1
|
Engelmaier A, Zimmermann M, Butterweck HA, Weber A. Clinical Study Support by Long-Term Stability Studies of Alpha 1-Proteinase Inhibitor and Urea in Relevant Biological Matrices. Pharmaceuticals (Basel) 2025; 18:572. [PMID: 40284007 PMCID: PMC12030187 DOI: 10.3390/ph18040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: According to recent guidelines, including the guideline on bioanalytical method validation issued by the European Medicine Agency, the stability of clinical analytes should be known. We summarize human α1-proteinase inhibitor (A1PI) and urea stability data in relevant matrices, as these analytes are usually measured in clinical A1PI studies. Methods: Stability samples with appropriate A1PI concentrations were prepared in a citrated human reference plasma pool and a matrix mimicking bronchoalveolar lavage (BAL) solution. These samples were kept at -20 °C and -60 °C for up to 24 months. A1PI protein was measured with a nephelometric method and an enzyme-linked immunosorbent assay using paired commercially available polyclonal antibodies. A1PI elastase inhibitory activity was determined with an elastase complex formation immunosorbent assay that combines A1PI complex formation with a solid phase-immobilized elastase and immunological detection of the formed A1PI-elastase complex and urea in samples kept at -20 °C only with an enzymatic assay. Results: Overall, the stability criterion (100 ± 20%) was met for the analytes A1PI protein and A1PI activity at both temperatures during storage of BAL-mimicking and plasma samples for 15 and 24 months, respectively; urea was stable in both matrices at -20 °C for 18 months. Plasma samples showed smaller drops in concentration over storage time than BAL-mimicking samples. As expected, the reduction of A1PI elastase inhibitory activity was more pronounced than that of A1PI protein. Interestingly, the analyte concentration did not significantly influence the results in either of the sample matrices. Conclusions: The data confirmed the appropriate stability of the three analytes in the matrices of citrated plasma and BAL-mimicking samples for at least up to 15 months.
Collapse
Affiliation(s)
- Andrea Engelmaier
- Pharmaceutical Science, Baxalta Innovations GmbH, Part of Takeda, 1220 Vienna, Austria;
| | - Martin Zimmermann
- Plasma Derived Therapies R&D, Baxalta Innovations GmbH, Part of Takeda, 1220 Vienna, Austria; (M.Z.); (H.A.B.)
| | - Harald A. Butterweck
- Plasma Derived Therapies R&D, Baxalta Innovations GmbH, Part of Takeda, 1220 Vienna, Austria; (M.Z.); (H.A.B.)
| | - Alfred Weber
- Plasma Derived Therapies R&D, Baxalta Innovations GmbH, Part of Takeda, 1220 Vienna, Austria; (M.Z.); (H.A.B.)
| |
Collapse
|
2
|
Erickson R, Huang C, Allen C, Ireland J, Roth G, Zou Z, Lu J, Lafont BAP, Garza NL, Brumbaugh B, Zhao M, Suzuki M, Olano L, Brzostowski J, Fischer ER, Twigg HL, Johnson RF, Sun PD. SARS-CoV-2 infection of human lung epithelial cells induces TMPRSS-mediated acute fibrin deposition. Nat Commun 2023; 14:6380. [PMID: 37821447 PMCID: PMC10567911 DOI: 10.1038/s41467-023-42140-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/27/2023] [Indexed: 10/13/2023] Open
Abstract
Severe COVID-associated lung injury is a major confounding factor of hospitalizations and death with no effective treatments. Here, we describe a non-classical fibrin clotting mechanism mediated by SARS-CoV-2 infected primary lung but not other susceptible epithelial cells. This infection-induced fibrin formation is observed in all variants of SARS-CoV-2 infections, and requires thrombin but is independent of tissue factor and other classical plasma coagulation factors. While prothrombin and fibrinogen levels are elevated in acute COVID BALF samples, fibrin clotting occurs only with the presence of viral infected but not uninfected lung epithelial cells. We suggest a viral-induced coagulation mechanism, in which prothrombin is activated by infection-induced transmembrane serine proteases, such as ST14 and TMPRSS11D, on NHBE cells. Our finding reveals the inefficiency of current plasma targeted anticoagulation therapy and suggests the need to develop a viral-induced ARDS animal model for treating respiratory airways with thrombin inhibitors.
Collapse
Affiliation(s)
- Rachel Erickson
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Chang Huang
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Cameron Allen
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Joanna Ireland
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Gwynne Roth
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Zhongcheng Zou
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Jinghua Lu
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Bernard A P Lafont
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nicole L Garza
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Beniah Brumbaugh
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Ming Zhao
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Motoshi Suzuki
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Lisa Olano
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Joseph Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Elizabeth R Fischer
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Homer L Twigg
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University Medical Center, 1120 West Michigan Street, CL 260A, Indianapolis, IN, 46202, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Peter D Sun
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA.
| |
Collapse
|
3
|
Payton AD, Perryman AN, Hoffman JR, Avula V, Wells H, Robinette C, Alexis NE, Jaspers I, Rager JE, Rebuli ME. Cytokine signature clusters as a tool to compare changes associated with tobacco product use in upper and lower airway samples. Am J Physiol Lung Cell Mol Physiol 2022; 322:L722-L736. [PMID: 35318855 PMCID: PMC9054348 DOI: 10.1152/ajplung.00299.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022] Open
Abstract
Inhalation exposure to cigarette smoke and e-cigarette aerosol is known to alter the respiratory immune system, particularly cytokine signaling. In assessments of health impacts of tobacco product use, cytokines are often measured using a variety of sample types, from serum to airway mucosa. However, it is currently unclear whether and how well cytokine levels from different sample types and the airway locations they represent are correlated, making comparing studies that utilize differing sample types challenging. To address this challenge, we compared baseline cytokine signatures in upper and lower airways and systemic samples and evaluated how groups of coexpressed cytokines change with tobacco product use. Matched nasal lavage fluid (NLF), nasal epithelial lining fluid (NELF), sputum, and circulating serum samples were collected from 14 nonsmokers, 13 cigarette smokers, and 17 e-cigarette users and analyzed for levels of 22 cytokines. Individual cytokine signatures were first compared across each sample type, followed by identification of cytokine clusters within each sample type. Identified clusters were then evaluated for potential alterations following tobacco product use using eigenvector analyses. Individual cytokine signatures in the respiratory tract were significantly correlated (NLF, NELF, and sputum) compared with randomly permutated signatures, whereas serum was not significantly different from random permutations. Cytokine clusters that were similar across airway sample types were modified by tobacco product use, particularly e-cigarettes, indicating a degree of uniformity in terms of how cytokine host defense and immune cell recruitment responses cooperate in the upper and lower airways. Overall, cluster-based analyses were found to be especially useful in small cohort assessments, providing higher sensitivity than individual signatures to detect biologically meaningful differences between tobacco use groups. This novel cluster analysis approach revealed that eigencytokine patterns in noninvasive upper airway samples simulate cytokine patterns in lower airways.
Collapse
Affiliation(s)
- Alexis D Payton
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alexia N Perryman
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jessica R Hoffman
- Curriculum for the Environment and Ecology, College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Vennela Avula
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Heather Wells
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Carole Robinette
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ilona Jaspers
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Julia E Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Meghan E Rebuli
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
4
|
Qin W, Wang T, Liu G, Sun L, Han W, Gao Y. Dynamic Urinary Proteome Changes in Ovalbumin-Induced Asthma Mouse Model Using Data-Independent Acquisition Proteomics. J Asthma Allergy 2021; 14:1355-1366. [PMID: 34785909 PMCID: PMC8590963 DOI: 10.2147/jaa.s330054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/30/2021] [Indexed: 01/09/2023] Open
Abstract
Background In this work, we aim to investigate dynamic urinary proteome changes during asthma development and to identify potential urinary protein biomarkers for the diagnosis of asthma. Methods An ovalbumin (OVA)-induced mouse model was used to mimic asthma. The urinary proteome from asthma and control mice was determined using data-independent acquisition combined with high-resolution tandem mass spectrometry. Results Overall, 331 proteins were identified, among which 53 were differentially expressed (26, 24, 14 and 20 on days 2, 8, 15 and 18, respectively; 1.5-fold change, adjust P<0.05). Gene Ontology annotation of the differential proteins showed that the acute-phase response, innate immune response, B cell receptor signaling pathway, and complement activation were significantly enriched. Protein–protein interaction network revealed that these differential proteins were partially biologically connected in OVA-induced asthma, as a group. On days 2 and 8, after two episodes of OVA sensitization, six differential proteins (CRAMP, ECP, HP, F2, AGP1, and CFB) were also reported to be closely associated with asthma. These proteins may hold the potential for the early screening of asthma. On days 15 and 18, after challenged with 1% OVA by inhalation, seven differential proteins (VDBP, HP, CTSE, PIGR, AAT, TRFE, and HPX) were also reported to be closely associated with asthma. Thus, these proteins hold the potential to be biomarkers for the diagnosis of asthma attack. Conclusion Our results indicate that the urinary proteome could reflect dynamic pathophysiological changes in asthma progression.
Collapse
Affiliation(s)
- Weiwei Qin
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, People's Republic of China.,Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Ting Wang
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Lixin Sun
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Wei Han
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, 100875, People's Republic of China
| |
Collapse
|
5
|
Covarrubias-Zambrano O, Motamedi M, Ameredes BT, Tian B, Calhoun WJ, Zhao Y, Brasier AR, Kalubowilage M, Malalasekera AP, Yapa AS, Wang H, Culbertson CT, Troyer DL, Bossmann SH. Optical biosensing of markers of mucosal inflammation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102476. [PMID: 34743019 DOI: 10.1016/j.nano.2021.102476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/13/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023]
Abstract
We report the design and adaptation of iron/iron oxide nanoparticle-based optical nanobiosensors for enzymes or cytokine/chemokines that are established biomarkers of lung diseases. These biomarkers comprise ADAM33, granzyme B, MMP-8, neutrophil elastase, arginase, chemokine (C-C motif) ligand 20 and interleukin-6. The synthesis of nanobiosensors for these seven biomarkers, their calibration with commercially available enzymes and cytokines/chemokines, as well as their validation using bronchoalveolar lavage (BAL) obtained from a mouse model of TLR3-mediated inflammation are discussed here. Exhaled Breath Condensate (EBC) is a minimally invasive approach for sampling airway fluid in the diagnosis and management of various lung diseases in humans (e.g., asthma, COPD and viral infections). We report the proof-of-concept of using human EBC in conjunction with nanobiosensors for diagnosis/monitoring airway inflammation. These findings suggest that, with nanosensor technology, human EBC can be utilized as a liquid biopsy to monitor inflammation/remodeling in lung disease.
Collapse
Affiliation(s)
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX, USA
| | - Bill T Ameredes
- Institute for Translational Sciences and Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX
| | - Bing Tian
- Institute for Translational Sciences and Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX
| | - William J Calhoun
- Institute for Translational Sciences and Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX
| | - Yingxin Zhao
- Institute for Translational Sciences and Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX
| | - Allan R Brasier
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI
| | | | - Aruni P Malalasekera
- Department of Chemistry, Southwestern College, 100 College Street, Winfield, KS, USA
| | - Asanka S Yapa
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | - Hongwang Wang
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | | | - Deryl L Troyer
- Department of Anatomy & Physiology, Kansas State University, Manhattan, KS, USA
| | - Stefan H Bossmann
- Department of Chemistry, Kansas State University, Manhattan, KS, USA; The University of Kansas Medical Center, Department of Cancer Biology and The University of Kansas Cancer Center, Kansas City, KS, USA.
| |
Collapse
|
6
|
Costa M, Páez A. Emerging insights into the role of albumin with plasma exchange in Alzheimer's disease management. Transfus Apher Sci 2021; 60:103164. [PMID: 34083161 DOI: 10.1016/j.transci.2021.103164] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative process that inexorably leads to progressive deterioration of cognition function and, ultimately, death. Central pathophysiologic features of AD include the accumulation of extracellular plaques comprised of amyloid-β peptide (Aβ) and the presence of intraneuronal neurofibrillary tangles. However, a large body of evidence suggests that oxidative stress and inflammation are major contributors to the pathogenesis and progression of AD. To date, available pharmacologic treatments are only symptomatic. Clinical trials focused on amyloid and non-amyloid-targeted treatments with small molecule pharmacotherapy and immunotherapies have accumulated a long list of failures. Considering that around 90 % of the circulating Aβ is bound to albumin, and that a dynamic equilibrium exists between peripheral and central Aβ, plasma exchange with albumin replacement has emerged as a new approach in a multitargeted AD therapeutic strategy (AMBAR Program). In plasma exchange, a patient's plasma is removed by plasmapheresis to eliminate toxic endogenous substances, including Aβ and functionally impaired albumin. The fluid replacement used is therapeutic albumin, which acts not only as a plasma volume expander but also has numerous pleiotropic functions (e.g., circulating Aβ- binding capacity, transporter, detoxifier, antioxidant) that are clinically relevant for the treatment of AD. Positive results from the AMBAR Program (phase 1, 2, an 2b/3 trials), i.e., slower decline or stabilization of disease symptoms in the most relevant clinical efficacy and safety endpoints, offer a glimmer of hope to both AD patients and caregivers.
Collapse
Affiliation(s)
| | - Antonio Páez
- Alzheimer's Research Group, Grifols, Barcelona, Spain.
| |
Collapse
|
7
|
Serum Lowers Bioactivity and Uptake of Synthetic Amorphous Silica by Alveolar Macrophages in a Particle Specific Manner. NANOMATERIALS 2021; 11:nano11030628. [PMID: 33802450 PMCID: PMC7999370 DOI: 10.3390/nano11030628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/20/2021] [Accepted: 02/26/2021] [Indexed: 12/02/2022]
Abstract
Various cell types are compromised by synthetic amorphous silica (SAS) if they are exposed to SAS under protein-free conditions in vitro. Addition of serum protein can mitigate most SAS effects, but it is not clear whether this is solely caused by protein corona formation and/or altered particle uptake. Because sensitive and reliable mass spectrometric measurements of SiO2 NP are cumbersome, quantitative uptake studies of SAS at the cellular level are largely missing. In this study, we combined the comparison of SAS effects on alveolar macrophages in the presence and absence of foetal calf serum with mass spectrometric measurement of 28Si in alkaline cell lysates. Effects on the release of lactate dehydrogenase, glucuronidase, TNFα and H2O2 of precipitated (SIPERNAT® 50, SIPERNAT® 160) and fumed SAS (AEROSIL® OX50, AEROSIL® 380 F) were lowered close to control level by foetal calf serum (FCS) added to the medium. Using a quantitative high resolution ICP-MS measurement combined with electron microscopy, we found that FCS reduced the uptake of particle mass by 9.9% (SIPERNAT® 50) up to 83.8% (AEROSIL® OX50). Additionally, larger particle agglomerates were less frequent in cells in the presence of FCS. Plotting values for lactate dehydrogenase (LDH), glucuronidase (GLU) or tumour necrosis factor alpha (TNFα) against the mean cellular dose showed the reduction of bioactivity with a particle sedimentation bias. As a whole, the mitigating effects of FCS on precipitated and fumed SAS on alveolar macrophages are caused by a reduction of bioactivity and by a lowered internalization, and both effects occur in a particle specific manner. The method to quantify nanosized SiO2 in cells is a valuable tool for future in vitro studies.
Collapse
|
8
|
Optimizations of In Vitro Mucus and Cell Culture Models to Better Predict In Vivo Gene Transfer in Pathological Lung Respiratory Airways: Cystic Fibrosis as an Example. Pharmaceutics 2020; 13:pharmaceutics13010047. [PMID: 33396283 PMCID: PMC7823756 DOI: 10.3390/pharmaceutics13010047] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 11/17/2022] Open
Abstract
The respiratory epithelium can be affected by many diseases that could be treated using aerosol gene therapy. Among these, cystic fibrosis (CF) is a lethal inherited disease characterized by airways complications, which determine the life expectancy and the effectiveness of aerosolized treatments. Beside evaluations performed under in vivo settings, cell culture models mimicking in vivo pathophysiological conditions can provide complementary insights into the potential of gene transfer strategies. Such models must consider multiple parameters, following the rationale that proper gene transfer evaluations depend on whether they are performed under experimental conditions close to pathophysiological settings. In addition, the mucus layer, which covers the epithelial cells, constitutes a physical barrier for gene delivery, especially in diseases such as CF. Artificial mucus models featuring physical and biological properties similar to CF mucus allow determining the ability of gene transfer systems to effectively reach the underlying epithelium. In this review, we describe mucus and cellular models relevant for CF aerosol gene therapy, with a particular emphasis on mucus rheology. We strongly believe that combining multiple pathophysiological features in single complex cell culture models could help bridge the gaps between in vitro and in vivo settings, as well as viral and non-viral gene delivery strategies.
Collapse
|
9
|
Yan C, Hu Y, Qiu G, Gong X, Elda D. The clinical safety and efficacy of flexible bronchoscopy in a neonatal intensive care unit. Exp Ther Med 2020; 20:95. [PMID: 32973944 DOI: 10.3892/etm.2020.9223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/22/2020] [Indexed: 12/22/2022] Open
Abstract
Flexible bronchoscopy (FB), developed in the 1960s, is widely used in the clinical practice of pediatrics and has demonstrated fundamental value in clinical diagnoses and treatment. However, as an invasive procedure, the use of FB is limited due to concerns regarding the tolerance of the procedure and the possible complications in neonatal units. Thus, the present study aimed to investigate the clinical safety and efficacy of flexible bronchoscopy (FB) in a neonatal intensive care unit (NICU). Neonates (n=54) who received FB in the NICU of Shanghai Children's Hospital between January 2012 and December 2016 were enrolled as the experimental group and another 54 neonates who required nebulization and tracheal secretion suction treatments were the control group. Indicators including blood gas, complete blood count, C-reactive protein (CRP), X-ray, patient breathing rate, temperature and blood pressure were monitored prior to and following the procedure. No significant differences in sex, gestational age, birth weight or postnatal age were observed between the experimental group and the control group (P>0.05). Among the 54 FB patients, several cases with side effect were identified, including 18 (33.3%) with respiratory tract stenosis, nine (16.7%) with malacia and stenosis and six (11.1%) with esophagotracheal fistula. Among the 54 members of the control group, 44 neonates (81.4%) were discharged with improved condition, five (9.3%) succumbed and five patients (9.3%) abandoned the treatment and left the hospital. Bronchoalveolar lavage demonstrated consistent results with respiratory secretion culture or tracheal tube culture. In comparison between the experimental and the control groups, no significant difference in pH, partial pressure of carbon dioxide (PCO2), partial pressure of oxygen (PO2) and HCO3 - was observed, while there were no statistical differences in the values of pH, PCO2 and HCO3 - (P>0.05). However, PO2 was significantly increased, and CRP was significantly reduced, following FB procedure compared with prior to FB (P<0.05). No pneumothorax, shock, other severe complications, fever or diffused pneumonia were observed during or after FB. The data from the present study demonstrated that FB is a safe and effective strategy for the diagnosis and differentiation of neonatal respiratory diseases in NICU.
Collapse
Affiliation(s)
- Chongbing Yan
- Department of Neonatology, Shanghai Children's Hospital, Shanghai 200062, P.R. China
| | - Yong Hu
- Department of Neonatology, Shanghai Children's Hospital, Shanghai 200062, P.R. China
| | - Gang Qiu
- Department of Neonatology, Shanghai Children's Hospital, Shanghai 200062, P.R. China
| | - Xiaohui Gong
- Department of Neonatology, Shanghai Children's Hospital, Shanghai 200062, P.R. China
| | - Dermyshi Elda
- Department of Neonatology, Shanghai Children's Hospital, Shanghai 200062, P.R. China
| |
Collapse
|
10
|
Zhang Y, Liu Z, Li S, Wang M, Dai D, Jing H, Liu L. Upregulation of E-cadherin in bronchoalveolar lavage fluid-derived exosomes in patients with lung cancer. Thorac Cancer 2019; 11:41-47. [PMID: 31696667 PMCID: PMC6938754 DOI: 10.1111/1759-7714.13220] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Lung cancer features extremely high rates of morbidity and mortality. Bronchoalveolar lavage fluid (BALF), obtained by bronchoscopy and bronchoalveolar perfusion, can provide information on the cellular components of the lung microenvironment to assist with diagnosis and treatment of lung cancer. Methods BALF was performed using a flexible bronchofiberscope. Exosomes were collected by ultracentrifugation. ELISA detected the amount of E‐cadherin. Transmission electron microscopic, ELISA and WB were conducted to identify the existence of the exosomes. Transwell and Wound healing assays were used to detect the ability of migration and invasion. Results We identified the existence of exosomes in BALF. Furthermore, we observed larger amounts of E‐cadherin in the BALF obtained from patients with lung cancer than in the control obtained from the healthy side of pneumonia. Exosomes from lung cancer groups promoted the migration and invasion of A549 cancer cells. Conclusion The exosomes from lung cancer BALF promoted the migration and invasion of A549 cancer cells by carrying E‐cadherin. E‐cadherin on the surface of exosomes may act through a VE‐cadherin dependent mechanism and induce lung cancer metastasis.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Science, Jilin University, Changchun, China.,The First Hospital of Jilin University, Department of Pediatrics, Jilin University, Changchun, China
| | - Ziyu Liu
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Science, Jilin University, Changchun, China
| | - Shanyu Li
- The First Hospital of Jilin University, Department of Pediatrics, Jilin University, Changchun, China
| | - Manning Wang
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Science, Jilin University, Changchun, China
| | - Dayou Dai
- Department of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongyu Jing
- The First Hospital of Jilin University, Department of Respiratory Medicine, Jilin University, Changchun, China
| | - Lingyun Liu
- The First Hospital of Jilin University, Department of Andrology, Jilin University, Changchun, China
| |
Collapse
|
11
|
Ma L, Cissé OH, Kovacs JA. A Molecular Window into the Biology and Epidemiology of Pneumocystis spp. Clin Microbiol Rev 2018; 31:e00009-18. [PMID: 29899010 PMCID: PMC6056843 DOI: 10.1128/cmr.00009-18] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pneumocystis, a unique atypical fungus with an elusive lifestyle, has had an important medical history. It came to prominence as an opportunistic pathogen that not only can cause life-threatening pneumonia in patients with HIV infection and other immunodeficiencies but also can colonize the lungs of healthy individuals from a very early age. The genus Pneumocystis includes a group of closely related but heterogeneous organisms that have a worldwide distribution, have been detected in multiple mammalian species, are highly host species specific, inhabit the lungs almost exclusively, and have never convincingly been cultured in vitro, making Pneumocystis a fascinating but difficult-to-study organism. Improved molecular biologic methodologies have opened a new window into the biology and epidemiology of Pneumocystis. Advances include an improved taxonomic classification, identification of an extremely reduced genome and concomitant inability to metabolize and grow independent of the host lungs, insights into its transmission mode, recognition of its widespread colonization in both immunocompetent and immunodeficient hosts, and utilization of strain variation to study drug resistance, epidemiology, and outbreaks of infection among transplant patients. This review summarizes these advances and also identifies some major questions and challenges that need to be addressed to better understand Pneumocystis biology and its relevance to clinical care.
Collapse
Affiliation(s)
- Liang Ma
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, Maryland, USA
| | - Ousmane H Cissé
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, Maryland, USA
| | - Joseph A Kovacs
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, Maryland, USA
| |
Collapse
|
12
|
Abstract
PURPOSE High levels of NaCl in the diet are associated with both cardiac and renal fibrosis, but whether salt intake affects pulmonary fibrosis has not been examined. AIM OF THE STUDY To test the hypothesis that salt intake might affect pulmonary fibrosis. MATERIALS AND METHODS Mice were fed low, normal, or high salt diets for 2 weeks, and then treated with oropharyngeal bleomycin to induce pulmonary fibrosis, or oropharyngeal saline as a control. RESULTS As determined by collagen staining of lung sections, and protein levels and cell numbers in the bronchoalveolar lavage (BAL) fluid at 21 days after bleomycin, the high salt diet did not exacerbate bleomycin-induced fibrosis, while the low salt diet attenuated fibrosis. For the bleomycin-treated mice, staining of the post-BAL lung sections indicated that compared to the regular salt diet, high salt increased the number of Ly6c-positive macrophages and decreased the number of CD11c and CD206-positive macrophages and dendritic cells. The low salt diet caused bleomycin-induced leukocyte numbers to be similar to control saline-treated mice, but reduced numbers of CD45/collagen-VI positive fibrocytes. In the saline controls, low dietary salt decreased CD11b and CD11c positive cells in lung sections, and high dietary salt increased fibrocytes. CONCLUSIONS Together, these data suggest the possibility that a low salt diet might attenuate pulmonary fibrosis.
Collapse
Affiliation(s)
- Wensheng Chen
- a Department of Biology , Texas A&M University , College Station , TX , USA.,b Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine , Hefei , China
| | - Darrell Pilling
- a Department of Biology , Texas A&M University , College Station , TX , USA
| | - Richard H Gomer
- a Department of Biology , Texas A&M University , College Station , TX , USA
| |
Collapse
|
13
|
Dalle-Donne I, Colombo G, Gornati R, Garavaglia ML, Portinaro N, Giustarini D, Bernardini G, Rossi R, Milzani A. Protein Carbonylation in Human Smokers and Mammalian Models of Exposure to Cigarette Smoke: Focus on Redox Proteomic Studies. Antioxid Redox Signal 2017; 26:406-426. [PMID: 27393565 DOI: 10.1089/ars.2016.6772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Oxidative stress is one mechanism whereby tobacco smoking affects human health, as reflected by increased levels of several biomarkers of oxidative stress/damage isolated from tissues and biological fluids of active and passive smokers. Many investigations of cigarette smoke (CS)-induced oxidative stress/damage have been carried out in mammalian animal and cellular models of exposure to CS. Animal models allow the investigation of many parameters that are similar to those measured in human smokers. In vitro cell models may provide new information on molecular and functional differences between cells of smokers and nonsmokers. Recent Advances: Over the past decade or so, a growing number of researches highlighted that CS induces protein carbonylation in different tissues and body fluids of smokers as well as in in vivo and in vitro models of exposure to CS. CRITICAL ISSUES We review recent findings on protein carbonylation in smokers and models thereof, focusing on redox proteomic studies. We also discuss the relevance and limitations of these models of exposure to CS and critically assess the congruence between the smoker's condition and laboratory models. FUTURE DIRECTIONS The identification of protein targets is crucial for understanding the mechanism(s) by which carbonylated proteins accumulate and potentially affect cellular functions. Recent progress in redox proteomics allows the enrichment, identification, and characterization of specific oxidative protein modifications, including carbonylation. Therefore, redox proteomics can be a powerful tool to gain new insights into the onset and/or progression of CS-related diseases and to develop strategies to prevent and/or treat them. Antioxid. Redox Signal. 26, 406-426.
Collapse
Affiliation(s)
| | - Graziano Colombo
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| | - Rosalba Gornati
- 2 Department of Biotechnology and Life Sciences, University of Insubria , Varese, Italy
| | - Maria L Garavaglia
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| | - Nicola Portinaro
- 3 Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano and Pediatric Orthopaedic Unit, Humanitas Clinical and Research Center , Rozzano (Milan), Italy
| | | | - Giovanni Bernardini
- 2 Department of Biotechnology and Life Sciences, University of Insubria , Varese, Italy
| | - Ranieri Rossi
- 4 Department of Life Sciences, University of Siena , Siena, Italy
| | - Aldo Milzani
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| |
Collapse
|
14
|
Carvalho AS, Matthiesen R. Bronchoalveolar Lavage: Quantitative Mass Spectrometry-Based Proteomics Analysis in Lung Diseases. Methods Mol Biol 2017; 1619:487-494. [PMID: 28674906 DOI: 10.1007/978-1-4939-7057-5_34] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Bronchoalveolar lavage (BAL) fluid, obtained by a relatively noninvasive procedure, is used as a practice for diagnosis of various lung diseases as source of cells for cytology analysis. The acellular component of BAL potentially can complement and be a key for the establishment of diagnostic or as a prognostic indicator. This chapter discusses the aspects of standardization of BAL sample preparation and processing and its implications on the BAL fluid proteome quantitative analysis by high-throughput mass spectrometry. The detailed conditions for quantitative analysis of BAL proteome in the context of biomarker discovery are introduced.
Collapse
Affiliation(s)
- Ana Sofia Carvalho
- Computational and Experimental Biology Group, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua Câmara Pestana nº 6, 6-A, Lisboa, 1150-082, Portugal.
| | - Rune Matthiesen
- Computational and Experimental Biology Group, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua Câmara Pestana nº 6, 6-A, Lisboa, 1150-082, Portugal
| |
Collapse
|
15
|
Kim N, Duncan GA, Hanes J, Suk JS. Barriers to inhaled gene therapy of obstructive lung diseases: A review. J Control Release 2016; 240:465-488. [PMID: 27196742 DOI: 10.1016/j.jconrel.2016.05.031] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 12/29/2022]
Abstract
Knowledge of genetic origins of obstructive lung diseases has made inhaled gene therapy an attractive alternative to the current standards of care that are limited to managing disease symptoms. Initial lung gene therapy clinical trials occurred in the early 1990s following the discovery of the genetic defect responsible for cystic fibrosis (CF), a monogenic disorder. However, despite over two decades of intensive effort, gene therapy has yet to help patients with CF or any other obstructive lung disease. The slow progress is due in part to poor understanding of the biological barriers to inhaled gene therapy. Encouragingly, clinical trials have shown that inhaled gene therapy with various viral vectors and non-viral gene vectors is well tolerated by patients, and continued research has provided valuable lessons and resources that may lead to future success of this therapeutic strategy. In this review, we first introduce representative obstructive lung diseases and examine limitations of currently available therapeutic options. We then review key components for successful execution of inhaled gene therapy, including gene delivery systems, primary physiological barriers and strategies to overcome them, and advances in preclinical disease models with which the most promising systems may be identified for human clinical trials.
Collapse
Affiliation(s)
- Namho Kim
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregg A Duncan
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Justin Hanes
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Environmental and Health Sciences, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jung Soo Suk
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
16
|
Carter CL, Jones JW, Farese AM, MacVittie TJ, Kane MA. Inflation-Fixation Method for Lipidomic Mapping of Lung Biopsies by Matrix Assisted Laser Desorption/Ionization-Mass Spectrometry Imaging. Anal Chem 2016; 88:4788-94. [PMID: 27028398 DOI: 10.1021/acs.analchem.6b00165] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chronic respiratory diseases are among the leading causes of deaths worldwide and major contributors of morbidity and global disease burden. To appropriately investigate lung disease, the respiratory airways must be fixed in their physiological orientation and should be inflated prior to investigations. We present an inflation-fixation method that enables lipidomic investigations of whole lung samples and resected biopsy specimens by matrix-assisted laser desorption/ionization-mass spectrometry imaging (MALDI-MSI). Formalin-inflation enables sample preparation to parallel standard clinical and surgical procedures, in addition to greatly reducing the complexity of analysis, by decreasing the number of analytes in the MALDI plume and reducing adduct formation in the resulting mass spectra. The reduced complexity increased sensitivity and enabled high-resolution imaging acquisitions without any loss in analyte detection at 10 and 20 μm scans. We present a detailed study of over 100 lipid ions detected in positive and negative ion modes covering the conducting and respiratory airways and parts of the peripheral nervous tissue running through the lungs. By defining the resolution required for clear definition of the alveolar space and thus the respiratory airways we have provided a guideline for MSI investigations of respiratory diseases involving the airways, including the interstitium. This study has provided a detailed map of lipid species and their localization within larger mammalian lung samples, for the first time, thus categorizing the lipidome for future MALDI-MSI studies of pulmonary diseases.
Collapse
Affiliation(s)
- Claire L Carter
- University of Maryland , School of Pharmacy, Department of Pharmaceutical Sciences, 20 North Pine Street, Baltimore, Maryland 21201, United States
| | - Jace W Jones
- University of Maryland , School of Pharmacy, Department of Pharmaceutical Sciences, 20 North Pine Street, Baltimore, Maryland 21201, United States
| | - Ann M Farese
- University of Maryland , School of Medicine, Department of Radiation Oncology, 655 West Baltimore Street, Baltimore, Maryland 21201, United States
| | - Thomas J MacVittie
- University of Maryland , School of Medicine, Department of Radiation Oncology, 655 West Baltimore Street, Baltimore, Maryland 21201, United States
| | - Maureen A Kane
- University of Maryland , School of Pharmacy, Department of Pharmaceutical Sciences, 20 North Pine Street, Baltimore, Maryland 21201, United States
| |
Collapse
|
17
|
Ortea I, Rodríguez-Ariza A, Chicano-Gálvez E, Arenas Vacas MS, Jurado Gámez B. Discovery of potential protein biomarkers of lung adenocarcinoma in bronchoalveolar lavage fluid by SWATH MS data-independent acquisition and targeted data extraction. J Proteomics 2016; 138:106-14. [PMID: 26917472 DOI: 10.1016/j.jprot.2016.02.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/01/2016] [Accepted: 02/13/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED Lung cancer currently ranks as the neoplasia with the highest global mortality rate. Although some improvements have been introduced in recent years, new advances in diagnosis are required in order to increase survival rates. New mildly invasive endoscopy-based diagnostic techniques include the collection of bronchoalveolar lavage fluid (BALF), which is discarded after using a portion of the fluid for standard pathological procedures. BALF proteomic analysis can contribute to clinical practice with more sensitive biomarkers, and can complement cytohistological studies by aiding in the diagnosis, prognosis, and subtyping of lung cancer, as well as the monitoring of treatment response. The range of quantitative proteomics methodologies used for biomarker discovery is currently being broadened with the introduction of data-independent acquisition (DIA) analysis-related approaches that address the massive quantitation of the components of a proteome. Here we report for the first time a DIA-based quantitative proteomics study using BALF as the source for the discovery of potential lung cancer biomarkers. The results have been encouraging in terms of the number of identified and quantified proteins. A panel of candidate protein biomarkers for adenocarcinoma in BALF is reported; this points to the activation of the complement network as being strongly over-represented and suggests this pathway as a potential target for lung cancer research. In addition, the results reported for haptoglobin, complement C4-A, and glutathione S-transferase pi are consistent with previous studies, which indicates that these proteins deserve further consideration as potential lung cancer biomarkers in BALF. Our study demonstrates that the analysis of BALF proteins by liquid chromatography-tandem mass spectrometry (LC-MS/MS), combining a simple sample pre-treatment and SWATH DIA MS, is a useful method for the discovery of potential lung cancer biomarkers. SIGNIFICANCE Bronchoalveolar lavage fluid (BALF) analysis can contribute to clinical practice with more sensitive biomarkers, thus complementing cytohistological studies in order to aid in the diagnosis, prognosis, and subtyping of lung cancer, as well as the monitoring of treatment response. Here we report a panel of candidate protein biomarkers for adenocarcinoma in BALF. Forty-four proteins showed a fold-change higher than 3.75 among adenocarcinoma patients compared with controls. This report is the first DIA-based quantitative proteomics study to use bronchoalveolar lavage fluid (BALF) as a matrix for discovering potential biomarkers. The results are encouraging in terms of the number of identified and quantified proteins, demonstrating that the analysis of BALF proteins by a SWATH approach is a useful method for the discovery of potential biomarkers of pulmonary diseases.
Collapse
Affiliation(s)
- I Ortea
- Proteomics Unit, IMIBIC, Maimonides Institute for Biomedical Research, Córdoba, Spain.
| | - A Rodríguez-Ariza
- Medical Oncology Department, Reina Sofia University Hospital and IMIBIC, Maimonides Institute for Biomedical Research, Córdoba, Spain
| | - E Chicano-Gálvez
- Proteomics Unit, IMIBIC, Maimonides Institute for Biomedical Research, Córdoba, Spain
| | - M S Arenas Vacas
- Department of Respiratory Medicine, Reina Sofia University Hospital and IMIBIC, Maimonides Institute for Biomedical Research, Córdoba, Spain
| | - B Jurado Gámez
- Department of Respiratory Medicine, Reina Sofia University Hospital and IMIBIC, Maimonides Institute for Biomedical Research, Córdoba, Spain
| |
Collapse
|
18
|
Perkins MW, Wong B, Tressler J, Coggins A, Rodriguez A, Devorak J, Sciuto AM. Assessment of inhaled acute ammonia-induced lung injury in rats. Inhal Toxicol 2016; 28:71-9. [DOI: 10.3109/08958378.2015.1136715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
19
|
Stephenson-Brown A, Acton AL, Preece JA, Fossey JS, Mendes PM. Selective glycoprotein detection through covalent templating and allosteric click-imprinting. Chem Sci 2015; 6:5114-5119. [PMID: 29142730 PMCID: PMC5666680 DOI: 10.1039/c5sc02031j] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/15/2015] [Indexed: 01/05/2023] Open
Abstract
A hierarchical bottom-up route exploiting reversible covalent interactions with boronic acids and so-called click chemistry for selective glycoprotein detection is described. The self-assembled and imprinted surfaces confer high binding affinities, nanomolar sensitivity, exceptional glycoprotein specificity and selectivity.
Many glycoproteins are intimately linked to the onset and progression of numerous heritable or acquired diseases of humans, including cancer. Indeed the recognition of specific glycoproteins remains a significant challenge in analytical method and diagnostic development. Herein, a hierarchical bottom-up route exploiting reversible covalent interactions with boronic acids and so-called click chemistry for the fabrication of glycoprotein selective surfaces that surmount current antibody constraints is described. The self-assembled and imprinted surfaces, containing specific glycoprotein molecular recognition nanocavities, confer high binding affinities, nanomolar sensitivity, exceptional glycoprotein specificity and selectivity with as high as 30 fold selectivity for prostate specific antigen (PSA) over other glycoproteins. This synthetic, robust and highly selective recognition platform can be used in complex biological media and be recycled multiple times with no performance decrement.
Collapse
Affiliation(s)
- Alexander Stephenson-Brown
- School of Chemical Engineering , University of Birmingham , Edgbaston , Birmingham , West Midlands B15 2TT , UK .
| | - Aaron L Acton
- School of Chemical Engineering , University of Birmingham , Edgbaston , Birmingham , West Midlands B15 2TT , UK .
| | - Jon A Preece
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham , West Midlands B15 2TT , UK .
| | - John S Fossey
- School of Chemistry , University of Birmingham , Edgbaston , Birmingham , West Midlands B15 2TT , UK .
| | - Paula M Mendes
- School of Chemical Engineering , University of Birmingham , Edgbaston , Birmingham , West Midlands B15 2TT , UK .
| |
Collapse
|
20
|
Nguyen EV, Gharib SA, Schnapp LM, Goodlett DR. Shotgun MS proteomic analysis of bronchoalveolar lavage fluid in normal subjects. Proteomics Clin Appl 2015; 8:737-47. [PMID: 24616423 DOI: 10.1002/prca.201300018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 12/10/2013] [Accepted: 01/13/2014] [Indexed: 01/09/2023]
Abstract
We provide a review of proteomic techniques used to characterize the bronchoalveolar lavage fluid (BALF) proteome of normal healthy subjects. Bronchoalveolar lavage (BAL) is the most common technique for sampling the components of the alveolar space. The proteomic techniques used to study normal BALF include protein separation by 2DE, whereby proteins were identified by comparison to a reference gel as well as high pressure liquid chromatography (HPLC)-MS/MS, also known as shotgun proteomics. We summarize recent progress using shotgun MS technologies to define the normal BALF proteome. Surprisingly, we find that despite advances in shotgun proteomic technologies over the course of the last 10 years, which have resulted in greater numbers of proteins being identified, the functional landscape of normal BALF proteome was similarly described by all methods examined.
Collapse
Affiliation(s)
- Elizabeth V Nguyen
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
21
|
Bhargava M, Higgins L, Wendt CH, Ingbar DH. Application of clinical proteomics in acute respiratory distress syndrome. Clin Transl Med 2014; 3:34. [PMID: 26932378 PMCID: PMC4883989 DOI: 10.1186/s40169-014-0034-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/18/2014] [Indexed: 12/25/2022] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a devastating cause of hypoxic respiratory failure, which continues to have high mortality. It is expected that a comprehensive systems- level approach will identify global and complex changes that contribute to the development of ARDS and subsequent repair of the damaged lung. In the last decade, powerful genome-wide analytical and informatics tools have been developed, that have provided valuable insights into the mechanisms of complex diseases such as ARDS. These tools include the rapid and precise measure of gene expression at the proteomic level. This article reviews the contemporary proteomics platforms that are available for comprehensive studies in ARDS. The challenges of various biofluids that could be investigated and some of the studies performed are also discussed.
Collapse
Affiliation(s)
- Maneesh Bhargava
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, USA.
| | - LeeAnn Higgins
- Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA.
| | - Christine H Wendt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, USA. .,Minneapolis Veterans Affairs Medical Center, Minneapolis, MN, USA.
| | - David H Ingbar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, USA.
| |
Collapse
|
22
|
Haenen S, Clynen E, Nemery B, Hoet PH, Vanoirbeek JA. Biomarker discovery in asthma and COPD: Application of proteomics techniques in human and mice. EUPA OPEN PROTEOMICS 2014. [DOI: 10.1016/j.euprot.2014.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
23
|
|
24
|
Wiktorowicz JE, Jamaluddin M. Proteomic analysis of the asthmatic airway. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 795:221-32. [PMID: 24162912 DOI: 10.1007/978-1-4614-8603-9_14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Proteomic investigations in general utilize varied technologies for sample preparation, separations, quantification, protein identification, and biological rationalization. Their applications range from pure discovery and mechanistic studies to biomarker discovery/verification/validation. In each specific case, the analytical strategy to be implemented is tailored to the type of sample that serves as the target of the investigations. Proteomic investigations take into consideration sample complexity, the cellular heterogeneity (particularly from tissues), the potential dynamic range of the protein and peptide abundance within the sample, the likelihood of posttranslational modifications (PTM), and other important factors that might influence the final output of the study. We describe the sample types typically used for proteomic investigations into the biology of asthma and review the most recent related publications with special attention to those that deal with the unique airway samples such as bronchoalveolar lavage fluids (BALF), epithelial lining fluid and cells (ELF), induced sputum (IS), and exhaled breath condensate (EBC). Finally, we describe the newest proteomics approaches to sample preparation of the unique airway samples, BALF and IS.
Collapse
Affiliation(s)
- John E Wiktorowicz
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, 2.208A Basic Science Bldg, 301 University Blvd, Galveston, TX, 77555-0635, USA,
| | | |
Collapse
|
25
|
Govender P, Baugh JA, Pennington SR, Dunn MJ, Donnelly SC. Role of proteomics in the investigation of pulmonary fibrosis. Expert Rev Proteomics 2014; 4:379-88. [PMID: 17552922 DOI: 10.1586/14789450.4.3.379] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pulmonary fibrosis arises as a consequence of aberrant remodeling and defective repair mechanisms within the lung. This destructive process is the cause of much of the morbidity and mortality in many pulmonary disorders. Unfortunately, therapeutic options are limited. A significant advancement in the management of patients with pulmonary fibrosis would be the identification of biomarkers for diagnosis, prognosis and prediction of patient response to therapy. Bronchoalveolar lavage is an ideal tissue target for the discovery of these potential biomarkers in pulmonary fibrosis. Integrative approaches using both gel- and mass spectrometry-based proteomic workflows will allow full coverage of this complex proteome, thereby unlocking this potential information as a clinical tool to aid diagnosis and guide treatment for individual patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Praveen Govender
- University College Dublin, School of Medicine & Medical Science, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | | | | | | | | |
Collapse
|
26
|
|
27
|
Parker JC. Acute lung injury and pulmonary vascular permeability: use of transgenic models. Compr Physiol 2013; 1:835-82. [PMID: 23737205 DOI: 10.1002/cphy.c100013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute lung injury is a general term that describes injurious conditions that can range from mild interstitial edema to massive inflammatory tissue destruction. This review will cover theoretical considerations and quantitative and semi-quantitative methods for assessing edema formation and increased vascular permeability during lung injury. Pulmonary edema can be quantitated directly using gravimetric methods, or indirectly by descriptive microscopy, quantitative morphometric microscopy, altered lung mechanics, high-resolution computed tomography, magnetic resonance imaging, positron emission tomography, or x-ray films. Lung vascular permeability to fluid can be evaluated by measuring the filtration coefficient (Kf) and permeability to solutes evaluated from their blood to lung clearances. Albumin clearances can then be used to calculate specific permeability-surface area products (PS) and reflection coefficients (σ). These methods as applied to a wide variety of transgenic mice subjected to acute lung injury by hyperoxic exposure, sepsis, ischemia-reperfusion, acid aspiration, oleic acid infusion, repeated lung lavage, and bleomycin are reviewed. These commonly used animal models simulate features of the acute respiratory distress syndrome, and the preparation of genetically modified mice and their use for defining specific pathways in these disease models are outlined. Although the initiating events differ widely, many of the subsequent inflammatory processes causing lung injury and increased vascular permeability are surprisingly similar for many etiologies.
Collapse
Affiliation(s)
- James C Parker
- Department of Physiology, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
28
|
Association of high levels of α-defensins and S100A proteins with Candida mannan detection in bronchoalveolar lavage fluid of preterm neonates. Pediatr Res 2013; 74:19-25. [PMID: 23575874 DOI: 10.1038/pr.2013.60] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 01/09/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Candida mannan (Mn) detection in bronchoalveolar lavage fluid (BALF) was shown to be useful for earlier identification and preemptive therapy targeting in preterm infants at high risk of invasive Candida infection. We investigated whether early detection of Candida Mn in BALF is associated with the presence of some neutrophilic products, as markers of prenatal infection/inflammation. METHODS BALF specimens were collected during the first 48 h of life from mechanically ventilated preterm newborns. Samples were analyzed by high-performance liquid chromatography-electrospray ionization-mass spectrometry. The relative amounts of α-defensins 1-4 and S100A proteins were measured by extracted ion current peak area. Absolute and differential white cell counts in BALF were obtained. Mn antigen concentrations were determined by the Platelia Candida antigen kit. RESULTS Twenty-five studied neonates were divided into two groups: Mn-positive group and Mn-negative group. Levels of α-defensins 1-4 and S100A12 were significantly higher in the Mn-positive group than in the Mn-negative group. Moreover, positive significant correlations between the absolute number of neutrophils and the levels of α-defensins 1-4 and S100A8 were observed. CONCLUSION The detection of Mn antigen in BALF of preterm infants is consistent with evidence of an innate immune response in their lungs as demonstrated by higher levels of α-defensins and S100A proteins.
Collapse
|
29
|
Foster MW, Thompson JW, Que LG, Yang IV, Schwartz DA, Moseley MA, Marshall HE. Proteomic analysis of human bronchoalveolar lavage fluid after subsgemental exposure. J Proteome Res 2013; 12:2194-205. [PMID: 23550723 DOI: 10.1021/pr400066g] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The analysis of airway fluid, as sampled by bronchoalveolar lavage (BAL), provides a minimally invasive route to interrogate lung biology in health and disease. Here, we used immunodepletion, coupled with gel- and label-free LC-MS/MS, for quantitation of the BAL fluid (BALF) proteome in samples recovered from human subjects following bronchoscopic instillation of saline, lipopolysaccharide (LPS) or house dust mite antigen into three distinct lung subsegments. Among more than 200 unique proteins quantified across nine samples, neutrophil granule-derived and acute phase proteins were most highly enriched in the LPS-exposed lobes. Of these, peptidoglycan response protein 1 was validated and confirmed as a novel marker of neutrophilic inflammation. Compared to a prior transcriptomic analysis of airway cells in this same cohort, the BALF proteome revealed a novel set of response factors. Independent of exposure, the enrichment of tracheal-expressed proteins in right lower lung lobes suggests a potential for constitutive intralobar variability in the BALF proteome; sampling of multiple lung subsegments also appears to aid in the identification of protein signatures that differentiate individuals at baseline. Collectively, this proof-of-concept study validates a robust workflow for BALF proteomics and demonstrates the complementary nature of proteomic and genomic techniques for investigating airway (patho)physiology.
Collapse
Affiliation(s)
- Matthew W Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Nyquist PA, Wang H, Suffredini AF. Protein biomarkers in patients with subarachnoid hemorrhage, vasospasm, and delayed ischemic neurological deficits. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 115:23-5. [PMID: 22890638 PMCID: PMC10820842 DOI: 10.1007/978-3-7091-1192-5_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating neurological disease. It has many sequelae, including vasospasm and delayed ischemic neurological deficits (DINDs). We explored the blood proteome in patients with aSAH using transcranial Doppler (TCD) velocity as a guide to patients who are at risk for symptomatic vasospasm and DIND. Blood was drawn on all days that patients were observed in the neurocritical care unit (NCCU) after aSAH. A team of neurologists and neurosurgeons identified patients with clinical evidence of vasospasm and DIND. Serum was fractionated using protein chips and surface-enhanced laser desorption and ionization time-of-flight mass spectrometry (SELDI-TOF MS). We detected a pattern of protein expression associated with those at risk for elevated TCD velocities by day 8, compared with blood collected in the presymptomatic stage (days 1-3). We further analyzed serum using pooled samples from study entry to the time of elevated TCD velocities using a protein microarray that analyzed 500 human proteins thematically oriented toward inflammation. After identifying several candidates with elevated concentrations in the pooled samples, we then used reverse protein arrays to quantitate the concentration of potential candidate proteins in the individual samples. Proteins with significantly elevated concentrations included apolipoprotein-E, apolipoprotein-A, serum amyloid protein-4, and serum amyloid protein-P. Future studies in larger sample populations are needed to evaluate these biomarkers further as representative of biosystems involved in vasospasm and DIND or as potential biomarkers predictive of risk associated with disease.
Collapse
Affiliation(s)
- Paul A Nyquist
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | | | | |
Collapse
|
31
|
Colombo G, Clerici M, Giustarini D, Rossi R, Milzani A, Dalle-Donne I. Redox albuminomics: oxidized albumin in human diseases. Antioxid Redox Signal 2012; 17:1515-27. [PMID: 22587567 DOI: 10.1089/ars.2012.4702] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Albumin is the major contributor to colloid oncotic pressure and also serves as an important carrier protein of many endogenous and exogenous molecules throughout the body. In blood and extravascular fluids, albumin is susceptible to different oxidative modifications, especially thiol oxidation and carbonylation. Because of its metal-binding properties and the redox properties of its Cys34 thiol, albumin displays an important antioxidant activity. As albumin is the predominant protein in most body fluids, its Cys34 represents the largest fraction of free thiols within body fluids. RECENT ADVANCES Evidence that albumin oxidation takes place in vivo has been reported only recently. Different redox proteomic, mass spectrometric, and chromatographic techniques have shown albumin redox modifications in various human pathophysiological conditions. As a whole, most data here presented demonstrate that massive albumin oxidation occurs in vivo in different biological fluids and, to some extent, that this process is correlated to organ dysfunction. CRITICAL ISSUES Recent reports suggest that the albumin redox state may serve as a global biomarker for the redox state in the body in various human diseases. However, further study is required to elucidate the exact relationship between albumin oxidation and pathology. In addition, it is unknown if some albumin oxidized forms may also have diagnostic uses. FUTURE DIRECTIONS Application of specific redox proteomics techniques for the characterization of oxidized albumin forms in screening studies is required. A further challenge will be to analyze how these oxidative albumin modifications are related to real impact to the body.
Collapse
|
32
|
Kim SH, Choi GS, Nam YH, Kim JH, Hur GY, Kim SH, Park SM, Park HS. Role of vitamin D-binding protein in isocyanate-induced occupational asthma. Exp Mol Med 2012; 44:319-29. [PMID: 22314196 PMCID: PMC3366325 DOI: 10.3858/emm.2012.44.5.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The development of a serological marker for early diagnosis of isocyanate-induced occupational asthma (isocyanate-OA) may improve clinical outcome. Our previous proteomic study found that expression of vitamin D-binding protein (VDBP) was upregulated in the patients with isocyanate-OA. In the present study, we evaluated the clinical relevance of VDBP as a serological marker in screening for isocyanate-OA among exposed workers and its role in the pathogenesis of isocyanate-OA. Three study groups including 61 patients with isocyanate-OA (group I), 180 asymptomatic exposed controls (AECs, group II), 58 unexposed healthy controls (NCs, group III) were enrolled in this study. The baseline serum VDBP level was significantly higher in group I compared with groups II and III. The sensitivity and specificity for predicting the phenotype of isocyanate-OA with VDBP were 69% and 81%, respectively. The group I subjects with high VDBP (≥ 311 µg/ml) had significantly lower PC20 methacholine levels than did subjects with low VDBP. The in vitro studies showed that TDI suppressed the uptake of VDBP into RLE-6TN cells, which was mediated by the downregulation of megalin, an endocytic receptor of the 25-hydroxycholecalciferol-VDBP complex. Furthermore, toluene diisocyanate (TDI) increased VEGF production and secretion from this epithelial cells by suppression of 1,25-dihydroxycholecalciferol [1,25(OH)2D3] production. The findings of this study suggest that the serum VDBP level may be used as a serological marker for the detection of isocyanate-OA among workers exposed to isocyanate. The TDI-induced VEGF production/secretion was reversed by 1,25(OH)2D3 treatment, which may provide a potential therapeutic strategy for patients with isocyanate-OA.
Collapse
Affiliation(s)
- Sung-Ho Kim
- Department of Pharmacology and Chronic Inflammatory, Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Fibrotic lung injury is often attributed to a myriad of factors, including environmental exposure, age, genetic predisposition, epigenetics, coexisting conditions, acute lung injury, and viral infection. No effective therapies, other than lung transplantation, have proven effective against lung fibrosis. Loss of cellular homeostasis mechanisms in alveolar epithelial type I cells and any inability of type II progenitor cells to resist and repair epithelial injury are indicators that impaired response to injury and regeneration is a critical component of this disorder. The alveolar epithelium has a limited repertoire of responses to injury, which are dictated by the alveolar milieu, a repository of cytokines and growth factors that affect recruitment of other cells to the site of injury, or the proliferation of resident cells at the site of injury. The identification and characterization of the cytokines, growth factors, and other biomarkers that dictate the response to disease is key to understanding, diagnosing, treating, and determining the trajectory of various lung disorders. Corrective therapy of the alveolar milieu may therefore prove to be beneficial in many presently serious and incurable lung diseases that likely begin and progress with injury to the alveolar epithelium.
Collapse
|
34
|
Shigemura M, Nasuhara Y, Konno S, Shimizu C, Matsuno K, Yamguchi E, Nishimura M. Effects of molecular structural variants on serum Krebs von den Lungen-6 levels in sarcoidosis. J Transl Med 2012; 10:111. [PMID: 22650152 PMCID: PMC3394217 DOI: 10.1186/1479-5876-10-111] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 05/31/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Serum Krebs von den Lungen-6 (KL-6), which is classified as human mucin-1 (MUC1), is used as a marker of sarcoidosis and other interstitial lung diseases. However, there remain some limitations due to a lack of information on the factors contributing to increased levels of serum KL-6. This study was designed to investigate the factors contributing to increased levels of serum KL-6 by molecular analysis. METHODS Western blot analysis using anti-KL-6 antibody was performed simultaneously on the bronchoalveolar lavage fluid (BALF) and serum obtained from 128 subjects with sarcoidosis. RESULTS KL-6/MUC1 in BALF showed three bands and five band patterns. These band patterns were associated with the MUC1 genotype and the KL-6 levels. KL-6/MUC1 band patterns in serum were dependent on molecular size class in BALF. Significantly increased levels of serum KL-6, serum/BALF KL-6 ratio and serum soluble interleukin 2 receptor were observed in the subjects with influx of high molecular size KL-6/MUC1 from the alveoli to blood circulation. The multivariate linear regression analysis involving potentially relevant variables such as age, gender, smoking status, lung parenchymal involvement based on radiographical stage and molecular size of KL-6/MUC1 in serum showed that the molecular size of KL-6/MUC1 in serum was significant independent determinant of serum KL-6 levels. CONCLUSIONS The molecular structural variants of KL-6/MUC1 and its leakage behavior affect serum levels of KL-6 in sarcoidosis. This information may assist in the interpretation of serum KL-6 levels in sarcoidosis.
Collapse
Affiliation(s)
- Masahiko Shigemura
- First Department of Medicine, Hokkaido University School of Medicine, and Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
35
|
Jones T, Bérubé K. The bioreactivity of the sub-10 μm component of volcanic ash: Soufrière Hills volcano, Montserrat. JOURNAL OF HAZARDOUS MATERIALS 2011; 194:128-134. [PMID: 21872393 DOI: 10.1016/j.jhazmat.2011.07.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 07/05/2011] [Accepted: 07/21/2011] [Indexed: 05/31/2023]
Abstract
With the recent eruption of the Icelandic volcano Eyafallajökull and resulting ash cloud over much of Europe there was considerable concern about possible respiratory hazards. Volcanic ash can contain minerals that are known human respiratory health hazards such as cristobalite. Short-term ash exposures can cause skin sores, respiratory and ocular irritations and exacerbation of pre-existing lung conditions such as asthma. Long-term occupational level exposures to crystalline silicon dioxide can cause lung inflammation, oedema, fibrosis and cancer. The potential health effects would be dependent on factors including mineralogy, surface chemistry, size, and levels and duration of exposure. Bulk ash from the Soufrière Hills volcano was sourced and inhalable (<2.5 μm) ash samples prepared and physicochemically characterised. The fine ash samples were tested for bioreactivity by SDS-PAGE which determined the strength of binding between mineral grains and lung proteins. Selected proteins bound tightly to cristobalite, and bound loosely to other ash components. A positive correlation was seen between the amount of SiO(2) in the sample and the strength of the binding. The strength of binding is a function of the mineral's bioreactivity, and therefore, a potential geo-biomarker of respiratory risk.
Collapse
Affiliation(s)
- Timothy Jones
- School of Earth and Ocean Sciences, Cardiff University, Wales CF10 3YE, UK.
| | | |
Collapse
|
36
|
Outcome prediction in pneumonia induced ALI/ARDS by clinical features and peptide patterns of BALF determined by mass spectrometry. PLoS One 2011; 6:e25544. [PMID: 21991318 PMCID: PMC3184998 DOI: 10.1371/journal.pone.0025544] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 09/05/2011] [Indexed: 01/11/2023] Open
Abstract
Background Peptide patterns of bronchoalveolar lavage fluid (BALF) were assumed to reflect the complex pathology of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) better than clinical and inflammatory parameters and may be superior for outcome prediction. Methodology/Principal Findings A training group of patients suffering from ALI/ARDS was compiled from equal numbers of survivors and nonsurvivors. Clinical history, ventilation parameters, Murray's lung injury severity score (Murray's LISS) and interleukins in BALF were gathered. In addition, samples of bronchoalveolar lavage fluid were analyzed by means of hydrophobic chromatography and MALDI-ToF mass spectrometry (MALDI-ToF MS). Receiver operating characteristic (ROC) analysis for each clinical and cytokine parameter revealed interleukin-6>interleukin-8>diabetes mellitus>Murray's LISS as the best outcome predictors. Outcome predicted on the basis of BALF levels of interleukin-6 resulted in 79.4% accuracy, 82.7% sensitivity and 76.1% specificity (area under the ROC curve, AUC, 0.853). Both clinical parameters and cytokines as well as peptide patterns determined by MALDI-ToF MS were analyzed by classification and regression tree (CART) analysis and support vector machine (SVM) algorithms. CART analysis including Murray's LISS, interleukin-6 and interleukin-8 in combination was correct in 78.0%. MALDI-ToF MS of BALF peptides did not reveal a single identifiable biomarker for ARDS. However, classification of patients was successfully achieved based on the entire peptide pattern analyzed using SVM. This method resulted in 90% accuracy, 93.3% sensitivity and 86.7% specificity following a 10-fold cross validation (AUC = 0.953). Subsequent validation of the optimized SVM algorithm with a test group of patients with unknown prognosis yielded 87.5% accuracy, 83.3% sensitivity and 90.0% specificity. Conclusions/Significance MALDI-ToF MS peptide patterns of BALF, evaluated by appropriate mathematical methods can be of value in predicting outcome in pneumonia induced ALI/ARDS.
Collapse
|
37
|
Cornelius RM, Macri J, Brash JL. Interfacial interactions of apolipoprotein AI and high density lipoprotein: Overlooked phenomena in blood-material contact. J Biomed Mater Res A 2011; 99:109-15. [DOI: 10.1002/jbm.a.33169] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 04/28/2011] [Accepted: 04/29/2011] [Indexed: 02/05/2023]
|
38
|
Fertsch-Gapp S, Semmler-Behnke M, Wenk A, Kreyling WG. Binding of polystyrene and carbon black nanoparticles to blood serum proteins. Inhal Toxicol 2011; 23:468-75. [DOI: 10.3109/08958378.2011.583944] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Marwitz S, Abdullah M, Vock C, Fine JS, Visvanathan S, Gaede KI, Hauber HP, Zabel P, Goldmann T. HOPE-BAL: improved molecular diagnostics by application of a novel technique for fixation and paraffin embedding. J Histochem Cytochem 2011; 59:601-14. [PMID: 21430262 DOI: 10.1369/0022155411404417] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The bronchoalveolar lavage (BAL) and its cells have been widely used as a support for clinical diagnosis and as a versatile tool for research questions since many years. Because there are no sufficient possibilities of long-term storage, the authors explore in this study the utility of a new fixative for fixation and paraffin embedding of human lavage cells with the possibility of implementing standard molecular biology techniques. HOPE-fixed, paraffin-embedded BAL cells of patients with different lung diseases (asthma, chronic obstructive pulmonary diseases, tuberculosis, sarcoidosis, emphysema, and fibrosis) were subjected to immunohistochemistry, in situ hybridization, quantitative polymerase chain reaction, and transcription microarray analysis. Furthermore, two-dimensional gel electrophoresis was conducted to evaluate the range of possible applications for research, diagnostics, and further implementing in biobanks. The authors show, by targeting some exemplary molecules, the power of screening and validating HOPE-BAL for new biomarkers. The transforming growth factor β signaling pathway may play a central role in immunomodulation upon infection as well as asthma. Furthermore, haptoglobin was overexpressed in asthma and sarcoidosis. Because of the excellent preservation of nucleic acids, protein, and morphologic structures, HOPE-BAL is a step forward into enhanced molecular diagnostics and biobanking in pulmonary medicine.
Collapse
Affiliation(s)
- Sebastian Marwitz
- Research Center Borstel, Clinical and Experimental Pathology, Borstel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gould NS, Day BJ. Targeting maladaptive glutathione responses in lung disease. Biochem Pharmacol 2011; 81:187-93. [PMID: 20951119 PMCID: PMC3039114 DOI: 10.1016/j.bcp.2010.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/28/2010] [Accepted: 10/01/2010] [Indexed: 11/20/2022]
Abstract
The lung is unique being exposed directly to the atmospheric environment containing xenobiotics, pathogens, and other agents which are continuously inhaled on a daily basis. Additionally, the lung is exposed to higher ambient oxygen levels which can promote the formation of a complex number of reactive oxygen and nitrogen species. Due to this constant barrage of potential damaging agents, the lung has developed a high degree of plasticity in dealing with ever changing conditions. In the present commentary, we will focus on glutathione (GSH) as a key antioxidant in the lung airways and discuss mechanisms by which the lung uses GSH to adapt to its rapidly changing environment. We will then examine the evidence on how defective and inadequate adaptive responses can lead to lung injury, inflammation and disease. Lastly, we will examine some of the recent attempts to alter lung GSH levels with therapies in a number of human lung diseases and discuss some of the limitations of such approaches.
Collapse
Affiliation(s)
- Neal S Gould
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | | |
Collapse
|
41
|
Bloemen K, Van Den Heuvel R, Govarts E, Hooyberghs J, Nelen V, Witters E, Desager K, Schoeters G. A new approach to study exhaled proteins as potential biomarkers for asthma. Clin Exp Allergy 2010; 41:346-56. [PMID: 21105917 DOI: 10.1111/j.1365-2222.2010.03638.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Asthma is a complex clinical disease characterized by airway inflammation. Recently, various studies reported on the analysis of exhaled breath condensate (EBC) in the search for potential biomarkers for asthma. However, in a complex disease such as asthma, one biomarker might not be enough for early diagnosis or follow-up. OBJECTIVE The use of proteome analysis may reveal disease-specific proteolytic peptide or protein patterns, and may lead to the identification of novel proteins for the detection of asthma. METHODS Liquid chromatography and mass spectrometry were used to separate and detect proteins (proteolytic peptides) present in EBC samples from 30 healthy children and 40 children with asthma in the age group of 6-12 years. RESULTS Support vector machine analysis resulted in differentiating profiles based on asthma status. These proteolytic peptide patterns were not correlated to some well known (spirometry, exhaled nitric oxide) and more recently described exhaled markers (EBC pH, LTB₄). The more abundant proteins in EBC were identified as cytokeratins, albumin, actin, haemoglobin, lysozyme, dermcidin, and calgranulin B. CONCLUSION Although the exact role in the disease development or physiological state of the airways of the proteins described in the presented pattern is not clear at this moment, this is an important step in the search for exhaled biomarkers for asthma. This study shows that EBC contains proteins that are of interest for future non-invasive asthma diagnosis or follow-up.
Collapse
Affiliation(s)
- K Bloemen
- Flemish Institute for Technological Research (VITO), Environmental Risk and Health, Toxicology, Mol, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Gould NS, Min E, Gauthier S, Chu HW, Martin R, Day BJ. Aging adversely affects the cigarette smoke-induced glutathione adaptive response in the lung. Am J Respir Crit Care Med 2010; 182:1114-22. [PMID: 20622027 PMCID: PMC3001254 DOI: 10.1164/rccm.201003-0442oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 07/08/2010] [Indexed: 01/25/2023] Open
Abstract
RATIONALE Cigarette smoke (CS) is the leading cause of chronic obstructive pulmonary disease, accounting for more than 90% of cases. The prevalence of chronic obstructive pulmonary disease is much higher in the elderly, suggesting an age dependency. A prominent defense against the oxidant burden caused by CS is the glutathione (GSH) adaptive response in the lung epithelial lining fluid (ELF) and tissue. However, as one ages the ability to maintain GSH levels declines. OBJECTIVES Examine the effect of aging on the GSH adaptive response to CS and resulting lung sensitization to inflammation and oxidation. METHODS Both young (2 mo old) and aged (8, 13, 19, and 26 mo old) mice were used to study the effects of age on the GSH adaptive response after an acute exposure to CS. MEASUREMENTS AND MAIN RESULTS Young mice had a robust sixfold increase in ELF GSH after a single exposure to CS. The GSH response to CS decreased as a function of age and diminishes in the older mice to only a twofold increase over air controls. As a consequence, levels of CS-induced tumor necrosis factor-α and nitric oxide synthase, markers of inflammation, and 8-hydroxy-2-deoxyguanosine, a marker of DNA oxidation, were elevated in the aged mice compared with the young mice. Additionally, depletion of ELF GSH with buthionine sulfoximine in young mice recapitulated changes in ELF tumor necrosis factor-α as seen in old mice. CONCLUSIONS These data suggest that the age-related maladaptive response to CS sensitizes the lung to both inflammation and oxidation potentially contributing to the development of CS-induced chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Neal S. Gould
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Pharmaceutical Sciences, Department of Medicine, and Department of Immunology, University of Colorado Denver, Aurora, Colorado
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Pharmaceutical Sciences, Department of Medicine, and Department of Immunology, University of Colorado Denver, Aurora, Colorado
| | - Steven Gauthier
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Pharmaceutical Sciences, Department of Medicine, and Department of Immunology, University of Colorado Denver, Aurora, Colorado
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Pharmaceutical Sciences, Department of Medicine, and Department of Immunology, University of Colorado Denver, Aurora, Colorado
| | - Richard Martin
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Pharmaceutical Sciences, Department of Medicine, and Department of Immunology, University of Colorado Denver, Aurora, Colorado
| | - Brian J. Day
- Department of Medicine, National Jewish Health, Denver, Colorado; Department of Pharmaceutical Sciences, Department of Medicine, and Department of Immunology, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
43
|
Haenen S, Vanoirbeek JAJ, De Vooght V, Maes E, Schoofs L, Nemery B, Hoet PHM, Clynen E. Proteome analysis of multiple compartments in a mouse model of chemical-induced asthma. J Proteome Res 2010; 9:5868-76. [PMID: 20860378 DOI: 10.1021/pr100638m] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Occupational asthma is the principal cause of work-related respiratory disease in the industrial world. Toluene-2,4-diisocyanate (TDI) is one of the most common respiratory sensitizers leading to occupational asthma. Using a mouse model of chemical-induced asthma, we explored proteome changes in multiple compartments of mice sensitized and challenged with TDI or acetone-olive oil (AOO; vehicle). Airway reactivity to methacholine and a bronchoalveolar lavage (BAL) cell count was assessed in treated and control mice, 1 day after challenge. Subsequently, two-dimensional differential gel electrophoresis (2D-DIGE) was performed on auricular lymph nodes, BAL, and serum comparing TDI-treated and vehicle-treated control mice. The differentially expressed proteins were identified by mass spectrometry and pathway analysis was performed. TDI-treated mice exhibit increased airway reactivity (2.6-fold increase) and a neutrophilic inflammation in the BAL fluid, compared to control mice. 2D-DIGE showed 53, 210, and 40 differentially expressed proteins in the auricular lymph nodes, BAL, and serum of TDI-treated versus vehicle-treated mice, respectively. Several of the identified proteins could be linked with inflammation, neutrophil chemotaxis, and/or oxidative stress. Physiologic and immunologic readouts of the asthmatic phenotype, such as inflammation, were confirmed in three compartments by several of the differentially expressed proteins via 2D-DIGE and computerized pathway analysis.
Collapse
Affiliation(s)
- Steven Haenen
- Katholieke Universiteit Leuven, Research Unit Lung Toxicology, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Proteomics has the goal of defining the complete protein complement of biological systems, which can then be analyzed in a comparative fashion to generate informative data regarding protein expression and function. Proteomic analyses can also facilitate the discovery of biomarkers that can be used to diagnose and monitor disease severity, activity and therapeutic response, as well as to identify new targets for drug development. A major challenge for proteomics, however, has been detecting low-abundance proteins in complex biological fluids. This review summarizes how proteomic analyses have advanced lung cell biology and facilitated the identification of new mechanisms of disease pathogenesis in respiratory disorders, such as asthma, cystic fibrosis, lung cancer, acute lung injury and sarcoidosis. The impact of nanotechnology and microfluidics, as well as studies of post-translational modifications and protein-protein interactions (the interactome), are considered. Furthermore, the application of systems-biology approaches to organize and analyze data regarding the lung proteome, interactome, genome, transcriptome, metabolome, glycome and small RNAome (regulatory RNAs), should facilitate future conceptual advances regarding lung cell biology, disease pathogenesis, biomarker discovery and drug development.
Collapse
Affiliation(s)
- Stewart J Levine
- National Institutes of Health, Pulmonary-Critical Care Medicine Branch, NHLBI, Building 10, Room 6D03, MSC 1590, Bethesda, MD 0892-1590, USA.
| |
Collapse
|
45
|
Abstract
Proteomic approaches have already been successfully implemented in areas such as cancer research. Surprisingly, only a few proteomics analyses have been published reporting on the protein profiles associated with asthma. Although proteomics has its limitations and experimental challenges, it can successfully contribute to the understanding of a complex disease such as asthma. We have reviewed the current literature that has reported the use of proteomic techniques to identify proteins that may contribute to altered lung function in asthma. Only a few of these studies have used proteomic techniques on human tissues associated with asthma, while most research has been performed with animal models of asthma. Proteomic applications have been used as a complimentary technique to verify the suspected candidate proteins involved in asthma. In addition, novel proteins have been identified as potential therapeutic targets. Future collaboration between the different scientific disciplines using proteomic studies of animal models of asthma and confirmation of these findings in human tissues will significantly contribute to the understanding of the etiology of asthma and lead to the development of new therapeutic strategies for this highly prevalent disease.
Collapse
Affiliation(s)
- Annette Osei-Kumah
- Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, University of Adelaide, SA 5005, Australia.
| | | | | |
Collapse
|
46
|
Chiu KH, Lee WLW, Chang CC, Chen SC, Chang YC, Ho MN, Hsu JF, Liao PC. A label-free differential proteomic analysis of mouse bronchoalveolar lavage fluid exposed to ultrafine carbon black. Anal Chim Acta 2010; 673:160-6. [PMID: 20599030 DOI: 10.1016/j.aca.2010.05.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 05/23/2010] [Accepted: 05/30/2010] [Indexed: 10/19/2022]
Abstract
Ultrafine carbon black (ufCB) is a potential hazard to the lung. It causes changes in protein expression and it increases alveolar-capillary permeability in the lung. Label-free quantitative proteomic methods allow a sensitive and accurate analytical method for identifying and quantifying proteins in a protein mixture without chemically modifying the proteins. We used a label-free quantitative proteomic approach that combined and aligned LC-MS and LC-MS/MS spectra to analyze mouse bronchoalveolar lavage fluid (BALF) protein changes associated with exposure to ufCB. We developed a simple normalization method for quantification without spiking the internal standard. The intensities of unchanged peptides were used as normalization factors based on a statistical method to avoid the influence of peptides changed because of ufCB. LC-MS/MS spectra and then database searching were used to identify proteins. The relative abundances of the aligned peptides of identified proteins were determined using LC-MS spectra. We identified 132 proteins, of which 77 are reported for the first time. In addition, the expression of 15 inflammatory proteins and surfactant-associated proteins was regulated (i.e., 7 upregulated and 8 downregulated) compared with the controls. Several proteins not previously reported provide complementary information on the proteins present in mouse BALF, and they are potential biomarkers for the understanding of mechanisms involved in ufCB-induced lung disorders hypothesize that using the label-free quantitative proteomic approach introduced here is well suited for more rigorous, large-scale quantitative analysis of biological samples. We hypothesize that this label-free quantitative proteomic approach will be suited for a large-scale quantitative analysis of biological samples.
Collapse
Affiliation(s)
- Kuo-Hsun Chiu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ahn KH, Kim SK, Lee JM, Jeon HJ, Lee DH, Kim DK. Proteomic Analysis of Bronchoalveolar Lavage Fluid Obtained from Rats Exposed to Formaldehyde. ACTA ACUST UNITED AC 2010. [DOI: 10.1248/jhs.56.287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kyong Hoon Ahn
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University
| | - Seok Kyun Kim
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University
| | - Jung Min Lee
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University
| | - Hyung Jun Jeon
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University
| | - Dong Hoon Lee
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University
| | - Dae Kyong Kim
- Department of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University
| |
Collapse
|
48
|
Abstract
Sputum is recognized as a sampling method for the monitoring and assessment of chronic lung diseases such as asthma, COPD (chronic obstructive pulmonary disease) and cystic fibrosis. Sputum samples the central airways and its protein components (e.g. mucins and cytokines), cellular components (e.g. eosinophils and neutrophils) and microbiological components (e.g. viruses and bacteria) can be used as markers of disease severity, exacerbation, susceptibility or progression. This paper describes the basic constituents of induced sputum and how these influence the quantification and identification of novel biomarkers of chronic lung diseases using techniques such as proteomics.
Collapse
|
49
|
Nicholas BL, O'Connor CD, Djukanovic R. From Proteomics to Prescription—The Search for COPD Biomarkers. COPD 2009; 6:298-303. [DOI: 10.1080/15412550903049140] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
50
|
|