos
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil.
- Hospital Sírio Libanês, São Paulo, SP, Brazil.
| | - Renato Fraga Righetti
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- Hospital Sírio Libanês, São Paulo, SP, Brazil
| | - Leandro do Nascimento Camargo
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- Hospital Sírio Libanês, São Paulo, SP, Brazil
| | | | - Silvia Fukuzaki
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
| | - Elaine Cristina de Campos
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- Hospital Sírio Libanês, São Paulo, SP, Brazil
| | | | | | | | | | | | - Carla Máximo Prado
- Department of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | | | | | | | | | - Isabela M Bensenor
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | | | | | | | | | - Paulo A Lotufo
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
5
|
Koziol-White C, Gebski E, Cao G, Panettieri RA. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res 2024; 25:231. [PMID: 38824592 PMCID: PMC11144351 DOI: 10.1186/s12931-024-02855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/03/2024] Open
Abstract
Precision Cut Lung Slices (PCLS) have emerged as a sophisticated and physiologically relevant ex vivo model for studying the intricacies of lung diseases, including fibrosis, injury, repair, and host defense mechanisms. This innovative methodology presents a unique opportunity to bridge the gap between traditional in vitro cell cultures and in vivo animal models, offering researchers a more accurate representation of the intricate microenvironment of the lung. PCLS require the precise sectioning of lung tissue to maintain its structural and functional integrity. These thin slices serve as invaluable tools for various research endeavors, particularly in the realm of airway diseases. By providing a controlled microenvironment, precision-cut lung slices empower researchers to dissect and comprehend the multifaceted interactions and responses within lung tissue, thereby advancing our understanding of pulmonary pathophysiology.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA.
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Gaoyaun Cao
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| |
Collapse
|
6
|
Putri KSS, Adhyatmika A, Boorsma CE, Habibie H, Ruigrok MJR, Heukels P, Timens W, de Jager MH, Hinrichs WLJ, Olinga P, Melgert BN. Osteoprotegerin is an Early Marker of the Fibrotic Process and of Antifibrotic Treatment Responses in Ex Vivo Lung Fibrosis. Lung 2024; 202:331-342. [PMID: 38642135 PMCID: PMC11143060 DOI: 10.1007/s00408-024-00691-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Lung fibrosis is a chronic lung disease with a high mortality rate with only two approved drugs (pirfenidone and nintedanib) to attenuate its progression. To date, there are no reliable biomarkers to assess fibrosis development and/or treatment effects for these two drugs. Osteoprotegerin (OPG) is used as a serum marker to diagnose liver fibrosis and we have previously shown it associates with lung fibrosis as well. METHODS Here we used murine and human precision-cut lung slices to investigate the regulation of OPG in lung tissue to elucidate whether it tracks with (early) fibrosis development and responds to antifibrotic treatment to assess its potential use as a biomarker. RESULTS OPG mRNA expression in murine lung slices was higher after treatment with profibrotic cytokines TGFβ1 or IL13, and closely correlated with Fn and PAI1 mRNA expression. More OPG protein was released from fibrotic human lung slices than from the control human slices and from TGFβ1 and IL13-stimulated murine lung slices compared to control murine slices. This OPG release was inhibited when murine slices were treated with pirfenidone or nintedanib. OPG release from human fibrotic lung slices was inhibited by pirfenidone treatment. CONCLUSION OPG can already be detected during the early stages of fibrosis development and responds, both in early- and late-stage fibrosis, to treatment with antifibrotic drugs currently on the market for lung fibrosis. Therefore, OPG should be further investigated as a potential biomarker for lung fibrosis and a potential surrogate marker for treatment effect.
Collapse
Affiliation(s)
- Kurnia S S Putri
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia
| | - Adhyatmika Adhyatmika
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- Drug Targeting and Personalized Medicine Research Group, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Carian E Boorsma
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Habibie Habibie
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Mitchel J R Ruigrok
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Peter Heukels
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
| | - Marina H de Jager
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Barbro N Melgert
- GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
7
|
Baglivo I, Quaranta VN, Dragonieri S, Colantuono S, Menzella F, Selvaggio D, Carpagnano GE, Caruso C. The New Paradigm: The Role of Proteins and Triggers in the Evolution of Allergic Asthma. Int J Mol Sci 2024; 25:5747. [PMID: 38891935 PMCID: PMC11171572 DOI: 10.3390/ijms25115747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Epithelial barrier damage plays a central role in the development and maintenance of allergic inflammation. Rises in the epithelial barrier permeability of airways alter tissue homeostasis and allow the penetration of allergens and other external agents. Different factors contribute to barrier impairment, such as eosinophilic infiltration and allergen protease action-eosinophilic cationic proteins' effects and allergens' proteolytic activity both contribute significantly to epithelial damage. In the airways, allergen proteases degrade the epithelial junctional proteins, allowing allergen penetration and its uptake by dendritic cells. This increase in allergen-immune system interaction induces the release of alarmins and the activation of type 2 inflammatory pathways, causing or worsening the main symptoms at the skin, bowel, and respiratory levels. We aim to highlight the molecular mechanisms underlying allergenic protease-induced epithelial barrier damage and the role of immune response in allergic asthma onset, maintenance, and progression. Moreover, we will explore potential clinical and radiological biomarkers of airway remodeling in allergic asthma patients.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Vitaliano Nicola Quaranta
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Silvano Dragonieri
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Francesco Menzella
- Pulmonology Unit, S. Valentino Hospital-AULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - David Selvaggio
- UOS di Malattie dell’Apparato Respiratorio Ospedale Cristo Re, 00167 Roma, Italy
| | - Giovanna Elisiana Carpagnano
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
8
|
Sharma R, Tiwari A, Kho AT, Wang AL, Srivastava U, Piparia S, Desai B, Wong R, Celedón JC, Peters SP, Smith LJ, Irvin CG, Castro M, Weiss ST, Tantisira KG, McGeachie MJ. Circulating MicroRNAs associated with Bronchodilator Response in Childhood Asthma. RESEARCH SQUARE 2023:rs.3.rs-3101724. [PMID: 37461659 PMCID: PMC10350209 DOI: 10.21203/rs.3.rs-3101724/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Rationale Bronchodilator response (BDR) is a measure of improvement in airway smooth muscle tone, inhibition of liquid accumulation and mucus section into the lumen in response to short-acting beta-2 agonists that varies among asthmatic patients. MicroRNAs (miRNAs) are well-known post-translational regulators. Identifying miRNAs associated with BDR could lead to a better understanding of the underlying complex pathophysiology. Objective The purpose of this study is to identify circulating miRNAs associated with bronchodilator response in asthma and decipher possible mechanism of bronchodilator response variation. Methods We used available small RNA sequencing on blood serum from 1,134 asthmatic children aged 6 to 14 years who participated in the Genetics of Asthma in Costa Rica Study (GACRS). We filtered the participants into high and low bronchodilator response (BDR) quartiles and used DeSeq2 to identify miRNAs with differential expression (DE) in high (N= 277) vs low (N= 278) BDR group. Replication was carried out in the Leukotriene modifier Or Corticosteroids or Corticosteroid-Salmeterol trial (LOCCS), an adult asthma cohort. The putative target genes of DE miRNAs were identified, and pathway enrichment analysis was performed. Results We identified 10 down-regulated miRNAs having odds ratios (OR) between 0.37 and 0.76 for a doubling of miRNA counts and one up-regulated miRNA (OR=2.26) between high and low BDR group. These were assessed for replication in the LOCCS cohort, where two miRNAs (miR-200b-3p and miR-1246) were associated. Further, functional annotation of 11 DE miRNAs were performed as well as of two replicated miRs. Target genes of these miRs were enriched in regulation of cholesterol biosynthesis by SREBPs, ESR-mediated signaling, G1/S transition, RHO GTPase cycle, and signaling by TGFB family pathways. Conclusion MiRNAs miR-1246 and miR-200b-3p are associated with both childhood and adult asthma BDR. Our findings add to the growing body of evidence that miRNAs play a significant role in the difference of asthma treatment response among patients as it points to genomic regulatory machinery underlying difference in bronchodilator response among patients. Trial registration LOCCS cohort [ClinicalTrials.gov number: NCT00156819], GACRS cohort [ClinicalTrials.gov number: NCT00021840].
Collapse
Affiliation(s)
- Rinku Sharma
- Brigham and Women's Hospital and Harvard Medical School
| | | | - Alvin T Kho
- Brigham and Women's Hospital and Harvard Medical School
| | | | | | | | - Brinda Desai
- University of California San Diego and Rady Children's Hospital
| | - Richard Wong
- University of California San Diego and Rady Children's Hospital
| | - Juan C Celedón
- University of Pittsburgh, UPMC Children's Hospital of Pittsburgh
| | | | | | | | | | - Scott T Weiss
- Brigham and Women's Hospital and Harvard Medical School
| | | | | |
Collapse
|
9
|
Tompkins E, Mimic B, Cuevas-Mora K, Schorsch H, Shah SD, Deshpande DA, Benovic JL, Penn RB, Pera T. PD 102807 Induces M3 mAChR-Dependent GRK-/Arrestin-Biased Signaling in Airway Smooth Muscle Cells. Am J Respir Cell Mol Biol 2022; 67:550-561. [PMID: 35944139 PMCID: PMC9651198 DOI: 10.1165/rcmb.2021-0320oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
G protein-coupled receptors (GPCRs) not only are turned on or off to control canonical G protein signaling but also may be fine-tuned to promote qualitative/biased signaling. Qualitative signaling by M3 muscarinic acetylcholine receptors (mAChRs) has been proposed, but its impact on physiologic systems remains unclear, and currently no biased M3 mAChR ligands have been described. Herein, we identify PD 102807 as a biased M3 ligand and delineate its signaling and function in human airway smooth muscle (ASM) cells. PD 102807 induced M3-mediated β-arrestin recruitment but not calcium mobilization. PD 102807 inhibited methacholine (MCh)-induced calcium mobilization in (M3-expressing) ASM cells. PD 102807 induced phosphorylation of AMP-activated protein kinase (AMPK) and the downstream effector acetyl-coenzyme A carboxylase (ACC). PD 102807- induced phosphorylated (p)-AMPK levels were greatly reduced in ASM cells with minimal M3 expression and were not inhibited by the Gq inhibitor YM-254890. Induction of p-AMPK and p-ACC was inhibited by β-arrestin 1 or GRK2/3 knockdown. Similarly, MCh induced phosphorylation of AMPK/ACC, but these effects were Gq dependent and unaffected by GRK2/3 knockdown. Consistent with the known ability of AMPK to inhibit transforming growth factor β (TGF-β)-mediated functions, PD 102807 inhibited TGF-β-induced SMAD-Luc activity, sm-α-actin expression, actin stress fiber formation, and ASM cell hypercontractility. These findings reveal that PD 102807 is a biased M3 ligand that inhibits M3-transduced Gq signaling but promotes Gq protein-independent, GRK-/arrestin-dependent, M3-mediated AMPK signaling, which in turn regulates ASM phenotype and contractile function. Consequently, biased M3 ligands hold significant promise as therapeutic agents capable of exploiting the pleiotropic nature of M3 signaling.
Collapse
Affiliation(s)
- Eric Tompkins
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Bogdana Mimic
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Karina Cuevas-Mora
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Hannah Schorsch
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Sushrut D. Shah
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Deepak A. Deshpande
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Jeffrey L. Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Raymond B. Penn
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| | - Tonio Pera
- Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Philadelphia, Pennsylvania; and
| |
Collapse
|
10
|
Joseph C, Tatler AL. Pathobiology of Airway Remodeling in Asthma: The Emerging Role of Integrins. J Asthma Allergy 2022; 15:595-610. [PMID: 35592385 PMCID: PMC9112045 DOI: 10.2147/jaa.s267222] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/25/2022] [Indexed: 12/19/2022] Open
Abstract
Airway remodeling is a complex clinical feature of asthma that involves long-term disruption and modification of airway architecture, which contributes significantly to airway hyperresponsiveness (AHR) and lung function decline. It is characterized by thickening of the airway smooth muscle layer, deposition of a matrix below the airway epithelium, resulting in subepithelial fibrosis, changes within the airway epithelium, leading to disruption of the barrier, and excessive mucous production and angiogenesis within the airway wall. Airway remodeling contributes to stiffer and less compliant airways in asthma and leads to persistent, irreversible airflow obstruction. Current asthma treatments aim to reduce airway inflammation and exacerbations but none are targeted towards airway remodeling. Inhibiting the development of airway remodeling or reversing established remodeling has the potential to dramatically improve symptoms and disease burden in asthmatic patients. Integrins are a family of transmembrane heterodimeric proteins that serve as the primary receptors for extracellular matrix (ECM) components, mediating cell-cell and cell-ECM interactions to initiate intracellular signaling cascades. Cells present within the lungs, including structural and inflammatory cells, express a wide and varying range of integrin heterodimer combinations and permutations. Integrins are emerging as an important regulator of inflammation, repair, remodeling, and fibrosis in the lung, particularly in chronic lung diseases such as asthma. Here, we provide a comprehensive summary of the current state of knowledge on integrins in the asthmatic airway and how these integrins promote the remodeling process, and emphasize their potential involvement in airway disease.
Collapse
Affiliation(s)
- Chitra Joseph
- Centre for Respiratory Research, National Institute for Health Research Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Amanda L Tatler
- Centre for Respiratory Research, National Institute for Health Research Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
11
|
Pera T, Loblundo C, Penn RB. Pharmacological Management of Asthma and COPD. COMPREHENSIVE PHARMACOLOGY 2022:762-802. [DOI: 10.1016/b978-0-12-820472-6.00095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Khan MM, Poeckel D, Halavatyi A, Zukowska-Kasprzyk J, Stein F, Vappiani J, Sevin DC, Tischer C, Zinn N, Eley JD, Gudmann NS, Muley T, Winter H, Fisher AJ, Nanthakumar CB, Bergamini G, Pepperkok R. An integrated multiomic and quantitative label-free microscopy-based approach to study pro-fibrotic signalling in ex vivo human precision-cut lung slices. Eur Respir J 2021; 58:2000221. [PMID: 33361096 PMCID: PMC8318569 DOI: 10.1183/13993003.00221-2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 12/09/2020] [Indexed: 12/17/2022]
Abstract
Fibrosis can affect any organ, resulting in the loss of tissue architecture and function with often life-threatening consequences. Pathologically, fibrosis is characterised by the expansion of connective tissue due to excessive deposition of extracellular matrix (ECM) proteins, including the fibrillar forms of collagen. A significant limitation for discovering cures for fibrosis is the availability of suitable human models and techniques to quantify mature fibrillar collagen deposition as close as possible to human physiological conditions.Here we have extensively characterised an ex vivo cultured human lung tissue-derived, precision-cut lung slices (hPCLS) model using label-free second harmonic generation (SHG) light microscopy to quantify fibrillar collagen deposition and mass spectrometry-based techniques to obtain a proteomic and metabolomic fingerprint of hPCLS in ex vivo culture.We demonstrate that hPCLS are viable and metabolically active, with mesenchymal, epithelial, endothelial and immune cell types surviving for at least 2 weeks in ex vivo culture. Analysis of hPCLS-conditioned supernatants showed a strong induction of pulmonary fibrosis-related ECM proteins upon transforming growth factor-β1 (TGF-β1) stimulation. This upregulation of ECM proteins was not translated into an increased deposition of fibrillar collagen. In support of this observation, we revealed the presence of a pro-ECM degradation activity in our ex vivo cultures of hPCLS, inhibition of which by a metalloproteinase inhibitor resulted in increased collagen deposition in response to TGF-β1 stimulation.Together the data show that an integrated approach of measuring soluble pro-fibrotic markers alongside quantitative SHG-based analysis of fibrillar collagen is a valuable tool for studying pro-fibrotic signalling and testing anti-fibrotic agents.
Collapse
Affiliation(s)
- Muzamil Majid Khan
- European Molecular Biology Laboratory, Heidelberg, Germany
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Daniel Poeckel
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
| | - Aliaksandr Halavatyi
- European Molecular Biology Laboratory, Heidelberg, Germany
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | | | - Frank Stein
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Daniel C Sevin
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
| | | | - Nico Zinn
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
| | | | | | - Thomas Muley
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Biobank Thoraxklinik, University Hospital Heidelberg, Heidelberg, Germany
| | - Hauke Winter
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Biobank Thoraxklinik, University Hospital Heidelberg, Heidelberg, Germany
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute and Institute of Transplantation, Newcastle upon Tyne Hospitals, Newcastle upon Tyne, UK
| | | | - Giovanna Bergamini
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
- G. Bergamini and R. Pepperkok contributed equally to this article as lead authors and supervised the work
| | - Rainer Pepperkok
- European Molecular Biology Laboratory, Heidelberg, Germany
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- G. Bergamini and R. Pepperkok contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
13
|
Stretch-activated calcium mobilization in airway smooth muscle and pathophysiology of asthma. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
14
|
Effects of (a Combination of) the Beta 2-Adrenoceptor Agonist Indacaterol and the Muscarinic Receptor Antagonist Glycopyrrolate on Intrapulmonary Airway Constriction. Cells 2021; 10:cells10051237. [PMID: 34069899 PMCID: PMC8157597 DOI: 10.3390/cells10051237] [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: 04/20/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/01/2022] Open
Abstract
Expression of bronchodilatory β2-adrenoceptors and bronchoconstrictive muscarinic M3-receptors alter with airway size. In COPD, (a combination of) β2-agonists and muscarinic M3-antagonists (anticholinergics) are used as bronchodilators. We studied whether differential receptor expression in large and small airways affects the response to β2-agonists and anticholinergics in COPD. Bronchoprotection by indacaterol (β2-agonist) and glycopyrrolate (anticholinergic) against methacholine- and EFS-induced constrictions of large and small airways was measured in guinea pig and human lung slices using video-assisted microscopy. In guinea pig lung slices, glycopyrrolate (1, 3 and 10 nM) concentration-dependently protected against methacholine- and EFS-induced constrictions, with no differences between large and small intrapulmonary airways. Indacaterol (0.01, 0.1, 1 and 10 μM) also provided concentration-dependent protection, which was greater in large airways against methacholine and in small airways against EFS. Indacaterol (10 μM) and glycopyrrolate (10 nM) normalized small airway hyperresponsiveness in COPD lung slices. Synergy of low indacaterol (10 nM) and glycopyrrolate (1 nM) concentrations was greater in LPS-challenged guinea pigs (COPD model) compared to saline-challenged controls. In conclusion, glycopyrrolate similarly protects large and small airways, whereas the protective effect of indacaterol in the small, but not the large, airways depends on the contractile stimulus used. Moreover, findings in a guinea pig model indicate that the synergistic bronchoprotective effect of indacaterol and glycopyrrolate is enhanced in COPD.
Collapse
|
15
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Wu X, Verschut V, Woest ME, Ng-Blichfeldt JP, Matias A, Villetti G, Accetta A, Facchinetti F, Gosens R, Kistemaker LEM. Rho-Kinase 1/2 Inhibition Prevents Transforming Growth Factor-β-Induced Effects on Pulmonary Remodeling and Repair. Front Pharmacol 2021; 11:609509. [PMID: 33551810 PMCID: PMC7855981 DOI: 10.3389/fphar.2020.609509] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor (TGF)-β-induced myofibroblast transformation and alterations in mesenchymal-epithelial interactions contribute to chronic lung diseases such as chronic obstructive pulmonary disease (COPD), asthma and pulmonary fibrosis. Rho-associated coiled-coil-forming protein kinase (ROCK) consists as two isoforms, ROCK1 and ROCK2, and both are playing critical roles in many cellular responses to injury. In this study, we aimed to elucidate the differential role of ROCK isoforms on TGF-β signaling in lung fibrosis and repair. For this purpose, we tested the effect of a non-selective ROCK 1 and 2 inhibitor (compound 31) and a selective ROCK2 inhibitor (compound A11) in inhibiting TGF-β-induced remodeling in lung fibroblasts and slices; and dysfunctional epithelial-progenitor interactions in lung organoids. Here, we demonstrated that the inhibition of ROCK1/2 with compound 31 represses TGF-β-driven actin remodeling as well as extracellular matrix deposition in lung fibroblasts and PCLS, whereas selective ROCK2 inhibition with compound A11 did not. Furthermore, the TGF-β induced inhibition of organoid formation was functionally restored in a concentration-dependent manner by both dual ROCK 1 and 2 inhibition and selective ROCK2 inhibition. We conclude that dual pharmacological inhibition of ROCK 1 and 2 counteracts TGF-β induced effects on remodeling and alveolar epithelial progenitor function, suggesting this to be a promising therapeutic approach for respiratory diseases associated with fibrosis and defective lung repair.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Manon E. Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- AQUILO BV, Groningen, Netherlands
| | - John-Poul Ng-Blichfeldt
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ana Matias
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gino Villetti
- Corporate Pre-Clinical R and D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Alessandro Accetta
- Corporate Pre-Clinical R and D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | | | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Loes E. M. Kistemaker
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- AQUILO BV, Groningen, Netherlands
| |
Collapse
|
17
|
Šutovská M, Kocmálová M, Kazimierová I, Forsberg CIN, Jošková M, Adamkov M, Fraňová S. Effects of Inhalation of STIM-Orai Antagonist SKF 96365 on Ovalbumin-Induced Airway Remodeling in Guinea Pigs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1335:87-101. [PMID: 33742420 DOI: 10.1007/5584_2021_633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Airway remodeling (AR) consists of wall thickening and hyperreactivity. STIM (stromal interaction molecule) and Orai protein pathways mediate extracellular Ca2+ signals involved in AR. This study aims to define the effects on AR of the STIM-Orai antagonist SKF 96365 given by inhalation in three increasing doses in ovalbumin-induced AR. In the control group, the antiasthmatic budesonide and salbutamol were given in the same model. The airway structure was evaluated by histological and immunohistochemistry and reactivity by specific airway resistance, contraction strength of isolated airway smooth muscles, and mucociliary clearance expressed by ciliary beating frequency. The immuno-biochemical markers of chronic inflammation were evaluated by BioPlex and ELISA assays. The AR was mediated by inflammatory cytokines and growth factors. The findings show significant anti-remodeling effects of SKF 96365, which were associated with a decrease in airway hyperreactivity. The anti-remodeling effect of SKF 96365 was mediated via the suppression of IL-4, IL-5, and IL-13 synthesis, and IL-12-INF-γ-TGF-β pathway. The budesonide-related AR suppression had to do with a decrease in proinflammatory cytokines and an increase in the anti-inflammatory IL-10, with negligible influence on growth factors synthesis and mucous glands activity.
Collapse
Affiliation(s)
- Martina Šutovská
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia
| | - Michaela Kocmálová
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia. .,Martin's Biomedical Center (BioMed), Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia.
| | - Ivana Kazimierová
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia.,Martin's Biomedical Center (BioMed), Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | | | - Marta Jošková
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia
| | - Marian Adamkov
- Institute of Histology and Embryology Jessenius Faculty of Medicine Comenius University, Martin, Slovakia
| | - Soňa Fraňová
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia
| |
Collapse
|
18
|
O'Sullivan MJ, Phung TKN, Park JA. Bronchoconstriction: a potential missing link in airway remodelling. Open Biol 2020; 10:200254. [PMID: 33259745 PMCID: PMC7776576 DOI: 10.1098/rsob.200254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
In asthma, progressive structural changes of the airway wall are collectively termed airway remodelling. Despite its deleterious effect on lung function, airway remodelling is incompletely understood. As one of the important causes leading to airway remodelling, here we discuss the significance of mechanical forces that are produced in the narrowed airway during asthma exacerbation, as a driving force of airway remodelling. We cover in vitro, ex vivo and in vivo work in this field, and discuss up-to-date literature supporting the idea that bronchoconstriction may be the missing link in a comprehensive understanding of airway remodelling in asthma.
Collapse
Affiliation(s)
| | | | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, USA
| |
Collapse
|
19
|
The anti-asthmatic potential of flavonol kaempferol in an experimental model of allergic airway inflammation. Eur J Pharmacol 2020; 891:173698. [PMID: 33129789 DOI: 10.1016/j.ejphar.2020.173698] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/17/2020] [Accepted: 10/27/2020] [Indexed: 12/29/2022]
Abstract
Flavonol kaempferol possesses a broad spectrum of potent pharmacological activities that seem to be effective in the modulation of allergic respiratory diseases. In our study, an experimental animal model of ovalbumin (OVA)-induced allergic airway inflammation in guinea pigs was used to determine the anti-asthmatic potential of kaempferol. The parameters of specific airway resistance (sRaw) and cough reflex response were evaluated in vivo. In vitro, an assessment of tracheal smooth muscle (TSM) contractility and analyses of inflammatory cytokines (IL-4, IL-5, IL-13, GM-CSF, IFN-γ), transforming growth factor (TGF-β1), immune cells count and ciliary beating frequency (CBF) were performed. Both single (6, 20 mg/kg b. w. p. o.) and long-term administered doses of kaempferol (20 mg/kg b. w. p. o., 21 days) suppressed sRaw provoked by histamine in conscious animals. The administration of kaempferol for 21 days attenuated histamine-induced TSM contractility in vitro and ameliorated the progression of chronic airway inflammation by decreasing the levels of IL-5, IL-13, GM-CSF, eosinophil count in bronchoalveolar lavage (BAL) fluid and TGF-β1 protein level in lung tissue. Kaempferol also eliminated the alterations in cough reflex sensitivity invoked by OVA-sensitization, but it did not affect CBF. The results demonstrate that flavonol kaempferol can modulate allergic airway inflammation and associated asthma features (AHR, aberrant stimulation of cough reflex).
Collapse
|
20
|
Veerati PC, Mitchel JA, Reid AT, Knight DA, Bartlett NW, Park JA, Grainge CL. Airway mechanical compression: its role in asthma pathogenesis and progression. Eur Respir Rev 2020; 29:190123. [PMID: 32759373 PMCID: PMC8008491 DOI: 10.1183/16000617.0123-2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/30/2020] [Indexed: 12/22/2022] Open
Abstract
The lung is a mechanically active organ, but uncontrolled or excessive mechanical forces disrupt normal lung function and can contribute to the development of disease. In asthma, bronchoconstriction leads to airway narrowing and airway wall buckling. A growing body of evidence suggests that pathological mechanical forces induced by airway buckling alone can perpetuate disease processes in asthma. Here, we review the data obtained from a variety of experimental models, including in vitro, ex vivo and in vivo approaches, which have been used to study the impact of mechanical forces in asthma pathogenesis. We review the evidence showing that mechanical compression alters the biological and biophysical properties of the airway epithelium, including activation of the epidermal growth factor receptor pathway, overproduction of asthma-associated mediators, goblet cell hyperplasia, and a phase transition of epithelium from a static jammed phase to a mobile unjammed phase. We also define questions regarding the impact of mechanical forces on the pathology of asthma, with a focus on known triggers of asthma exacerbations such as viral infection.
Collapse
Affiliation(s)
- Punnam Chander Veerati
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Jennifer A Mitchel
- Molecular and Integrative Physiological Sciences Program, Dept of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Reid
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Dept of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
- Research and Academic Affairs, Providence Health Care Research Institute, Vancouver, Canada
| | - Nathan W Bartlett
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| | - Jin-Ah Park
- Molecular and Integrative Physiological Sciences Program, Dept of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chris L Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
- Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| |
Collapse
|
21
|
Wortley MA, Bonvini SJ. Transforming Growth Factor-β1: A Novel Cause of Resistance to Bronchodilators in Asthma? Am J Respir Cell Mol Biol 2019; 61:134-135. [PMID: 30768914 PMCID: PMC6670036 DOI: 10.1165/rcmb.2019-0020ed] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | - Sara J Bonvini
- 1Respiratory PharmacologyImperial CollegeLondon, United Kingdom
| |
Collapse
|
22
|
Benam KH, Königshoff M, Eickelberg O. Breaking the In Vitro Barrier in Respiratory Medicine. Engineered Microphysiological Systems for Chronic Obstructive Pulmonary Disease and Beyond. Am J Respir Crit Care Med 2019; 197:869-875. [PMID: 29262260 DOI: 10.1164/rccm.201709-1795pp] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Kambez H Benam
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; and.,2 Department of Bioengineering, University of Colorado Denver, Aurora, Colorado
| | - Melanie Königshoff
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; and
| | - Oliver Eickelberg
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado; and
| |
Collapse
|
23
|
Wu X, van Dijk EM, Bos IST, Kistemaker LEM, Gosens R. Mouse Lung Tissue Slice Culture. Methods Mol Biol 2019; 1940:297-311. [PMID: 30788834 DOI: 10.1007/978-1-4939-9086-3_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Precision-cut lung slices (PCLS) represent an ex vivo model widely used in visualizing interactions between lung structure and function. The major advantage of this technique is that the presence, differentiation state, and localization of the more than 40 cell types that make up the lung are in accordance with the physiological situation found in lung tissue, including the right localization and patterning of extracellular matrix elements. Here we describe the methodology involved in preparing and culturing PCLS followed by detailed practical information about their possible applications.
Collapse
Affiliation(s)
- Xinhui Wu
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eline M van Dijk
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - I Sophie T Bos
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Loes E M Kistemaker
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
24
|
Adler KB. Does Transforming Growth Factor-β Induce Persistent Airway Obstruction after Asthma Exacerbations? Am J Respir Cell Mol Biol 2019; 58:543-544. [PMID: 29714630 DOI: 10.1165/rcmb.2017-0339ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Kenneth B Adler
- 1 College of Veterinary Medicine North Carolina State University Raleigh, North Carolina
| |
Collapse
|
25
|
Kim SH, Mitchel JA, McGill M, Cremona TP, Baek JW, Kasahara DI, Anathy V, Israel E, Park JA. Increased extracellular maspin levels after mechanical compression in vitro or allergen challenge in vivo. J Allergy Clin Immunol 2019; 144:1116-1118.e4. [PMID: 31228474 DOI: 10.1016/j.jaci.2019.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/05/2019] [Accepted: 06/10/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Sae-Hoon Kim
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass; Department of Internal Medicine, Seoul National University Bundang University Hospital, Seoul, Korea
| | - Jennifer A Mitchel
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Maureen McGill
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Tiziana P Cremona
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Ji Won Baek
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - David I Kasahara
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | | | - Jin-Ah Park
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass.
| |
Collapse
|
26
|
Veerati PC, Grainge C. Peering deeper into asthmatic lungs. Respirology 2019; 24:1037-1038. [PMID: 31216084 DOI: 10.1111/resp.13625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 05/28/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Punnam C Veerati
- Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Chris Grainge
- Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
27
|
Irons L, Owen MR, O'Dea RD, Brook BS. Effect of Loading History on Airway Smooth Muscle Cell-Matrix Adhesions. Biophys J 2019; 114:2679-2690. [PMID: 29874617 DOI: 10.1016/j.bpj.2018.04.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 01/06/2023] Open
Abstract
Integrin-mediated adhesions between airway smooth muscle (ASM) cells and the extracellular matrix (ECM) regulate how contractile forces generated within the cell are transmitted to its external environment. Environmental cues are known to influence the formation, size, and survival of cell-matrix adhesions, but it is not yet known how they are affected by dynamic fluctuations associated with tidal breathing in the intact airway. Here, we develop two closely related theoretical models to study adhesion dynamics in response to oscillatory loading of the ECM, representing the dynamic environment of ASM cells in vivo. Using a discrete stochastic-elastic model, we simulate individual integrin binding and rupture events and observe two stable regimes in which either bond formation or bond rupture dominate, depending on the amplitude of the oscillatory loading. These regimes have either a high or low fraction of persistent adhesions, which could affect the level of strain transmission between contracted ASM cells and the airway tissue. For intermediate loading, we observe a region of bistability and hysteresis due to shared loading between existing bonds; the level of adhesion depends on the loading history. These findings are replicated in a related continuum model, which we use to investigate the effect of perturbations mimicking deep inspirations (DIs). Because of the bistability, a DI applied to the high adhesion state could either induce a permanent switch to a lower adhesion state or allow a return of the system to the high adhesion state. Transitions between states are further influenced by the frequency of oscillations, cytoskeletal or ECM stiffnesses, and binding affinities, which modify the magnitudes of the stable adhesion states as well as the region of bistability. These findings could explain (in part) the transient bronchodilatory effect of a DI observed in asthmatics compared to a more sustained effect in normal subjects.
Collapse
Affiliation(s)
- Linda Irons
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom.
| | - Markus R Owen
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Reuben D O'Dea
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Bindi S Brook
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
28
|
Li N, He Y, Yang G, Yu Q, Li M. Role of TRPC1 channels in pressure-mediated activation of airway remodeling. Respir Res 2019; 20:91. [PMID: 31092255 PMCID: PMC6518742 DOI: 10.1186/s12931-019-1050-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Bronchoconstriction and cough, a characteristic of the asthmatic response, leads to development of compressive stresses in the airway wall. We hypothesized that progressively pathological high mechanical stress could act on mechanosensitive cation channels, such as transient receptor potential channel 1 (TRPC1) and then contributes to airway remodeling. METHODS We imitate the pathological airway pressure in vitro using cyclic stretch at 10 and 15% elongation. Ca2+ imaging was applied to measure the activity of TRPC1 after bronchial epithelial cells exposed to cyclic stretch for 0, 0.5, 1, 1.5, 2, 2.5 h. To further clarify the function of channnel TRPC1 in the process of mechano-transduction in airway remodeling, the experiment in vivo was implemented. The TRPC1 siRNA and budesonide were applied separately to asthmatic models. The morphological changes were measured by HE and Massion method. The expression levels of TRPC1 were evaluated by real-time PCR, western blot and immunohistochemistry. The protein expression level of IL-13, TGF-β1 and MMP-9 in BALF were measured by ELISA. RESULTS The result showed that cyclic stretch for 15% elongation at 1.5 h could maximize the activity of TRPC1 channel. This influx in Ca2+ was blocked by TRPC1 siRNA. Higher TRPC1 expression was observed in the bronchial epithelial layer of ovalbumin induced asthmatic models. The knockdown of TRPC1 with TRPC1 siRNA was associated with a hampered airway remodeling process, such as decreased bronchial wall thickness and smooth muscle hypertrophy/hyperplasia, a decreased ECM deposition area and inflammation infiltration around airway wall. Meantime, expression of IL-13, TGF-β1 and MMP-9 in OVA+TRPC1 siRNA also showed reduced level. TRPC1 intervention treatment showed similar anti-remodeling therapeutic effect with budesonide. CONCLUSIONS These results demonstrate that most TRPC1 channels expressed in bronchial epithelial cells mediate the mechanotransduction mechanism. TRPC1 inducing abnormal Ca2+ signal mediates receptor-stimulated and mechanical stimulus-induced airway remodeling. The inhibition of TRPC1 channel could produce similar therapeutic effect as glucocortisteroid to curb the development of asthmatic airway remodeling.
Collapse
Affiliation(s)
- Na Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Ye He
- Department of Geriatrics, Sichuan Provincial People’s Hospital, Sichuan Academy of Medical Science, Chengdu, Sichuan Province 610072 People’s Republic of China
| | - Gang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Qian Yu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Minchao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| |
Collapse
|
29
|
Vasconcelos LHC, Silva MDCC, Costa AC, de Oliveira GA, de Souza ILL, Queiroga FR, Araujo LCDC, Cardoso GA, Righetti RF, Silva AS, da Silva PM, Carvalho CRDO, Vieira GC, Tibério IDFLC, Cavalcante FDA, da Silva BA. A Guinea Pig Model of Airway Smooth Muscle Hyperreactivity Induced by Chronic Allergic Lung Inflammation: Contribution of Epithelium and Oxidative Stress. Front Pharmacol 2019; 9:1547. [PMID: 30814952 PMCID: PMC6353839 DOI: 10.3389/fphar.2018.01547] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 12/18/2018] [Indexed: 11/17/2022] Open
Abstract
Asthma is a heterogeneous disease of the airways characterized by chronic inflammation associated with bronchial and smooth muscle hyperresponsiveness. Currently, different murine models for the study of asthma show poor bronchial hyperresponsiveness due to a scarcity of smooth muscle and large airways, resulting in a failure to reproduce smooth muscle hyperreactivity. Thus, we aimed to standardize a guinea pig model of chronic allergic lung inflammation mimicking airway smooth muscle hyperreactivity observed in asthmatics (Asth). Animals were randomly divided into a control group (Ctrl), which received saline (0.9% NaCl), and the Asth group, subjected to in vivo sensitization with ovalbumin (OVA) nebulization. Morphological analysis was performed by hematoxylin-eosin staining. Bronchial hyperresponsiveness was evaluated by nebulization time in the fifth, sixth, and seventh inhalations (NT5-7) and tracheal isometric contractions were assessed by force transducer. Total antioxidant capacity was measured by the 2,2-diphenyl-1-picrylhydrazyl (DPPH) method and protein expression by Western blot. Histologically, the Asth group developed peribronchial cellular infiltrate, epithelial hyperplasia and smooth muscle thickening. After the fourth nebulization, the Asth group developed bronchial hyperreactivity. The trachea from the Asth group contracted after in vitro stimulation with OVA, differing from the Ctrl group, which showed no response. Additionally, airway smooth muscle hyperreactivity to carbachol and histamine was observed in the Asth group only in intact epithelium preparations, but not to KCl, and this effect was associated with an augmented production of reactive oxygen species. Moreover, lung inflammation impaired the relaxant potency of isoproterenol only in intact epithelium preparations, without interfering with nifedipine, and it was found to be produced by transforming growth factor-β negative modulation of β adrenergic receptors and, furthermore, big-conductance Ca2+-sensitive K+ channels. These effects were also associated with increased levels of phosphatidylinositol 3-kinases but not extracellular signal-regulated kinases 1/2 or phosphorylation, and augmented α-actin content as well, explaining the increased smooth muscle mass. Furthermore, pulmonary antioxidant capacity was impaired in the Asth group. Therefore, we developed a standardized and easy-to-use, reproducible guinea pig model of lung inflammation that mimics airway smooth muscle hypercontractility, facilitating the investigation of the mechanisms of bronchial hyperresponsiveness in asthma and new therapeutic alternatives.
Collapse
Affiliation(s)
- Luiz Henrique César Vasconcelos
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Maria da Conceição Correia Silva
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Alana Cristina Costa
- Graduação em Farmácia, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Giuliana Amanda de Oliveira
- Graduação em Farmácia, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Iara Leão Luna de Souza
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Fernando Ramos Queiroga
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Layanne Cabral da Cunha Araujo
- Programa de Pós graduação em Ciências (Fisiologia Humana), Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, Brazil
| | - Glêbia Alexa Cardoso
- Programa Associado de Pós graduação em Educação Física, Universidade Federal da Paraíba/Universidade do Pernambuco, João Pessoa, Brazil
| | - Renato Fraga Righetti
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- Hospital Sírio Libanês, São Paulo, Brazil
| | - Alexandre Sérgio Silva
- Programa Associado de Pós graduação em Educação Física, Universidade Federal da Paraíba/Universidade do Pernambuco, João Pessoa, Brazil
- Departamento de Educação Física, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Patrícia Mirella da Silva
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
- Departamento de Biologia Molecular, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Carla Roberta de Oliveira Carvalho
- Programa de Pós graduação em Ciências (Fisiologia Humana), Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Biofísica e Fisiologia, Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, Brazil
| | - Giciane Carvalho Vieira
- Departamento de Morfologia/Centro de Ciências da Saúde/Universidade Federal da Paraíba, João Pessoa, Brazil
| | | | - Fabiana de Andrade Cavalcante
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
- Departamento de Fisiologia e Patologia/Centro de Ciências da Saúde/Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
- Departamento de Ciências Farmacêuticas/Centro de Ciências da Saúde/Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
30
|
Maarsingh H, Bidan CM, Brook BS, Zuidhof AB, Elzinga CRS, Smit M, Oldenburger A, Gosens R, Timens W, Meurs H. Small airway hyperresponsiveness in COPD: relationship between structure and function in lung slices. Am J Physiol Lung Cell Mol Physiol 2019; 316:L537-L546. [PMID: 30628486 PMCID: PMC6459292 DOI: 10.1152/ajplung.00325.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The direct relationship between pulmonary structural changes and airway hyperresponsiveness (AHR) in chronic obstructive pulmonary disease (COPD) is unclear. We investigated AHR in relation to airway and parenchymal structural changes in a guinea pig model of COPD and in COPD patients. Precision-cut lung slices (PCLS) were prepared from guinea pigs challenged with lipopolysaccharide or saline two times weekly for 12 wk. Peripheral PCLS were obtained from patients with mild to moderate COPD and non-COPD controls. AHR to methacholine was measured in large and small airways using video-assisted microscopy. Airway smooth muscle mass and alveolar airspace size were determined in the same slices. A mathematical model was used to identify potential changes in biomechanical properties underlying AHR. In guinea pigs, lipopolysaccharide increased the sensitivity of large (>150 μm) airways toward methacholine by 4.4-fold and the maximal constriction of small airways (<150 μm) by 1.5-fold. Similarly increased small airway responsiveness was found in COPD patients. In both lipopolysaccharide-challenged guinea pigs and patients, airway smooth muscle mass was unaltered, whereas increased alveolar airspace correlated with small airway hyperresponsiveness in guinea pigs. Fitting the parameters of the model indicated that COPD weakens matrix mechanical properties and enhances stiffness differences between the airway and the parenchyma, in both species. In conclusion, this study demonstrates small airway hyperresponsiveness in PCLS from COPD patients. These changes may be related to reduced parenchymal retraction forces and biomechanical changes in the airway wall. PCLS from lipopolysaccharide-exposed guinea pigs may be useful to study mechanisms of small airway hyperresponsiveness in COPD.
Collapse
Affiliation(s)
- Harm Maarsingh
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University , West Palm Beach, Florida.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Cécile M Bidan
- Laboratoire Interdisciplinaire de Physique, Centre for Scientific Research, Université Grenoble Alpes , Grenoble , France.,Department of Biomaterials, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany
| | - Bindi S Brook
- School of Mathematical Sciences, University of Nottingham , Nottingham , United Kingdom
| | - Annet B Zuidhof
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Carolina R S Elzinga
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Marieke Smit
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Department of Pathology and Medical Biology, University Medical Center Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Anouk Oldenburger
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
31
|
Rosethorne EM, Charlton SJ. Airway remodeling disease: primary human structural cells and phenotypic and pathway assays to identify targets with potential to prevent or reverse remodeling. J Exp Pharmacol 2018; 10:75-85. [PMID: 30568517 PMCID: PMC6276605 DOI: 10.2147/jep.s159124] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Airway remodeling is a characteristic of many chronic respiratory diseases and occurs when there are significant changes to the architecture of the small and large airways leading to progressive loss of lung function. Some common features include airway smooth muscle and goblet cell hyperplasia, basement membrane thickening and subepithelial fibrosis. To explore the mechanisms driving airway remodeling and identify novel targets to treat this aspect of respiratory disease, appropriate models must be used that will accurately predict the pathology of disease. Phenotypic assays can be used in primary human lung cells to measure changes in cell behavior that are associated with particular disease pathology. This is becoming increasingly popular when targeting chronic pathologies such as airway remodeling, where phenotypic assays are likely to model disease in vitro more accurately than traditional second messenger assays. Here we review the use of primary human lung structural cells in a range of disease-relevant chronic phenotypic assays, and how they may be used in target identification/validation and drug discovery.
Collapse
Affiliation(s)
- Elizabeth M Rosethorne
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK, .,Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK, .,Excellerate Bioscience Ltd, MediCity, Nottingham NG7 2UH, UK,
| | - Steven J Charlton
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK, .,Centre of Membrane and Protein and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK, .,Excellerate Bioscience Ltd, MediCity, Nottingham NG7 2UH, UK,
| |
Collapse
|
32
|
Gosens R, Gross N. The mode of action of anticholinergics in asthma. Eur Respir J 2018; 52:13993003.01247-2017. [PMID: 30115613 PMCID: PMC6340638 DOI: 10.1183/13993003.01247-2017] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 08/09/2018] [Indexed: 01/25/2023]
Abstract
Acetylcholine binds to muscarinic receptors to play a key role in the pathophysiology of asthma, leading to bronchoconstriction, increased mucus secretion, inflammation and airway remodelling. Anticholinergics are muscarinic receptor antagonists that are used in the treatment of chronic obstructive pulmonary disease and asthma. Recent in vivo and in vitro data have increased our understanding of how acetylcholine contributes to the disease manifestations of asthma, as well as elucidating the mechanism of action of anticholinergics. This review assesses the latest literature on acetylcholine in asthma pathophysiology, with a closer look at its role in airway inflammation and remodelling. New insights into the mechanism of action of anticholinergics, their effects on airway remodelling, and a review of the efficacy and safety of long-acting anticholinergics in asthma treatment will also be covered, including a summary of the latest clinical trial data. Pre-clinical data suggest that anticholinergics can reduce acetylcholine-induced airway inflammation and remodellinghttp://ow.ly/xqAQ30loP8F
Collapse
Affiliation(s)
| | - Nicholas Gross
- University Medical Research LLC, St Francis Hospital, Hartford, CT, USA
| |
Collapse
|
33
|
Hill MR, Philp CJ, Billington CK, Tatler AL, Johnson SR, O'Dea RD, Brook BS. A theoretical model of inflammation- and mechanotransduction-driven asthmatic airway remodelling. Biomech Model Mechanobiol 2018; 17:1451-1470. [PMID: 29968161 PMCID: PMC6154265 DOI: 10.1007/s10237-018-1037-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/22/2018] [Indexed: 12/28/2022]
Abstract
Inflammation, airway hyper-responsiveness and airway remodelling are well-established hallmarks of asthma, but their inter-relationships remain elusive. In order to obtain a better understanding of their inter-dependence, we develop a mechanochemical morphoelastic model of the airway wall accounting for local volume changes in airway smooth muscle (ASM) and extracellular matrix in response to transient inflammatory or contractile agonist challenges. We use constrained mixture theory, together with a multiplicative decomposition of growth from the elastic deformation, to model the airway wall as a nonlinear fibre-reinforced elastic cylinder. Local contractile agonist drives ASM cell contraction, generating mechanical stresses in the tissue that drive further release of mitogenic mediators and contractile agonists via underlying mechanotransductive signalling pathways. Our model predictions are consistent with previously described inflammation-induced remodelling within an axisymmetric airway geometry. Additionally, our simulations reveal novel mechanotransductive feedback by which hyper-responsive airways exhibit increased remodelling, for example, via stress-induced release of pro-mitogenic and pro-contractile cytokines. Simulation results also reveal emergence of a persistent contractile tone observed in asthmatics, via either a pathological mechanotransductive feedback loop, a failure to clear agonists from the tissue, or a combination of both. Furthermore, we identify various parameter combinations that may contribute to the existence of different asthma phenotypes, and we illustrate a combination of factors which may predispose severe asthmatics to fatal bronchospasms.
Collapse
Affiliation(s)
- Michael R Hill
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Room C25, Mathematical Sciences Building, University Park, Nottingham, NG7 2RD, UK.
| | - Christopher J Philp
- Division of Respiratory Medicine, Nottingham Biomedical Research Centre, University of Nottingham, D Floor, South Block, Queen's Medical Centre Campus, Nottingham, NG7 2UH, UK
| | - Charlotte K Billington
- Division of Respiratory Medicine, Nottingham Biomedical Research Centre, University of Nottingham, D Floor, South Block, Queen's Medical Centre Campus, Nottingham, NG7 2UH, UK
| | - Amanda L Tatler
- Division of Respiratory Medicine, Nottingham Biomedical Research Centre, Nottingham City Hospital, University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Simon R Johnson
- Division of Respiratory Medicine, Nottingham Biomedical Research Centre, University of Nottingham, D Floor, South Block, Queen's Medical Centre Campus, Nottingham, NG7 2UH, UK
| | - Reuben D O'Dea
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Room C28, Mathematical Sciences Building, University Park, Nottingham, NG7 2RD, UK
| | - Bindi S Brook
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Room C26, Mathematical Sciences Building, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
34
|
Zuo H, Han B, Poppinga WJ, Ringnalda L, Kistemaker LEM, Halayko AJ, Gosens R, Nikolaev VO, Schmidt M. Cigarette smoke up-regulates PDE3 and PDE4 to decrease cAMP in airway cells. Br J Pharmacol 2018; 175:2988-3006. [PMID: 29722436 PMCID: PMC6016635 DOI: 10.1111/bph.14347] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 03/16/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE cAMP is a central second messenger that broadly regulates cell function and can underpin pathophysiology. In chronic obstructive pulmonary disease, a lung disease primarily provoked by cigarette smoke (CS), the activation of cAMP-dependent pathways, via inhibition of hydrolyzing PDEs, is a major therapeutic strategy. Mechanisms that disrupt cAMP signalling in airway cells, in particular regulation of endogenous PDEs, are poorly understood. EXPERIMENTAL APPROACH We used a novel Förster resonance energy transfer (FRET) based cAMP biosensor in mice in vivo, ex vivo precision cut lung slices (PCLS) and in human cell models, in vitro, to track the effects of CS exposure. KEY RESULTS Under fenoterol stimulation, FRET responses to cilostamide were significantly increased in in vivo, ex vivo PCLS exposed to CS and in human airway smooth muscle cells exposed to CS extract. FRET signals to rolipram were only increased in the in vivo CS model. Under basal conditions, FRET responses to cilostamide and rolipram were significantly increased in in vivo, ex vivo PCLS exposed to CS. Elevated FRET signals to rolipram correlated with a protein up-regulation of PDE4 subtypes. In ex vivo PCLS exposed to CS extract, rolipram reversed down-regulation of ciliary beating frequency, whereas only cilostamide significantly increased airway relaxation of methacholine pre-contracted airways. CONCLUSION AND IMPLICATIONS Exposure to CS, in vitro or in vivo, up-regulated expression and activity of both PDE3 and PDE4, which affected real-time cAMP dynamics. These mechanisms determine the availability of cAMP and can contribute to CS-induced pulmonary pathophysiology.
Collapse
Affiliation(s)
- Haoxiao Zuo
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands.,Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Bing Han
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - Wilfred J Poppinga
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - Lennard Ringnalda
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Hamburg, Germany
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| |
Collapse
|
35
|
Huo Y, Xu J, Guan L, Wu W, Guo B, Yang Y, Lin L, Ou Y, Jiang F, Zhou L, Chen R. Methacholine induces extracellular matrix production by human airway smooth muscle cells through β-catenin signaling. Respir Physiol Neurobiol 2018; 254:55-63. [PMID: 29715518 DOI: 10.1016/j.resp.2018.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 04/21/2018] [Accepted: 04/28/2018] [Indexed: 11/28/2022]
Abstract
Altered extracellular matrix (ECM) production by airway smooth muscle cells (ASMCs) is an important feature of airway remodeling. Muscarinic receptor agonists contribute to ECM production in vivo, but the mechanisms involved remain unclear. This study attempted to investigate the role of methacholine in promoting ECM production by human ASMCs (HASMCs) and the underlying mechanism. We found that methacholine induced the expression of collagen I protein and multiple ECM genes. β-catenin signaling was activated in this process upon GSK3β phosphorylation, leading to upregulation of total and active β-catenin. Silencing β-catenin by specific small interfering RNA (siRNA) or with the β-catenin inhibitor, PKF115-584, decreased collagen I expression. Conversely, overexpression of active β-catenin by adenoviruses carrying the S33Y-β-catenin mutant increased the methacholine-induced collagen I expression. Furthermore, methacholine induced TGF-β expression in HASMCs, while pan-TGF-β-neutralizing antibody only partially decreased collagen I expression. These findings suggest that methacholine induced ECM production through β-catenin signaling and partially through TGF-β.
Collapse
Affiliation(s)
- Yating Huo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Jiawen Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Lili Guan
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Weiliang Wu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Bingpeng Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Yuqiong Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Lin Lin
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Yonger Ou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Fangfang Jiang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Luqian Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Rongchang Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| |
Collapse
|
36
|
Ojiaku CA, Cao G, Zhu W, Yoo EJ, Shumyatcher M, Himes BE, An SS, Panettieri RA. TGF-β1 Evokes Human Airway Smooth Muscle Cell Shortening and Hyperresponsiveness via Smad3. Am J Respir Cell Mol Biol 2018; 58:575-584. [PMID: 28984468 PMCID: PMC5946330 DOI: 10.1165/rcmb.2017-0247oc] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/05/2017] [Indexed: 01/10/2023] Open
Abstract
Transforming growth factor β1 (TGF-β1), a cytokine whose levels are elevated in the airways of patients with asthma, perpetuates airway inflammation and modulates airway structural cell remodeling. However, the role of TGF-β1 in excessive airway narrowing in asthma, or airway hyperresponsiveness (AHR), remains unclear. In this study, we set out to investigate the direct effects of TGF-β1 on human airway smooth muscle (HASM) cell shortening and hyperresponsiveness. The dynamics of AHR and single-cell excitation-contraction coupling were measured in human precision-cut lung slices and in isolated HASM cells using supravital microscopy and magnetic twisting cytometry, respectively. In human precision-cut lung slices, overnight treatment with TGF-β1 significantly augmented basal and carbachol-induced bronchoconstriction. In isolated HASM cells, TGF-β1 increased basal and methacholine-induced cytoskeletal stiffness in a dose- and time-dependent manner. TGF-β1-induced single-cell contraction was corroborated by concomitant increases in myosin light chain and myosin phosphatase target subunit 1 phosphorylation levels, which were attenuated by small interfering RNA-mediated knockdown of Smad3 and pharmacological inhibition of Rho kinase. Strikingly, these physiological effects of TGF-β1 occurred through a RhoA-independent mechanism, with little effect on HASM cell [Ca2+]i levels. Together, our data suggest that TGF-β1 enhances HASM excitation-contraction coupling pathways to induce HASM cell shortening and hyperresponsiveness. These findings reveal a potential link between airway injury-repair responses and bronchial hyperreactivity in asthma, and define TGF-β1 signaling as a potential target to reduce AHR in asthma.
Collapse
Affiliation(s)
- Christie A. Ojiaku
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, and
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey; and
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey; and
| | - Wanqu Zhu
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, and
| | - Edwin J. Yoo
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, and
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey; and
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven S. An
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, and
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Reynold A. Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey; and
| |
Collapse
|
37
|
Mailhot-Larouche S, Deschênes L, Gazzola M, Lortie K, Henry C, Brook BS, Morissette MC, Bossé Y. Repeated airway constrictions in mice do not alter respiratory function. J Appl Physiol (1985) 2018; 124:1483-1490. [PMID: 29470147 DOI: 10.1152/japplphysiol.01073.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
It is suggested that the frequent strain the airways undergo in asthma because of repeated airway smooth muscle (ASM)-mediated constrictions contributes to airway wall remodeling. However, the effects of repeated constrictions on airway remodeling, as well as the ensuing impact of this presumptive remodeling on respiratory mechanics, have never been investigated in subjects without asthma. In this study, we set out to determine whether repeated constrictions lead to features that are reminiscent of asthma in mice without asthma. BALB/c mice were subjected to a 30-min constriction elicited by aerosolized methacholine every other day over 6 wk. Forty-eight hours after the last constriction, the mechanics of the respiratory system was evaluated at baseline and in response to incremental doses of nebulized methacholine with the flexiVent. The whole-lung lavages, the tracheas, and the lungs were also collected to evaluate inflammation, the contractile capacity of ASM, and the structural components of the airway wall, respectively. The resistance and the compliance of the respiratory system, as well as the Newtonian resistance and the resistive and elastic properties of the lung tissue, were not affected by repeated constrictions, both at baseline and in response to methacholine. All the other examined features also remained unaltered, except the number of goblet cells in the epithelium and the number of macrophages in the whole-lung lavages, which both increased with repeated constrictions. This study demonstrates that, despite causing goblet cell hyperplasia and a mild macrophagic inflammation, repeated constrictions with methacholine do not lead to structural changes that adversely impact the physiology. NEW & NOTEWORTHY Repeated airway constrictions led to signs of remodeling that are typically observed in asthma, which neither altered respiratory mechanics nor the contractile capacity of airway smooth muscle. These findings shed light on a debate between those claiming that constrictions induce remodeling and those convinced that methacholine challenges are harmless. Insofar as our results with mice relate to humans, the findings indicate that repeated challenges with methacholine can be performed safely.
Collapse
Affiliation(s)
| | - Louis Deschênes
- Quebec Heart and Lung Institute, Université Laval , Quebec, QC , Canada
| | - Morgan Gazzola
- Quebec Heart and Lung Institute, Université Laval , Quebec, QC , Canada
| | - Katherine Lortie
- Quebec Heart and Lung Institute, Université Laval , Quebec, QC , Canada
| | - Cyndi Henry
- Quebec Heart and Lung Institute, Université Laval , Quebec, QC , Canada
| | - Bindi S Brook
- School of Mathematical Sciences, The University of Nottingham , Nottingham , United Kingdom
| | | | - Ynuk Bossé
- Quebec Heart and Lung Institute, Université Laval , Quebec, QC , Canada
| |
Collapse
|
38
|
Löfdahl A, Wenglén C, Rydell-Törmänen K, Westergren-Thorsson G, Larsson-Callerfelt AK. Effects of 5-Hydroxytryptamine Class 2 Receptor Antagonists on Bronchoconstriction and Pulmonary Remodeling Processes. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1113-1119. [PMID: 29454752 DOI: 10.1016/j.ajpath.2018.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/02/2018] [Accepted: 01/18/2018] [Indexed: 12/11/2022]
Abstract
Serotonin [5-hydroxytryptamine (5-HT)] is associated with several chronic pulmonary diseases, recognizing 5-HT2 receptor antagonists as potential inhibitors of tissue remodeling. However, the effects of 5-HT2 receptors, especially 5-HT2B receptors on airway function and remodeling, are unclear. We investigated the role of 5-HT2B receptors on airway smooth muscle contractility and remodeling processes. Murine precision-cut lung slices were pretreated with 5-HT2B receptor antagonists (EXT5, EXT9, RS 127445, and PRX 08066), as well as ketanserin (5-HT2A/2C receptor antagonist) (1, 10 μmol/L), before addition of cumulative concentrations of 5-HT to induce bronchoconstriction. Remodeling effects after treatment with 10 μmol/L 5-HT and 5-HT2 receptor antagonists were further studied in distal lung tissue by examining release of profibrotic transforming growth factor (TGF)-β1 and proliferation of human bronchial smooth muscle cells (HBSMCs). 5-HT-induced bronchoconstriction was significantly reduced by EXT5, EXT9, and ketanserin, but not by RS 127445 or PRX 08066. The 5-HT2B receptor antagonists significantly reduced TGF-β1 release. 5-HT, in combination with TGF-β1, increased proliferation of HBSMCs, a process reduced by EXT5 and EXT9. Our results indicate that EXT5 and EXT9 may relieve bronchoconstriction in murine airways and serve as an add-on effect in attenuating pulmonary remodeling by improving airway function. The antiproliferative effect on HBSMCs and the inhibition of TGF-β1 release further support a role of 5-HT2B receptors in pathologic remodeling processes.
Collapse
Affiliation(s)
- Anna Löfdahl
- Lung Biology Group, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | | | | | | | | |
Collapse
|
39
|
Shaifta Y, MacKay CE, Irechukwu N, O'Brien KA, Wright DB, Ward JPT, Knock GA. Transforming growth factor-β enhances Rho-kinase activity and contraction in airway smooth muscle via the nucleotide exchange factor ARHGEF1. J Physiol 2017; 596:47-66. [PMID: 29071730 PMCID: PMC5746525 DOI: 10.1113/jp275033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/19/2017] [Indexed: 01/16/2023] Open
Abstract
Key points Transforming growth‐factor‐β (TGF‐β) and RhoA/Rho‐kinase are independently implicated in the airway hyper‐responsiveness associated with asthma, but how these proteins interact is not fully understood. We examined the effects of pre‐treatment with TGF‐β on expression and activity of RhoA, Rho‐kinase and ARHGEF1, an activator of RhoA, as well as on bradykinin‐induced contraction, in airway smooth muscle. TGF‐β enhanced bradykinin‐induced RhoA translocation, Rho‐kinase‐dependent phosphorylation and contraction, but partially suppressed bradykinin‐induced RhoA activity (RhoA‐GTP content). TGF‐β enhanced the expression of ARHGEF1, while a small interfering RNA against ARHGEF1 and a RhoGEF inhibitor prevented the effects of TGF‐β on RhoA and Rho‐kinase activity and contraction, respectively. ARHGEF1 expression was also enhanced in airway smooth muscle from asthmatic patients and ovalbumin‐sensitized mice. ARHGEF1 is a key TGF‐β target gene, an important regulator of Rho‐kinase activity and therefore a potential therapeutic target for the treatment of asthmatic airway hyper‐responsiveness.
Abstract Transforming growth factor‐β (TGF‐β), RhoA/Rho‐kinase and Src‐family kinases (SrcFK) have independently been implicated in airway hyper‐responsiveness, but how they interact to regulate airway smooth muscle contractility is not fully understood. We found that TGF‐β pre‐treatment enhanced acute contractile responses to bradykinin (BK) in isolated rat bronchioles, and inhibitors of RhoGEFs (Y16) and Rho‐kinase (Y27632), but not the SrcFK inhibitor PP2, prevented this enhancement. In cultured human airway smooth muscle cells (hASMCs), TGF‐β pre‐treatment enhanced the protein expression of the Rho guanine nucleotide exchange factor ARHGEF1, MLC20, MYPT‐1 and the actin‐severing protein cofilin, but not of RhoA, ROCK2 or c‐Src. In hASMCs, acute treatment with BK triggered subcellular translocation of ARHGEF1 and RhoA and enhanced auto‐phosphorylation of SrcFK and phosphorylation of MYPT1 and MLC20, but induced de‐phosphorylation of cofilin. TGF‐β pre‐treatment amplified the effects of BK on RhoA translocation and MYPT1/MLC20 phosphorylation, but suppressed the effects of BK on RhoA‐GTP content, SrcFK auto‐phosphorylation and cofilin de‐phosphorylation. In hASMCs, an ARHGEF1 small interfering RNA suppressed the effects of BK and TGF‐β on RhoA‐GTP content, RhoA translocation and MYPT1 and MLC20 phosphorylation, but minimally influenced the effects of TGF‐β on cofilin expression and phosphorylation. ARHGEF1 expression was also enhanced in ASMCs of asthmatic patients and in lungs of ovalbumin‐sensitized mice. Our data indicate that TGF‐β enhances BK‐induced contraction, RhoA translocation and Rho‐kinase activity in airway smooth muscle largely via ARHGEF1, but independently of SrcFK and total RhoA‐GTP content. A role for smooth muscle ARHGEF1 in asthmatic airway hyper‐responsiveness is worthy of further investigation. Transforming growth‐factor‐β (TGF‐β) and RhoA/Rho‐kinase are independently implicated in the airway hyper‐responsiveness associated with asthma, but how these proteins interact is not fully understood. We examined the effects of pre‐treatment with TGF‐β on expression and activity of RhoA, Rho‐kinase and ARHGEF1, an activator of RhoA, as well as on bradykinin‐induced contraction, in airway smooth muscle. TGF‐β enhanced bradykinin‐induced RhoA translocation, Rho‐kinase‐dependent phosphorylation and contraction, but partially suppressed bradykinin‐induced RhoA activity (RhoA‐GTP content). TGF‐β enhanced the expression of ARHGEF1, while a small interfering RNA against ARHGEF1 and a RhoGEF inhibitor prevented the effects of TGF‐β on RhoA and Rho‐kinase activity and contraction, respectively. ARHGEF1 expression was also enhanced in airway smooth muscle from asthmatic patients and ovalbumin‐sensitized mice. ARHGEF1 is a key TGF‐β target gene, an important regulator of Rho‐kinase activity and therefore a potential therapeutic target for the treatment of asthmatic airway hyper‐responsiveness.
Collapse
Affiliation(s)
- Yasin Shaifta
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Charles E MacKay
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Nneka Irechukwu
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Katie A O'Brien
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - David B Wright
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Jeremy P T Ward
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| | - Greg A Knock
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, UK
| |
Collapse
|
40
|
Hussain M, Xu C, Lu M, Wu X, Tang L, Wu X. Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3226-3242. [PMID: 28866134 DOI: 10.1016/j.bbadis.2017.08.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/29/2017] [Indexed: 12/23/2022]
Abstract
Embryonic lung development requires reciprocal endodermal-mesodermal interactions; mediated by various signaling proteins. Wnt/β-catenin is a signaling protein that exhibits the pivotal role in lung development, injury and repair while aberrant expression of Wnt/β-catenin signaling leads to asthmatic airway remodeling: characterized by hyperplasia and hypertrophy of airway smooth muscle cells, alveolar and vascular damage goblet cells metaplasia, and deposition of extracellular matrix; resulting in decreased lung compliance and increased airway resistance. The substantial evidence suggests that Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Here, we summarized the recent advances related to the mechanistic role of Wnt/β-catenin signaling in lung development, consequences of aberrant expression or deletion of Wnt/β-catenin signaling in expansion and progression of asthmatic airway remodeling, and linking early-impaired pulmonary development and airway remodeling later in life. Finally, we emphasized all possible recent potential therapeutic significance and future prospectives, that are adaptable for therapeutic intervention to treat asthmatic airway remodeling.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| | - Chengyun Xu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China.
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| |
Collapse
|
41
|
Nhu QM, Aceves SS. Tissue Remodeling in Chronic Eosinophilic Esophageal Inflammation: Parallels in Asthma and Therapeutic Perspectives. Front Med (Lausanne) 2017; 4:128. [PMID: 28831387 PMCID: PMC5549614 DOI: 10.3389/fmed.2017.00128] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic eosinophilic inflammation is associated with tissue remodeling and fibrosis in a number of chronic T-helper 2 (Th2)-mediated diseases including eosinophilic esophagitis (EoE) and asthma. Chronic inflammation results in dysregulated tissue healing, leading to fibrosis and end organ dysfunction, manifesting clinically as irreversible airway obstruction in asthma and as esophageal rigidity, strictures, narrowing, dysmotility, dysphagia, and food impactions in EoE. Current therapies for EoE and asthma center on reducing inflammation-driven tissue remodeling and fibrosis with corticosteroids, coupled with symptomatic control and allergen avoidance. Additional control of Th2 inflammation can be achieved in select asthma patients with biologic therapies such as anti-IL-5 and anti-IL-13 antibodies, which have also been trialed in EoE. Recent molecular analysis suggests an emerging role for structural cell dysfunction, either inherited or acquired, in the pathogenesis and progression of EoE and asthma tissue remodeling. In addition, new data suggest that inflammation-independent end organ rigidity can alter structural cell function. Herein, we review emerging data and concepts for the pathogenesis of tissue remodeling and fibrosis primarily in EoE and relevant pathogenetic parallels in asthma, focusing additionally on emerging disease-specific therapies and the ability of these therapies to reduce tissue remodeling in subsets of patients.
Collapse
Affiliation(s)
- Quan M Nhu
- Scripps Translational Science Institute, The Scripps Research Institute, La Jolla, CA, United States.,Division of Gastroenterology and Hepatology, Department of Medicine, Scripps Clinic - Scripps Green Hospital, La Jolla, CA, United States.,Division of Allergy and Immunology, Department of Medicine, Scripps Clinic-Scripps Green Hospital, La Jolla, CA, United States.,Division of Allergy and Immunology, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Division of Allergy and Immunology, Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Seema S Aceves
- Division of Allergy and Immunology, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Division of Allergy and Immunology, Department of Medicine, University of California, San Diego, La Jolla, CA, United States.,Rady Children's Hospital - San Diego, San Diego, CA, United States
| |
Collapse
|
42
|
Ojiaku CA, Yoo EJ, Panettieri RA. Transforming Growth Factor β1 Function in Airway Remodeling and Hyperresponsiveness. The Missing Link? Am J Respir Cell Mol Biol 2017; 56:432-442. [PMID: 27854509 DOI: 10.1165/rcmb.2016-0307tr] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of asthma includes a complex interplay among airway inflammation, hyperresponsiveness, and remodeling. Current evidence suggests that airway structural cells, including bronchial smooth muscle cells, myofibroblasts, fibroblasts, and epithelial cells, mediate all three aspects of asthma pathogenesis. Although studies show a connection between airway remodeling and changes in bronchomotor tone, the relationship between the two remains unclear. Transforming growth factor β1 (TGF-β1), a growth factor elevated in the airway of patients with asthma, plays a role in airway remodeling and in the shortening of various airway structural cells. However, the role of TGF-β1 in mediating airway hyperresponsiveness remains unclear. In this review, we summarize the literature addressing the role of TGF-β1 in airway remodeling and shortening. Through our review, we aim to further elucidate the role of TGF-β1 in asthma pathogenesis and the link between airway remodeling and airway hyperresponsiveness in asthma and to define TGF-β1 as a potential therapeutic target for reducing asthma morbidity and mortality.
Collapse
Affiliation(s)
- Christie A Ojiaku
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Edwin J Yoo
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Reynold A Panettieri
- 2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
43
|
Kistemaker LEM, Oenema TA, Baarsma HA, Bos IST, Schmidt M, Facchinetti F, Civelli M, Villetti G, Gosens R. The PDE4 inhibitor CHF-6001 and LAMAs inhibit bronchoconstriction-induced remodeling in lung slices. Am J Physiol Lung Cell Mol Physiol 2017; 313:L507-L515. [PMID: 28596292 DOI: 10.1152/ajplung.00069.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/17/2017] [Accepted: 06/01/2017] [Indexed: 11/22/2022] Open
Abstract
Combination therapy of PDE4 inhibitors and anticholinergics induces bronchoprotection in COPD. Mechanical forces that arise during bronchoconstriction may contribute to airway remodeling. Therefore, we investigated the impact of PDE4 inhibitors and anticholinergics on bronchoconstriction-induced remodeling. Because of the different mechanism of action of PDE4 inhibitors and anticholinergics, we hypothesized functional interactions of these two drug classes. Guinea pig precision-cut lung slices were preincubated with the PDE4 inhibitors CHF-6001 or roflumilast and/or the anticholinergics tiotropium or glycopyorrolate, followed by stimulation with methacholine (10 μM) or TGF-β1 (2 ng/ml) for 48 h. The inhibitory effects on airway smooth muscle remodeling, airway contraction, and TGF-β release were investigated. Methacholine-induced protein expression of smooth muscle-myosin was fully inhibited by CHF-6001 (0.3-100 nM), whereas roflumilast (1 µM) had smaller effects. Tiotropium and glycopyrrolate fully inhibited methacholine-induced airway remodeling (0.1-30 nM). The combination of CHF-6001 and tiotropium or glycopyrrolate, in concentrations partially effective by themselves, fully inhibited methacholine-induced remodeling in combination. CHF-6001 did not affect airway closure and had limited effects on TGF-β1-induced remodeling, but rather, it inhibited methacholine-induced TGF-β release. The PDE4 inhibitor CHF-6001, and to a lesser extent roflumilast, and the LAMAs tiotropium and glycopyrrolate inhibit bronchoconstriction-induced remodeling. The combination of CHF-6001 and anticholinergics was more effective than the individual compounds. This cooperativity might be explained by the distinct mechanisms of action inhibiting TGF-β release and bronchoconstriction.
Collapse
Affiliation(s)
- Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; .,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Research and Development, Aquilo, Groningen, The Netherlands; and
| | - Tjitske A Oenema
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hoeke A Baarsma
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - I Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Fabrizio Facchinetti
- Corporate Pre-clinical Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Maurizio Civelli
- Corporate Pre-clinical Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Gino Villetti
- Corporate Pre-clinical Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
44
|
Van Dijk EM, Culha S, Menzen MH, Bidan CM, Gosens R. Elastase-Induced Parenchymal Disruption and Airway Hyper Responsiveness in Mouse Precision Cut Lung Slices: Toward an Ex vivo COPD Model. Front Physiol 2017; 7:657. [PMID: 28101062 PMCID: PMC5209351 DOI: 10.3389/fphys.2016.00657] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022] Open
Abstract
Background: COPD is a progressive lung disease characterized by emphysema and enhanced bronchoconstriction. Current treatments focused on bronchodilation can delay disease progression to some extent, but recovery or normalization of loss of lung function is impossible. Therefore, novel therapeutic targets are needed. The importance of the parenchyma in airway narrowing is increasingly recognized. In COPD, the parenchyma and extracellular matrix are altered, possibly affecting airway mechanics and enhancing bronchoconstriction. Our aim was to set up a comprehensive ex vivo Precision Cut Lung Slice (PCLS) model with a pathophysiology resembling that of COPD and integrate multiple readouts in order to study the relationship between parenchyma, airway functionality, and lung repair processes. Methods: Lungs of C57Bl/6J mice were sliced and treated ex vivo with elastase (2.5 μg/ml) or H2O2 (200 μM) for 16 h. Following treatment, parenchymal structure, airway narrowing, and gene expression levels of alveolar Type I and II cell repair were assessed. Results: Following elastase, but not H2O2 treatment, slices showed a significant increase in mean linear intercept (Lmi), reflective of emphysema. Only elastase-treated slices showed disorganization of elastin and collagen fibers. In addition, elastase treatment lowered both alveolar Type I and II marker expression, whereas H2O2 stimulation lowered alveolar Type I marker expression only. Furthermore, elastase-treated slices showed enhanced methacholine-induced airway narrowing as reflected by increased pEC50 (5.87 at basal vs. 6.50 after elastase treatment) and Emax values (47.96 vs. 67.30%), and impaired chloroquine-induced airway opening. The increase in pEC50 correlated with an increase in mean Lmi. Conclusion: Using this model, we show that structural disruption of elastin fibers leads to impaired alveolar repair, disruption of the parenchymal compartment, and altered airway biomechanics, enhancing airway contraction. This finding may have implications for COPD, as the amount of elastin fiber and parenchymal tissue disruption is associated with disease severity. Therefore, we suggest that PCLS can be used to model certain aspects of COPD pathophysiology and that the parenchymal tissue damage observed in COPD contributes to lung function decline by disrupting airway biomechanics. Targeting the parenchymal compartment may therefore be a promising therapeutic target in the treatment of COPD.
Collapse
Affiliation(s)
- Eline M Van Dijk
- Department of Molecular Pharmacology, University of GroningenGroningen, Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Sule Culha
- Department of Molecular Pharmacology, University of GroningenGroningen, Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Mark H Menzen
- Department of Molecular Pharmacology, University of GroningenGroningen, Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Cécile M Bidan
- Université Grenoble Alpes, Centre National de la Recherche Scientifique, LIPhy Grenoble, France
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of GroningenGroningen, Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| |
Collapse
|
45
|
Lilburn DML, Tatler AL, Six JS, Lesbats C, Habgood A, Porte J, Hughes-Riley T, Shaw DE, Jenkins G, Meersmann T. Investigating lung responses with functional hyperpolarized xenon-129 MRI in an ex vivo rat model of asthma. Magn Reson Med 2016; 76:1224-35. [PMID: 26507239 PMCID: PMC5026173 DOI: 10.1002/mrm.26003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 08/26/2015] [Accepted: 09/08/2015] [Indexed: 01/08/2023]
Abstract
PURPOSE Asthma is a disease of increasing worldwide importance that calls for new investigative methods. Ex vivo lung tissue is being increasingly used to study functional respiratory parameters independent of confounding systemic considerations but also to reduce animal numbers and associated research costs. In this work, a straightforward laboratory method is advanced to probe dynamic changes in gas inhalation patterns by using an ex vivo small animal ovalbumin (OVA) model of human asthma. METHODS Hyperpolarized (hp) (129) Xe was actively inhaled by the excised lungs exposed to a constant pressure differential that mimicked negative pleural cavity pressure. The method enabled hp (129) Xe MRI of airway responsiveness to intravenous methacholine (MCh) and airway challenge reversal through salbutamol. RESULTS Significant differences were demonstrated between control and OVA challenged animals on global lung hp (129) Xe gas inhalation with P < 0.05 at MCh dosages above 460 μg. Spatial mapping of the regional hp gas distribution revealed an approximately three-fold increase in heterogeneity for the asthma model organs. CONCLUSION The experimental results from this proof of concept work suggest that the ex vivo hp noble gas imaging arrangement and the applied image analysis methodology may be useful as an adjunct to current diagnostic techniques. Magn Reson Med 76:1224-1235, 2016. © 2015 The Authors. Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Collapse
Affiliation(s)
- David M L Lilburn
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Amanda L Tatler
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Joseph S Six
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Clémentine Lesbats
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Anthony Habgood
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Joanne Porte
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Theodore Hughes-Riley
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Dominick E Shaw
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Gisli Jenkins
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Thomas Meersmann
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
46
|
Hiorns JE, Bidan CM, Jensen OE, Gosens R, Kistemaker LEM, Fredberg JJ, Butler JP, Krishnan R, Brook BS. Airway and Parenchymal Strains during Bronchoconstriction in the Precision Cut Lung Slice. Front Physiol 2016; 7:309. [PMID: 27559314 PMCID: PMC4989902 DOI: 10.3389/fphys.2016.00309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/07/2016] [Indexed: 01/25/2023] Open
Abstract
The precision-cut lung slice (PCLS) is a powerful tool for studying airway reactivity, but biomechanical measurements to date have largely focused on changes in airway caliber. Here we describe an image processing tool that reveals the associated spatio-temporal changes in airway and parenchymal strains. Displacements of sub-regions within the PCLS are tracked in phase-contrast movies acquired after addition of contractile and relaxing drugs. From displacement maps, strains are determined across the entire PCLS or along user-specified directions. In a representative mouse PCLS challenged with 10(-4)M methacholine, as lumen area decreased, compressive circumferential strains were highest in the 50 μm closest to the airway lumen while expansive radial strains were highest in the region 50-100 μm from the lumen. However, at any given distance from the airway the strain distribution varied substantially in the vicinity of neighboring small airways and blood vessels. Upon challenge with the relaxant agonist chloroquine, although most strains disappeared, residual positive strains remained a long time after addition of chloroquine, predominantly in the radial direction. Taken together, these findings establish strain mapping as a new tool to elucidate local dynamic mechanical events within the constricting airway and its supporting parenchyma.
Collapse
Affiliation(s)
- Jonathan E Hiorns
- School of Mathematical Sciences, University of Nottingham Nottingham, UK
| | - Cécile M Bidan
- Laboratoire Interdisciplinaire de Physique, Centre National de la Recherche Scientifique, Université Grenoble AlpesGrenoble, France; Department of Molecular Pharmacology, University of GroningenGroningen, Netherlands; Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Oliver E Jensen
- School of Mathematics, University of Manchester Manchester, UK
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands
| | - Jeffrey J Fredberg
- Department of Environmental Health, Harvard School of Public Health Boston, MA, USA
| | - Jim P Butler
- Department of Environmental Health, Harvard School of Public Health Boston, MA, USA
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School Boston, MA, USA
| | - Bindi S Brook
- School of Mathematical Sciences, University of Nottingham Nottingham, UK
| |
Collapse
|
47
|
Eleftheriadis N, Poelman H, Leus NGJ, Honrath B, Neochoritis CG, Dolga A, Dömling A, Dekker FJ. Design of a novel thiophene inhibitor of 15-lipoxygenase-1 with both anti-inflammatory and neuroprotective properties. Eur J Med Chem 2016; 122:786-801. [PMID: 27477687 DOI: 10.1016/j.ejmech.2016.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 01/18/2023]
Abstract
The enzyme 15-lipoxygenase-1 (15-LOX-1) plays a dual role in diseases with an inflammatory component. On one hand 15-LOX-1 plays a role in pro-inflammatory gene expression and on the other hand it has been shown to be involved in central nervous system (CNS) disorders by its ability to mediate oxidative stress and damage of mitochondrial membranes under hypoxic conditions. In order to further explore applications in the CNS, novel 15-LOX-1 inhibitors with favorable physicochemical properties need to be developed. Here, we present Substitution Oriented Screening (SOS) in combination with Multi Component Chemistry (MCR) as an effective strategy to identify a diversely substituted small heterocyclic inhibitors for 15-LOX-1, denoted ThioLox, with physicochemical properties superior to previously identified inhibitors. Ex vivo biological evaluation in precision-cut lung slices (PCLS) showed inhibition of pro-inflammatory gene expression and in vitro studies on neuronal HT-22 cells showed a strong protection against glutamate toxicity for this 15-LOX-1 inhibitor. This provides a novel approach to identify novel small with favorable physicochemical properties for exploring 15-LOX-1 as a drug target in inflammatory diseases and neurodegeneration.
Collapse
Affiliation(s)
- Nikolaos Eleftheriadis
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Hessel Poelman
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Niek G J Leus
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Birgit Honrath
- Institute of Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Constantinos G Neochoritis
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Amalia Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
48
|
Kistemaker LEM, Bos IST, Menzen MH, Maarsingh H, Meurs H, Gosens R. Combination therapy of tiotropium and ciclesonide attenuates airway inflammation and remodeling in a guinea pig model of chronic asthma. Respir Res 2016; 17:13. [PMID: 26846267 PMCID: PMC4743207 DOI: 10.1186/s12931-016-0327-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/24/2016] [Indexed: 12/18/2022] Open
Abstract
Background The long-acting anticholinergic tiotropium has recently been registered for the treatment of asthma, and its use is associated with a reduction in exacerbation frequency. Anti-inflammatory and anti-remodeling effects of tiotropium have been demonstrated in in vitro and in vivo models. Because tiotropium treatment is used in combination with inhaled corticosteroids, potential additive effects between the two would be clinically relevant. Therefore, the aim of this study was to investigate additive effects between tiotropium and ciclesonide on airway inflammation and remodeling in guinea pig models of asthma. Methods Guinea pigs (n = 3–8/group) were sensitized and challenged with ovalbumin in an acute (single challenge) and a chronic model (12 weekly challenges) of allergic asthma. Animals were treated with vehicle, nebulized tiotropium (0.01–0.3 mM) and/or intranasally instilled ciclesonide (0.001–1 mg/kg) before each challenge. Bronchoalveolar lavage fluid and lungs were collected for analysis of airway inflammation and remodeling. Results Tiotropium and ciclesonide treatment, alone or in combination, did not inhibit airway inflammation in the acute asthma model. In a dose-finding study, low doses of tiotropium and ciclesonide inhibited airway eosinophilia and airway smooth muscle thickening in the chronic asthma model. Threshold doses of 0.01 mM tiotropium (nebulizer concentration) and 0.01 mg/kg ciclesonide were selected to investigate potential additive effects between both drugs. At these doses, tiotropium and ciclesonide did not inhibit airway eosinophilia or airway smooth muscle thickening when administered alone, but significantly inhibited these allergen-induced responses when administered in combination. Conclusions Combined treatment with low doses of tiotropium and ciclesonide inhibits airway inflammation and remodeling in a guinea pig model of chronic asthma, suggesting that combined treatment with anticholinergics and corticosteroids may have anti-inflammatory and anti-remodeling activity in allergic airway diseases. Since tiotropium is registered as a therapy for asthma added on to corticosteroid treatment, these beneficial effects of the combination therapy may be clinically relevant. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0327-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands. .,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - I Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Mark H Menzen
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harm Maarsingh
- Department of Pharmaceutical Sciences, Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, FL, USA
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
49
|
Li M, de Graaf IAM, Groothuis GMM. Precision-cut intestinal slices: alternative model for drug transport, metabolism, and toxicology research. Expert Opin Drug Metab Toxicol 2016; 12:175-90. [DOI: 10.1517/17425255.2016.1125882] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Ming Li
- Pharmacokinetics, Toxicology & Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Inge A. M. de Graaf
- Pharmacokinetics, Toxicology & Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Geny M. M. Groothuis
- Pharmacokinetics, Toxicology & Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
50
|
Eleftheriadis N, Neochoritis CG, Leus NGJ, van der Wouden PE, Dömling A, Dekker FJ. Rational Development of a Potent 15-Lipoxygenase-1 Inhibitor with in Vitro and ex Vivo Anti-inflammatory Properties. J Med Chem 2015; 58:7850-62. [PMID: 26331552 DOI: 10.1021/acs.jmedchem.5b01121] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human 15-lipoxygenase-1 (h-15-LOX-1) is a mammalian lipoxygenase and plays an important role in several inflammatory lung diseases such as asthma, COPD, and chronic bronchitis. Novel potent inhibitors of h-15-LOX-1 are required to explore the role of this enzyme further and to enable drug discovery efforts. In this study, we applied an approach in which we screened a fragment collection that is focused on a diverse substitution pattern of nitrogen-containing heterocycles such as indoles, quinolones, pyrazoles, and others. We denoted this approach substitution-oriented fragment screening (SOS) because it focuses on the identification of novel substitution patterns rather than on novel scaffolds. This approach enabled the identification of hits with good potency and clear structure-activity relationships (SAR) for h-1-5-LOX-1 inhibition. Molecular modeling enabled the rationalization of the observed SAR and supported structure-based design for further optimization to obtain inhibitor 14 d that binds with a Ki of 36 nM to the enzyme. In vitro and ex vivo biological evaluations of our best inhibitor demonstrate a significant increase of interleukin-10 (IL-10) gene expression, which indicates its anti-inflammatory properties.
Collapse
Affiliation(s)
- Nikolaos Eleftheriadis
- Department of Pharmaceutical Gene Modulation, ‡Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen , Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Constantinos G Neochoritis
- Department of Pharmaceutical Gene Modulation, ‡Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen , Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Niek G J Leus
- Department of Pharmaceutical Gene Modulation, ‡Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen , Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Petra E van der Wouden
- Department of Pharmaceutical Gene Modulation, ‡Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen , Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Alexander Dömling
- Department of Pharmaceutical Gene Modulation, ‡Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen , Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Frank J Dekker
- Department of Pharmaceutical Gene Modulation, ‡Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen , Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|