1
|
Magallón M, Castillo-Corullón S, Bañuls L, Romero T, Pellicer D, Herrejón A, Navarro-García MM, González C, Dasí F. Impact of Hypoxia on Neutrophil Degranulation and Inflammatory Response in Alpha-1 Antitrypsin Deficiency Patients. Antioxidants (Basel) 2024; 13:1071. [PMID: 39334730 PMCID: PMC11428696 DOI: 10.3390/antiox13091071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Alpha-1 antitrypsin deficiency (AATD) is an inflammatory disorder where neutrophils play a key role. Excessive neutrophil activation leads to local hypoxia and tissue damage. Most research on neutrophil function has been conducted under atmospheric conditions (21% O2), which may not represent physiological or pathological conditions. This study aimed to determine the effects of hypoxia on neutrophil degranulation and cytokine production in AATD patients. METHODS Neutrophils isolated from 54 AATD patients (31 MZ; 8 SZ; 15 ZZ) and 7 controls (MM) were exposed to hypoxia (1% O2) for 4 h. Neutrophil degranulation was assessed by measuring elastase (NE), myeloperoxidase (MPO), lactoferrin, and matrix metalloproteinase-9 (MMP-9) levels using immunoassay-based methods. Pro-inflammatory (IL-8, IL-1 beta, IL-6, and TNF-alpha) and anti-inflammatory (IL-4 and IL-10) cytokine levels were assessed by a Luminex-based method. RESULTS Our results indicate a significantly increased release of NE (p = 0.015), MPO (p = 0.042), lactoferrin (p = 0.015), and MMP-9 (p = 0.001) compared to controls. Pro-inflammatory cytokines show a significant rise in IL-8 (p = 0.019), a trend towards increased IL-1 beta (p = 0.3196), no change in IL-6 (p = 0.7329), and reduced TNF-alpha (p = 0.006). Anti-inflammatory cytokines show increased IL-4 (p = 0.057) and decreased IL-10 (p = 0.05703). CONCLUSIONS Increased neutrophil degranulation and inflammatory phenotype are observed in AATD neutrophils under physiological hypoxia.
Collapse
Affiliation(s)
- María Magallón
- Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (M.M.); (L.B.); (D.P.)
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
| | - Silvia Castillo-Corullón
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
- Paediatrics Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
- Department of Paediatrics, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Lucía Bañuls
- Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (M.M.); (L.B.); (D.P.)
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
| | - Teresa Romero
- Pediatrics Unit, Hospital de Manises, Avda. Generalitat Valenciana, 50, 46940 Manises, Spain;
| | - Daniel Pellicer
- Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (M.M.); (L.B.); (D.P.)
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
| | - Alberto Herrejón
- Pulmonology Unit, Hospital Universitario Doctor Peset, Avda. Gaspar Aguilar, 90, 46017 Valencia, Spain;
| | - María Mercedes Navarro-García
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
| | - Cruz González
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
- Pulmonology Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - Francisco Dasí
- Department of Physiology, School of Medicine, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain; (M.M.); (L.B.); (D.P.)
- Instituto de Investigación Sanitaria INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain; (S.C.-C.); (M.M.N.-G.); (C.G.)
| |
Collapse
|
2
|
Magallón M, Castillo-Corullón S, Bañuls L, Pellicer D, Romero T, Martínez-Ferraro C, Navarro-García MM, Herrejón A, González C, Dasí F. Hypoxia Enhances Oxidative Stress in Neutrophils from ZZ Alpha-1 Antitrypsin Deficiency Patients. Antioxidants (Basel) 2023; 12:antiox12040872. [PMID: 37107247 PMCID: PMC10135227 DOI: 10.3390/antiox12040872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is a neutrophilic inflammatory disorder that may result in local hypoxia, reactive oxygen and nitrogen species (ROS/RNS) production, and increased damage in adjacent tissues. This study aims to determine the impact of hypoxia on neutrophil oxidative stress profile in AATD patients. Neutrophils were isolated from AATD patients and control volunteers and exposed to hypoxia (1% O2 for 4 h), ROS/RNS, mitochondrial parameters, and non-enzymatic antioxidant defenses measured by flow cytometry. The expression of enzymatic antioxidant defenses was determined by qRT-PCR. Our results indicate that ZZ-AATD neutrophils produce higher amounts of hydrogen peroxide, peroxynitrite, and nitric oxide and decreased levels of the antioxidant enzymes catalase, superoxide dismutase, and glutathione reductase. Likewise, our results show a decrease in mitochondrial membrane potential, indicating that this organelle could be involved in the production of the reactive species observed. No decrease in glutathione and thiol levels were observed. The accumulation of substances with high oxidative capacity would explain the greater oxidative damage observed in proteins and lipids. In conclusion, our results indicate that, compared to MM control individuals, ZZ-AATD neutrophils show increased ROS/RNS production under hypoxic conditions opening a new rationale for using antioxidant therapies to treat the disease.
Collapse
Affiliation(s)
- María Magallón
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| | - Silvia Castillo-Corullón
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
- Pediatrics Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
- School of Medicine, Department of Paediatrics, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Lucía Bañuls
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| | - Daniel Pellicer
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| | - Teresa Romero
- Pediatrics Unit, Hospital de Manises, Avda. Generalitat Valenciana, 50, 46940 Manises, Spain
| | - Carlos Martínez-Ferraro
- Pediatrics Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | | | - Alberto Herrejón
- Pulmonology Unit, Hospital Doctor Peset, Avda. Gaspar Aguilar, 90, 46017 Valencia, Spain
| | - Cruz González
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
- Pulmonology Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez, 17, 46010 Valencia, Spain
| | - Francisco Dasí
- School of Medicine, Department of Physiology, University of Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
- IIS INCLIVA, Rare Respiratory Diseases Group, Avda. Menéndez y Pelayo, 4, 46010 Valencia, Spain
| |
Collapse
|
3
|
Kheradmand F, Zhang Y, Corry DB. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol Rev 2023; 103:1059-1093. [PMID: 36201635 PMCID: PMC9886356 DOI: 10.1152/physrev.00036.2021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/01/2023] Open
Abstract
The pathophysiology of chronic obstructive pulmonary disease (COPD) and the undisputed role of innate immune cells in this condition have dominated the field in the basic research arena for many years. Recently, however, compelling data suggesting that adaptive immune cells may also contribute to the progressive nature of lung destruction associated with COPD in smokers have gained considerable attention. The histopathological changes in the lungs of smokers can be limited to the large or small airways, but alveolar loss leading to emphysema, which occurs in some individuals, remains its most significant and irreversible outcome. Critically, however, the question of why emphysema progresses in a subset of former smokers remained a mystery for many years. The recognition of activated and organized tertiary T- and B-lymphoid aggregates in emphysematous lungs provided the first clue that adaptive immune cells may play a crucial role in COPD pathophysiology. Based on these findings from human translational studies, experimental animal models of emphysema were used to determine the mechanisms through which smoke exposure initiates and orchestrates adaptive autoreactive inflammation in the lungs. These models have revealed that T helper (Th)1 and Th17 subsets promote a positive feedback loop that activates innate immune cells, confirming their role in emphysema pathogenesis. Results from genetic studies and immune-based discoveries have further provided strong evidence for autoimmunity induction in smokers with emphysema. These new findings offer a novel opportunity to explore the mechanisms underlying the inflammatory landscape in the COPD lung and offer insights for development of precision-based treatment to halt lung destruction.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| | - Yun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
4
|
Pariano M, Puccetti M, Stincardini C, Napolioni V, Gatticchi L, Galarini R, Renga G, Barola C, Bellet MM, D'Onofrio F, Nunzi E, Bartoli A, Antognelli C, Cariani L, Russo M, Porcaro L, Colombo C, Majo F, Lucidi V, Montemitro E, Fiscarelli E, Ellemunter H, Lass-Flörl C, Ricci M, Costantini C, Giovagnoli S, Romani L. Aryl Hydrocarbon Receptor Agonism Antagonizes the Hypoxia-driven Inflammation in Cystic Fibrosis. Am J Respir Cell Mol Biol 2023; 68:288-301. [PMID: 36252182 DOI: 10.1165/rcmb.2022-0196oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hypoxia contributes to the exaggerated yet ineffective airway inflammation that fails to oppose infections in cystic fibrosis (CF). However, the potential for impairment of essential immune functions by HIF-1α (hypoxia-inducible factor 1α) inhibition demands a better comprehension of downstream hypoxia-dependent pathways that are amenable for manipulation. We assessed here whether hypoxia may interfere with the activity of AhR (aryl hydrocarbon receptor), a versatile environmental sensor highly expressed in the lungs, where it plays a homeostatic role. We used murine models of Aspergillus fumigatus infection in vivo and human cells in vitro to define the functional role of AhR in CF, evaluate the impact of hypoxia on AhR expression and activity, and assess whether AhR agonism may antagonize hypoxia-driven inflammation. We demonstrated that there is an important interferential cross-talk between the AhR and HIF-1α signaling pathways in murine and human CF, in that HIF-1α induction squelched the normal AhR response through an impaired formation of the AhR:ARNT (aryl hydrocarbon receptor nuclear translocator)/HIF-1β heterodimer. However, functional studies and analysis of the AhR genetic variability in patients with CF proved that AhR agonism could prevent hypoxia-driven inflammation, restore immune homeostasis, and improve lung function. This study emphasizes the contribution of environmental factors, such as infections, in CF disease progression and suggests the exploitation of hypoxia:xenobiotic receptor cross-talk for antiinflammatory therapy in CF.
Collapse
Affiliation(s)
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | | | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Roberta Galarini
- Istituto Zooprofilattico Sperimentale dell' Umbria e delle Marche "Togo Rosati," Perugia, Italy
| | | | - Carolina Barola
- Istituto Zooprofilattico Sperimentale dell' Umbria e delle Marche "Togo Rosati," Perugia, Italy
| | | | | | | | | | | | - Lisa Cariani
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico di Milano, Italy
| | - Maria Russo
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico di Milano, Italy
| | - Luigi Porcaro
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico di Milano, Italy
| | - Carla Colombo
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico di Milano, Italy
| | | | | | | | | | - Helmut Ellemunter
- Cystic Fibrosis Centre, Medical University Innsbruck, Innsbruck, Austria
| | | | - Maurizio Ricci
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | | | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | | |
Collapse
|
5
|
Puccetti M, Pariano M, Stincardini C, Wojtylo P, Schoubben A, Nunzi E, Ricci M, Romani L, Giovagnoli S. Pulmonary drug delivery technology enables anakinra repurposing in cystic fibrosis. J Control Release 2023; 353:1023-1036. [PMID: 36442616 DOI: 10.1016/j.jconrel.2022.11.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
Inflammation is a key pathological driver in cystic fibrosis (CF). Current therapies are ineffective in treating and preventing the escalation of inflammatory events often exacerbated by superimposed infection. In this work, we propose a novel treatment based on the pulmonary administration of anakinra, a non-glycosylated recombinant form of IL-1Ra. An inhalable dry powder of anakinra was successfully developed to meet the specific needs of lung drug delivery. The new formulation was investigated in vitro for aerodynamic performances and activity and in vivo for its pharmacological profile, including the pharmacokinetics, treatment schedule, antimicrobial and anti-inflammatory activity and systemic toxicity. The protein was structurally preserved inside the formulation and retained its pharmacological activity in vitro immediately after preparation and over time when stored at ambient conditions. Anakinra when delivered to the lungs showed an improved and extended therapeutic efficacy in CF models in vivo as well as higher potency compared to systemic delivery. Peripheral side effects were significantly reduced and correlated with lower serum levels compared to systemic treatment. These findings provide proof-of-concept demonstration for anakinra repurposing in CF through the pulmonary route.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy.
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, piazzale Lucio Severi 1, 06132 Perugia, Italy
| | - Claudia Stincardini
- Department of Medicine and Surgery, University of Perugia, piazzale Lucio Severi 1, 06132 Perugia, Italy
| | - Paulina Wojtylo
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy
| | - Aurelie Schoubben
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, piazzale Lucio Severi 1, 06132 Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, piazzale Lucio Severi 1, 06132 Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123 Perugia, Italy.
| |
Collapse
|
6
|
Scavello F, Piacentini L, Castiglione S, Zeni F, Macrì F, Casaburo M, Vinci MC, Colombo GI, Raucci A. Effects of RAGE Deletion on the Cardiac Transcriptome during Aging. Int J Mol Sci 2022; 23:ijms231911130. [PMID: 36232442 PMCID: PMC9569842 DOI: 10.3390/ijms231911130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiac aging is characterized by increased cardiomyocyte hypertrophy, myocardial stiffness, and fibrosis, which enhance cardiovascular risk. The receptor for advanced glycation end-products (RAGE) is involved in several age-related diseases. RAGE knockout (Rage−/−) mice show an acceleration of cardiac dimension changes and interstitial fibrosis with aging. This study identifies the age-associated cardiac gene expression signature induced by RAGE deletion. We analyzed the left ventricle transcriptome of 2.5-(Young), 12-(Middle age, MA), and 21-(Old) months-old female Rage−/− and C57BL/6N (WT) mice. By comparing Young, MA, and Old Rage−/− versus age-matched WT mice, we identified 122, 192, and 12 differently expressed genes, respectively. Functional inference analysis showed that RAGE deletion is associated with: (i) down-regulation of genes involved in antigen processing and presentation of exogenous antigen, adaptive immune response, and cellular responses to interferon beta and gamma in Young animals; (ii) up-regulation of genes related to fatty acid oxidation, cardiac structure remodeling and cellular response to hypoxia in MA mice; (iii) up-regulation of few genes belonging to complement activation and triglyceride biosynthetic process in Old animals. Our findings show that the age-dependent cardiac phenotype of Rage−/− mice is associated with alterations of genes related to adaptive immunity and cardiac stress pathways.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Luca Piacentini
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Federica Macrì
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Manuel Casaburo
- Animal Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Maria Cristina Vinci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Gualtiero I. Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Correspondence: (G.I.C.); (A.R.); Tel.: +39-025-800-2464 (G.I.C.); +39-025-800-2802 (A.R.); Fax: +39-025-800-2342 (G.I.C. & A.R.)
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Animal Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Correspondence: (G.I.C.); (A.R.); Tel.: +39-025-800-2464 (G.I.C.); +39-025-800-2802 (A.R.); Fax: +39-025-800-2342 (G.I.C. & A.R.)
| |
Collapse
|
7
|
Zhou W, Yu T, Hua Y, Hou Y, Ding Y, Nie H. Effects of Hypoxia on Respiratory Diseases: Perspective View of Epithelial Ion Transport. Am J Physiol Lung Cell Mol Physiol 2022; 323:L240-L250. [PMID: 35819839 DOI: 10.1152/ajplung.00065.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The balance of gas exchange and lung ventilation is essential for the maintenance of body homeostasis. There are many ion channels and transporters in respiratory epithelial cells, including epithelial sodium channel, Na,K-ATPase, cystic fibrosis transmembrane conductance regulator, and some transporters. These ion channels/transporters maintain the capacity of liquid layer on the surface of respiratory epithelial cells, and provide an immune barrier for the respiratory system to clear off foreign pathogens. However, in some harmful external environment and/or pathological conditions, the respiratory epithelium is prone to hypoxia, which would destroy the ion transport function of the epithelium and unbalance the homeostasis of internal environment, triggering a series of pathological reactions. Many respiratory diseases associated with hypoxia manifest an increased expression of hypoxia-inducible factor-1, which mediates the integrity of the epithelial barrier and affects epithelial ion transport function. It is important to study the relationship between hypoxia and ion transport function, whereas the mechanism of hypoxia-induced ion transport dysfunction in respiratory diseases is not clear. This review focuses on the relationship of hypoxia and respiratory diseases, as well as dysfunction of ion transport and tight junctions in respiratory epithelial cells under hypoxia.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Hua
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Identification of RAGE and OSM as New Prognosis Biomarkers of Severe Pneumonia. Can Respir J 2022; 2022:3854191. [PMID: 35035643 PMCID: PMC8759921 DOI: 10.1155/2022/3854191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Objective To investigate efficiency of RAGE and OSM as new prognosis biomarkers of severe pneumonia. Methods Eligible patients were classified into hypoxemia and nonhypoxemia groups. Meanwhile, the same cohort was divided into survival and nonsurvival groups after a post-hospital stay of 30 days. We analyzed risk factors for the hypoxia and death among these patients. Results Compared with nonsurvival group, significant increase was noticed in PH, lymphocyte, albumin and platelet level in survival group, while significant decline was noticed in neutrophils, RBC, hemoglobin, hematocrit, creatinine, total bilirubin, CRP, PCT, OSM, RAGE and neutrophils/lymphocyte level. Oxygenation index level was related to APACHE II, LIS, SOFA, NUTRIC score, WBC, neutrophils, lymphocyte, RAGE, and albumin level (p < 0.05). LIS, SOFA, NUTRIC score, lac, lymphocyte, platelet, BUN, total bilirubin, PCT, and OSM levels were associated with mortality rate (p < 0.05). Conclusions RAGE and OSM may serve as a new biomarker for poor prognosis in pneumonia patients.
Collapse
|
9
|
van de Veerdonk FL, Renga G, Pariano M, Bellet MM, Servillo G, Fallarino F, De Luca A, Iannitti RG, Piobbico D, Gargaro M, Manni G, D'Onofrio F, Stincardini C, Sforna L, Borghi M, Castelli M, Pieroni S, Oikonomou V, Villella VR, Puccetti M, Giovagnoli S, Galarini R, Barola C, Maiuri L, Maria Agnese DF, Cellini B, Talesa V, Dinarello CA, Costantini C, Romani L. Anakinra restores cellular proteostasis by coupling mitochondrial redox balance to autophagy. J Clin Invest 2021; 132:144983. [PMID: 34847078 PMCID: PMC8759782 DOI: 10.1172/jci144983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/24/2021] [Indexed: 12/09/2022] Open
Abstract
Autophagy selectively degrades aggregation-prone misfolded proteins caused by defective cellular proteostasis. However, the complexity of autophagy may prevent the full appreciation of how its modulation could be used as a therapeutic strategy in disease management. Here we define a molecular pathway through which recombinant interleukin-1 receptor antagonist (IL-1Ra, anakinra) affects cellular proteostasis independently from the IL-1 receptor (IL-1R1). Anakinra promoted H2O2-driven autophagy through a xenobiotic sensing pathway involving the aryl hydrocarbon receptor that, activated through the indoleamine 2,3-dioxygenase 1-kynurenine pathway, transcriptionally activates NADPH Oxidase 4 independent of the IL-1R1. By coupling the mitochondrial redox balance to autophagy, anakinra improved the dysregulated proteostasis network in murine and human cystic fibrosis. We anticipate that anakinra may represent a therapeutic option in addition to its IL-1R1 dependent anti-inflammatory properties by acting at the intersection of mitochondrial oxidative stress and autophagy with the capacity to restore conditions in which defective proteostasis leads to human disease.
Collapse
Affiliation(s)
| | - Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giuseppe Servillo
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Antonella De Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rossana G Iannitti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Danilo Piobbico
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giorgia Manni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Fiorella D'Onofrio
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luigi Sforna
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Monica Borghi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Castelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefania Pieroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Valeria R Villella
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Roberta Galarini
- Centro Sviluppo e Validazione Metodi, Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche, Perugia, Italy
| | - Carolina Barola
- Centro Sviluppo e Validazione Metodi, Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche, Perugia, Italy
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | | | - Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Vincenzo Talesa
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, Denver, United States of America
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Pariano M, Costantini C, Santarelli I, Puccetti M, Giovagnoli S, Talesa VN, Romani L, Antognelli C. Defective Glyoxalase 1 Contributes to Pathogenic Inflammation in Cystic Fibrosis. Vaccines (Basel) 2021; 9:vaccines9111311. [PMID: 34835243 PMCID: PMC8625157 DOI: 10.3390/vaccines9111311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder that affects multiple organs, although a decline in respiratory function represents the major cause of morbidity and mortality. The airways of CF patients are characterized by a chronic inflammatory state to which the receptor for advanced glycation end-products greatly contributes. Glyoxalase 1 (GLO1) is the major enzyme metabolizing methylglyoxal, a potent precursor of advanced glycation end-products. Its role in CF has never been investigated. We herein resorted to murine and human preclinical models of CF to define the contribution of GLO1 to inflammatory pathology. We found that the expression and activity of GLO1, measured by real-time PCR and Western blot or a specific spectrophotometric assay, respectively, are defective in mice and human bronchial cells from CF patients exposed to Aspergillus fumigatus, a common pathogen in CF, but could be restored upon blockade of interleukin-1 receptor signaling by anakinra in mice. This study suggests that GLO1 contributes to pathology in CF and may be potentially targeted to mitigate inflammation.
Collapse
Affiliation(s)
- Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (C.C.); (I.S.); (V.N.T.); (L.R.)
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (C.C.); (I.S.); (V.N.T.); (L.R.)
| | - Ilaria Santarelli
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (C.C.); (I.S.); (V.N.T.); (L.R.)
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.G.)
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.G.)
| | - Vincenzo N. Talesa
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (C.C.); (I.S.); (V.N.T.); (L.R.)
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (C.C.); (I.S.); (V.N.T.); (L.R.)
| | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (M.P.); (C.C.); (I.S.); (V.N.T.); (L.R.)
- Correspondence: ; Tel.:+39-075-585-8354
| |
Collapse
|
11
|
Scavello F, Tedesco CC, Castiglione S, Maciag A, Sangalli E, Veglia F, Spinetti G, Puca AA, Raucci A. Modulation of soluble receptor for advanced glycation end products isoforms and advanced glycation end products in long-living individuals. Biomark Med 2021; 15:785-796. [PMID: 34236256 DOI: 10.2217/bmm-2020-0856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/15/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Circulating levels of soluble receptor for advanced glycation end products (sRAGE) and advanced glycation end products (AGEs) correlate with aging/cardiovascular risk, which is delayed in long-living individuals (LLIs). AGEs/sRAGE isoforms (cleaved RAGE [cRAGE] and secretory RAGE [esRAGE]) ratio is a valuable marker for disease risk. Results: We evaluated circulating sRAGE isoforms, and AGEs in LLIs (n = 95; 90-105 years) and controls (n = 94; 11-89 years). cRAGE decreased with age in controls and further declined in LLIs. esRAGE increased in LLIs. AGEs rose with age in controls and decreased in LLIs that were characterized by a lower AGEs/sRAGE ratio. Notably, cRAGE and AGE/esRAGE ratio better discriminated controls from LLIs. Conclusion: circulating cRAGE could be considered a reliable marker of chronological age while esRAGE a protective factor for longevity.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology & Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Calogero C Tedesco
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology & Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Anna Maciag
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
| | - Elena Sangalli
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| | - Gaia Spinetti
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
| | - Annibale A Puca
- Cardiovascular Research Unit, IRCCS MultiMedica, Milan, 20138, Italy
- Department of Medicine, Surgery & Dentistry, 'Scuola Medica Salernitana', University of Salerno, Baronissi, 84081, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology & Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, 20138, Italy
| |
Collapse
|
12
|
Chiappalupi S, Salvadori L, Donato R, Riuzzi F, Sorci G. Hyperactivated RAGE in Comorbidities as a Risk Factor for Severe COVID-19-The Role of RAGE-RAS Crosstalk. Biomolecules 2021; 11:biom11060876. [PMID: 34204735 PMCID: PMC8231494 DOI: 10.3390/biom11060876] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
The receptor for advanced glycation-end products (RAGE) is a multiligand receptor with a role in inflammatory and pulmonary pathologies. Hyperactivation of RAGE by its ligands has been reported to sustain inflammation and oxidative stress in common comorbidities of severe COVID-19. RAGE is essential to the deleterious effects of the renin-angiotensin system (RAS), which participates in infection and multiorgan injury in COVID-19 patients. Thus, RAGE might be a major player in severe COVID-19, and appears to be a useful therapeutic molecular target in infections by SARS-CoV-2. The role of RAGE gene polymorphisms in predisposing patients to severe COVID-19 is discussed. .
Collapse
Affiliation(s)
- Sara Chiappalupi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (S.C.); (F.R.)
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy;
| | - Laura Salvadori
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy;
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Rosario Donato
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy;
- Correspondence: (R.D.); (G.S.); Tel.: +39-075-585-8258 (G.S.)
| | - Francesca Riuzzi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (S.C.); (F.R.)
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy;
- Consorzio Interuniversitario Biotecnologie (CIB), 34127 Trieste, Italy
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (S.C.); (F.R.)
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy;
- Consorzio Interuniversitario Biotecnologie (CIB), 34127 Trieste, Italy
- Centro Universitario di Ricerca Sulla Genomica Funzionale (CURGeF), University of Perugia, 06132 Perugia, Italy
- Correspondence: (R.D.); (G.S.); Tel.: +39-075-585-8258 (G.S.)
| |
Collapse
|
13
|
Scavello F, Zeni F, Milano G, Macrì F, Castiglione S, Zuccolo E, Scopece A, Pezone G, Tedesco CC, Nigro P, Degani G, Gambini E, Veglia F, Popolo L, Pompilio G, Colombo GI, Bianchi ME, Raucci A. Soluble Receptor for Advanced Glycation End-products regulates age-associated Cardiac Fibrosis. Int J Biol Sci 2021; 17:2399-2416. [PMID: 34326683 PMCID: PMC8315019 DOI: 10.7150/ijbs.56379] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
Myocardial aging increases the cardiovascular risk in the elderly. The Receptor for Advanced Glycation End-products (RAGE) is involved in age-related disorders. The soluble isoform (sRAGE) acts as a scavenger blocking the membrane-bound receptor activation. This study aims at investigating RAGE contribution to age-related cardiac remodeling. We analyzed the cardiac function of three different age groups of female Rage-/- and C57BL/6N (WT) mice: 2.5- (Young), 12- (Middle-age, MA) and 21-months (Old) old. While aging, Rage-/- mice displayed an increase in left ventricle (LV) dimensions compared to age-matched WT animals, with the main differences observed in the MA groups. Rage-/- mice showed higher fibrosis and a larger number of α-Smooth Muscle Actin (SMA)+ cells with age, along with increased expression of pro-fibrotic Transforming Growth Factor (TGF)-β1 pathway components. RAGE isoforms were undetectable in LV of WT mice, nevertheless, circulating sRAGE declined with aging and inversely associated with LV diastolic dimensions. Human cardiac fibroblasts stimulated with sRAGE exhibited a reduction in proliferation, pro-fibrotic proteins and TGF-beta Receptor 1 (TGFbR1) expression and Smad2-3 activation. Finally, sRAGE administration to MA WT animals reduced cardiac fibrosis. Hence, our work shows that RAGE associates with age-dependent myocardial changes and indicates sRAGE as an inhibitor of cardiac fibroblasts differentiation and age-dependent cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giuseppina Milano
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Federica Macrì
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Estella Zuccolo
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Alessandro Scopece
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giovanni Pezone
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | - Patrizia Nigro
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Genny Degani
- Department of Biosciences, University of Milan, Milan, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Laura Popolo
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Gualtiero I. Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Marco E. Bianchi
- Chromatin Dynamics Unit, San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| |
Collapse
|
14
|
Chiappalupi S, Salvadori L, Vukasinovic A, Donato R, Sorci G, Riuzzi F. Targeting RAGE to prevent SARS-CoV-2-mediated multiple organ failure: Hypotheses and perspectives. Life Sci 2021; 272:119251. [PMID: 33636175 PMCID: PMC7900755 DOI: 10.1016/j.lfs.2021.119251] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
A novel infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in December 2019 and declared as a global pandemic by the World Health. Approximately 15% of patients with COVID-19 progress to severe pneumonia and eventually develop acute respiratory distress syndrome (ARDS), septic shock and/or multiple organ failure with high morbidity and mortality. Evidence points towards a determinant pathogenic role of members of the renin-angiotensin system (RAS) in mediating the susceptibility, infection, inflammatory response and parenchymal injury in lungs and other organs of COVID-19 patients. The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, has important roles in pulmonary pathological states, including fibrosis, pneumonia and ARDS. RAGE overexpression/hyperactivation is essential to the deleterious effects of RAS in several pathological processes, including hypertension, chronic kidney and cardiovascular diseases, and diabetes, all of which are major comorbidities of SARS-CoV-2 infection. We propose RAGE as an additional molecular target in COVID-19 patients for ameliorating the multi-organ pathology induced by the virus and improving survival, also in the perspective of future infections by other coronaviruses.
Collapse
Affiliation(s)
- Sara Chiappalupi
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Laura Salvadori
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy
| | - Aleksandra Vukasinovic
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Rosario Donato
- Interuniversity Institute of Myology (IIM), Perugia 06132, Italy
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy; Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia 06132, Italy
| | - Francesca Riuzzi
- Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Interuniversity Institute of Myology (IIM), Perugia 06132, Italy.
| |
Collapse
|
15
|
Perkins TN, Donnell ML, Oury TD. The axis of the receptor for advanced glycation endproducts in asthma and allergic airway disease. Allergy 2021; 76:1350-1366. [PMID: 32976640 DOI: 10.1111/all.14600] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/31/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
Asthma is a generalized term that describes a scope of distinct pathologic phenotypes of variable severity, which share a common complication of reversible airflow obstruction. Asthma is estimated to affect almost 400 million people worldwide, and nearly ten percent of asthmatics have what is considered "severe" disease. The majority of moderate to severe asthmatics present with a "type 2-high" (T2-hi) phenotypic signature, which pathologically is driven by the type 2 cytokines Interleukin-(IL)-4, IL-5, and IL-13. However, "type 2-low" (T2-lo) phenotypic signatures are often associated with more severe, steroid-refractory neutrophilic asthma. A wide range of clinical and experimental studies have found that the receptor for advanced glycation endproducts (RAGE) plays a significant role in the pathogenesis of asthma and allergic airway disease (AAD). Current experimental data indicates that RAGE is a critical mediator of the type 2 inflammatory reactions which drive the development of T2-hi AAD. However, clinical studies demonstrate that increased RAGE ligands and signaling strongly correlate with asthma severity, especially in severe neutrophilic asthma. This review presents an overview of the current understandings of RAGE in asthma pathogenesis, its role as a biomarker of disease, and future implications for mechanistic studies, and potential therapeutic intervention strategies.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mason L Donnell
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Dang H, Polineni D, Pace RG, Stonebraker JR, Corvol H, Cutting GR, Drumm ML, Strug LJ, O’Neal WK, Knowles MR. Mining GWAS and eQTL data for CF lung disease modifiers by gene expression imputation. PLoS One 2020; 15:e0239189. [PMID: 33253230 PMCID: PMC7703903 DOI: 10.1371/journal.pone.0239189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Genome wide association studies (GWAS) have identified several genomic loci with candidate modifiers of cystic fibrosis (CF) lung disease, but only a small proportion of the expected genetic contribution is accounted for at these loci. We leveraged expression data from CF cohorts, and Genotype-Tissue Expression (GTEx) reference data sets from multiple human tissues to generate predictive models, which were used to impute transcriptional regulation from genetic variance in our GWAS population. The imputed gene expression was tested for association with CF lung disease severity. By comparing and combining results from alternative approaches, we identified 379 candidate modifier genes. We delved into 52 modifier candidates that showed consensus between approaches, and 28 of them were near known GWAS loci. A number of these genes are implicated in the pathophysiology of CF lung disease (e.g., immunity, infection, inflammation, HLA pathways, glycosylation, and mucociliary clearance) and the CFTR protein biology (e.g., cytoskeleton, microtubule, mitochondrial function, lipid metabolism, endoplasmic reticulum/Golgi, and ubiquitination). Gene set enrichment results are consistent with current knowledge of CF lung disease pathogenesis. HLA Class II genes on chr6, and CEP72, EXOC3, and TPPP near the GWAS peak on chr5 are most consistently associated with CF lung disease severity across the tissues tested. The results help to prioritize genes in the GWAS regions, predict direction of gene expression regulation, and identify new candidate modifiers throughout the genome for potential therapeutic development.
Collapse
Affiliation(s)
- Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Deepika Polineni
- University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Rhonda G. Pace
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Jaclyn R. Stonebraker
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Harriet Corvol
- Pediatric Pulmonary Department, Assistance Publique-Hôpitaux sde Paris (AP-HP), Hôpital Trousseau, Institut National de la Santé et la Recherche Médicale (INSERM) U938, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris 6, Paris, France
| | - Garry R. Cutting
- McKusick-Nathans Institute of Genetic Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mitchell L. Drumm
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Lisa J. Strug
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Michael R. Knowles
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
17
|
Patregnani JT, Fujiogi M, Camargo CA, Brooks BA, Hoptay CE, Mansbach JM, Teach SJ, Freishtat RJ, Hasegawa K. Serum soluble receptor for advanced glycation end-products (sRAGE) in infants with bronchiolitis: Associations with acute severity and recurrent wheeze. Clin Infect Dis 2020; 73:e2665-e2672. [PMID: 33173945 DOI: 10.1093/cid/ciaa1700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/04/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Although bronchiolitis contributes to substantial acute (e.g., intensive care use) and chronic (e.g., recurrent wheeze and infections) morbidities in young children, the pathobiology remains uncertain. We examined relations of serum soluble receptor for advanced glycation end-products (sRAGE) with acute and chronic morbidities of bronchiolitis and whether the effect of serum sRAGE on development of recurrent wheeze is mediated through acute severity. METHODS A multi-center, multi-year, prospective cohort study of infants hospitalized for bronchiolitis was analyzed. We measured serum sRAGE level at acute hospitalization and examined its association with intensive care use (use of mechanical ventilation and/or admission to intensive care unit) and development of recurrent wheeze by age 3 years. We performed causal mediation analysis to estimate indirect (mediation) and direct effects of sRAGE on recurrent wheeze. RESULTS In 886 infants with bronchiolitis, median age was 2.9 months. Overall, 15% underwent intensive care and 32% developed recurrent wheeze by age 3 years. In the multivariable model adjusting for 11 confounders, higher presenting sRAGE level was associated with significantly lower risk of intensive care use (OR for each one-log increment, 0.39; 95%CI 0.16-0.91; P=0.03) and significantly lower rate of recurrent wheeze (HR 0.58; 95%CI 0.36-0.94; P=0.03). In mediation analysis, the direct effect was significant (HR 0.60; 95%CI 0.37-0.97; P=0.04) while the indirect effect was not (P=0.30). CONCLUSIONS Serum sRAGE levels were inversely associated with acute and chronic morbidities of bronchiolitis. Effect of sRAGE on development of recurrent wheeze is potentially driven through pathways other than acute severity of bronchiolitis.
Collapse
Affiliation(s)
- Jason T Patregnani
- Division of Cardiac Critical Care Medicine, Children's National Hospital, Washington, DC, United States.,Department of Genomics and Precision Medicine, George Washington University, Washington, DC, United States.,Division of Pediatric Critical Care Medicine, Maine Medical Center, Portland, Maine; Tufts University, Medford, MA, United States
| | - Michimasa Fujiogi
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Bonnie A Brooks
- Division of Cardiac Critical Care Medicine, Children's National Hospital, Washington, DC, United States
| | - Claire E Hoptay
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, United States
| | - Jonathan M Mansbach
- Division of General Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Stephen J Teach
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, United States
| | - Robert J Freishtat
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, United States.,Division of Emergency Medicine, Children's National Hospital, Washington, DC, United States
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Abstract
Receptor for advanced glycation end products (RAGE) is an immunoglobulin-like receptor present on cell surface. RAGE binds to an array of structurally diverse ligands, acts as a pattern recognition receptor (PRR) and is expressed on cells of different origin performing different functions. RAGE ligation leads to the initiation of a cascade of signaling events and is implicated in diseases, such as inflammation, cancer, diabetes, vascular dysfunctions, retinopathy, and neurodegenerative diseases. Because of the significant involvement of RAGE in the progression of numerous diseases, RAGE signaling has been targeted through use of inhibitors and anti-RAGE antibodies as a treatment strategy and therapy. Here in this review, we have summarized the physical and physiological aspects of RAGE biology in mammalian system and the importance of targeting this molecule in the treatment of various RAGE mediated pathologies. Highlights Receptor for advanced glycation end products (RAGE) is a member of immunoglobulin superfamily of receptors and involved in many pathophysiological conditions. RAGE ligation with its ligands leads to initiation of distinct signaling cascades and activation of numerous transcription factors. Targeting RAGE signaling through inhibitors and anti-RAGE antibodies can be promising treatment strategy.
Collapse
Affiliation(s)
- Nitish Jangde
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, India.,Manipal Academy of Higher Education, Manipal, India
| | - Rashmi Ray
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, India
| | - Vivek Rai
- Laboratory of Vascular Immunology, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
19
|
Costantini C, Puccetti M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Bellet MM, Cellini B, Giovagnoli S, Romani L. Selectively targeting key inflammatory pathways in cystic fibrosis. Eur J Med Chem 2020; 206:112717. [PMID: 32823008 DOI: 10.1016/j.ejmech.2020.112717] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/10/2020] [Accepted: 07/21/2020] [Indexed: 01/04/2023]
Abstract
Cystic fibrosis (CF) is a rare genetic disorder caused by a defect in the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR), resulting in ionic imbalance of surface fluid. Although affecting multiple organs, the progressive deterioration of respiratory function by recurrent infections and chronic inflammation represents the main cause of morbidity and mortality in CF patients. The development of modulators targeting the basic defect of CFTR has represented a major breakthrough in CF therapy, but the impact on inflammation has remained enigmatic. The emerging scenario taking hold in the field points to inflammation as a major, somehow missed, therapeutic target for prevention of lung decline. Not surprisingly, the development of anti-inflammatory drugs is taking its share in the drug development pipeline. But the path is not straightforward and targeting inflammation should be balanced with the increased risk of infection. The strategy to restore the homeostatic regulation of inflammation to efficiently respond to infection while preventing lung damage needs to be based on identifying and targeting endogenous immunoregulatory pathways that are defective in CF. We herein provide an overview of anti-inflammatory drugs currently approved or under investigation in CF patients, and present our recent studies on how the knowledge on defective immune pathways in CF may translate into innovative and selective anti-inflammatory therapeutics. Through the discovery of naturally occurring molecules or their synthetic mimics, this review emphasizes the critical importance of selectively targeting key inflammatory pathways to preserve immunocompetence in CF patients.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, 06132, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Claudia Stincardini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Barbara Cellini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, Perugia, 06132, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| |
Collapse
|
20
|
Renga G, Oikonomou V, Moretti S, Stincardini C, Bellet MM, Pariano M, Bartoli A, Brancorsini S, Mosci P, Finocchi A, Rossi P, Costantini C, Garaci E, Goldstein AL, Romani L. Thymosin β4 promotes autophagy and repair via HIF-1α stabilization in chronic granulomatous disease. Life Sci Alliance 2019; 2:2/6/e201900432. [PMID: 31719116 PMCID: PMC6851533 DOI: 10.26508/lsa.201900432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
This study demonstrates that thymosin β4 stabilizes HIF-1a to promote autophagy and up-regulate genes involved in tissue and mucosal barrier protection in chronic granulomatous disease. Chronic granulomatous disease (CGD) is a genetic disorder of the NADPH oxidase characterized by increased susceptibility to infections and hyperinflammation associated with defective autophagy and increased inflammasome activation. Herein, we demonstrate that thymosin β4 (Tβ4), a g-actin sequestering peptide with multiple and diverse intracellular and extracellular activities affecting inflammation, wound healing, fibrosis, and tissue regeneration, promoted in human and murine cells noncanonical autophagy, a form of autophagy associated with phagocytosis and limited inflammation via the death-associated protein kinase 1. We further show that the hypoxia inducible factor-1 (HIF-1)α was underexpressed in CGD but normalized by Tβ4 to promote autophagy and up-regulate genes involved in mucosal barrier protection. Accordingly, inflammation and granuloma formation were impaired and survival increased in CGD mice with colitis or aspergillosis upon Tβ4 treatment or HIF-1α stabilization. Thus, the promotion of endogenous pathways of inflammation resolution through HIF-1α stabilization is druggable in CGD by Tβ4.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Paolo Mosci
- Internal Medicine, Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paolo Rossi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Rome, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, the George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
21
|
DiGiacomo JW, Gilkes DM. Tumor Hypoxia As an Enhancer of Inflammation-Mediated Metastasis: Emerging Therapeutic Strategies. Target Oncol 2019; 13:157-173. [PMID: 29423593 DOI: 10.1007/s11523-018-0555-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Metastasis is the leading cause of cancer-related deaths. Recent research has implicated tumor inflammation as a promoter of metastasis. Myeloid, lymphoid, and mesenchymal cells in the tumor microenvironment promote inflammatory signaling amongst each other and together with cancer cells to modulate sustained inflammation, which may enhance cancer invasiveness. Tumor hypoxia, a state of reduced available oxygen present in the majority of solid tumors, acts as a prognostic factor for a worse outcome and is known to have a role in tumor inflammation through the regulation of inflammatory mediator signals in both cancer and neighboring cells in the microenvironment. Multiple methods to target tumor hypoxia have been developed and tested in clinical trials, and still more are emerging as the impacts of hypoxia become better understood. These strategies include mechanistic inhibition of the hypoxia inducible factor signaling pathway and hypoxia activated pro-drugs, leading to both anti-tumor and anti-inflammatory effects. This prompts a need for further research on the prevention of hypoxia-mediated inflammation in cancer. Hypoxia-targeting strategies seem to have the most potential for therapeutic benefit when combined with traditional chemotherapy agents. This paper will serve to summarize the role of the inflammatory response in metastasis, to discuss how hypoxia can enable or enhance inflammatory signaling, and to review established and emerging strategies to target the hypoxia-inflammation-metastasis axis.
Collapse
Affiliation(s)
- Josh W DiGiacomo
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.,Breast & Ovarian Cancer Program, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Daniele M Gilkes
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Breast & Ovarian Cancer Program, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
22
|
Jia D, He Y, Zhu Q, Liu H, Zuo C, Chen G, Yu Y, Lu A. RAGE-mediated extracellular matrix proteins accumulation exacerbates HySu-induced pulmonary hypertension. Cardiovasc Res 2018; 113:586-597. [PMID: 28407046 DOI: 10.1093/cvr/cvx051] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 03/16/2017] [Indexed: 01/05/2023] Open
Abstract
Aims Extracellular matrix (ECM) proteins accumulation contributes to the progression of pulmonary arterial hypertension (PAH), a rare and fatal cardiovascular condition defined by high pulmonary arterial pressure, whether primary, idiopathic, or secondary to other causes. The receptor for advanced glycation end products (RAGE) is constitutively expressed in the lungs and plays an important role in ECM deposition. Nonetheless, the mechanisms by which RAGE mediates ECM deposition/formation in pulmonary arteries and its roles in PAH progression remain unclear. Methods and results Expression of RAGE and its activating ligands, S100/calgranulins and high mobility group box 1 (HMGB1), were increased in both human and mouse pulmonary arterial smooth muscle cells (PASMCs) under hypoxic conditions and were also strikingly upregulated in pulmonary arteries in hypoxia plus SU5416 (HySu)-induced PAH in mice. RAGE deletion alleviated pulmonary arterial pressure and restrained extracellular matrix accumulation in pulmonary arteries in HySu-induced PAH murine model. Moreover, blocking RAGE activity with a neutralizing antibody in human PASMCs, or RAGE deficiency in mouse PASMCs exposed to hypoxia, suppressed the expression of fibrotic proteins by reducing TGF-β1 expression. RAGE reconstitution in deficient mouse PASMCs restored hypoxia-stimulated TGF-β1 production via ERK1/2 and p38 MAPK pathway activation and subsequently increased ECM protein expression. Interestingly, HMGB1 acting on RAGE, not toll-like receptor 4 (TLR4), induced ECM deposition in PASMCs. Finally, in both idiopathic PAH patients and HySu-induced PAH mice, soluble RAGE (sRAGE) levels in serum were significantly elevated compared to those in controls. Conclusions Activation of RAGE facilitates the development of hypoxia-induced pulmonary hypertension by increase of ECM deposition in pulmonary arteries. Our results indicate that sRAGE may be a potential biomarker for PAH diagnosis and disease severity, and that RAGE may be a promising target for PAH treatment.
Collapse
Affiliation(s)
- Daile Jia
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijiner Road, Shanghai 200025, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 yueyang Road, Shanghai 200031, China
| | - Yuhu He
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijiner Road, Shanghai 200025, China
| | - Qian Zhu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijiner Road, Shanghai 200025, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 yueyang Road, Shanghai 200031, China
| | - Huan Liu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijiner Road, Shanghai 200025, China
| | - Caojian Zuo
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijiner Road, Shanghai 200025, China
| | - Guilin Chen
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 yueyang Road, Shanghai 200031, China
| | - Ying Yu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 yueyang Road, Shanghai 200031, China
| | - Ankang Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijiner Road, Shanghai 200025, China
| |
Collapse
|
23
|
Towards Targeting the Aryl Hydrocarbon Receptor in Cystic Fibrosis. Mediators Inflamm 2018; 2018:1601486. [PMID: 29670460 PMCID: PMC5835240 DOI: 10.1155/2018/1601486] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/05/2017] [Indexed: 01/28/2023] Open
Abstract
Tryptophan (trp) metabolism is an important regulatory component of gut mucosal homeostasis and the microbiome. Metabolic pathways targeting the trp can lead to a myriad of metabolites, of both host and microbial origins, some of which act as endogenous low-affinity ligands for the aryl hydrocarbon receptor (AhR), a cytosolic, ligand-operated transcription factor that is involved in many biological processes, including development, cellular differentiation and proliferation, xenobiotic metabolism, and the immune response. Low-level activation of AhR by endogenous ligands is beneficial in the maintenance of immune health and intestinal homeostasis. We have defined a functional node whereby certain bacteria species contribute to host/microbial symbiosis and mucosal homeostasis. A microbial trp metabolic pathway leading to the production of indole-3-aldehyde (3-IAld) by lactobacilli provided epithelial protection while inducing antifungal resistance via the AhR/IL-22 axis. In this review, we highlight the role of AhR in inflammatory lung diseases and discuss the possible therapeutic use of AhR ligands in cystic fibrosis.
Collapse
|
24
|
Matou-Nasri S, Sharaf H, Wang Q, Almobadel N, Rabhan Z, Al-Eidi H, Yahya WB, Trivilegio T, Ali R, Al-Shanti N, Ahmed N. Biological impact of advanced glycation endproducts on estrogen receptor-positive MCF-7 breast cancer cells. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2808-2820. [PMID: 28712835 DOI: 10.1016/j.bbadis.2017.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023]
Abstract
Diabetes mellitus potentiates the risk of breast cancer. We have previously described the pro-tumorigenic effects of advanced glycation endproducts (AGEs) on estrogen receptor (ER)-negative MDA-MB-231 breast cancer cell line mediated through the receptor for AGEs (RAGE). However, a predominant association between women with ER-positive breast cancer and type 2 diabetes mellitus has been reported. Therefore, we have investigated the biological impact of AGEs on ER-positive human breast cancer cell line MCF-7 using in vitro cell-based assays including cell count, migration, and invasion assays. Western blot, FACS analyses and quantitative real time-PCR were also performed. We found that AGEs at 50-100μg/mL increased MCF-7 cell proliferation and cell migration associated with an enhancement of pro-matrix metalloproteinase (MMP)-9 activity, without affecting their poor invasiveness. However, 200μg/mL AGEs inhibited MCF-7 cell proliferation through induction of apoptosis indicated by caspase-3 cleavage detected using Western blotting. A phospho-protein array analysis revealed that AGEs mainly induce the phosphorylation of extracellular-signal regulated kinase (ERK)1/2 and cAMP response element binding protein-1 (CREB1), both signaling molecules considered as key regulators of AGEs pro-tumorigenic effects. We also showed that AGEs up-regulate RAGE and ER expression at the protein and transcript levels in MCF-7 cells, in a RAGE-dependent manner after blockade of AGEs/RAGE interaction using neutralizing anti-RAGE antibody. Throughout the study, BSA had no effect on cellular processes. These findings pave the way for future studies investigating whether the exposure of AGEs-treated ER-positive breast cancer cells to estrogen could lead to a potentiation of breast cancer development and progression.
Collapse
Affiliation(s)
- Sabine Matou-Nasri
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia.
| | - Hana Sharaf
- School of Healthcare Science, Manchester Metropolitan University, Manchester, M1 5GD, United Kingdom
| | - Qiuyu Wang
- School of Healthcare Science, Manchester Metropolitan University, Manchester, M1 5GD, United Kingdom
| | - Nasser Almobadel
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Zaki Rabhan
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Hamad Al-Eidi
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Wesam Bin Yahya
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Thadeo Trivilegio
- Core Facility, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Rizwan Ali
- Core Facility, King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Nasser Al-Shanti
- School of Healthcare Science, Manchester Metropolitan University, Manchester, M1 5GD, United Kingdom
| | - Nessar Ahmed
- School of Healthcare Science, Manchester Metropolitan University, Manchester, M1 5GD, United Kingdom..
| |
Collapse
|
25
|
Oczypok EA, Perkins TN, Oury TD. All the "RAGE" in lung disease: The receptor for advanced glycation endproducts (RAGE) is a major mediator of pulmonary inflammatory responses. Paediatr Respir Rev 2017; 23:40-49. [PMID: 28416135 PMCID: PMC5509466 DOI: 10.1016/j.prrv.2017.03.012] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation endproducts (RAGE) is a pro-inflammatory pattern recognition receptor (PRR) that has been implicated in the pathogenesis of numerous inflammatory diseases. It was discovered in 1992 on endothelial cells and was named for its ability to bind advanced glycation endproducts and promote vascular inflammation in the vessels of patients with diabetes. Further studies revealed that RAGE is most highly expressed in lung tissue and spurred numerous explorations into RAGE's role in the lung. These studies have found that RAGE is an important mediator in allergic airway inflammation (AAI) and asthma, pulmonary fibrosis, lung cancer, chronic obstructive pulmonary disease (COPD), acute lung injury, pneumonia, cystic fibrosis, and bronchopulmonary dysplasia. RAGE has not yet been targeted in the lungs of paediatric or adult clinical populations, but the development of new ways to inhibit RAGE is setting the stage for the emergence of novel therapeutic agents for patients suffering from these pulmonary conditions.
Collapse
Affiliation(s)
| | | | - Tim D. Oury
- Corresponding author. Tel.: +1 412 648 9659; Fax: +1 412 648 9527
| |
Collapse
|
26
|
Desoubeaux G, Cray C. Rodent Models of Invasive Aspergillosis due to Aspergillus fumigatus: Still a Long Path toward Standardization. Front Microbiol 2017; 8:841. [PMID: 28559881 PMCID: PMC5432554 DOI: 10.3389/fmicb.2017.00841] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023] Open
Abstract
Invasive aspergillosis has been studied in laboratory by the means of plethora of distinct animal models. They were developed to address pathophysiology, therapy, diagnosis, or miscellaneous other concerns associated. However, there are great discrepancies regarding all the experimental variables of animal models, and a thorough focus on them is needed. This systematic review completed a comprehensive bibliographic analysis specifically-based on the technical features of rodent models infected with Aspergillus fumigatus. Out the 800 articles reviewed, it was shown that mice remained the preferred model (85.8% of the referenced reports), above rats (10.8%), and guinea pigs (3.8%). Three quarters of the models involved immunocompromised status, mainly by steroids (44.4%) and/or alkylating drugs (42.9%), but only 27.7% were reported to receive antibiotic prophylaxis to prevent from bacterial infection. Injection of spores (30.0%) and inhalation/deposition into respiratory airways (66.9%) were the most used routes for experimental inoculation. Overall, more than 230 distinct A. fumigatus strains were used in models. Of all the published studies, 18.4% did not mention usage of any diagnostic tool, like histopathology or mycological culture, to control correct implementation of the disease and to measure outcome. In light of these findings, a consensus discussion should be engaged to establish a minimum standardization, although this may not be consistently suitable for addressing all the specific aspects of invasive aspergillosis.
Collapse
Affiliation(s)
- Guillaume Desoubeaux
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of MiamiMiami, FL, USA.,Service de Parasitologie-Mycologie-Médecine tropicale, Centre Hospitalier Universitaire de ToursTours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR) Institut National de la Santé et de la Recherche Médicale U1100/Équipe 3, Université François-RabelaisTours, France
| | - Carolyn Cray
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of MiamiMiami, FL, USA
| |
Collapse
|
27
|
Antonelli A, Di Maggio S, Rejman J, Sanvito F, Rossi A, Catucci A, Gorzanelli A, Bragonzi A, Bianchi ME, Raucci A. The shedding-derived soluble receptor for advanced glycation endproducts sustains inflammation during acute Pseudomonas aeruginosa lung infection. Biochim Biophys Acta Gen Subj 2017; 1861:354-364. [DOI: 10.1016/j.bbagen.2016.11.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 01/07/2023]
|
28
|
Margaroli C, Tirouvanziam R. Neutrophil plasticity enables the development of pathological microenvironments: implications for cystic fibrosis airway disease. Mol Cell Pediatr 2016; 3:38. [PMID: 27868161 PMCID: PMC5136534 DOI: 10.1186/s40348-016-0066-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/04/2016] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION The pathological course of several chronic inflammatory diseases, including cystic fibrosis, chronic obstructive pulmonary disease, and rheumatoid arthritis, features an aberrant innate immune response dominated by neutrophils. In cystic fibrosis, neutrophil burden and activity of neutrophil elastase in the extracellular fluid have been identified as strong predictors of lung disease severity. REVIEW Although neutrophils are generally considered to be rigid, pre-programmed effector leukocytes, recent studies suggest extensive plasticity in how neutrophil functions unfold upon recruitment to peripheral tissues, and how they choose their ultimate fate. Indeed, upon migration to cystic fibrosis airways, neutrophils display dysregulated lifespan, metabolic activation, and altered effector and regulatory functions, consistent with profound adaptation and phenotypic reprogramming. Licensed by signals present in cystic fibrosis airway microenvironment to survive and develop these novel functions, neutrophils orchestrate, in partnership with the epithelium and with the resident microbiota, the evolution of a pathological microenvironment. This microenvironment is defined by altered proteolytic, redox, and metabolic balance and the presence of stable luminal structures in which neutrophils and microbes coexist. CONCLUSIONS The elucidation of molecular mechanisms driving neutrophil plasticity in vivo will open new treatment opportunities designed to modulate, rather than block, the crucial adaptive functions fulfilled by neutrophils. This review aims to outline emerging mechanisms of neutrophil plasticity and their participation in the building of pathological microenvironments in the context of cystic fibrosis and other diseases with similar features.
Collapse
Affiliation(s)
- Camilla Margaroli
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
- Emory + Children's Center, 2015 Uppergate Dr NE, Rm 344, Atlanta, GA, 30322-1014, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
- Emory + Children's Center, 2015 Uppergate Dr NE, Rm 344, Atlanta, GA, 30322-1014, USA.
| |
Collapse
|
29
|
Soluble Receptor for Advanced Glycation End Product Ameliorates Chronic Intermittent Hypoxia Induced Renal Injury, Inflammation, and Apoptosis via P38/JNK Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1015390. [PMID: 27688824 PMCID: PMC5027322 DOI: 10.1155/2016/1015390] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/12/2016] [Accepted: 07/25/2016] [Indexed: 01/11/2023]
Abstract
Obstructive sleep apnea (OSA) associated chronic kidney disease is mainly caused by chronic intermittent hypoxia (CIH) triggered tissue damage. Receptor for advanced glycation end product (RAGE) and its ligand high mobility group box 1 (HMGB1) are expressed on renal cells and mediate inflammatory responses in OSA-related diseases. To determine their roles in CIH-induced renal injury, soluble RAGE (sRAGE), the RAGE neutralizing antibody, was intravenously administered in a CIH model. We also evaluated the effect of sRAGE on inflammation and apoptosis. Rats were divided into four groups: (1) normal air (NA), (2) CIH, (3) CIH+sRAGE, and (4) NA+sRAGE. Our results showed that CIH accelerated renal histological injury and upregulated RAGE-HMGB1 levels involving inflammatory (NF-κB, TNF-α, and IL-6), apoptotic (Bcl-2/Bax), and mitogen-activated protein kinases (phosphorylation of P38, ERK, and JNK) signal transduction pathways, which were abolished by sRAGE but p-ERK. Furthermore, sRAGE ameliorated renal dysfunction by attenuating tubular endothelial apoptosis determined by immunofluorescence staining of CD31 and TUNEL. These findings suggested that RAGE-HMGB1 activated chronic inflammatory transduction cascades that contributed to the pathogenesis of the CIH-induced renal injury. Inhibition of RAGE ligand interaction by sRAGE provided a therapeutic potential for CIH-induced renal injury, inflammation, and apoptosis through P38 and JNK pathways.
Collapse
|
30
|
Hunt WR, Helfman BR, McCarty NA, Hansen JM. Advanced glycation end products are elevated in cystic fibrosis-related diabetes and correlate with worse lung function. J Cyst Fibros 2016; 15:681-8. [DOI: 10.1016/j.jcf.2015.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/09/2015] [Accepted: 12/14/2015] [Indexed: 11/27/2022]
|
31
|
Williams AE, Chambers RC. Neutrophils and tissue damage: is hypoxia the key to excessive degranulation? Thorax 2016; 71:977-978. [DOI: 10.1136/thoraxjnl-2016-208879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
32
|
Identification and Characterization of a Novel Aspergillus fumigatus Rhomboid Family Putative Protease, RbdA, Involved in Hypoxia Sensing and Virulence. Infect Immun 2016; 84:1866-1878. [PMID: 27068092 DOI: 10.1128/iai.00011-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/04/2016] [Indexed: 12/22/2022] Open
Abstract
Aspergillus fumigatus is the most common pathogenic mold infecting humans and a significant cause of morbidity and mortality in immunocompromised patients. In invasive pulmonary aspergillosis, A. fumigatus spores are inhaled into the lungs, undergoing germination and invasive hyphal growth. The fungus occludes and disrupts the blood vessels, leading to hypoxia and eventual tissue necrosis. The ability of this mold to adapt to hypoxia is regulated in part by the sterol regulatory element binding protein (SREBP) SrbA and the DscA to DscD Golgi E3 ligase complex critical for SREBP activation by proteolytic cleavage. Loss of the genes encoding these proteins results in avirulence. To identify novel regulators of hypoxia sensing, we screened the Neurospora crassa gene deletion library under hypoxia and identified a novel rhomboid family protease essential for hypoxic growth. Deletion of the A. fumigatus rhomboid homolog rbdA resulted in an inability to grow under hypoxia, hypersensitivity to CoCl2, nikkomycin Z, fluconazole, and ferrozine, abnormal swollen tip morphology, and transcriptional dysregulation-accurately phenocopying deletion of srbA. In vivo, rbdA deletion resulted in increased sensitivity to phagocytic killing, a reduced inflammatory Th1 and Th17 response, and strongly attenuated virulence. Phenotypic rescue of the ΔrbdA mutant was achieved by expression and nuclear localization of the N terminus of SrbA, including its HLH domain, further indicating that RbdA and SrbA act in the same signaling pathway. In summary, we have identified RbdA, a novel putative rhomboid family protease in A. fumigatus that mediates hypoxia adaptation and fungal virulence and that is likely linked to SrbA cleavage and activation.
Collapse
|
33
|
Ramasamy R, Shekhtman A, Schmidt AM. The multiple faces of RAGE--opportunities for therapeutic intervention in aging and chronic disease. Expert Opin Ther Targets 2015; 20:431-46. [PMID: 26558318 DOI: 10.1517/14728222.2016.1111873] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION This review focuses on the multi-ligand receptor of the immunoglobulin superfamily--receptor for advanced glycation endproducts (RAGE). The accumulation of the multiple ligands of RAGE in cellular stress milieux links RAGE to the pathobiology of chronic disease and natural aging. AREAS COVERED In this review, we present a discussion on the ligands of RAGE and the implications of these ligand families in disease. We review the recent literature on the role of ligand-RAGE interaction in the consequences of natural aging; the macro- and microvascular complications of diabetes; obesity and insulin resistance; autoimmune disorders and chronic inflammation; and tumors and Alzheimer's disease. We discuss the mechanisms of RAGE signaling through its intracellular binding effector molecule--the formin DIAPH1. Physicochemical evidence of how the RAGE cytoplasmic domain binds to the FH1 (formin homology 1) domain of DIAPH1, and the consequences thereof, are also reviewed. EXPERT OPINION We discuss the modalities of RAGE antagonism currently in preclinical and clinical studies. Finally, we present the rationale behind potentially targeting the RAGE cytoplasmic domain-DIAPH1 interaction as a logical strategy for therapeutic intervention in the pathological settings of chronic diseases and aging wherein RAGE ligands accumulate and signal.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| | - Alexander Shekhtman
- b Department of Chemistry , University at Albany, State University of New York , Albany , NY 12222 , USA
| | - Ann Marie Schmidt
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| |
Collapse
|
34
|
Gopal P, Gosker HR, Theije CCD, Eurlings IM, Sell DR, Monnier VM, Reynaert NL. Effect of chronic hypoxia on RAGE and its soluble forms in lungs and plasma of mice. Biochim Biophys Acta Mol Basis Dis 2015; 1852:992-1000. [PMID: 25703138 DOI: 10.1016/j.bbadis.2015.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 01/30/2015] [Accepted: 02/12/2015] [Indexed: 12/24/2022]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor. Alternative splicing and enzymatic shedding produce soluble forms that protect against damage by ligands including Advanced Glycation End products (AGEs). A link between RAGE and oxygen levels is evident from studies showing RAGE-mediated injury following hyperoxia. The effect of hypoxia on pulmonary RAGE expression and circulating sRAGE levels is however unknown. Therefore mice were exposed to chronic hypoxia for 21 d and expression of RAGE, sheddases in lungs and circulating sRAGE were determined. In addition, accumulation of AGEs in lungs and expression of the AGE detoxifying enzyme GLO1 and receptors were evaluated. In lung tissue gene expression of total RAGE, variants 1 and 3 were elevated in mice exposed to hypoxia, whereas mRAGE and sRAGE protein levels were decreased. In the hypoxic group plasma sRAGE levels were enhanced. Although the levels of pro-ADAM10 were elevated in lungs of hypoxia exposed mice, the relative amount of the active form was decreased and gelatinase activity unaffected. In the lungs, the RAGE ligand HMGB1 was decreased and of the AGEs, only LW-1 was increased by chronic hypoxia. Gene expression of AGE receptors 2 and 3 was significantly upregulated. Chronic hypoxia is associated with downregulation of pulmonary RAGE protein levels, but a relative increase in sRAGE. These alterations might be part of the adaptive and protective response mechanism to chronic hypoxia and are not associated with AGE formation except for the fluorophore LW-1 which emerges as a novel marker of tissue hypoxia.
Collapse
Affiliation(s)
- P Gopal
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - H R Gosker
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - C C de Theije
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - I M Eurlings
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands
| | - D R Sell
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - V M Monnier
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - N L Reynaert
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
35
|
McIntyre BAS, Kushwah R, Mechael R, Shapovalova Z, Alev C, Bhatia M. Innate immune response of human pluripotent stem cell-derived airway epithelium. Innate Immun 2014; 21:504-11. [PMID: 25261966 DOI: 10.1177/1753425914551074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 08/20/2014] [Indexed: 01/08/2023] Open
Abstract
The acquisition of innate immune response is requisite to having bona fide differentiation of airway epithelium. Procedures developed to differentiate lung airway from human pluripotent stem cells (hPSCs) have demonstrated anecdotal evidence for innate immune response, but an in-depth exploration of response levels is lacking. Herein, using an established method of airway epithelial generation from hPSCs, we show that hPSC-derived epithelial cells are able to up-regulate expression of TNFα, IL8 and IL1β in response to challenge with bacterial endotoxin LPS, but lack response from genes associated with innate immune response in other cell types. Further, stimulation of cells with TNF-α resulted in auto-induction of TNFα transcript, as well as cytokine responses of IL8 and IL1β. The demonstration of innate immune induction in hPSC-derived airway epithelia gives further strength to the functionality of in vitro protocols aimed at generating differentiated airway cells that can potentially be used in a translational setting. Finally, we propose that innate immune challenge of airway epithelium from human pluripotent stem cell sources be used as a robust validation of functional in vitro differentiation.
Collapse
Affiliation(s)
- Brendan A S McIntyre
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada
| | - Rahul Kushwah
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada
| | - Rami Mechael
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Zoya Shapovalova
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada
| | - Cantas Alev
- Laboratory for Early Embryogenesis, RIKEN Center for Developmental Biology (CDB), Kobe, Hyogo, Japan
| | - Mickie Bhatia
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada
| |
Collapse
|
36
|
Affiliation(s)
- Manu Jain
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | |
Collapse
|
37
|
Cell death-associated molecular-pattern molecules: inflammatory signaling and control. Mediators Inflamm 2014; 2014:821043. [PMID: 25140116 PMCID: PMC4130149 DOI: 10.1155/2014/821043] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/09/2014] [Indexed: 12/27/2022] Open
Abstract
Apoptosis, necroptosis, and pyroptosis are different cellular death programs characterized in organs and tissues as consequence of microbes infection, cell stress, injury, and chemotherapeutics exposure. Dying and death cells release a variety of self-proteins and bioactive chemicals originated from cytosol, nucleus, endoplasmic reticulum, and mitochondria. These endogenous factors are named cell death-associated molecular-pattern (CDAMP), damage-associated molecular-pattern (DAMP) molecules, and alarmins. Some of them cooperate or act as important initial or delayed inflammatory mediators upon binding to diverse membrane and cytosolic receptors coupled to signaling pathways for the activation of the inflammasome platforms and NF-κB multiprotein complexes. Current studies show that the nonprotein thiols and thiol-regulating enzymes as well as highly diffusible prooxidant reactive oxygen and nitrogen species released together in extracellular inflammatory milieu play essential role in controlling pro- and anti-inflammatory activities of CDAMP/DAMP and alarmins. Here, we provide an overview of these emerging concepts and mechanisms of triggering and maintenance of tissue inflammation under massive death of cells.
Collapse
|
38
|
McGuire JK. Hypoxia, Receptor for Advanced Glycation End Products, and Cystic Fibrosis: A Pathway to Chronic Inflammation? Am J Respir Crit Care Med 2013; 188:1280-1. [DOI: 10.1164/rccm.201310-1908ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|