1
|
Höglund N, Rossi H, Javela HM, Oikari S, Nieminen P, Mustonen AM, Airas N, Kärjä V, Mykkänen A. The amount of hyaluronic acid and airway remodelling increase with the severity of inflammation in neutrophilic equine asthma. BMC Vet Res 2024; 20:273. [PMID: 38918797 PMCID: PMC11197223 DOI: 10.1186/s12917-024-04136-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Equine asthma (EA) is a chronic lower airway inflammation that leads to structural and functional changes. Hyaluronic acid (HA) has crucial functions in the extracellular matrix homeostasis and inflammatory mediator activity. HA concentration in the lungs increases in several human airway diseases. However, its associations with naturally occurring EA and airway remodelling have not been previously studied. Our aim was to investigate the association of equine neutrophilic airway inflammation (NAI) severity, airway remodelling, and HA concentration in horses with naturally occurring EA. We hypothesised that HA concentration and airway remodelling would increase with the severity of NAI. HA concentrations of bronchoalveolar lavage fluid supernatant (SUP) and plasma of 27 neutrophilic EA horses, and 28 control horses were measured. Additionally, remodelling and HA staining intensity were assessed from endobronchial biopsies from 10 moderate NAI horses, 5 severe NAI horses, and 15 control horses. RESULTS The HA concentration in SUP was higher in EA horses compared to controls (p = 0.007). Plasma HA concentrations were not different between the groups. In the endobronchial biopsies, moderate NAI horses showed epithelial hyperplasia and inflammatory cell infiltrate, while severe NAI horses also showed fibrosis and desquamation of the epithelium. The degree of remodelling was higher in severe NAI compared to moderate NAI (p = 0.048) and controls (p = 0.016). Intense HA staining was observed in bronchial cell membranes, basement membranes, and connective tissue without significant differences between the groups. CONCLUSION The release of HA to the airway lumen increases in naturally occurring neutrophilic EA without clear changes in its tissue distribution, and significant airway remodelling only develops in severe NAI.
Collapse
Affiliation(s)
- Nina Höglund
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, FI-00014, Finland.
| | - Heini Rossi
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hanna-Maaria Javela
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, FI-00014, Finland
| | - Sanna Oikari
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
| | - Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
- Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, Joensuu, FI-80101, Finland
| | - Niina Airas
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, FI-00014, Finland
| | - Vesa Kärjä
- Department of Clinical Pathology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, FI-70210, Finland
| | - Anna Mykkänen
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
2
|
Sallee CJ, Hippensteel JA, Miller KR, Oshima K, Pham AT, Richter RP, Belperio J, Sierra YL, Schwingshackl A, Mourani PM, Schmidt EP, Sapru A, Maddux AB. Endothelial Glycocalyx Degradation Patterns in Sepsis-Associated Pediatric Acute Respiratory Distress Syndrome: A Single Center Retrospective Observational Study. J Intensive Care Med 2024; 39:277-287. [PMID: 37670670 PMCID: PMC10845819 DOI: 10.1177/08850666231200162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Sepsis-associated destruction of the pulmonary microvascular endothelial glycocalyx (EGCX) creates a vulnerable endothelial surface, contributing to the development of acute respiratory distress syndrome (ARDS). Constituents of the EGCX shed into circulation, glycosaminoglycans and proteoglycans, may serve as biomarkers of endothelial dysfunction. We sought to define the patterns of plasma EGCX degradation products in children with sepsis-associated pediatric ARDS (PARDS), and test their association with clinical outcomes. METHODS We retrospectively analyzed a prospective cohort (2018-2020) of children (≥1 month to <18 years of age) receiving invasive mechanical ventilation for acute respiratory failure for ≥72 h. Children with and without sepsis-associated PARDS were selected from the parent cohort and compared. Blood was collected at time of enrollment. Plasma glycosaminoglycan disaccharide class (heparan sulfate, chondroitin sulfate, and hyaluronan) and sulfation subtypes (heparan sulfate and chondroitin sulfate) were quantified using liquid chromatography tandem mass spectrometry. Plasma proteoglycans (syndecan-1) were measured through an immunoassay. RESULTS Among the 39 mechanically ventilated children (29 with and 10 without sepsis-associated PARDS), sepsis-associated PARDS patients demonstrated higher levels of heparan sulfate (median 639 ng/mL [interquartile range, IQR 421-902] vs 311 [IQR 228-461]) and syndecan-1 (median 146 ng/mL [IQR 32-315] vs 8 [IQR 8-50]), both p = 0.01. Heparan sulfate subtype analysis demonstrated greater proportions of N-sulfated disaccharide levels among children with sepsis-associated PARDS (p = 0.01). Increasing N-sulfated disaccharide levels by quartile were associated with severe PARDS (n = 9/29) with the highest quartile including >60% of the severe PARDS patients (test for trend, p = 0.04). Higher total heparan sulfate and N-sulfated disaccharide levels were independently associated with fewer 28-day ventilator-free days in children with sepsis-associated PARDS (all p < 0.05). CONCLUSIONS Children with sepsis-associated PARDS exhibited higher plasma levels of heparan sulfate disaccharides and syndecan-1, suggesting that EGCX degradation biomarkers may provide insights into endothelial dysfunction and PARDS pathobiology.
Collapse
Affiliation(s)
- Colin J. Sallee
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children's Hospital, Los Angeles, CA, USA
| | - Joseph A. Hippensteel
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen R. Miller
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Kaori Oshima
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Andrew T. Pham
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert P. Richter
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - John Belperio
- Department of Medicine, Division of Pulmonary Critical Care and Sleep Medicine, David Geffen School of Medicine at University of California Los Angeles and Ronald Reagan Medical Center, Los Angeles, CA, USA
| | - Yamila L. Sierra
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Andreas Schwingshackl
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children's Hospital, Los Angeles, CA, USA
| | - Peter M. Mourani
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR, USA
| | - Eric P. Schmidt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Anil Sapru
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children's Hospital, Los Angeles, CA, USA
| | - Aline B. Maddux
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
3
|
Yang T, Liu LL, Wu XH, Xue JG, He CY. Serum hyaluronic acid and procollagen III, N-terminal propeptide levels are highly associated with disease severity and predict the progression of COVID-19. Front Cell Infect Microbiol 2023; 13:1249038. [PMID: 37860066 PMCID: PMC10582934 DOI: 10.3389/fcimb.2023.1249038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/04/2023] [Indexed: 10/21/2023] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic is a rapidly evolving global emergency and continuously poses a serious threat to public health, highlighting the urgent need of identifying biomarkers for disease severity and progression. In order to early identify severe and critical patients, we retrospectively analyze the clinical characteristics and risk indicators of severe disease in patients with corona virus disease 2019 (COVID-19). Methods A total of 420 confirmed COVID-19 patients were included in the study. According to the "Diagnosis and Treatment of novel coronavirus Pneumonia (10th Edition)", the cases were divided into mild group (n = 243) and severe group (n =177). Laboratory parameters were analyzed in combination with clinical data. Results Male patients over 46 years who have smoking habits were more likely to suffer from severe COVID-19. Critically ill patients had lower lymphocyte counts and red blood cell counts, and higher white blood cell counts (P<0.05). Expectedly, serum inflammatory factors (NLR, PLR, LMR, CLR, PCT, CRP), coagulation markers (APTT, PT, TT, FIB, D-Dimer), Myocardial damage markers (hs-TNT, LDH) were significantly increased (P<0.05) in severe COVID-19 patients. Surprisedly, those patients showed obviously elevated levels of common tumor markers (ProGRP, CYFRA21-1, SCC, NSE) (P<0.05). In this case, the levels of tumor marker reflected more the condition of inflammation than the growth of tumor. More importantly, HA and PIIIN-P were highly associated with COVID-19 severity. The AUC of the ROC curve for the diagnosis of severe COVID-19 by HA and PIIIN-P was 0.826. Meanwhile, HA was positively correlated with myocardial damage markers (hs-TNT, LDH). PIIIN-P was positively correlated with myocardial damage markers (hs-TNT, LDH) and inflammatory factors (NLR, PLR, LMR, CLR, ProGRP, SCC, PCT, CRP). On the contrary, PIIIN-P was negatively correlated with pulmonary function indexes (oxygenation index and oxygen saturation of hemoglobin). Conclusion HA and PIIIN-P are highly associated with disease severity and progression of COVID-19 and can be used as new markers for the prediction of severe COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Chun Yan He
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| |
Collapse
|
4
|
McCarthy SD, Tilbury MA, Masterson CH, MacLoughlin R, González HE, Laffey JG, Wall JG, O'Toole D. Aerosol Delivery of a Novel Recombinant Modified Superoxide Dismutase Protein Reduces Oxidant Injury and Attenuates Escherichia coli Induced Lung Injury in Rats. J Aerosol Med Pulm Drug Deliv 2023; 36:246-256. [PMID: 37638822 DOI: 10.1089/jamp.2022.0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a life-threatening respiratory failure syndrome with diverse etiologies characterized by increased permeability of alveolar-capillary membranes, pulmonary edema, and acute onset hypoxemia. During the ARDS acute phase, neutrophil infiltration into the alveolar space results in uncontrolled release of reactive oxygen species (ROS) and proteases, overwhelming antioxidant defenses and causing alveolar epithelial and lung endothelial injury. Objectives: To investigate the therapeutic potential of a novel recombinant human Cu-Zn-superoxide dismutase (SOD) fusion protein in protecting against ROS injury and for aerosolized SOD delivery to treat Escherichia coli induced ARDS. Methods: Fusion proteins incorporating human Cu-Zn-SOD (hSOD1), with (pep1-hSOD1-his) and without (hSOD1-his) a fused hyaluronic acid-binding peptide, were expressed in E. coli. Purified proteins were evaluated in in vitro assays with human bronchial epithelial cells and through aerosolized delivery to the lung of an E. coli-induced ARDS rat model. Results: SOD proteins exhibited high SOD activity in vitro and protected bronchial epithelial cells from oxidative damage. hSOD1-his and pep1-hSOD1-his retained SOD activity postnebulization and exhibited no adverse effects in the rat. Pep1-hSOD1-his administered through instillation or nebulization to the lung of an E. coli-induced pneumonia rat improved arterial oxygenation and lactate levels compared to vehicle after 48 hours. Static lung compliance was improved when the pep1-hSOD1-his protein was delivered by instillation. White cell infiltration to the lung was significantly reduced by aerosolized delivery of protein, and reduction of cytokine-induced neutrophil chemoattractant-1, interferon-gamma, and interleukin 6 pro-inflammatory cytokine concentrations in bronchoalveolar lavage was observed. Conclusions: Aerosol delivery of a novel recombinant modified SOD protein reduces oxidant injury and attenuates E. coli induced lung injury in rats. The results provide a strong basis for further investigation of the therapeutic potential of hSOD1 in the treatment of ARDS.
Collapse
Affiliation(s)
- Sean D McCarthy
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Discipline of Anaesthesia, School of Medicine, University of Galway, Galway, Ireland
| | - Maura A Tilbury
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Claire H Masterson
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | | | - Héctor E González
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Discipline of Anaesthesia, School of Medicine, University of Galway, Galway, Ireland
| | - John G Laffey
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Discipline of Anaesthesia, School of Medicine, University of Galway, Galway, Ireland
| | - J Gerard Wall
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Daniel O'Toole
- SFI Centre for Medical Devices (CÚRAM), University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Discipline of Anaesthesia, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
5
|
Barnes HW, Demirdjian S, Haddock NL, Kaber G, Martinez HA, Nagy N, Karmouty-Quintana H, Bollyky PL. Hyaluronan in the pathogenesis of acute and post-acute COVID-19 infection. Matrix Biol 2023; 116:49-66. [PMID: 36750167 PMCID: PMC9899355 DOI: 10.1016/j.matbio.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) recently emerged as the cause of a global pandemic. Infection with SARS-CoV-2 can result in COVID-19 with both acute and chronic disease manifestations that continue to impact many patients long after the resolution of viral replication. There is therefore great interest in understanding the host factors that contribute to COVID-19 pathogenesis. In this review, we address the role of hyaluronan (HA), an extracellular matrix polymer with roles in inflammation and cellular metabolism, in COVID-19 and critically evaluate the hypothesis that HA promotes COVID-19 pathogenesis. We first provide a brief overview of COVID-19 infection. Then we briefly summarize the known roles of HA in airway inflammation and immunity. We then address what is known about HA and the pathogenesis of COVID-19 acute respiratory distress syndrome (COVID-19 ARDS). Next, we examine potential roles for HA in post-acute SARS-CoV-2 infection (PASC), also known as "long COVID" as well as in COVID-associated fibrosis. Finally, we discuss the potential therapeutics that target HA as a means to treat COVID-19, including the repurposed drug hymecromone (4-methylumbelliferone). We conclude that HA is a promising potential therapeutic target for the treatment of COVID-19.
Collapse
Affiliation(s)
- Henry W Barnes
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA
| | - Sally Demirdjian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA
| | - Naomi L Haddock
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA
| | - Hunter A Martinez
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA
| | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, Texas, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Beckman Center, 279 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Guan T, Zhou X, Zhou W, Lin H. Regulatory T cell and macrophage crosstalk in acute lung injury: future perspectives. Cell Death Dis 2023; 9:9. [PMID: 36646692 PMCID: PMC9841501 DOI: 10.1038/s41420-023-01310-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
Acute lung injury (ALI) describes the injury to endothelial cells in the lungs and associated vessels due to various factors. Furthermore, ALI accompanied by inflammation and thrombosis has been reported as a common complication of SARS-COV-2 infection. It is widely accepted that inflammation and the cytokine storm are main causes of ALI. Two classical anti-inflammatory cell types, regulatory T cells (Tregs) and M2 macrophages, are theoretically capable of resisting uncontrolled inflammation. Recent studies have indicated possible crosstalk between Tregs and macrophages involving their mutual activation. In this review, we discuss the current findings related to ALI pathogenesis and the role of Tregs and macrophages. In particular, we review the molecular mechanisms underlying the crosstalk between Tregs and macrophages in ALI pathogenesis. Understanding the role of Tregs and macrophages will provide the potential targets for treating ALI.
Collapse
Affiliation(s)
- Tianshu Guan
- grid.260463.50000 0001 2182 8825Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, 330006 Nanchang, Jiangxi China ,grid.260463.50000 0001 2182 8825Queen Mary university, Nanchang University, 330006 Nanchang, Jiangxi Province China
| | - Xv Zhou
- grid.260463.50000 0001 2182 8825Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, 330006 Nanchang, Jiangxi China ,grid.260463.50000 0001 2182 8825Queen Mary university, Nanchang University, 330006 Nanchang, Jiangxi Province China
| | - Wenwen Zhou
- grid.260463.50000 0001 2182 8825Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, 330006 Nanchang, Jiangxi China
| | - Hui Lin
- grid.260463.50000 0001 2182 8825Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, 330006 Nanchang, Jiangxi China
| |
Collapse
|
7
|
Joglekar MM, Nizamoglu M, Fan Y, Nemani SSP, Weckmann M, Pouwels SD, Heijink IH, Melgert BN, Pillay J, Burgess JK. Highway to heal: Influence of altered extracellular matrix on infiltrating immune cells during acute and chronic lung diseases. Front Pharmacol 2022; 13:995051. [PMID: 36408219 PMCID: PMC9669433 DOI: 10.3389/fphar.2022.995051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/19/2022] [Indexed: 10/31/2023] Open
Abstract
Environmental insults including respiratory infections, in combination with genetic predisposition, may lead to lung diseases such as chronic obstructive pulmonary disease, lung fibrosis, asthma, and acute respiratory distress syndrome. Common characteristics of these diseases are infiltration and activation of inflammatory cells and abnormal extracellular matrix (ECM) turnover, leading to tissue damage and impairments in lung function. The ECM provides three-dimensional (3D) architectural support to the lung and crucial biochemical and biophysical cues to the cells, directing cellular processes. As immune cells travel to reach any site of injury, they encounter the composition and various mechanical features of the ECM. Emerging evidence demonstrates the crucial role played by the local environment in recruiting immune cells and their function in lung diseases. Moreover, recent developments in the field have elucidated considerable differences in responses of immune cells in two-dimensional versus 3D modeling systems. Examining the effect of individual parameters of the ECM to study their effect independently and collectively in a 3D microenvironment will help in better understanding disease pathobiology. In this article, we discuss the importance of investigating cellular migration and recent advances in this field. Moreover, we summarize changes in the ECM in lung diseases and the potential impacts on infiltrating immune cell migration in these diseases. There has been compelling progress in this field that encourages further developments, such as advanced in vitro 3D modeling using native ECM-based models, patient-derived materials, and bioprinting. We conclude with an overview of these state-of-the-art methodologies, followed by a discussion on developing novel and innovative models and the practical challenges envisaged in implementing and utilizing these systems.
Collapse
Affiliation(s)
- Mugdha M. Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - YiWen Fan
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Sai Sneha Priya Nemani
- Department of Paediatric Pneumology &Allergology, University Children’s Hospital, Schleswig-Holstein, Campus Lübeck, Germany
- Epigenetics of Chronic Lung Disease, Priority Research Area Chronic Lung Diseases; Leibniz Lung Research Center Borstel; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Markus Weckmann
- Department of Paediatric Pneumology &Allergology, University Children’s Hospital, Schleswig-Holstein, Campus Lübeck, Germany
- Epigenetics of Chronic Lung Disease, Priority Research Area Chronic Lung Diseases; Leibniz Lung Research Center Borstel; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Simon D. Pouwels
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Irene H. Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Barbro N. Melgert
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, Groningen, Netherlands
| | - Janesh Pillay
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Critical Care, Groningen, Netherlands
| | - Janette K. Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, Netherlands
| |
Collapse
|
8
|
Zhao F, Barber CJ, Sammani S, Wan L, Miller BW, Furenlid LR, Li Z, Gotur DB, Barrios R, Woolfenden JM, Martin DR, Liu Z. Use of radiolabeled hyaluronic acid for preclinical assessment of inflammatory injury and acute respiratory distress syndrome. Nucl Med Biol 2022; 114-115:86-98. [PMID: 36270074 PMCID: PMC9562607 DOI: 10.1016/j.nucmedbio.2022.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 12/27/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is accompanied by a dramatic increase in lung hyaluronic acid (HA), leading to a dose-dependent reduction of pulmonary oxygenation. This pattern is associated with severe infections, such as COVID-19, and other important lung injury etiologies. HA actively participates in molecular pathways involved in the cytokine storm of COVID-19-induced ARDS. The objective of this study was to evaluate an imaging approach of radiolabeled HA for assessment of dysregulated HA deposition in mouse models with skin inflammation and lipopolysaccharide (LPS)-induced ARDS using a novel portable intensified Quantum Imaging Detector (iQID) gamma camera system. METHODS HA of 10 kDa molecular weight (HA10) was radiolabeled with 125I and 99mTc respectively to produce [125I]I-HA10 and [99mTc]Tc-HA10, followed by comparative studies on stability, in vivo biodistribution, and uptake at inflammatory skin sites in mice with 12-O-tetradecanoylphorbol-13-acetate (TPA)-inflamed ears. [99mTc]Tc-HA10 was used for iQID in vivo dynamic imaging of mice with ARDS induced by intratracheal instillation of LPS. RESULTS [99mTc]Tc-HA10 and [125I]I-HA10 had similar biodistribution and localization at inflammatory sites. [99mTc]Tc-HA10 was shown to be feasible in measuring skin injury and monitoring skin wound healing. [99mTc]Tc-HA10 dynamic pulmonary images yielded good visualization of radioactive uptake in the lungs. There was significantly increased lung uptake and slower lung washout in mice with LPS-induced ARDS than in control mice. Postmortem biodistribution measurement of [99mTc]TcHA10 (%ID/g) was 11.0 ± 3.9 vs. 1.3 ± 0.3 in the ARDS mice (n = 6) and controls (n = 6) (P < 0.001), consistent with upregulated HA expression as determined by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) staining. CONCLUSIONS [99mTc]Tc-HA10 is promising as a biomarker for evaluating HA dysregulation that contributes to pulmonary injury in ARDS. Rapid iQID imaging of [99mTc]Tc-HA10 clearance from injured lungs may provide a functional template for timely assessment and quantitative monitoring of pulmonary pathophysiology and intervention in ARDS.
Collapse
Affiliation(s)
- Fangyuan Zhao
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; Food Science and Engineering College, Qingdao Agricultural University, China
| | - Christy J Barber
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Saad Sammani
- Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Li Wan
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Brian W Miller
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; College of Optical Sciences, University of Arizona, Tucson, AZ, United States of America
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; College of Optical Sciences, University of Arizona, Tucson, AZ, United States of America
| | - Zheng Li
- Department of Radiology, Houston Methodist Hospital, Houston, TX, United States of America
| | - Deepa B Gotur
- Department of Medicine, Houston Methodist Hospital, Houston, TX, United States of America
| | - Roberto Barrios
- Department of Pathology, Houston Methodist Hospital, Houston, TX, United States of America
| | - James M Woolfenden
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America
| | - Diego R Martin
- Department of Radiology, Houston Methodist Hospital, Houston, TX, United States of America
| | - Zhonglin Liu
- Department of Medical Imaging, University of Arizona, Tucson, AZ, United States of America; Department of Radiology, Houston Methodist Hospital, Houston, TX, United States of America.
| |
Collapse
|
9
|
Kratochvil MJ, Kaber G, Demirdjian S, Cai PC, Burgener EB, Nagy N, Barlow GL, Popescu M, Nicolls MR, Ozawa MG, Regula DP, Pacheco-Navarro AE, Yang S, de Jesus Perez VA, Karmouty-Quintana H, Peters AM, Zhao B, Buja ML, Johnson PY, Vernon RB, Wight TN, Milla CE, Rogers AJ, Spakowitz AJ, Heilshorn SC, Bollyky PL. Biochemical, biophysical, and immunological characterization of respiratory secretions in severe SARS-CoV-2 infections. JCI Insight 2022; 7:152629. [PMID: 35730564 PMCID: PMC9309048 DOI: 10.1172/jci.insight.152629] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 05/10/2022] [Indexed: 01/27/2023] Open
Abstract
Thick, viscous respiratory secretions are a major pathogenic feature of COVID-19, but the composition and physical properties of these secretions are poorly understood. We characterized the composition and rheological properties (i.e., resistance to flow) of respiratory secretions collected from intubated COVID-19 patients. We found the percentages of solids and protein content were greatly elevated in COVID-19 compared with heathy control samples and closely resembled levels seen in cystic fibrosis, a genetic disease known for thick, tenacious respiratory secretions. DNA and hyaluronan (HA) were major components of respiratory secretions in COVID-19 and were likewise abundant in cadaveric lung tissues from these patients. COVID-19 secretions exhibited heterogeneous rheological behaviors, with thicker samples showing increased sensitivity to DNase and hyaluronidase treatment. In histologic sections from these same patients, we observed increased accumulation of HA and the hyaladherin versican but reduced tumor necrosis factor-stimulated gene-6 staining, consistent with the inflammatory nature of these secretions. Finally, we observed diminished type I interferon and enhanced inflammatory cytokines in these secretions. Overall, our studies indicated that increases in HA and DNA in COVID-19 respiratory secretion samples correlated with enhanced inflammatory burden and suggested that DNA and HA may be viable therapeutic targets in COVID-19 infection.
Collapse
Affiliation(s)
- Michael J. Kratochvil
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.,Department of Materials Science and Engineering and
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Sally Demirdjian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Pamela C. Cai
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | | | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Graham L. Barlow
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Medeea Popescu
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Mark R. Nicolls
- Department of Pulmonology, Allergy and Critical Care Medicine
| | | | | | | | - Samuel Yang
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology;,Divisions of Critical Care Medicine and Pulmonary and Sleep Medicine, Department of Internal Medicine
| | | | - Bihong Zhao
- Department of Pathology and Laboratory Medicine; and,Department of Internal Medicine, University of Texas Health Science Center — McGovern Medical School, Houston, Texas, USA
| | - Maximilian L. Buja
- Department of Pathology and Laboratory Medicine; and,Department of Internal Medicine, University of Texas Health Science Center — McGovern Medical School, Houston, Texas, USA
| | - Pamela Y. Johnson
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Robert B. Vernon
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington, USA
| | | | - Carlos E. Milla
- Center for Excellence in Pulmonary Biology, Department of Pediatrics
| | | | - Andrew J. Spakowitz
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | | | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
10
|
Crotty KM, Yeligar SM. Hyaladherins May be Implicated in Alcohol-Induced Susceptibility to Bacterial Pneumonia. Front Immunol 2022; 13:865522. [PMID: 35634317 PMCID: PMC9133445 DOI: 10.3389/fimmu.2022.865522] [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: 01/30/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Although the epidemiology of bacterial pneumonia and excessive alcohol use is well established, the mechanisms by which alcohol induces risk of pneumonia are less clear. Patterns of alcohol misuse, termed alcohol use disorders (AUD), affect about 15 million people in the United States. Compared to otherwise healthy individuals, AUD increase the risk of respiratory infections and acute respiratory distress syndrome (ARDS) by 2-4-fold. Levels and fragmentation of hyaluronic acid (HA), an extracellular glycosaminoglycan of variable molecular weight, are increased in chronic respiratory diseases, including ARDS. HA is largely involved in immune-assisted wound repair and cell migration. Levels of fragmented, low molecular weight HA are increased during inflammation and decrease concomitant with leukocyte levels following injury. In chronic respiratory diseases, levels of fragmented HA and leukocytes remain elevated, inflammation persists, and respiratory infections are not cleared efficiently, suggesting a possible pathological mechanism for prolonged bacterial pneumonia. However, the role of HA in alcohol-induced immune dysfunction is largely unknown. This mini literature review provides insights into understanding the role of HA signaling in host immune defense following excessive alcohol use. Potential therapeutic strategies to mitigate alcohol-induced immune suppression in bacterial pneumonia and HA dysregulation are also discussed.
Collapse
Affiliation(s)
- Kathryn M Crotty
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States.,Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Samantha M Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States.,Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| |
Collapse
|
11
|
Rosser JI, Nagy N, Goel R, Kaber G, Demirdjian S, Saxena J, Bollyky JB, Frymoyer AR, Pacheco-Navarro AE, Burgener EB, Rajadas J, Wang Z, Arbach O, Dunn CE, Kalinowski A, Milla CE, Bollyky PL. Oral hymecromone decreases hyaluronan in human study participants. J Clin Invest 2022; 132:e157983. [PMID: 35499083 PMCID: PMC9057598 DOI: 10.1172/jci157983] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDHyaluronan (HA), an extracellular matrix glycosaminoglycan, has been implicated in the pathophysiology of COVID-19 infection, pulmonary hypertension, pulmonary fibrosis, and other diseases, but is not targeted by any approved drugs. We asked whether hymecromone (4-methylumbelliferone [4-MU]), an oral drug approved in Europe for biliary spasm treatment that also inhibits HA in vitro and in animal models, could be repurposed as an inhibitor of HA synthesis in humans.METHODSWe conducted an open-label, single-center, dose-response study of hymecromone in healthy adults. Subjects received hymecromone at 1200 (n = 8), 2400 (n = 9), or 3600 (n = 9) mg/d divided into 3 doses daily, administered orally for 4 days. We assessed safety and tolerability of hymecromone and analyzed HA, 4-MU, and 4-methylumbelliferyl glucuronide (4-MUG; the main metabolite of 4-MU) concentrations in sputum and serum.RESULTSHymecromone was well tolerated up to doses of 3600 mg/d. Both sputum and serum drug concentrations increased in a dose-dependent manner, indicating that higher doses lead to greater exposures. Across all dose arms combined, we observed a significant decrease in sputum HA from baseline after 4 days of treatment. We also observed a decrease in serum HA. Additionally, higher baseline sputum HA levels were associated with a greater decrease in sputum HA.CONCLUSIONAfter 4 days of exposure to oral hymecromone, healthy human subjects experienced a significant reduction in sputum HA levels, indicating this oral therapy may have potential in pulmonary diseases where HA is implicated in pathogenesis.TRIAL REGISTRATIONClinicalTrials.gov NCT02780752.FUNDINGStanford Medicine Catalyst, Stanford SPARK, Stanford Innovative Medicines Accelerator program, NIH training grants 5T32AI052073-14 and T32HL129970.
Collapse
Affiliation(s)
- Joelle I. Rosser
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Riya Goel
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Sally Demirdjian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | - Jamie Saxena
- Division of Infectious Diseases, Department of Pediatrics
| | | | | | | | | | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute & Pulmonary and Critical Care, Department of Medicine, Stanford University, Stanford, California, USA
- Bioengineering and Therapeutic Sciences, UCSF School of Pharmacy, San Francisco, California, USA
| | - Zhe Wang
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute & Pulmonary and Critical Care, Department of Medicine, Stanford University, Stanford, California, USA
| | - Olga Arbach
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Colleen E. Dunn
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, and
| | - Anissa Kalinowski
- Department of Epidemiology, Stanford University, Stanford, California, USA
| | - Carlos E. Milla
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, and
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| |
Collapse
|
12
|
Albtoush N, Petrey AC. The role of Hyaluronan synthesis and degradation in the critical respiratory illness COVID-19. Am J Physiol Cell Physiol 2022; 322:C1037-C1046. [PMID: 35442830 PMCID: PMC9126216 DOI: 10.1152/ajpcell.00071.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyaluronan (HA) is a polysaccharide found in all tissues as an integral component of the extracellular matrix (ECM) that plays a central regulatory role in inflammation. In fact, HA matrices are increasingly considered as a barometer of inflammation. A number of proteins specifically recognize the HA structure and these interactions modify cell behavior and control the stability of the ECM. Moreover, inflamed airways are remarkably rich with HA and are associated with various inflammatory diseases including cystic fibrosis, influenza, sepsis, and more recently coronavirus disease 2019 (COVID-19). COVID-19 is a worldwide pandemic caused by a novel coronavirus called SARS-CoV-2, and infected individuals have a wide range of disease manifestations ranging from asymptomatic to severe illness. Critically ill COVID-19 patient cases are frequently complicated by development of acute respiratory distress syndrome (ARDS), which typically leads to poor outcomes with high mortality rate. In general, ARDS is characterized by poor oxygenation accompanied with severe lung inflammation, damage, and vascular leakage and has been suggested to be linked to an accumulation of HA within the airways. Here, we provide a succinct overview of known inflammatory mechanisms regulated by HA in general, and those both observed and postulated in critically ill patients with COVID-19.
Collapse
Affiliation(s)
- Nansy Albtoush
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States
| | - Aaron C Petrey
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States.,Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
13
|
Kratochvil MJ, Kaber G, Demirdjian S, Cai PC, Burgener EB, Nagy N, Barlow GL, Popescu M, Nicolls MR, Ozawa MG, Regula DP, Pacheco-navarro AE, Yang S, de Jesus Perez VA, Karmouty-quintana H, Peters AM, Zhao B, Buja ML, Johnson PY, Vernon RB, Wight TN, Milla CE, Rogers AJ, Spakowitz AJ, Heilshorn SC, Bollyky PL, Stanford COVID-19 Biobank Study Group. Biochemical, Biophysical, and Immunological Characterization of Respiratory Secretions in Severe SARS-CoV-2 (COVID-19) Infections.. [PMID: 35411348 PMCID: PMC8996635 DOI: 10.1101/2022.03.28.22272848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Thick, viscous respiratory secretions are a major pathogenic feature of COVID-19 disease, but the composition and physical properties of these secretions are poorly understood. We characterized the composition and rheological properties (i.e. resistance to flow) of respiratory secretions collected from intubated COVID-19 patients. We find the percent solids and protein content are greatly elevated in COVID-19 compared to heathy control samples and closely resemble levels seen in cystic fibrosis, a genetic disease known for thick, tenacious respiratory secretions. DNA and hyaluronan (HA) are major components of respiratory secretions in COVID-19 and are likewise abundant in cadaveric lung tissues from these patients. COVID-19 secretions exhibit heterogeneous rheological behaviors with thicker samples showing increased sensitivity to DNase and hyaluronidase treatment. In histologic sections from these same patients, we observe increased accumulation of HA and the hyaladherin versican but reduced tumor necrosis factor–stimulated gene-6 (TSG6) staining, consistent with the inflammatory nature of these secretions. Finally, we observed diminished type I interferon and enhanced inflammatory cytokines in these secretions. Overall, our studies indicate that increases in HA and DNA in COVID-19 respiratory secretion samples correlate with enhanced inflammatory burden and suggest that DNA and HA may be viable therapeutic targets in COVID-19 infection.
Collapse
|
14
|
Hymecromone: a clinical prescription hyaluronan inhibitor for efficiently blocking COVID-19 progression. Signal Transduct Target Ther 2022; 7:91. [PMID: 35304437 PMCID: PMC8931182 DOI: 10.1038/s41392-022-00952-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/18/2022] [Accepted: 03/03/2022] [Indexed: 01/08/2023] Open
Abstract
Currently, there is no effective drugs for treating clinically COVID-19 except dexamethasone. We previously revealed that human identical sequences of SARS-CoV-2 promote the COVID-19 progression by upregulating hyaluronic acid (HA). As the inhibitor of HA synthesis, hymecromone is an approved prescription drug used for treating biliary spasm. Here, we aimed to investigate the relation between HA and COVID-19, and evaluate the therapeutic effects of hymecromone on COVID-19. Firstly, HA was closely relevant to clinical parameters, including lymphocytes (n = 158; r = −0.50; P < 0.0001), C-reactive protein (n = 156; r = 0.55; P < 0.0001), D-dimer (n = 154; r = 0.38; P < 0.0001), and fibrinogen (n = 152; r = 0.37; P < 0.0001), as well as the mass (n = 78; r = 0.43; P < 0.0001) and volume (n = 78; r = 0.41; P = 0.0002) of ground-glass opacity, the mass (n = 78; r = 0.48; P < 0.0001) and volume (n = 78; r = 0.47; P < 0.0001) of consolidation in patient with low level of hyaluronan (HA < 48.43 ng/mL). Furthermore, hyaluronan could directly cause mouse pulmonary lesions. Besides, hymecromone remarkably reduced HA via downregulating HAS2/HAS3 expression. Moreover, 89% patients with hymecromone treatment had pulmonary lesion absorption while only 42% patients in control group had pulmonary lesion absorption (P < 0.0001). In addition, lymphocytes recovered more quickly in hymecromone-treated patients (n = 8) than control group (n = 5) (P < 0.05). These findings suggest that hymecromone is a promising drug for COVID-19 and deserves our further efforts to determine its effect in a larger cohort.
Collapse
|
15
|
Queisser KA, Mellema RA, Middleton EA, Portier I, Manne BK, Denorme F, Beswick EJ, Rondina MT, Campbell RA, Petrey AC. COVID-19 generates hyaluronan fragments that directly induce endothelial barrier dysfunction. JCI Insight 2021; 6:147472. [PMID: 34314391 PMCID: PMC8492325 DOI: 10.1172/jci.insight.147472] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
Vascular injury has emerged as a complication contributing to morbidity in coronavirus disease 2019 (COVID-19). The glycosaminoglycan hyaluronan (HA) is a major component of the glycocalyx, a protective layer of glycoconjugates that lines the vascular lumen and regulates key endothelial cell functions. During critical illness, as in the case of sepsis, enzymes degrade the glycocalyx, releasing fragments with pathologic activities into circulation and thereby exacerbating disease. Here, we analyzed levels of circulating glycosaminoglycans in 46 patients with COVID-19 ranging from moderate to severe clinical severity and measured activities of corresponding degradative enzymes. This report provides evidence that the glycocalyx becomes significantly damaged in patients with COVID-19 and corresponds with severity of disease. Circulating HA fragments and hyaluronidase, 2 signatures of glycocalyx injury, strongly associate with sequential organ failure assessment scores and with increased inflammatory cytokine levels in patients with COVID-19. Pulmonary microvascular endothelial cells exposed to COVID-19 milieu show dysregulated HA biosynthesis and degradation, leading to production of pathological HA fragments that are released into circulation. Finally, we show that HA fragments present at high levels in COVID-19 patient plasma can directly induce endothelial barrier dysfunction in a ROCK- and CD44-dependent manner, indicating a role for HA in the vascular pathology of COVID-19.
Collapse
Affiliation(s)
| | | | - Elizabeth A. Middleton
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Division of General Internal Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Bhanu Kanth Manne
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Ellen J. Beswick
- Department of Pathology and
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Matthew T. Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Pathology and
- Division of General Internal Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Geriatric Research, Education, and Clinical Center and
- Department of Internal Medicine, George E. Wahlen Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Aaron C. Petrey
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Pathology and
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
16
|
Che Ani AA, Mohd Akhir S, Chiang Li-Xue W, Shahidan NZ, Abd Hamid A. Diffuse alveolar haemorrhage and myocardial infarction: life-threatening effects from self-injected hyaluronic acid dermal filler. Monaldi Arch Chest Dis 2021; 91. [PMID: 34296835 DOI: 10.4081/monaldi.2021.1845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022] Open
Abstract
Hyaluronic acid (HA) is a widely used dermal filler for soft tissue augmentation. We described a case of a 38-year-old transwoman who presented with sudden onset of severe respiratory distress following self-injection of HA dermal filler. She developed multiple episodes of pulmonary haemorrhage, and her chest X-ray showed diffuse ground-glass opacities consistent with diffuse alveolar haemorrhage (DAH). There were no relevant drugs or past medical histories. Anti-nuclear antibodies and rheumatoid factor were negative. Initially, the pulmonary haemorrhage episodes and ventilation requirement improved with systemic steroid, however she subsequently developed acute myocardial infarction with progressive clinical deterioration leading to death. To the best of our knowledge, this is the first HA-related DAH with myocardial infarction reported with a fatal outcome. This case highlights the importance of awareness and the necessity of having a high suspicion of DAH in patients with history of illicit HA dermal filler use.
Collapse
Affiliation(s)
- Amira Aishah Che Ani
- Department of Anaesthesiology and Critical Care, Hospital Tuanku Fauziah, Perlis, Ministry of Health.
| | - Sakinah Mohd Akhir
- Department of Anaesthesiology and Critical Care, Hospital Tuanku Fauziah, Perlis, Ministry of Health.
| | - Winnie Chiang Li-Xue
- Department of Anaesthesiology and Critical Care, Hospital Tuanku Fauziah, Perlis, Ministry of Health.
| | - Nur Zurairah Shahidan
- Department of Anaesthesiology and Critical Care, Hospital Tuanku Fauziah, Perlis, Ministry of Health.
| | - Azman Abd Hamid
- Department of Anaesthesiology and Critical Care, Hospital Tuanku Fauziah, Perlis, Ministry of Health.
| |
Collapse
|
17
|
Mosleh G, Badr P, Zaeri M, Mohagheghzadeh A. Potentials of Antitussive Traditional Persian Functional Foods for COVID-19 Therapy †. Front Pharmacol 2021; 12:624006. [PMID: 34335237 PMCID: PMC8322585 DOI: 10.3389/fphar.2021.624006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 06/21/2021] [Indexed: 01/22/2023] Open
Abstract
Coronavirus disease 2019 is a worldwide pandemic resulting in a severe acute respiratory syndrome. Remdesivir is the only FDA-approved drug for hospitalized patients older than age 12. It shows the necessity of finding new therapeutic strategies. Functional foods (FFs) could have co-therapeutic and protective effects against COVID-19 infection. Traditional Persian medicine (TPM), one of the safest and most popular schools of medicine for hundreds of years, has recommended potential FF candidates to manage such a global pandemic. To reveal the potential of TPM in terms of antitussive FFs, traditional Persian pharmacopoeia "Qarabadin-e-Salehi" was searched using the keywords "Soaal" and "Sorfeh." Also, a search of MEDLINE, PubMed Central, Google Scholar, and Science Direct was performed for the relevant literature published from the inception up to March 2021. A combination of search terms including "cough, antitussive, antioxidant, anti-inflammation, antiviral, COVID-19, mucoactive, mucolytic, expectorant, and mucoregulatory" was also applied. The potential mechanism of action in SARS-CoV-2 infection was discussed. Twelve TPM FFs were found including Laooqs, Morabbas, a Saviq, a soup, and a syrup. They are combinations of two to seven ingredients. Natural compounds of mentioned formulations have the main pharmacological mechanisms including antiviral, anti-inflammatory, antioxidant, antihistamine, bronchodilator, immunomodulatory, and mucoactive effects as well as central or peripheral antitussive activities. FFs are cost-effective, easily accessible, and safe options for both treatment and prevention of COVID-19. They might have positive psychological effects along with their pharmacological effects and nutritional virtues. They could also manage persistent respiratory discomforts after recovery from COVID-19.
Collapse
Affiliation(s)
- Ghazaleh Mosleh
- Phytopharmaceutical Technology and Traditional Medicine Incubator, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parmis Badr
- Phytopharmaceutical Technology and Traditional Medicine Incubator, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Meysam Zaeri
- Department of Phytopharmaceuticals (Traditional Pharmacy), School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolali Mohagheghzadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Phytopharmaceuticals (Traditional Pharmacy), School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Pandey A, Kulshrestha R, Bansal SK. Dynamic role of LMW-hyaluronan fragments and Toll-like receptors 2,4 in progression of bleomycin induced lung parenchymal injury to fibrosis. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2021. [PMCID: PMC8138115 DOI: 10.1186/s43168-021-00073-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Pulmonary fibrosis (PF) is a progressive and lethal lung disease of elderly whose incidence has been increasing following the Covid-19 pandemic caused by severe acute respiratory syndrome corona virus 2 (SARS-CoV-2). PF immunopathogenesis involves progressive alveolar epithelial cell damage, release of damage-associated molecular patterns (DAMPs), and extracellular matrix (ECM) injury. We assessed the dynamic role of LMW-hyaluronan (LMW-HA) as DAMP in initiation of host immune TLR-2,4 responses and as determinant in progression of ECM injury to fibrosis. Male Wistar rats were divided into Group I (saline control, n = 24) and Group II (intratracheal bleomycin, 7 U/kg/animal, n = 24). Animals were euthanized on 0, 7, 14, and 28 days. The time course of release of LMW-HA, TLR-2,4 mRNA and protein levels, and NF-κB-p65 levels after bleomycin injury were correlated with the development of parenchymal inflammation, remodelling, and fibrosis. Results Acute lung injury caused by bleomycin significantly increases the pro-inflammatory LMW-HA levels and elevates TLR-2,4 levels on day 7. Subsequently, TLR-2 upregulation, TLR-4 downregulation, and NF-κB signalling follow on days 14 and 28. This results in progressive tissue inflammation, alveolar and interstitial macrophage accumulation, and fibrosis. Conclusions LMW-HA significantly increases in PF caused by non-infectious and infectious (Covid-19) etiologies. The accumulating HA fragments function as endogenous DAMPs and trigger inflammatory responses, through differential TLR2 and TLR4 signalling, thus promoting inflammation and macrophage influx. LMW-HA are reflective of the state of ongoing tissue inflammation and may be considered as a natural biosensor for fibrotic lung diseases and as potential therapeutic targets.
Collapse
|
19
|
Moallemian Isfahani M, Emam-Djomeh Z, Rao IM, Rezaei N. Nutrition and Immunity in COVID-19. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:485-497. [PMID: 33973196 DOI: 10.1007/978-3-030-63761-3_28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nutrition can strongly influence infection trajectories by either boosting or suppressing the immune system. During the recently emerged pandemic of coronavirus disease 2019 (COVID-19), individuals who possess diets high in fat, refined carbohydrates, and sugars have shown to be highly prone to the disease and associated adverse outcomes. Both micronutrients and macronutrients provide benefits at different stages of the infection. Thus, using appropriate nutritional recommendations and interventions is necessary to combat the infection in patients with COVID-19 in both outpatient and inpatient settings.
Collapse
Affiliation(s)
- Marjan Moallemian Isfahani
- Dietetics and Nutrition Experts Team (DiNET), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Emam-Djomeh
- Dietetics and Nutrition Experts Team (DiNET), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Food Science and Engineering, University College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Idupulapati M Rao
- Centro Internacional de Agricultura Tropical, Santiago de Cali, Colombia
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
20
|
Ontong P, Prachayasittikul V. Unraveled roles of hyaluronan in severe COVID-19. EXCLI JOURNAL 2021; 20:117-125. [PMID: 33564281 PMCID: PMC7868638 DOI: 10.17179/excli2020-3215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/11/2021] [Indexed: 12/23/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic viral pneumonia caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2). Most of the severe COVID-19 patients come up with trouble breathing, persistent pressure in the chest and developing to acute respiratory distress syndrome (ARDS) with a high mortality rate. Infected lung brings about uncontrolled inflammation followed by the fluid leakage and accumulation of extracellular matrix. Hyaluronan (HA) is an essential component of the extracellular matrix (ECM) and plays crucial roles in both biological and pathological states. It is also primarily located within the respiratory airways and is uprising during COVID-19 infection. Hitherto, the association between COVID-19 pathophysiology and HA is still unclear. Herein, we provide an overview of the pathophysiology of SARS-CoV-2 infection in conjunction with the involvement of HA and the diminution of HA for therapeutic potential of COVID-19. For severe patients, HA depletion may be beneficial for preventing ARDS while monitoring and managing HA level in lung may improve survival rate of patients.
Collapse
Affiliation(s)
- Pawared Ontong
- Department of Community Medical Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
21
|
Chang R, Mamun A, Dominic A, Le NT. SARS-CoV-2 Mediated Endothelial Dysfunction: The Potential Role of Chronic Oxidative Stress. Front Physiol 2021; 11:605908. [PMID: 33519510 PMCID: PMC7844210 DOI: 10.3389/fphys.2020.605908] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells have emerged as key players in SARS-CoV-2 infection and COVID-19 inflammatory pathologies. Dysfunctional endothelial cells can promote chronic inflammation and disease processes like thrombosis, atherosclerosis, and lung injury. In endothelial cells, mitochondria regulate these inflammatory pathways via redox signaling, which is primarily achieved through mitochondrial reactive oxygen species (mtROS). Excess mtROS causes oxidative stress that can initiate and exacerbate senescence, a state that promotes inflammation and chronic endothelial dysfunction. Oxidative stress can also activate feedback loops that perpetuate mitochondrial dysfunction, mtROS overproduction, and inflammation. In this review, we provide an overview of phenotypes mediated by mtROS in endothelial cells - such as mitochondrial dysfunction, inflammation, and senescence - as well as how these chronic states may be initiated by SARS-CoV-2 infection of endothelial cells. We also propose that SARS-CoV-2 activates mtROS-mediated feedback loops that cause long-term changes in host redox status and endothelial function, promoting cardiovascular disease and lung injury after recovery from COVID-19. Finally, we discuss the implications of these proposed pathways on long-term vascular health and potential treatments to address these chronic conditions.
Collapse
Affiliation(s)
- Ryan Chang
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Abrar Mamun
- Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Abishai Dominic
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
22
|
Dabbish AM, Yonis N, Salama M, Essa MM, Qoronfleh MW. Inflammatory pathways and potential therapies for COVID-19: A mini review. EUR J INFLAMM 2021; 19. [DOI: 10.1177/20587392211002986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
Abstract
The public health crisis of the novel coronavirus disease (COVID-19) is alarming since January 2020. COVID-19 genome (SARS-CoV-2) is related to other highly pathogenic coronaviruses SARS-CoV (severe acute respiratory syndrome coronavirus) and MERS-CoV (Middle East respiratory syndrome coronavirus). Amino acid substitutions in some of SARS-CoV-2 proteins resulted in mutations proposing more virulent and contagious properties for this novel virus. Coronavirus penetrates the host cell via endocytosis and once infected, immune responses are triggered to fight against the pathogen. Innate immune response activates major transcription factors to secrete proinflammatory cytokines and type 1 interferon response (T1INF) to induce antiviral immunity. While adaptive immunity initiates cascade of B-cells antibody mediated and T-cells cellular mediate immunities, several mechanisms are raised by SARS-CoV-2 to evade host immune response. Consequently, a surge of proinflammatory cytokines, known as cytokine storm (CS) are released. Failure to manage CS results in several pathological complications as acute respiratory distress syndrome (ARDS). Although researches have not discovered an effective treatment against SARS-CoV-2, recent therapeutic approaches recommending the use of anti-inflammatories in combination with antivirals and some repurposed drugs for COVID-19 patients. Future medications should be designed to target essential hallmarks in the CS to improve clinical outcomes.
Collapse
Affiliation(s)
- Areeg M Dabbish
- Biotechnology Graduate Program, School of Science and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Nouran Yonis
- Institute of Global Health and Human Ecology (IGHHE) Graduate Program, The American University in Cairo, New Cairo, Egypt
| | - Mohamed Salama
- Institute of Global Health and Human Ecology (IGHHE), The American University in Cairo, New Cairo, Egypt
| | - Musthafa M Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| | - M Walid Qoronfleh
- Research & Policy Department, World Innovation Summit for Health (WISH), Qatar Foundation, Doha, Qatar
| |
Collapse
|
23
|
Benatti MN, Fabro AT, Miranda CH. Endothelial glycocalyx shedding in the acute respiratory distress syndrome after flu syndrome. J Intensive Care 2020; 8:72. [PMID: 32974033 PMCID: PMC7503444 DOI: 10.1186/s40560-020-00488-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/30/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Scientific evidence indicates that endothelial glycocalyx (EG) shedding contributes to the pathophysiological installation of acute respiratory distress syndrome (ARDS) after bacterial sepsis. The aim was to evaluate the EG shedding in ARDS installation after flu syndrome. METHODS This cross-sectional study included patients with flu syndrome during the influenza outbreak divided into two groups: patients with and without ARDS. Healthy subjects without flu syndrome were included in a control group. We measured EG damage biomarkers (hyaluronan, syndecan-1) and endothelial cell injury biomarker (soluble thrombomodulin) during the first medical evaluation. Histological assessment of the perimeter of the hyaline membrane and the number of neutrophils infiltrated in the alveolar septum was performed in patients who died. RESULTS ARDS group had 30 patients (44 ± 16 years old, 57% men), the non-ARDS group had 36 patients (39 ± 17 years old, 42% men), and the control group had 35 individuals (44 ± 9 years old, 51% men). Hyaluronan levels were significantly higher in the ARDS group than the two groups [31 ng/ml (interquartile range-IQR 12-56) vs. 5 ng/ml (IQR 3-10) vs. 5 ng/ml (IQR 2-8); p < 0.0001]. Hyaluronan levels above 19 ng/ml in patients with flu syndrome were associated with a significant increase in 28-day mortality rate: relative risk (RR): 6.95; (95% confidence interval 1.88-25.67); p = 0.0017. A positive correlation was observed between hyaline membrane perimeter and soluble thrombomodulin levels (r = 0.89; p = 0.05) as well as between the number of neutrophils in the alveolar septum and hyaluronan levels (r = 0.89; p = 0.05). CONCLUSIONS Evidence of EG shedding was found in ARDS established after flu syndrome.
Collapse
Affiliation(s)
- Maira Nilson Benatti
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Rua Bernardino de Campos, 1000, Ribeirão Preto, São Paulo, 14020-670 Brazil
| | - Alexandre Todorovic Fabro
- Department of Pathology and Legal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Ribeirão Preto, São Paulo, Brazil
| | - Carlos Henrique Miranda
- Division of Emergency Medicine, Department of Internal Medicine, Ribeirão Preto School of Medicine, São Paulo University, Rua Bernardino de Campos, 1000, Ribeirão Preto, São Paulo, 14020-670 Brazil
| |
Collapse
|
24
|
Kaber G, Kratochvil MJ, Burgener EB, Peltan EL, Barlow G, Yang S, Nicolls MR, de Jesus Perez V, Rosser JI, Wardle AJ, Kalinowski A, Ozawa MG, Regula DP, Nagy N, Heilshorn SC, Milla CE, Rogers AJ, Bollyky PL. Hyaluronan is abundant in COVID-19 respiratory secretions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 32935110 DOI: 10.1101/2020.09.11.20191692] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
COVID-19 respiratory infections are associated with copious, adherent respiratory secretions that prolong chronic ventilation and contribute to the morbidity and mortality caused by the disease. We hypothesized that hyaluronan, an extracellular matrix glycosaminoglycan produced at sites of active inflammation that promotes edema in other settings, might be a component of these secretions. To interrogate this, we examined the respiratory secretions collected from eight intubated patients with COVID-19, six control patients with cystic fibrosis (CF), a different respiratory disease also associated with thick adherent secretions, and eight healthy controls. In this sample set we found that hyaluronan content is increased approximately 20-fold in both CF and COVID-19 patients compared to healthy controls. The hyaluronan in COVID-19 samples was comprised of low-molecular weight fragments, the hyaluronan form most strongly linked with pro-inflammatory functions. Hyaluronan is similarly abundant in histologic sections from cadaveric lung tissue from COVID-19 patients. These findings implicate hyaluronan in the thick respiratory secretions characteristic of COVID-19 infection. Therapeutic strategies targeting hyaluronan should be investigated further for potential use in patients with COVID-19.
Collapse
|
25
|
Hohn A, Baumann A, Pietroschinsky E, Franklin J, Illerhaus A, Buchwald D, Hinkelbein J, Zahn PK, Annecke T. Hemoadsorption: effective in reducing circulating fragments of the endothelial glycocalyx during cardiopulmonary bypass in patients undergoing on-pump cardiac surgery? Minerva Anestesiol 2020; 87:35-42. [PMID: 32643361 DOI: 10.23736/s0375-9393.20.14525-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The vascular endothelial glycocalyx is susceptible to ischemia and hypoxia. Released soluble components of the endothelial glycocalyx (EG) have been identified as potential damage associated molecular patterns (DAMPs) able to enhance an ongoing inflammatory response. Shedding of the EG has been associated with released atrial-natriuretic peptide (ANP) during cardiac surgery procedures. A novel hemoadsorption technique (CytoSorb®) has been shown to effectively remove molecules up to 55 kDa unspecifically from circulation. It is not known whether ANP or glycocalyx components can be removed successfully by this technique. METHODS In 15 patients undergoing on-pump cardiac surgery, the hemoadsorption device was integrated in the cardiopulmonary bypass (CPB) circuit. Pre- and post-adsorber concentrations of ANP, heparan sulphate (HEP), syndecan-1 (SYN) and hyaluronan (HYA) were measured at 10 (T1), 30 (T2), and 60 (T3) minutes after aortic cross-clamping and complete CPB. RESULTS Hemoadsorption significantly reduced mean HEP concentrations (-157.5 [333.4] ng/mL; P<0.001) post adsorber. For ANP and SYN no statistically significant changes were detected whereas mean [SD] HYA concentrations even increased significantly (+21.6 [43.0] ng/mL; P<0.001) post adsorber. CONCLUSIONS In this study representing a real-life scenario, we could demonstrate that the novel hemoadsorption device (CytoSorb®) was able to effectively adsorb HEP from the circulation if integrated in a CPB circuit. However, blood concentrations of HYA, SYN, and ANP could not be reduced during CPB in our investigation.
Collapse
Affiliation(s)
- Andreas Hohn
- Faculty of Medicine, University of Cologne, Cologne, Germany - .,Department of Anesthesiology and Intensive Care Medicine, Cologne University Hospital, Cologne, Germany - .,Department of Anesthesiology and Intensive Care Medicine, Kliniken Maria Hilf GmbH, Moenchengladbach, Germany -
| | - Andreas Baumann
- Department of Anesthesiology, Intensive Care, Palliative Care and Pain Medicine, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Eva Pietroschinsky
- Faculty of Medicine, University of Cologne, Cologne, Germany.,Department of Anesthesiology and Intensive Care Medicine, Cologne University Hospital, Cologne, Germany
| | - Jeremy Franklin
- Institute of Medical Statistics, Informatics and Epidemiology, University of Cologne, Cologne, Germany
| | - Anja Illerhaus
- Faculty of Medicine, University of Cologne, Cologne, Germany.,Department of Dermatology, Cologne University Hospital, Cologne, Germany
| | - Dirk Buchwald
- Department of Cardiac and Thoracic Surgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Jochen Hinkelbein
- Faculty of Medicine, University of Cologne, Cologne, Germany.,Department of Anesthesiology and Intensive Care Medicine, Cologne University Hospital, Cologne, Germany
| | - Peter K Zahn
- Department of Anesthesiology, Intensive Care, Palliative Care and Pain Medicine, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Thorsten Annecke
- Faculty of Medicine, University of Cologne, Cologne, Germany.,Department of Anesthesiology and Intensive Care Medicine, Cologne University Hospital, Cologne, Germany.,Department of Anesthesiology and Intensive Care Medicine, Kliniken der Stadt Köln GmbH, University of Witten Herdecke, Cologne, Germany
| |
Collapse
|
26
|
Liu A, Park JH, Zhang X, Sugita S, Naito Y, Lee JH, Kato H, Hao Q, Matthay MA, Lee JW. Therapeutic Effects of Hyaluronic Acid in Bacterial Pneumonia in Ex Vivo Perfused Human Lungs. Am J Respir Crit Care Med 2019; 200:1234-1245. [PMID: 31390880 PMCID: PMC6857490 DOI: 10.1164/rccm.201812-2296oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 08/07/2019] [Indexed: 12/18/2022] Open
Abstract
Rationale: Recent studies have demonstrated that extracellular vesicles (EVs) released during acute lung injury (ALI) were inflammatory.Objectives: The current study was undertaken to test the role of EVs induced and released from severe Escherichia coli pneumonia (E. coli EVs) in the pathogenesis of ALI and to determine whether high-molecular-weight (HMW) hyaluronic acid (HA) administration would suppress lung injury from E. coli EVs or bacterial pneumonia.Methods:E. coli EVs were collected from the perfusate of an ex vivo perfused human lung injured with intrabronchial E. coli bacteria for 6 hours by ultracentrifugation and then given intrabronchially or intravenously to naive human lungs. One hour later, HMW HA was instilled into the perfusate (n = 5-6). In separate experiments, HMW HA was given after E. coli bacterial pneumonia (n = 6-10). In vitro experiments were conducted to evaluate binding of EVs to HMW HA and uptake of EVs by human monocytes.Measurements and Main Results: Administration of HMW HA ameliorated the impairment of alveolar fluid clearance, protein permeability, and acute inflammation from E. coli EVs or pneumonia and reduced total bacteria counts after E. coli pneumonia. HMW HA bound to E. coli EVs, inhibiting the uptake of EVs by human monocytes, an effect associated with reduced TNFα (tumor necrosis factor α) secretion. Surprisingly, HMW HA increased E. coli bacteria phagocytosis by monocytes.Conclusions: EVs induced and released during severe bacterial pneumonia were inflammatory and induced ALI, and HMW HA administration was effective in inhibiting the uptake of EVs by target cells and decreasing lung injury from E. coli EVs or bacterial pneumonia.
Collapse
Affiliation(s)
- Airan Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Jeong-Hyun Park
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Xiwen Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; and
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Shinji Sugita
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Yoshifumi Naito
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Jae-Hoon Lee
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Hideya Kato
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Qi Hao
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Michael A. Matthay
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Jae-Woo Lee
- Department of Anesthesiology and
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
27
|
Mai N, Miller-Rhodes K, Knowlden S, Halterman MW. The post-cardiac arrest syndrome: A case for lung-brain coupling and opportunities for neuroprotection. J Cereb Blood Flow Metab 2019; 39:939-958. [PMID: 30866740 PMCID: PMC6547189 DOI: 10.1177/0271678x19835552] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic inflammation and multi-organ failure represent hallmarks of the post-cardiac arrest syndrome (PCAS) and predict severe neurological injury and often fatal outcomes. Current interventions for cardiac arrest focus on the reversal of precipitating cardiac pathologies and the implementation of supportive measures with the goal of limiting damage to at-risk tissue. Despite the widespread use of targeted temperature management, there remain no proven approaches to manage reperfusion injury in the period following the return of spontaneous circulation. Recent evidence has implicated the lung as a moderator of systemic inflammation following remote somatic injury in part through effects on innate immune priming. In this review, we explore concepts related to lung-dependent innate immune priming and its potential role in PCAS. Specifically, we propose and investigate the conceptual model of lung-brain coupling drawing from the broader literature connecting tissue damage and acute lung injury with cerebral reperfusion injury. Subsequently, we consider the role that interventions designed to short-circuit lung-dependent immune priming might play in improving patient outcomes following cardiac arrest and possibly other acute neurological injuries.
Collapse
Affiliation(s)
- Nguyen Mai
- 1 Department of Neuroscience, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA.,2 Center for Neurotherapeutics Discovery, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA
| | - Kathleen Miller-Rhodes
- 1 Department of Neuroscience, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA.,2 Center for Neurotherapeutics Discovery, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA
| | - Sara Knowlden
- 2 Center for Neurotherapeutics Discovery, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA.,3 Department of Neurology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA
| | - Marc W Halterman
- 1 Department of Neuroscience, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA.,2 Center for Neurotherapeutics Discovery, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA.,3 Department of Neurology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY, USA
| |
Collapse
|
28
|
Cui Z, Liao J, Cheong N, Longoria C, Cao G, DeLisser HM, Savani RC. The Receptor for Hyaluronan-Mediated Motility (CD168) promotes inflammation and fibrosis after acute lung injury. Matrix Biol 2018; 78-79:255-271. [PMID: 30098420 PMCID: PMC6368477 DOI: 10.1016/j.matbio.2018.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/09/2018] [Accepted: 08/04/2018] [Indexed: 12/15/2022]
Abstract
Acute lung injury results in early inflammation and respiratory distress, and later fibrosis. The glycosaminoglycan hyaluronan (HA) and the Receptor for Hyaluronan-Mediated Motility (RHAMM, CD168) have been implicated in the response to acute lung injury. We hypothesized that, compared to wild type (WT) mice, RHAMM knockout (KO) mice would be protected from, whereas mice with macrophage-specific transgenic overexpression of RHAMM (TG) would have worse inflammation, respiratory distress and fibrosis after intratracheal (IT) bleomycin. Compared to WT mice, 10 days after IT bleomycin, RHAMM KO mice had less weight loss, less increase in respiratory rate, and fewer CD45+ cells in the lung. At day 28, compared to injured WT animals, injured RHAMM KO mice had lower M1 macrophage content, as well as decreased fibrosis as determined by trichrome staining, Ashcroft scores and lung HPO content. Four lines of transgenic mice with selective overexpression of RHAMM in macrophages were generated using the Scavenger Receptor A promoter driving a myc-tagged full length RHAMM cDNA. Baseline expression of RHAMM and CD44 was the same in WT and TG mice. By flow cytometry, TG bone marrow-derived macrophages (BMDM) had increased cell surface RHAMM and myc, but equal CD44 expression. TG BMDM also had 2-fold increases in both chemotaxis to HA and proliferation in fetal bovine serum. In TG mice, increased inflammation after thioglycollate-induced peritonitis was restricted to macrophages and not neutrophils. For lung injury studies, non-transgenic mice given bleomycin had respiratory distress with increased respiratory rates from day 7 to 21. However, TG mice had higher respiratory rates from 4 days after bleomycin and continued to increase respiratory rates up to day 21. At 21 days after IT bleomycin, TG mice had increased lung macrophage accumulation. Lavage HA concentrations were 6-fold higher in injured WT mice, but 30-fold higher in injured TG mice. At 21 days after IT bleomycin, WT mice had developed fibrosis, but TG mice showed exaggerated fibrosis with increased Ashcroft scores and HPO content. We conclude that RHAMM is a critical component of the inflammatory response, respiratory distress and fibrosis after acute lung injury. We speculate that RHAMM is a potential therapeutic target to limit the consequences of acute lung injury.
Collapse
Affiliation(s)
- Zheng Cui
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Jie Liao
- Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Naeun Cheong
- Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher Longoria
- Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gaoyuan Cao
- Perelmen Center for Advanced Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Horace M DeLisser
- Perelmen Center for Advanced Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Rashmin C Savani
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA; Center for Pulmonary & Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
29
|
Zhou T, Yu Z, Jian MY, Ahmad I, Trempus C, Wagener BM, Pittet JF, Aggarwal S, Garantziotis S, Song W, Matalon S. Instillation of hyaluronan reverses acid instillation injury to the mammalian blood gas barrier. Am J Physiol Lung Cell Mol Physiol 2018; 314:L808-L821. [PMID: 29368549 DOI: 10.1152/ajplung.00510.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Acid (HCl) aspiration during anesthesia may lead to acute lung injury. There is no effective therapy. We hypothesized that HCl instilled intratracheally in C57BL/6 mice results in the formation of low-molecular weight hyaluronan (L-HA), which activates RhoA and Rho kinase (ROCK), causing airway hyperresponsiveness (AHR) and increased permeability. Furthermore, instillation of high-molecular weight hyaluronan (H-HA; Yabro) will reverse lung injury. We instilled HCl in C57BL/6 wild-type (WT), myeloperoxidase gene-deficient (MPO-/-) mice, and CD44 gene-deficient (CD44-/-) mice. WT mice were also instilled intranasally with H-HA (Yabro) at 1 and 23 h post-HCl. All measurements were performed at 1, 5, or 24 h post-HCl. Instillation of HCl in WT but not in CD44-/- resulted in increased inflammation, AHR, lung injury, and L-HA in the bronchoalveolar lavage fluid (BALF) 24 h post-HCl; L-HA levels and lung injury were significantly lower in HCl-instilled MPO-/- mice. Isolated perfused lungs of HCl instilled WT but not of CD44-/- mice had elevated values of the filtration coefficient ( Kf). Addition of L-HA on the apical surface of human primary bronchial epithelial cell monolayer decreased barrier resistance ( RT). H-HA significantly mitigated inflammation, AHR, and pulmonary vascular leakage at 24 h after HCl instillation and mitigated the increase of Kf and RT, as well as ROCK2 phosphorylation. Increased H- and L-HA levels were found in the BALF of mechanically ventilated patients but not in healthy volunteers. HCl instillation-induced lung injury is mediated by the L-HA-CD44-RhoA-ROCK2 signaling pathway, and H-HA is a potential novel therapeutic agent for acid aspiration-induced lung injury.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Ming-Yuan Jian
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Israr Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Carol Trempus
- Matrix Biology Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences , Research Triangle Park, North Carolina
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Saurabh Aggarwal
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Stavros Garantziotis
- Matrix Biology Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences , Research Triangle Park, North Carolina
| | - Weifeng Song
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|