1
|
Bartlett JA, Huntemann ED, Krishnamurthy S, Hartwig SM, Pewa A, Thurman AL, Chimenti MS, Taylor EB, Varga SM, McCray PB. CF airway epithelia display exaggerated host defense responses and prolonged cilia loss during RSV infection. J Cyst Fibros 2025:S1569-1993(25)00055-4. [PMID: 39956716 DOI: 10.1016/j.jcf.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND In individuals with cystic fibrosis (CF), respiratory viral infections frequently result in hospitalization and have been linked to secondary bacterial infection and colonization, highlighting viral infections as possible contributors to CF lung disease progression. We hypothesized that expression of antiviral host defense genes is dysregulated in CF airway epithelia. METHODS We infected primary CF and Non-CF airway epithelia with respiratory syncytial virus (RSV) and characterized their responses at 12 hr, 24 hr, 48 hr, 72 hr, and 120 hr post infection (hpi) by RNA sequencing (RNAseq). RESULTS Our analysis revealed strikingly different gene expression profiles for the CF and Non-CF epithelia over the course of the infection. While both CF and Non-CF cells exhibited an early signature for interferon signaling and antiviral defense pathways, this response was relatively exaggerated and sustained in CF epithelia. We also observed, in both genotypes, a transient downregulation of cilia-associated genes and loss of ciliary activity by 72 hpi. Interestingly, recovery of cilia activity was delayed in the CF epithelia. CONCLUSIONS These findings further our understanding of innate immune dysfunction in the CF airway epithelium and suggest that virus-induced cilia injury may further compromise host defenses in CF airways.
Collapse
Affiliation(s)
- Jennifer A Bartlett
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Eric D Huntemann
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sateesh Krishnamurthy
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Stacey M Hartwig
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Alvin Pewa
- Fraternal Order of Eagles Diabetes Research Center Metabolomics Core Facility, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Andrew L Thurman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Eric B Taylor
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Steven M Varga
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Paul B McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
2
|
Basu R, Elmendorf AJ, Lorentz B, Mahler CA, Lazzaro O, App B, Zhou S, Yamamoto Y, Suber M, Wann JC, Roh HC, Sheets PL, Johnson TS, Flak JN. Ventromedial hypothalamic nucleus subset stimulates tissue thermogenesis via preoptic area outputs. Mol Metab 2024; 84:101951. [PMID: 38729241 PMCID: PMC11112375 DOI: 10.1016/j.molmet.2024.101951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/20/2024] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVE Hypothalamic signals potently stimulate energy expenditure by engaging peripheral mechanisms to restore energy homeostasis. Previous studies have identified several critical hypothalamic sites (e.g. preoptic area (POA) and ventromedial hypothalamic nucleus (VMN)) that could be part of an interconnected neurocircuit that controls tissue thermogenesis and essential for body weight control. However, the key neurocircuit that can stimulate energy expenditure has not yet been established. METHODS Here, we investigated the downstream mechanisms by which VMN neurons stimulate adipose tissue thermogenesis. We manipulated subsets of VMN neurons acutely as well as chronically and studied its effect on tissue thermogenesis and body weight control, using Sf1Cre and Adcyap1Cre mice and measured physiological parameters under both high-fat diet and standard chow diet conditions. To determine the node efferent to these VMN neurons, that is involved in modulating energy expenditure, we employed electrophysiology and optogenetics experiments combined with measurements using tissue-implantable temperature microchips. RESULTS Activation of the VMN neurons that express the steroidogenic factor 1 (Sf1; VMNSf1 neurons) reduced body weight, adiposity and increased energy expenditure in diet-induced obese mice. This function is likely mediated, at least in part, by the release of the pituitary adenylate cyclase-activating polypeptide (PACAP; encoded by the Adcyap1 gene) by the VMN neurons, since we previously demonstrated that PACAP, at the VMN, plays a key role in energy expenditure control. Thus, we then shifted focus to the subpopulation of VMNSf1 neurons that contain the neuropeptide PACAP (VMNPACAP neurons). Since the VMN neurons do not directly project to the peripheral tissues, we traced the location of the VMNPACAP neurons' efferents. We identified that VMNPACAP neurons project to and activate neurons in the caudal regions of the POA whereby these projections stimulate tissue thermogenesis in brown and beige adipose tissue. We demonstrated that selective activation of caudal POA projections from VMNPACAP neurons induces tissue thermogenesis, most potently in negative energy balance and activating these projections lead to some similar, but mostly unique, patterns of gene expression in brown and beige tissue. Finally, we demonstrated that the activation of the VMNPACAP neurons' efferents that lie at the caudal POA are necessary for inducing tissue thermogenesis in brown and beige adipose tissue. CONCLUSIONS These data indicate that VMNPACAP connections with the caudal POA neurons impact adipose tissue function and are important for induction of tissue thermogenesis. Our data suggests that the VMNPACAP → caudal POA neurocircuit and its components are critical for controlling energy balance by activating energy expenditure and body weight control.
Collapse
Affiliation(s)
- Rashmita Basu
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Andrew J Elmendorf
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Betty Lorentz
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Connor A Mahler
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Olivia Lazzaro
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Britany App
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Shudi Zhou
- Department of Medical Neuroscience, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yura Yamamoto
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Mya Suber
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Jamie C Wann
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hyun Cheol Roh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Patrick L Sheets
- Department of Medical Neuroscience, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Travis S Johnson
- Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA; Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA; Melvin and Bren Simon Comprehensive Cancer Centre, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonathan N Flak
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Lilly Diabetes Research Center, Indiana Biosciences Research Institute, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Bae H, Kim BR, Jung S, Le J, van der Heide D, Yu W, Park SH, Hilkin BM, Gansemer ND, Powers LS, Kang T, Meyerholz DK, Schuster VL, Jang C, Welsh MJ. Arteriovenous metabolomics in pigs reveals CFTR regulation of metabolism in multiple organs. J Clin Invest 2024; 134:e174500. [PMID: 38743489 PMCID: PMC11213515 DOI: 10.1172/jci174500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), a multiorgan disease that is characterized by diverse metabolic defects. However, other than specific CFTR mutations, the factors that influence disease progression and severity remain poorly understood. Aberrant metabolite levels have been reported, but whether CFTR loss itself or secondary abnormalities (infection, inflammation, malnutrition, and various treatments) drive metabolic defects is uncertain. Here, we implemented comprehensive arteriovenous metabolomics in newborn CF pigs, and the results revealed CFTR as a bona fide regulator of metabolism. CFTR loss impaired metabolite exchange across organs, including disruption of lung uptake of fatty acids, yet enhancement of uptake of arachidonic acid, a precursor of proinflammatory cytokines. CFTR loss also impaired kidney reabsorption of amino acids and lactate and abolished renal glucose homeostasis. These and additional unexpected metabolic defects prior to disease manifestations reveal a fundamental role for CFTR in controlling multiorgan metabolism. Such discovery informs a basic understanding of CF, provides a foundation for future investigation, and has implications for developing therapies targeting only a single tissue.
Collapse
Affiliation(s)
- Hosung Bae
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Bo Ram Kim
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | | | - Wenjie Yu
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Sang Hee Park
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Brieanna M. Hilkin
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Nicholas D. Gansemer
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Linda S. Powers
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Taekyung Kang
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - David K. Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Victor L. Schuster
- Department of Internal Medicine, Albert Einstein College of Medicine, Bronx, New York, New York, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
- Center for Complex Biological Systems and
- Center for Epigenetics and Metabolism, University of California – Irvine, Irvine, California, USA
| | - Michael J. Welsh
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
- Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
4
|
Zhang Z, Wang J, Li Y, Liu F, Chen L, He S, Lin F, Wei X, Fang Y, Li Q, Zhou J, Lu W. Proteomics and metabolomics profiling reveal panels of circulating diagnostic biomarkers and molecular subtypes in stable COPD. Respir Res 2023; 24:73. [PMID: 36899372 PMCID: PMC10007826 DOI: 10.1186/s12931-023-02349-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/27/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a complex and heterogeneous disease with high morbidity and mortality, especially in advanced patients. We aimed to develop multi-omics panels of biomarkers for the diagnosis and explore its molecular subtypes. METHODS A total of 40 stable patients with advanced COPD and 40 controls were enrolled in the study. Proteomics and metabolomics techniques were applied to identify potential biomarkers. An additional 29 COPD and 31 controls were enrolled for validation of the obtained proteomic signatures. Information on demographic, clinical manifestation, and blood test were collected. The ROC analyses were carried out to evaluate the diagnostic performance, and experimentally validated the final biomarkers on mild-to-moderate COPD. Next, molecular subtyping was performed using proteomics data. RESULTS Theophylline, palmitoylethanolamide, hypoxanthine, and cadherin 5 (CDH5) could effectively diagnose advanced COPD with high accuracy (auROC = 0.98, sensitivity of 0.94, and specificity of 0.95). The performance of the diagnostic panel was superior to that of other single/combined results and blood tests. Proteome based stratification of COPD revealed three subtypes (I-III) related to different clinical outcomes and molecular feature: simplex COPD, COPD co-existing with bronchiectasis, and COPD largely co-existing with metabolic syndrome, respectively. Two discriminant models were established using the auROC of 0.96 (Principal Component Analysis, PCA) and 0.95 (the combination of RRM1 + SUPV3L1 + KRT78) in differentiating COPD and COPD with co-morbidities. Theophylline and CDH5 were exclusively elevated in advanced COPD but not in its mild form. CONCLUSIONS This integrative multi-omics analysis provides a more comprehensive understanding of the molecular landscape of advanced COPD, which may suggest molecular targets for specialized therapy.
Collapse
Affiliation(s)
- Zili Zhang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Laboratory, Guangzhou, 510005, Guangdong, China
| | - Yuanyuan Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fei Liu
- Department of Respiratory and Critical Care, Shaoguan First People's Hospital, Shaoguan, Guangdong, China
| | - Lingdan Chen
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shunping He
- Department of Respiratory and Critical Care, Shaoguan First People's Hospital, Shaoguan, Guangdong, China
| | - Fanjie Lin
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinguang Wei
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yaowei Fang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiongqiong Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Juntuo Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100083, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Weber R, Perkins N, Bruderer T, Micic S, Moeller A. Identification of Exhaled Metabolites in Children with Cystic Fibrosis. Metabolites 2022; 12:980. [PMID: 36295881 PMCID: PMC9611656 DOI: 10.3390/metabo12100980] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
The early detection of inflammation and infection is important to prevent irreversible lung damage in cystic fibrosis. Novel and non-invasive monitoring tools would be of high benefit for the quality of life of patients. Our group previously detected over 100 exhaled mass-to-charge (m/z) features, using on-line secondary electrospray ionization high-resolution mass spectrometry (SESI-HRMS), which distinguish children with cystic fibrosis from healthy controls. The aim of this study was to annotate as many m/z features as possible with putative chemical structures. Compound identification was performed by applying a rigorous workflow, which included the analysis of on-line MS2 spectra and a literature comparison. A total of 49 discriminatory exhaled compounds were putatively identified. A group of compounds including glycolic acid, glyceric acid and xanthine were elevated in the cystic fibrosis group. A large group of acylcarnitines and aldehydes were found to be decreased in cystic fibrosis. The proposed compound identification workflow was used to identify signatures of volatile organic compounds that discriminate children with cystic fibrosis from healthy controls, which is the first step for future non-invasive and personalized applications.
Collapse
Affiliation(s)
- Ronja Weber
- Department of Respiratory Medicine and Childhood Research Center, University Children’s Hospital Zurich, Steinwiesstrasse 75, 8032 Zurich, Switzerland
| | - Nathan Perkins
- Division of Clinical Chemistry and Biochemistry, University of Zurich, Steinwiesstrasse 75, 8032 Zurich, Switzerland
| | - Tobias Bruderer
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy
| | - Srdjan Micic
- Department of Respiratory Medicine and Childhood Research Center, University Children’s Hospital Zurich, Steinwiesstrasse 75, 8032 Zurich, Switzerland
| | - Alexander Moeller
- Department of Respiratory Medicine and Childhood Research Center, University Children’s Hospital Zurich, Steinwiesstrasse 75, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Raemistrasse 71, 8006 Zurich, Switzerland
| |
Collapse
|
6
|
Woodley FW, Gecili E, Szczesniak RD, Shrestha CL, Nemastil CJ, Kopp BT, Hayes D. Sweat metabolomics before and after intravenous antibiotics for pulmonary exacerbation in people with cystic fibrosis. Respir Med 2022; 191:106687. [PMID: 34864373 PMCID: PMC8810598 DOI: 10.1016/j.rmed.2021.106687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/06/2021] [Accepted: 11/20/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND People with cystic fibrosis (PWCF) suffer from acute unpredictable reductions in pulmonary function associated with a pulmonary exacerbation (PEx) that may require hospitalization. PEx symptoms vary between PWCF without universal diagnostic criteria for diagnosis and response to treatment. RESEARCH QUESTION We characterized sweat metabolomes before and after intravenous (IV) antibiotics in PWCF hospitalized for PEx to determine feasibility and define biological alterations by IV antibiotics for PEx. STUDY DESIGN AND METHODS PWCF with PEx requiring hospitalization for IV antibiotics were recruited from clinic. Sweat samples were collected using the Macroduct® Sweat Collection System at admission prior to initiation of IV antibiotics and after completion prior to discharge. Samples were analyzed for metabolite changes using ultra-high-performance liquid chromatography/tandem accurate mass spectrometry. RESULTS Twenty-six of 29 hospitalized PWCF completed the entire study. A total of 326 compounds of known identity were detected in sweat samples. Of detected metabolites, 147 were significantly different between pre-initiation and post-completion of IV antibiotics for PEx (average treatment 14 days). Global sweat metabolomes changed from before and after IV antibiotic treatment. We discovered specific metabolite profiles predictive of PEx status as well as enriched biologic pathways associated with PEx. However, metabolomic changes were similar in PWCF who failed to return to baseline pulmonary function and those who did not. INTERPRETATION Our findings demonstrate the feasibility of non-invasive sweat metabolomic profiling in PWCF and the potential for sweat metabolomics as a prospective diagnostic and research tool to further advance our understanding of PEx in PWCF.
Collapse
Affiliation(s)
- Frederick W. Woodley
- Division of Gastroenterology, Hepatology and Nutrition, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Emrah Gecili
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rhonda D. Szczesniak
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA,Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chandra L. Shrestha
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Christopher J. Nemastil
- Division of Pulmonary Medicine, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Benjamin T. Kopp
- Division of Pulmonary Medicine, Nationwide Children’s Hospital and The Ohio State University College of Medicine, Columbus, OH, USA,Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Don Hayes
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
7
|
Van den Bossche S, De Broe E, Coenye T, Van Braeckel E, Crabbé A. The cystic fibrosis lung microenvironment alters antibiotic activity: causes and effects. Eur Respir Rev 2021; 30:30/161/210055. [PMID: 34526313 DOI: 10.1183/16000617.0055-2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic airway colonisation by Pseudomonas aeruginosa, a hallmark of cystic fibrosis (CF) lung disease, is associated with increased morbidity and mortality and despite aggressive antibiotic treatment, P. aeruginosa is able to persist in CF airways. In vitro antibiotic susceptibility assays are poor predictors of antibiotic efficacy to treat respiratory tract infections in the CF patient population and the selection of the antibiotic(s) is often made on an empirical base. In the current review, we discuss the factors that are responsible for the discrepancies between antibiotic activity in vitro and clinical efficacy in vivo We describe how the CF lung microenvironment, shaped by host factors (such as iron, mucus, immune mediators and oxygen availability) and the microbiota, influences antibiotic activity and varies widely between patients. A better understanding of the CF microenvironment and population diversity may thus help improve in vitro antibiotic susceptibility testing and clinical decision making, in turn increasing the success rate of antibiotic treatment.
Collapse
Affiliation(s)
| | - Emma De Broe
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Eva Van Braeckel
- Dept of Respiratory Medicine, Cystic Fibrosis Reference Centre, Ghent University Hospital, Ghent, Belgium.,Dept of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
8
|
Wang T, Li S, Wu Y, Yan X, Zhu Y, Jiang Y, Jiang F, Liu W. Mechanistic Investigation of Xuebijing for Treatment of Paraquat-Induced Pulmonary Fibrosis by Metabolomics and Network Pharmacology. ACS OMEGA 2021; 6:19717-19730. [PMID: 34368559 PMCID: PMC8340419 DOI: 10.1021/acsomega.1c02370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
After paraquat (PQ) poisoning, it is difficult to accurately diagnose patients' condition by only measuring their blood PQ concentration. Therefore, it is important to establish an accurate method to assist in the diagnosis of PQ poisoning, especially in the early stages. In this study, a gas chromatography-mass spectrometry (GC-MS) metabonomics strategy was established to obtain metabolite information. A random forest algorithm was used to search for potential biomarkers of PQ poisoning, and data mining and network pharmacological analysis were used to evaluate the active components, drug-disease targets, and key pathways of Xuebijing (XBJ) injection in the treatment of PQ-induced pulmonary fibrosis. Targets from the network pharmacology analysis and metabolites from plasma metabolomics were jointly analyzed to select crucial metabolic pathways. Finally, molecular docking technology and in vitro experiments were used to verify the pathway targets to further reveal the potential mechanisms underlying the antipulmonary fibrosis effect of XBJ. Metabonomics studies showed that l-valine, glycine, citric acid, d-mannose, d-galactose, maltose, l-tryptophan, and arachidonic acid contributed more to the differentiation of different groups than other metabolites. Compared with the control group, the PQ poisoning group had higher levels of l-valine, glycine, citric acid, l-tryptophan, and arachidonic acid, and lower levels of d-mannose, d-galactose, and maltose. After treatment with XBJ injection, the relative levels of these metabolites were reversed. The network pharmacological analysis screened a total of 180 targets, mainly involving multiple signaling pathways and metabolic pathways, which jointly played an antipulmonary fibrosis effect. Based on the combined analysis of 180 targets and 8 different metabolites, arachidonic acid metabolism was selected as the key metabolic pathway. Molecular docking analysis showed that the XBJ compound had strong binding activity with the target protein. Western blot results showed that XBJ injection could reduce the inflammatory response by downregulating the expressions of p-p65, p-IKBα, and p-IKKβ, thus inhibiting the development of PQ-induced pulmonary fibrosis. In summary, the combined results from metabolomics and network pharmacology studies showed that Xuebijing has the characteristics of multitarget, multichannel, and multicomponent action in the treatment of pulmonary fibrosis caused by PQ.
Collapse
Affiliation(s)
- Tongtong Wang
- Department
of Pharmacy, The First Affiliate Hospital
of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China
| | - Sha Li
- Department
of Pharmacy, Changsha Stomatological Hospital, Changsha 410005, China
| | - Yangke Wu
- Department
of Pharmacy, The First Affiliate Hospital
of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China
| | - Xiao Yan
- Department
of Pharmacy, The First Affiliate Hospital
of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China
| | - Yiming Zhu
- Department
of Pharmacy, The First Affiliate Hospital
of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China
| | - Yu Jiang
- Hunan
Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Changsha 410005, China
| | - Feiya Jiang
- Department
of Pharmacy, The First Affiliate Hospital
of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China
| | - Wen Liu
- Department
of Pharmacy, The First Affiliate Hospital
of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha 410005, China
| |
Collapse
|
9
|
Masood A, Jacob M, Gu X, Abdel Jabar M, Benabdelkamel H, Nizami I, Li L, Dasouki M, Abdel Rahman AM. Distinctive metabolic profiles between Cystic Fibrosis mutational subclasses and lung function. Metabolomics 2021; 17:4. [PMID: 33394183 DOI: 10.1007/s11306-020-01760-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a lethal multisystemic disease of a monogenic origin with numerous mutations. Functional defects in the cystic fibrosis transmembrane conductance receptor (CFTR) protein based on these mutations are categorised into distinct classes having different clinical presentations and disease severity. OBJECTIVES The present study aimed to create a comprehensive metabolomic profile of altered metabolites in patients with CF, among different classes and in relation to lung function. METHODS A chemical isotope labeling liquid chromatography-mass spectrometry metabolomics was used to study the serum metabolic profiles of young and adult CF (n = 39) patients and healthy controls (n = 30). Comparisons were made at three levels, CF vs. controls, among mutational classes of CF, between CF class III and IV, and correlated the lung function findings. RESULTS A distinctive metabolic profile was observed in the three analyses. 78, 20, and 13 significantly differentially dysregulated metabolites were identified in the patients with CF, among the different classes and between class III and IV, respectively. The significantly identified metabolites included amino acids, di-, and tri-peptides, glutathione, glutamine, glutamate, and arginine metabolism. The top significant metabolites include 1-Aminopropan-2-ol, ophthalmate, serotonin, cystathionine, and gamma-glutamylglutamic acid. Lung function represented by an above-average FEV1% level was associated with decreased glutamic acid and increased guanosine levels. CONCLUSION Metabolomic profiling identified alterations in different amino acids and dipeptides, involved in regulating glutathione metabolism. Two metabolites, 3,4-dihydroxymandelate-3-O-sulfate and 5-Aminopentanoic acid, were identified in common between the three anlayses and may represent as highly sensitive biomarkers for CF.
Collapse
Affiliation(s)
- Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, PO. Box 2925 (98), Riyadh, 11461, Saudi Arabia
| | - Minnie Jacob
- Metabolomics Section, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, PO. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Xinyun Gu
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Mai Abdel Jabar
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, PO. Box 2925 (98), Riyadh, 11461, Saudi Arabia
| | - Imran Nizami
- Lung Transplant Section, Organ Transplant Center, King Faisal Specialist Hospital and Research Center, Zahrawi Street, Al Maather, Riyadh, 11211, Saudi Arabia
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Majed Dasouki
- Metabolomics Section, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, PO. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Anas M Abdel Rahman
- Metabolomics Section, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, PO. Box 3354, Riyadh, 11211, Saudi Arabia.
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh, Saudi Arabia.
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X7, Canada.
| |
Collapse
|
10
|
Wisniewski BL, Shrestha CL, Zhang S, Thompson R, Gross M, Groner JA, Uppal K, Ramilo O, Mejias A, Kopp BT. Metabolomics profiling of tobacco exposure in children with cystic fibrosis. J Cyst Fibros 2020; 19:791-800. [PMID: 32487493 PMCID: PMC7492400 DOI: 10.1016/j.jcf.2020.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Inflammation is integral to early disease progression in children with CF. The effect of modifiable environmental factors on infection and inflammation in persons with CF is poorly understood. Our prior studies determined that secondhand smoke exposure (SHSe) is highly prevalent in young children with CF. SHSe is associated with increased inflammation, heightened bacterial burden, and worsened clinical outcomes. However, the specific metabolite and signaling pathways that regulate responses to SHSe in CF are relatively unknown. METHODS High-resolution metabolomics was performed on plasma samples from infants (n = 25) and children (n = 40) with CF compared to non-CF controls (n = 15). CF groups were stratified according to infant or child age and SHSe status. RESULTS Global metabolomic profiles segregated by age and SHSe status. SHSe in CF was associated with changes in pathways related to steroid biosynthesis, fatty acid metabolism, cysteine metabolism, and oxidative stress. CF infants with SHSe demonstrated enrichment for altered metabolite localization to the small intestine, liver, and striatum. CF children with SHSe demonstrated metabolite enrichment for organs/tissues associated with oxidative stress including mitochondria, peroxisomes, and the endoplasmic reticulum. In a confirmatory analysis, SHSe was associated with changes in biomarkers of oxidative stress and cellular adhesion including MMP-9, MPO, and ICAM-1. CONCLUSIONS SHSe in young children and infants with CF is associated with altered global metabolomics profiles and specific biochemical pathways, including enhanced oxidative stress. SHSe remains an important but understudied modifiable variable in early CF disease.
Collapse
Affiliation(s)
- Benjamin L Wisniewski
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, W510, 700 Children's Drive, Columbus, OH 43205, USA
| | - Chandra L Shrestha
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, W510, 700 Children's Drive, Columbus, OH 43205, USA
| | - Shuzhong Zhang
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, W510, 700 Children's Drive, Columbus, OH 43205, USA
| | - Rohan Thompson
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Myron Gross
- Minnesota CHEAR Exposure Assessment Hub, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Judith A Groner
- Section of Ambulatory Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| | - Karan Uppal
- National Exposure Assessment Laboratory at Emory, Emory University, Atlanta, GA, USA
| | - Octavio Ramilo
- Division of Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, USA; Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Asuncion Mejias
- Division of Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, USA; Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Benjamin T Kopp
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, W510, 700 Children's Drive, Columbus, OH 43205, USA.
| |
Collapse
|
11
|
Ling KM, Garratt LW, Gill EE, Lee AHY, Agudelo-Romero P, Sutanto EN, Iosifidis T, Rosenow T, Turvey SE, Lassmann T, Hancock REW, Kicic A, Stick SM. Rhinovirus Infection Drives Complex Host Airway Molecular Responses in Children With Cystic Fibrosis. Front Immunol 2020; 11:1327. [PMID: 32765492 PMCID: PMC7378398 DOI: 10.3389/fimmu.2020.01327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/26/2020] [Indexed: 01/22/2023] Open
Abstract
Early-life viral infections are responsible for pulmonary exacerbations that can contribute to disease progression in young children with cystic fibrosis (CF). The most common respiratory viruses detected in the CF airway are human rhinoviruses (RV), and augmented airway inflammation in CF has been attributed to dysregulated airway epithelial responses although evidence has been conflicting. Here, we exposed airway epithelial cells from children with and without CF to RV in vitro. Using RNA-Seq, we profiled the transcriptomic differences of CF and non-CF airway epithelial cells at baseline and in response to RV. There were only modest differences between CF and non-CF cells at baseline. In response to RV, there were 1,442 and 896 differentially expressed genes in CF and non-CF airway epithelial cells, respectively. The core antiviral responses in CF and non-CF airway epithelial cells were mediated through interferon signaling although type 1 and 3 interferon signaling, when measured, were reduced in CF airway epithelial cells following viral challenge consistent with previous reports. The transcriptional responses in CF airway epithelial cells were more complex than in non-CF airway epithelial cells with diverse over-represented biological pathways, such as cytokine signaling and metabolic and biosynthetic pathways. Network analysis highlighted that the differentially expressed genes of CF airway epithelial cells' transcriptional responses were highly interconnected and formed a more complex network than observed in non-CF airway epithelial cells. We corroborate observations in fully differentiated air–liquid interface (ALI) cultures, identifying genes involved in IL-1 signaling and mucin glycosylation that are only dysregulated in the CF airway epithelial response to RV infection. These data provide novel insights into the CF airway epithelial cells' responses to RV infection and highlight potential pathways that could be targeted to improve antiviral and anti-inflammatory responses in CF.
Collapse
Affiliation(s)
- Kak-Ming Ling
- Paediatrics, Medical School, Faculty of Healthy and Medical Science, The University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Luke W Garratt
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Erin E Gill
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy H Y Lee
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Patricia Agudelo-Romero
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Thomas Iosifidis
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Tim Rosenow
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Timo Lassmann
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
12
|
Liessi N, Pedemonte N, Armirotti A, Braccia C. Proteomics and Metabolomics for Cystic Fibrosis Research. Int J Mol Sci 2020; 21:ijms21155439. [PMID: 32751630 PMCID: PMC7432297 DOI: 10.3390/ijms21155439] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/18/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of this review article is to introduce the reader to the state-of-the-art of the contribution that proteomics and metabolomics sciences are currently providing for cystic fibrosis (CF) research: from the understanding of cystic fibrosis transmembrane conductance regulator (CFTR) biology to biomarker discovery for CF diagnosis. Our work particularly focuses on CFTR post-translational modifications and their role in cellular trafficking as well as on studies that allowed the identification of CFTR molecular interactors. We also show how metabolomics is currently helping biomarker discovery in CF. The most recent advances in these fields are covered by this review, as well as some considerations on possible future scenarios for new applications.
Collapse
Affiliation(s)
- Nara Liessi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
| | - Nicoletta Pedemonte
- U.O.C. Genetica Medica, IRCCS Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Andrea Armirotti
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
- Correspondence: ; Tel.: +39-010-2896-938
| | - Clarissa Braccia
- D3PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
| |
Collapse
|
13
|
Al-Qahtani W, Abdel Jabar M, Masood A, Jacob M, Nizami I, Dasouki M, Abdel Rahman AM. Dried Blood Spot-Based Metabolomic Profiling in Adults with Cystic Fibrosis. J Proteome Res 2020; 19:2346-2357. [PMID: 32312052 DOI: 10.1021/acs.jproteome.0c00031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mucoviscidosis of the respiratory, gastrointestinal, and genitourinary tracts is the major pathology in patients with cystic fibrosis (CF), a lethal monogenic panethnic and multisystemic disease most commonly identified in Caucasians. Currently, the measurement of immuno reactive trypsinogen in dry blood spots (DBSs) is the gold-standard method for initial newborn screening for CF, followed by targeted CF transmembrane regulator (CFTR) mutation analysis, and ultimate confirmation with abnormally elevated sweat chloride. Previous metabolomics studies in patients with CF reported on different biomarkers such as breath 2-aminoacetophenone produced during acute and chronic infection in human tissues, including the lungs of CF patients. Herein, we used liquid and gas chromatography-mass spectrometry-based targeted metabolomics profiling to identify potentially reliable, sensitive, and specific biomarkers in DBSs collected from 69 young and adult people including CF patients (n = 39) and healthy control (n = 30). A distinctive metabolic profile including 26 significantly differentially expressed metabolites involving amino acids, glycolysis, mitochondrial and peroxisomal metabolism, and sorbitol pathways was identified. Specifically, the osmolyte (sorbitol) was remarkably downregulated in CF patients compared to healthy controls indicating perturbation in the sorbitol pathway, which may be responsible for the mucoviscidosis seen in patients with CF. The significance of our findings is supported by the clinical utility of inhaled mannitol and hypertonic saline in patients with CF. The systemic administration of sorbitol in such patients may confer additional benefits beyond the respiratory system, especially in those with misfolded CFTR proteins.
Collapse
Affiliation(s)
- Wafa Al-Qahtani
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh 11533, Saudi Arabia
| | - Mai Abdel Jabar
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia
| | - Minnie Jacob
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Imran Nizami
- Lung Transplant Section, Organ Transplant Center, King Faisal Specialist Hospital and Research Center, Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Majed Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Anas M Abdel Rahman
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh 11533, Saudi Arabia
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland A1B 3X7, Canada
| |
Collapse
|
14
|
Viswan A, Singh C, Kayastha AM, Azim A, Sinha N. An NMR based panorama of the heterogeneous biology of acute respiratory distress syndrome (ARDS) from the standpoint of metabolic biomarkers. NMR IN BIOMEDICINE 2020; 33:e4192. [PMID: 31733128 DOI: 10.1002/nbm.4192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/16/2019] [Accepted: 09/05/2019] [Indexed: 06/10/2023]
Abstract
Acute respiratory distress syndrome (ARDS), manifested by intricate etiology and pathophysiology, demands careful clinical surveillance due to its high mortality and imminent life support measures. NMR based metabolomics provides an approach for ARDS which culminates from a wide spectrum of illness thereby confounding early manifestation and prognosis predictors. 1 H NMR with its manifold applications in critical disease settings can unravel the biomarker of ARDS thus holding potent implications by providing surrogate endpoints of clinical utility. NMR metabolomics which is the current apogee platform of omics trilogy is contributing towards the possible panacea of ARDS by subsequent validation of biomarker credential on larger datasets. In the present review, the physiological derangements that jeopardize the whole metabolic functioning in ARDS are exploited and the biomarkers involved in progression are addressed and substantiated. The following sections of the review also outline the clinical spectrum of ARDS from the standpoint of NMR based metabolomics which is an emerging element of systems biology. ARDS is the main premise of intensivists textbook, which has been thoroughly reviewed along with its incidence, progressive stages of severity, new proposed diagnostic definition, and the preventive measures and the current pitfalls of clinical management. The advent of new therapies, the need for biomarkers, the methodology and the contemporary promising approaches needed to improve survival and address heterogeneity have also been evaluated. The review has been stepwise illustrated with potent biometrics employed to selectively pool out differential metabolites as diagnostic markers and outcome predictors. The following sections have been drafted with an objective to better understand ARDS mechanisms with predictive and precise biomarkers detected so far on the basis of underlying physiological parameters having close proximity to diseased phenotype. The aim of this review is to stimulate interest in conducting more studies to help resolve the complex heterogeneity of ARDS with biomarkers of clinical utility and relevance.
Collapse
Affiliation(s)
- Akhila Viswan
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS) - Campus, Lucknow, Uttar Pradesh, India
- Faculty of Engineering and Technology, Dr. A. P. J Abdul Kalam Technical University, Lucknow, India
| | - Chandan Singh
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS) - Campus, Lucknow, Uttar Pradesh, India
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Arvind M Kayastha
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Afzal Azim
- Critical Care Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Neeraj Sinha
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS) - Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
15
|
Inflammation in CF: Key Characteristics and Therapeutic Discovery. Respir Med 2020. [DOI: 10.1007/978-3-030-42382-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
16
|
Favia M, de Bari L, Bobba A, Atlante A. An Intriguing Involvement of Mitochondria in Cystic Fibrosis. J Clin Med 2019; 8:jcm8111890. [PMID: 31698802 PMCID: PMC6912654 DOI: 10.3390/jcm8111890] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) occurs when the cystic fibrosis transmembrane conductance regulator (CFTR) protein is not synthetized and folded correctly. The CFTR protein helps to maintain the balance of salt and water on many body surfaces, such as the lung surface. When the protein is not working correctly, chloride becomes trapped in cells, then water cannot hydrate the cellular surface and the mucus covering the cells becomes thick and sticky. Furthermore, a defective CFTR appears to produce a redox imbalance in epithelial cells and extracellular fluids and to cause an abnormal generation of reactive oxygen species: as a consequence, oxidative stress has been implicated as a causative factor in the aetiology of the process. Moreover, massive evidences show that defective CFTR gives rise to extracellular GSH level decrease and elevated glucose concentrations in airway surface liquid (ASL), thus encouraging lung infection by pathogens in the CF advancement. Recent research in progress aims to rediscover a possible role of mitochondria in CF. Here the latest new and recent studies on mitochondrial bioenergetics are collected. Surprisingly, they have enabled us to ascertain that mitochondria have a leading role in opposing the high ASL glucose level as well as oxidative stress in CF.
Collapse
Affiliation(s)
- Maria Favia
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari—CNR, Via G. Amendola 122/O, 70126 Bari, Italy; (L.d.B.); (A.B.)
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via E. Orabona 4, 70126 Bari, Italy
- Correspondence: (M.F.); (A.A.)
| | - Lidia de Bari
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari—CNR, Via G. Amendola 122/O, 70126 Bari, Italy; (L.d.B.); (A.B.)
| | - Antonella Bobba
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari—CNR, Via G. Amendola 122/O, 70126 Bari, Italy; (L.d.B.); (A.B.)
| | - Anna Atlante
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari—CNR, Via G. Amendola 122/O, 70126 Bari, Italy; (L.d.B.); (A.B.)
- Correspondence: (M.F.); (A.A.)
| |
Collapse
|
17
|
Zang X, Monge ME, Gaul DA, McCarty NA, Stecenko A, Fernández FM. Early Detection of Cystic Fibrosis Acute Pulmonary Exacerbations by Exhaled Breath Condensate Metabolomics. J Proteome Res 2019; 19:144-152. [PMID: 31621328 DOI: 10.1021/acs.jproteome.9b00443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The most common cause of death in cystic fibrosis (CF) patients is progressive lung function decline, which is punctuated by acute pulmonary exacerbations (APEs). A major challenge is to discover biomarkers for detecting an oncoming APE and allow for pre-emptive clinical interventions. Metabolic profiling of exhaled breath condensate (EBC) samples collected from CF patients before, during, and after APEs and under stable conditions (n = 210) was performed using ultraperformance liquid chromatography (UPLC) coupled to Orbitrap mass spectrometry (MS). Negative ion mode MS data showed that classification between metabolic profiles from "pre-APE" (pending APE before the CF patient had any signs of illness) and stable CF samples was possible with good sensitivities (85.7 and 89.5%), specificities (88.4 and 84.1%), and accuracies (87.7 and 85.7%) for pediatric and adult patients, respectively. Improved classification performance was achieved by combining positive with negative ion mode data. Discriminant metabolites included two potential biomarkers identified in a previous pilot study: lactic acid and 4-hydroxycyclohexylcarboxylic acid. Some of the discriminant metabolites had microbial origins, indicating a possible role of bacterial metabolism in APE progression. The results show promise for detecting an oncoming APE using EBC metabolites, thus permitting early intervention to abort such an event.
Collapse
Affiliation(s)
- Xiaoling Zang
- School of Chemistry and Biochemistry , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - María Eugenia Monge
- Centro de Investigaciones en Bionanociencias (CIBION) , Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Godoy Cruz 2390 , Ciudad de Buenos Aires C1425FQD , Argentina
| | - David A Gaul
- School of Chemistry and Biochemistry , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Nael A McCarty
- Emory + Children's Center for Cystic Fibrosis and Airways Disease Research and Department of Pediatrics , Emory University School of Medicine and Children's Healthcare of Atlanta , Atlanta , Georgia 30322 , United States
| | - Arlene Stecenko
- Emory + Children's Center for Cystic Fibrosis and Airways Disease Research and Department of Pediatrics , Emory University School of Medicine and Children's Healthcare of Atlanta , Atlanta , Georgia 30322 , United States
| | - Facundo M Fernández
- School of Chemistry and Biochemistry , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| |
Collapse
|
18
|
Muhlebach MS, Sha W, MacIntosh B, Kelley TJ, Muenzer J. Metabonomics reveals altered metabolites related to inflammation and energy utilization at recovery of cystic fibrosis lung exacerbation. Metabol Open 2019; 3:100010. [PMID: 32812947 PMCID: PMC7424819 DOI: 10.1016/j.metop.2019.100010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/31/2019] [Accepted: 06/01/2019] [Indexed: 02/07/2023] Open
Abstract
Background Cystic fibrosis lung disease is characterized by chronic bacterial infections in the setting of mucus abnormalities. Patients experience periodic exacerbations that manifest with increased respiratory symptoms that require intensification of therapy with enhanced airway clearance and intravenous (IV) antibiotics. Objectives In an observational study we tested if the profile of metabolites in serum distinguished the pre-from post-exacerbation state and which systemically measurable pathways were affected during the process to recovery. Methods Serum collected within 48 h of start and completion, respectively of IV antibiotics was collected from people with CF ages 6–30 years. Three day food records were collected prior to each sample. To reduce variation between subjects only subjects who had pancreatic insufficiency, had similar CF mutations, and did not have CF liver disease or diabetes were included. Metabolomic profiling was conducted by Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy with metabolites being identified based on retention time/index, mass to charge ratio and comparison to known compounds. Biostatistical analyses used paired t-test with correction for multiple comparisons and orthogonal partial least square discriminant analysis. Results Thirty subjects (20 male) with a mean ± SEM age of 15.3 ± 1.2 years participated, 17 of whom had matched food-records. Lung function was significantly improved post-therapy compared to pre-therapy, (mean ± SEM) 75 ± 4% vs. 68 ± 4% predicted (n = 26). Serum metabonomics showed distinction of the pre-vs. post-therapy groups with 123 compounds contributing to the differentiation pre-versus post-antibiotics by multiple biostatistical analyses. Compounds and pathways affected included bile acids and microbial derived amino acid metabolites, increases in lipid classes of the glycerophospholipid, glycerolipids, cholesterol, phopsholipids, and most pronounced, the class of sphingolipids. Changes in n6/n3 fatty acids, decreased polyamines but increased metabolites in the nitric oxide pathway, and changes in the tryptophan-kynurenine pathway indicated decreased inflammation at resolution of exacerbation. Conclusions Changes in serum metabolites that distinguished CF pulmonary exacerbation vs. resolution of symptoms showed evidence of decreased inflammation and improvement from a catabolic state. Serum metabonomics distinguishes cystic fibrosis exacerbation to resolution state. Distinguishing pathways indicate alterations in (gut) microbiome and bile acids. Serum metabolites at resolution show decreased inflammation and oxidative stress. Lipid markers indicate improvement from a catabolic state at exacerbation.
Collapse
Key Words
- AA, arachidonate
- ARG, arginase
- BA, bile acids
- BHBA, 3-hydroxybutyrate
- CF, cystic fibrosis
- CFTR, Cystic Fibrosis Transmembrane Regulator
- CRP, C-reactive protein
- DHA, docosahexaenoate
- ESI, electrospray ionization
- FDR, false discovery rate
- FEV1, forced expiratory volume in 1st second
- IDO, indoleamine-2-3-dioxygenase
- IV, intravenous
- NOS, nitric oxide synthase
- ODC, ornithine decarboxylase
- OPLS-DA, orthogonal partial least square discriminant analysis
- QC, quality control
- RI, retention time/index
- UNC, University of North Carolina at Chapel Hill
- UPLC, ultrahigh performance liquid chromatography-tandem mass spectroscopy
- VIP, variable influence on projection score
- n3-DPA, docosapentaenoate
- q, significance at a 5% FDR cut-off
Collapse
Affiliation(s)
- Marianne S Muhlebach
- Dept. Pediatrics, Division Pulmonary Medicine, University of North Carolina at Chapel Hill, 450 MacNider, 330 S. Columbia Road, Chapel Hill, NC, 27599-7217, USA.,Marsico Lung Institute, Chapel Hill, NC, USA
| | - Wei Sha
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, 150 Research Campus Dr, Kannapolis, NC 28081, NC, USA
| | - Beth MacIntosh
- Metabolic and Nutrition Research Core, UNC Healthcare, Nutrition and Food Services Department, 102 Mason Farm Rd, CB#7777, Chapel Hill, NC, USA
| | - Thomas J Kelley
- Departments of Pediatrics/Pharmacology, Case Western Reserve University, 833 BRB, 10900, Euclid Ave, Cleveland, OH, USA
| | - Joseph Muenzer
- Dept. Pediatrics, Division Metabolism and Genetics, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
19
|
Crabbé A, Ostyn L, Staelens S, Rigauts C, Risseeuw M, Dhaenens M, Daled S, Van Acker H, Deforce D, Van Calenbergh S, Coenye T. Host metabolites stimulate the bacterial proton motive force to enhance the activity of aminoglycoside antibiotics. PLoS Pathog 2019; 15:e1007697. [PMID: 31034512 PMCID: PMC6508747 DOI: 10.1371/journal.ppat.1007697] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 05/09/2019] [Accepted: 03/11/2019] [Indexed: 11/27/2022] Open
Abstract
Antibiotic susceptibility of bacterial pathogens is typically evaluated using in vitro assays that do not consider the complex host microenvironment. This may help explaining a significant discrepancy between antibiotic efficacy in vitro and in vivo, with some antibiotics being effective in vitro but not in vivo or vice versa. Nevertheless, it is well-known that antibiotic susceptibility of bacteria is driven by environmental factors. Lung epithelial cells enhance the activity of aminoglycoside antibiotics against the opportunistic pathogen Pseudomonas aeruginosa, yet the mechanism behind is unknown. The present study addresses this gap and provides mechanistic understanding on how lung epithelial cells stimulate aminoglycoside activity. To investigate the influence of the local host microenvironment on antibiotic activity, an in vivo-like three-dimensional (3-D) lung epithelial cell model was used. We report that conditioned medium of 3-D lung cells, containing secreted but not cellular components, potentiated the bactericidal activity of aminoglycosides against P. aeruginosa, including resistant clinical isolates, and several other pathogens. In contrast, conditioned medium obtained from the same cell type, but grown as conventional (2-D) monolayers did not influence antibiotic efficacy. We found that 3-D lung cells secreted endogenous metabolites (including succinate and glutamate) that enhanced aminoglycoside activity, and provide evidence that bacterial pyruvate metabolism is linked to the observed potentiation of antimicrobial activity. Biochemical and phenotypic assays indicated that 3-D cell conditioned medium stimulated the proton motive force (PMF), resulting in increased bacterial intracellular pH. The latter stimulated antibiotic uptake, as determined using fluorescently labelled tobramycin in combination with flow cytometry analysis. Our findings reveal a cross-talk between host and bacterial metabolic pathways, that influence downstream activity of antibiotics. Understanding the underlying basis of the discrepancy between the activity of antibiotics in vitro and in vivo may lead to improved diagnostic approaches and pave the way towards novel means to stimulate antibiotic activity. There is a poor correlation between the activity of antibiotics in the laboratory and in patients, including in several infectious diseases of the respiratory tract. What may help explaining differences between antibiotic activity in vitro and in vivo is that current antibiotic susceptibility tests do not consider the in vivo lung environment. The lung environment contains many factors that may influence bacterial susceptibility to antibiotics. This includes lung epithelial cells, which have been shown to improve the activity of aminoglycoside antibiotics. Yet, how lung epithelial cells increase aminoglycoside activity is currently unknown. Here, we cultured lung epithelial cells in an in vivo-like model and found that they secrete metabolites that enhance the activity of aminoglycoside antibiotics. We found that host cell secretions increased antibiotic uptake through stimulation of bacterial metabolism, which in turn resulted in enhanced activity. Our findings highlight that cross-talk between host and bacterial metabolisms contributes to the efficacy of antibiotic treatment. Understanding how the host metabolism influences antibiotic activity may open up therapeutic avenues to exploit host metabolism for improving antibiotic activity and help explaining discrepancies between antibiotic efficacy in vitro and in vivo.
Collapse
Affiliation(s)
- Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
- * E-mail:
| | - Lisa Ostyn
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Sorien Staelens
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Charlotte Rigauts
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Martijn Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Ghent, Belgium
| | - Maarten Dhaenens
- ProGenTomics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Simon Daled
- ProGenTomics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Heleen Van Acker
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Dieter Deforce
- ProGenTomics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | | | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
Modulation of glucose-related metabolic pathways controls glucose level in airway surface liquid and fight oxidative stress in cystic fibrosis cells. J Bioenerg Biomembr 2019; 51:203-218. [PMID: 31030390 DOI: 10.1007/s10863-019-09797-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/09/2019] [Indexed: 10/26/2022]
Abstract
Direct and indirect evidences show that elevated glucose concentrations in airway surface liquid (ASL) promote lung infection by pathogens, playing a role in the progression of the Cystic Fibrosis (CF) disease. The joint action of transporter/s for glucose and of the cellular enzymes is essential in order to try to lower ASL glucose level. Inside the cell, the glycolysis and the pentose phosphate pathway (PPP) compete for the utilization of glucose-6-phosphate (G6P), the product in which glucose, after entry within the cell and phosphorylation, is trapped. The study aims to clarify whether, modulating the activity of enzymatic proteins and/or the level of metabolites/cofactors, involved in intracellular glucose utilization, a lowering of the extracellular glucose level in CF occurs. Biochemical approaches have enabled us to understand i) how G6P is shunted between glycolysis and PPP and ii) that mitochondria, more than enzymes/cofactors participating to the two cell glucose utilization pathways, are protagonists of the scene in counteracting the high ASL glucose level as well as oxidative stress in CF.
Collapse
|
21
|
Valdivieso ÁG, Clauzure M, Massip-Copiz MM, Cancio CE, Asensio CJA, Mori C, Santa-Coloma TA. Impairment of CFTR activity in cultured epithelial cells upregulates the expression and activity of LDH resulting in lactic acid hypersecretion. Cell Mol Life Sci 2019; 76:1579-1593. [PMID: 30599064 PMCID: PMC11105536 DOI: 10.1007/s00018-018-3001-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/23/2018] [Accepted: 12/17/2018] [Indexed: 12/31/2022]
Abstract
Mutations in the gene encoding the CFTR chloride channel produce cystic fibrosis (CF). CF patients are more susceptible to bacterial infections in lungs. The most accepted hypothesis sustains that a reduction in the airway surface liquid (ASL) volume favor infections. Alternatively, it was postulated that a reduced HCO3- transport through CFTR leads to a decreased ASL pH, favoring bacterial colonization. The issue is controversial, since recent data from cultured primary cells and CF children showed normal pH values in the ASL. We have reported previously a decreased mitochondrial Complex I (mCx-I) activity in cultured cells with impaired CFTR activity. Thus, we hypothesized that the reduced mCx-I activity could lead to increased lactic acid production (Warburg-like effect) and reduced extracellular pH (pHe). In agreement with this idea, we report here that cells with impaired CFTR function (intestinal Caco-2/pRS26, transfected with an shRNA-CFTR, and lung IB3-1 CF cells) have a decreased pHe. These cells showed increased lactate dehydrogenase (LDH) activity, LDH-A expression, and lactate secretion. Similar effects were reproduced in control cells stimulated with recombinant IL-1β. The c-Src and JNK inhibitors PP2 and SP600125 were able to increase the pHe, although the differences between control and CFTR-impaired cells were not fully compensated. Noteworthy, the LDH inhibitor oxamate completely restored the pHe of the intestinal Caco-2/pRS26 cells and have a significant effect in lung IB3-1 cells; therefore, an increased lactic acid secretion seems to be the key factor that determine a reduced pHe in these epithelial cells.
Collapse
Affiliation(s)
- Ángel G Valdivieso
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina.
| | - Mariángeles Clauzure
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina
| | - María M Massip-Copiz
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina
| | - Carla E Cancio
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina
| | - Cristian J A Asensio
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina
| | - Consuelo Mori
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina
| | - Tomás A Santa-Coloma
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), and The National Scientific and Technical Research Council of Argentina (CONICET), Alicia Moreau de Justo 1600, 1107, Buenos Aires, Argentina.
| |
Collapse
|
22
|
DiBattista A, McIntosh N, Lamoureux M, Al-Dirbashi OY, Chakraborty P, Britz-McKibbin P. Metabolic Signatures of Cystic Fibrosis Identified in Dried Blood Spots For Newborn Screening Without Carrier Identification. J Proteome Res 2019; 18:841-854. [PMID: 30507207 DOI: 10.1021/acs.jproteome.8b00351] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cystic fibrosis (CF) is a complex multiorgan disorder that is among the most common fatal genetic diseases benefiting from therapeutic interventions early in life. Newborn screening (NBS) for presymptomatic detection of CF currently relies on a two-stage immunoreactive trypsinogen (IRT) and cystic fibrosis transmembrane conductance regulator (CFTR) mutation panel algorithm that is sensitive but not specific for identifying affected neonates with a low positive predictive value. For the first time, we report the discovery of a panel of CF-specific metabolites from a single 3.2 mm diameter dried blood spot (DBS) punch when using multisegment injection-capillary electrophoresis-mass spectrometry (MS) as a high-throughput platform for nontargeted metabolite profiling from volume-restricted/biobanked specimens with quality control. This retrospective case-control study design identified 32 metabolites, including a series of N-glycated amino acids, oxidized glutathione disulfide, and nicotinamide that were differentially expressed in normal birth weight CF neonates without meconium ileus ( n = 36) as compared to gestational age/sex-matched screen-negative controls ( n = 44) after a false discovery rate adjustment ( q < 0.05). Also, 16 metabolites from DBS extracts allowed for discrimination of true CF cases from presumptive screen-positive carriers with one identified CFTR mutation and transient neonatal hypertrypsinogenemic neonates ( n = 72), who were later confirmed as unaffected due to a low sweat chloride (<29 mM) test result. Importantly, six CF-specific biomarker candidates satisfying a Bonferroni adjustment ( p < 7.25 × 10-5) from three independent batches of DBS specimens included several amino acids depleted in circulation (Tyr, Ser, Thr, Pro, Gly) likely reflecting protein maldigestion/malabsorption. Additionally, CF neonates had lower ophthalmic acid as an indicator of oxidative stress due to impaired glutathione efflux from exocrine/epithelial tissue and elevation of an unknown trivalent peptide that was directly correlated with IRT (ρ = 0.332, p = 4.55 × 10-4). Structural elucidation of unknown metabolites was performed by high-resolution MS/MS, whereas biomarker validation was realized when comparing a subset of metabolites from matching neonatal DBS specimens independently analyzed by direct infusion-MS/MS at an accredited NBS facility. This work sheds new light into the metabolic phenotype of CF early in life, which is required for better functional understanding of CFTR mutations of unknown clinical consequence and the development of more accurate yet cost-effective strategies for CF screening.
Collapse
Affiliation(s)
- Alicia DiBattista
- Department of Chemistry and Chemical Biology , McMaster University , Hamilton L8S 4M1 , Canada
| | | | | | - Osama Y Al-Dirbashi
- College of Medicine and Health Sciences , United Arab Emirates University , Al Ain 15551 , United Arab Emirates
| | | | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology , McMaster University , Hamilton L8S 4M1 , Canada
| |
Collapse
|
23
|
Kopp BT, Joseloff E, Goetz D, Ingram B, Heltshe SL, Leung DH, Ramsey BW, McCoy K, Borowitz D. Urinary metabolomics reveals unique metabolic signatures in infants with cystic fibrosis. J Cyst Fibros 2018; 18:507-515. [PMID: 30477895 DOI: 10.1016/j.jcf.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/25/2018] [Accepted: 10/28/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Biologic pathways and metabolic mechanisms underpinning early systemic disease in cystic fibrosis (CF) are poorly understood. The Baby Observational and Nutrition Study (BONUS) was a prospective multi-center study of infants with CF with a primary aim to examine the current state of nutrition in the first year of life. Its secondary aim was to prospectively explore concurrent nutritional, metabolic, respiratory, infectious, and inflammatory characteristics associated with early CF anthropometric measurements. We report here metabolomics differences within the urine of these infants as compared to infants without CF. METHODS Urine metabolomics was performed for 85 infants with predefined clinical phenotypes at approximately one year of age enrolled in BONUS via Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS). Samples were stratified by disease status (non-CF controls (n = 22); CF (n = 63, All-CF)) and CF clinical phenotype: respiratory hospitalization (CF Resp, n = 22), low length (CF LL, n = 23), and low weight (CF LW, n = 15). RESULTS Global urine metabolomics profiles in CF were heterogeneous, however there were distinct metabolic differences between the CF and non-CF groups. Top pathways altered in CF included tRNA charging and methionine degradation. ADCYAP1 and huntingtin were identified as predicted unique regulators of altered metabolic pathways in CF compared to non-CF. Infants with CF displayed alterations in metabolites associated with bile acid homeostasis, pentose sugars, and vitamins. CONCLUSIONS Predicted metabolic pathways and regulators were identified in CF infants compared to non-CF, but metabolic profiles were unable to discriminate between CF phenotypes. Targeted metabolomics provides an opportunity for further understanding of early CF disease. TRIAL REGISTRATION United States ClinicalTrials.Gov registry NCT01424696 (clinicaltrials.gov).
Collapse
Affiliation(s)
- B T Kopp
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA.
| | - E Joseloff
- Cystic Fibrosis Foundation, Bethesda, MD, USA
| | - D Goetz
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | | | - S L Heltshe
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, WA, USA; University of Washington, Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle, WA, USA
| | - D H Leung
- Department of Pediatrics, Baylor College of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - B W Ramsey
- Cystic Fibrosis Foundation Therapeutics Development Network Coordinating Center, Seattle Children's Research Institute, Seattle, WA, USA; University of Washington, Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle, WA, USA
| | - K McCoy
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - D Borowitz
- Cystic Fibrosis Foundation, Bethesda, MD, USA; Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
24
|
Kopp BT, McCulloch S, Shrestha CL, Zhang S, Sarzynski L, Woodley FW, Hayes D. Metabolomic responses to lumacaftor/ivacaftor in cystic fibrosis. Pediatr Pulmonol 2018; 53:583-591. [PMID: 29461009 DOI: 10.1002/ppul.23972] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/30/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is a life-limiting disease caused by a defect in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Lumacaftor/Ivacaftor is a novel CFTR modulator approved for patients that are homozygous for Phe508del CFTR, but its clinical effectiveness varies amongst patients, making it difficult to determine clinical responders. Therefore, identifying biochemical biomarkers associated with drug response are clinically important for follow-up studies. METHODS Serum metabolomics was performed on twenty patients with CF pre- and 6-month post-Lumacaftor/Ivacaftor response via Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS). Correlation with clinical variables was performed. RESULTS Metabolomics analysis demonstrated 188 differentially regulated metabolites between patients pre- and post-Lumacaftor/Ivacaftor initiation, with a predominance of lipid and amino acid alterations. The top 30 metabolites were able to differentiate pre- and post-Lumacaftor/Ivacaftor status in greater than 90% of patients via a random-forest confusion matrix. Alterations in bile acids, phospholipids, and bacteria-associated metabolites were the predominant changes associated with drug response. Importantly, changes in metabolic patterns were associated with clinical responders. CONCLUSIONS Selected key lipid and amino acid metabolic pathways were significantly affected by Lumacaftor/Ivacaftor initiation and similar pathways were affected in clinical responders. Targeted metabolomics may provide useful and relevant biomarkers of CFTR modulator responses.
Collapse
Affiliation(s)
- Benjamin T Kopp
- Department of Pediatrics, The Ohio State University College of Medicine Columbus, Columbus, Ohio.,Division of Pulmonary Medicine, Nationwide Children's Hospital Columbus, Columbus, Ohio.,Center for Microbial Pathogenesis, Nationwide Children's Hospital Columbus, Columbus, Ohio
| | | | - Chandra L Shrestha
- Center for Microbial Pathogenesis, Nationwide Children's Hospital Columbus, Columbus, Ohio
| | - Shuzhong Zhang
- Center for Microbial Pathogenesis, Nationwide Children's Hospital Columbus, Columbus, Ohio
| | - Lisa Sarzynski
- Department of Pediatrics, The Ohio State University College of Medicine Columbus, Columbus, Ohio.,Division of Pulmonary Medicine, Nationwide Children's Hospital Columbus, Columbus, Ohio
| | - Frederick W Woodley
- Department of Pediatrics, The Ohio State University College of Medicine Columbus, Columbus, Ohio.,Division of Gastroenterology, Nationwide Children's Hospital Columbus, Columbus, Ohio
| | - Don Hayes
- Department of Pediatrics, The Ohio State University College of Medicine Columbus, Columbus, Ohio.,Division of Pulmonary Medicine, Nationwide Children's Hospital Columbus, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University College of Medicine Columbus, Columbus, Ohio
| |
Collapse
|
25
|
Severity of allergic rhinitis assessed by using urine metabolomic profiling: Proof of concept. J Allergy Clin Immunol 2018; 142:687-689.e6. [PMID: 29678748 DOI: 10.1016/j.jaci.2018.02.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/09/2018] [Accepted: 02/21/2018] [Indexed: 12/25/2022]
|
26
|
Aberrant GSH reductase and NOX activities concur with defective CFTR to pro-oxidative imbalance in cystic fibrosis airways. J Bioenerg Biomembr 2018. [PMID: 29524019 DOI: 10.1007/s10863-018-9748-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Cystic fibrosis (CF) is associated to impaired Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) channel also causing decreased glutathione (GSH) secretion, defective airway bacterial clearance and inflammation. Here we checked the main ROS-producing and ROS-scavenging enzymes as potential additional factors involved in CF pathogenesis. We found that CFBE41o-cells, expressing F508del CFTR, have increased NADPH oxidase (NOX) activity and expression level, mainly responsible of the increased ROS production, and decreased glutathione reductase (GR) activity, not dependent on GR protein level decrease. Furthermore, defective CFTR proved to cause both extracellular and intracellular GSH level decrease, probably by reducing the amount of extracellular GSH-derived cysteine required for cytosolic GSH synthesis. Importantly, we provide evidence that defective CFTR and NOX/GR activity imbalance both contribute to NADPH and GSH level decrease and ROS overproduction in CF cells.
Collapse
|
27
|
Gookin JL, Mathews KG, Cullen J, Seiler G. Qualitative metabolomics profiling of serum and bile from dogs with gallbladder mucocele formation. PLoS One 2018; 13:e0191076. [PMID: 29324798 PMCID: PMC5764353 DOI: 10.1371/journal.pone.0191076] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/26/2017] [Indexed: 01/20/2023] Open
Abstract
Mucocele formation is characterized by secretion of abnormally thick mucus by the gallbladder epithelium of dogs that may cause obstruction of the bile duct or rupture of the gallbladder. The disease is increasingly recognized and is associated with a high morbidity and mortality. The cause of gallbladder mucocele formation in dogs is unknown. There is a strong breed predisposition and affected dogs have a high incidence of concurrent endocrinopathy or hyperlipidemia. These observations suggest a significant influence of both genetic and metabolic factors on disease pathogenesis. In this study, we investigated a theory that mucocele formation is associated with a syndrome of metabolic disruption. We surmised that a global, untargeted metabolomics approach could provide unique insight into the systemic pathogenesis of gallbladder mucocele formation and identify specific compounds as candidate biomarkers or treatment targets. Moreover, concurrent examination of the serum and hepatic duct bile metabolome would enable the construction of mechanism-based theories or identification of specific compounds responsible for altered function of the gallbladder epithelium. Abnormalities observed in dogs with gallbladder mucocele formation, including a 33-fold decrease in serum adenosine 5’-monophosphate (AMP), lower quantities of precursors required for synthesis of energy transporting nucleotides, and increases in citric acid cycle intermediates, suggest excess metabolic energy and a carbon surplus. Altered quantities of compounds involved in protein translation and RNA turnover, together with accumulation of gamma-glutamylated and N-acetylated amino acids in serum suggest abnormal regulation of protein and amino acid metabolism. Increases in lathosterol and 7α-hydroxycholesterol suggest a primary increase in cholesterol synthesis and diversion to bile acid formation. A number of specific biomarker compounds were identified for their ability to distinguish between control dogs and those that formed a gallbladder mucocele. Particularly noteworthy was a significant decrease in quantity of biologically active compounds that stimulate biliary ductal fluid secretion including adenosine, cAMP, taurolithocholic acid, and taurocholic acid. These findings support the presence of significant metabolic disruption in dogs with mucocele formation. A targeted, quantitative analysis of the identified serum biomarkers is warranted to determine their utility for diagnosis of this disease. Finally, repletion of compounds whose biological activity normally promotes biliary ductal secretion should be examined for any therapeutic impact for resolution or prevention of mucocele formation.
Collapse
Affiliation(s)
- Jody L Gookin
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Kyle G Mathews
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - John Cullen
- Department of Population Health and Pathobiology, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Gabriela Seiler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| |
Collapse
|
28
|
Zang X, Pérez JJ, Jones CM, Monge ME, McCarty NA, Stecenko AA, Fernández FM. Comparison of Ambient and Atmospheric Pressure Ion Sources for Cystic Fibrosis Exhaled Breath Condensate Ion Mobility-Mass Spectrometry Metabolomics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2017; 28:1489-1496. [PMID: 28364225 DOI: 10.1007/s13361-017-1660-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/01/2017] [Accepted: 03/12/2017] [Indexed: 06/07/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the gene that encodes the cystic fibrosis transmembrane conductance regulator (CFTR) protein. The vast majority of the mortality is due to progressive lung disease. Targeted and untargeted CF breath metabolomics investigations via exhaled breath condensate (EBC) analyses have the potential to expose metabolic alterations associated with CF pathology and aid in assessing the effectiveness of CF therapies. Here, transmission-mode direct analysis in real time traveling wave ion mobility spectrometry time-of-flight mass spectrometry (TM-DART-TWIMS-TOF MS) was tested as a high-throughput alternative to conventional direct infusion (DI) electrospray ionization (ESI) and atmospheric pressure chemical ionization (APCI) methods, and a critical comparison of the three ionization methods was conducted. EBC was chosen as the noninvasive surrogate for airway sampling over expectorated sputum as EBC can be collected in all CF subjects regardless of age and lung disease severity. When using pooled EBC collected from a healthy control, ESI detected the most metabolites, APCI a log order less, and TM-DART the least. TM-DART-TWIMS-TOF MS was used to profile metabolites in EBC samples from five healthy controls and four CF patients, finding that a panel of three discriminant EBC metabolites, some of which had been previously detected by other methods, differentiated these two classes with excellent cross-validated accuracy. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Xiaoling Zang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - José J Pérez
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Christina M Jones
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- National Institute of Standards and Technology, Chemical Science Division, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC, 29412, USA
| | - María Eugenia Monge
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD, Ciudad de Buenos Aires, Argentina
| | - Nael A McCarty
- Emory+Children's Center for Cystic Fibrosis and Airways Disease Research and Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
- Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Arlene A Stecenko
- Emory+Children's Center for Cystic Fibrosis and Airways Disease Research and Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
29
|
Affiliation(s)
- Liuqing Wen
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Kenneth Huang
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Yunpeng Liu
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
- National
Glycoengineering Research Center, Shandong University, Jinan 250100, China
| | - Peng George Wang
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
- National
Glycoengineering Research Center, Shandong University, Jinan 250100, China
| |
Collapse
|
30
|
Metabolic alterations in the sera of Chinese patients with mild persistent asthma: a GC-MS-based metabolomics analysis. Acta Pharmacol Sin 2015; 36:1356-66. [PMID: 26526201 DOI: 10.1038/aps.2015.102] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/11/2015] [Indexed: 12/31/2022]
Abstract
AIM To character the specific metabolomics profiles in the sera of Chinese patients with mild persistent asthma and to explore potential metabolic biomarkers. METHODS Seventeen Chinese patients with mild persistent asthma and age- and sex-matched healthy controls were enrolled. Serum samples were collected, and serum metabolites were analyzed using GC-MS coupled with a series of multivariate statistical analyses. RESULTS Clear intergroup separations existed between the asthmatic patients and control subjects. A list of differential metabolites and several top altered metabolic pathways were identified. The levels of succinate (an intermediate in tricarboxylic acid cycle) and inosine were highly upregulated in the asthmatic patients, suggesting a greater effort to breathe during exacerbation and hypoxic stress due to asthma. Other differential metabolites, such as 3,4-dihydroxybenzoic acid and phenylalanine, were also identified. Furthermore, the differential metabolites possessed higher values of area under the ROC curve (AUC), suggesting an excellent clinical ability for the prediction of asthma. CONCLUSION Metabolic activity is significantly altered in the sera of Chinese patients with mild persistent asthma. The data might be helpful for identifying novel biomarkers and therapeutic targets for asthma.
Collapse
|
31
|
Muhlebach MS, Sha W. Lessons learned from metabolomics in cystic fibrosis. Mol Cell Pediatr 2015; 2:9. [PMID: 26542299 PMCID: PMC4883209 DOI: 10.1186/s40348-015-0020-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/09/2015] [Indexed: 11/10/2022] Open
Abstract
Cystic fibrosis is a mono-genetic multi-system disease; however, respiratory manifestations cause the main morbidity and mortality where chronic bacterial infections lead to bronchiectasis and ultimately respiratory failure. Metabolomics allows a relatively complete snapshot of metabolic processes in a sample using different mass spectrometry methods. Sample types used for discovery of biomarkers or pathomechanisms in cystic fibrosis (CF) have included blood, respiratory secretions, and exhaled breath to date. Metabolomics has shown distinction of CF vs. non-CF for matrices of blood, exhaled breath, and respiratory epithelial cultures, each showing different pathways. Severity of lung disease has been addressed by studies in bronchoalveolar lavage and exhaled breath condensate showing separation by metabolites that the authors of each study related to inflammation; e.g., ethanol, acetone, purines. Lipidomics has been applied to blood and sputum samples showing associations with lung function and Pseudomonas aeruginosa infection status. Finally, studies of bacteria grown in vitro showed differences of bacterial metabolites to be associated with clinical parameters. Metabolomics, in the sense of global metabolomic profiling, is a powerful technique that has allowed discovery of pathways that had not previously been implicated in CF. These may include purines, mitochondrial pathways, and different aspects of glucose metabolism besides the known differences in lipid metabolism in CF. However, targeted studies to validate such potential metabolites and pathways of interest are necessary. Studies evaluating metabolites of bacterial origin are in their early stages. Thus further well-designed studies could be envisioned.
Collapse
Affiliation(s)
- Marianne S Muhlebach
- Department Pediatrics, Division Pulmonology, UNC Chapel Hill 430 MacNider, CB 7217, Chapel Hill, NC, 27599-7217, USA.
| | - Wei Sha
- Bioinformatics Services Division, University of North Carolina at Charlotte, Kannapolis, NC, 28081, USA.
| |
Collapse
|
32
|
Laguna TA, Reilly CS, Williams CB, Welchlin C, Wendt CH. Metabolomics analysis identifies novel plasma biomarkers of cystic fibrosis pulmonary exacerbation. Pediatr Pulmonol 2015; 50:869-77. [PMID: 26115542 PMCID: PMC5553866 DOI: 10.1002/ppul.23225] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/29/2015] [Accepted: 05/04/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) lung disease is characterized by infection, inflammation, lung function decline, and intermittent pulmonary exacerbations. However, the link between pulmonary exacerbation and lung disease progression remains unclear. Global metabolomic profiling can provide novel mechanistic insight into a disease process in addition to putative biomarkers for future study. Our objective was to investigate how the plasma metabolomic profile changes between CF pulmonary exacerbation and a clinically well state. METHODS Plasma samples and lung function data were collected from 25 CF patients during hospitalization for a pulmonary exacerbation and during quarterly outpatient clinic visits. In collaboration with Metabolon, Inc., the metabolomic profiles of matched pair plasma samples, one during exacerbation and one at a clinic visit, were analyzed using gas and liquid chromatography coupled with mass spectrometry. Compounds were identified by comparison to a library of standards. Mixed effects models that controlled for nutritional status and lung function were used to test for differences and principal components analysis was performed. RESULTS Our population had a median age of 27 years (14-39) and had a median FEV1 % predicted of 65% (23-105%). 398 total metabolites were identified and after adjustment for confounders, five metabolites signifying perturbations in nucleotide (hypoxanthine), nucleoside (N4-acetylcytidine), amino acid (N-acetylmethionine), carbohydrate (mannose), and steroid (cortisol) metabolism were identified. Principal components analysis provided good separation between the two clinical phenotypes. CONCLUSIONS Our findings provide putative metabolite biomarkers for future study and allow for hypothesis generation about the pathophysiology of CF pulmonary exacerbation.
Collapse
Affiliation(s)
- Theresa A Laguna
- Department of Pediatrics, University of Minnesota Medical School and The University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - Cavan S Reilly
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota
| | - Cynthia B Williams
- Department of Pediatrics, University of Minnesota Medical School and The University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - Cole Welchlin
- Department of Pediatrics, University of Minnesota Medical School and The University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - Chris H Wendt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Minnesota School of Medicine and Veterans Administration Medical Center, Minneapolis, Minnesota
| |
Collapse
|
33
|
Zhao J, Evans CR, Carmody LA, LiPuma JJ. Impact of storage conditions on metabolite profiles of sputum samples from persons with cystic fibrosis. J Cyst Fibros 2015; 14:468-73. [PMID: 25725986 DOI: 10.1016/j.jcf.2015.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Although recent studies have begun to elucidate how airway microbial community structure relates to lung disease in cystic fibrosis (CF), microbial community activity and the host's response to changes in this activity are poorly understood. Metabolomic profiling provides a means to investigate microbial activity and human cell activity within diseased airways. However, variables in sample storage and shipping likely affect downstream analyses and standards for sample handling are lacking. METHODS We assessed the impact of sample storage conditions on liquid chromatography mass spectrometry analysis of CF sputum samples. RESULTS Significant changes in global metabolomic profiles occurred in samples stored at room temperature or at 4°C for longer than one day. Untargeted metabolomic profiles were stable in sputum samples stored at -20°C or -80°C for at least 28 days. Quorum sensing molecules and phenazines, both considered important to the in vivo activity of Pseudomonas during airway infection, were detected after sample storage at room temperature for five days. CONCLUSIONS Sputum samples can be stored at -20°C or -80°C for weeks with minimal effect on global metabolomic profiles. This observation provides guidance in designing metabolomic studies that have the potential to deepen our understanding of how airway microbial communities impact lung disease progression in CF.
Collapse
Affiliation(s)
- Jiangchao Zhao
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Charles R Evans
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Lisa A Carmody
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - John J LiPuma
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
34
|
Zheng Y, Yu B, Alexander D, Couper DJ, Boerwinkle E. Medium-term variability of the human serum metabolome in the Atherosclerosis Risk in Communities (ARIC) study. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:364-73. [PMID: 24910946 DOI: 10.1089/omi.2014.0019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Metabolomics is a systems biology tool providing small molecule signatures of disease etiology. In order to estimate the biologic variability of the human serum metabolome, this study calculated intraclass correlation coefficients (ICCs) for 178 stably-detected metabolites measured by untargeted chromatography/mass spectrometry. We studied a subsample of 60 participants (57% males, 70% Caucasians, aged 73.77±5.3 years) in the Atherosclerosis Risk in Communities (ARIC) Study who provided two fasting serum samples 4-6 weeks apart. The median ICC across all metabolites was 0.60, and 82% of metabolites had at least fair variability (i.e., ICC>= 0.40). There was variation in the medium-term variability among metabolites, with those in the pathways of amino acid and lipid metabolism showing relatively high ICCs, and those in the carbohydrate pathway showing relatively low ICCs. The results of this study provide a valuable resource for future study design and outcome interpretation of mass spectrometry-based metabolomic studies in epidemiology.
Collapse
Affiliation(s)
- Yan Zheng
- 1 Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston , Houston, Texas
| | | | | | | | | |
Collapse
|
35
|
Nobakht M Gh BF, Aliannejad R, Rezaei-Tavirani M, Taheri S, Oskouie AA. The metabolomics of airway diseases, including COPD, asthma and cystic fibrosis. Biomarkers 2014; 20:5-16. [PMID: 25403491 DOI: 10.3109/1354750x.2014.983167] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD), asthma and cystic fibrosis (CF) are characterized by airway obstruction and an inflammatory process. Reaching early diagnosis and discrimination of subtypes of these respiratory diseases are quite a challenging task than other chronic illnesses. Metabolomics is the study of metabolic pathways and the measurement of unique biochemical molecules generated in a living system. In the last decade, metabolomics has already proved to be useful for the characterization of several pathological conditions and offers promises as a clinical tool. In this article, we review the current state of the metabolomics of COPD, asthma and CF with a focus on the different methods and instrumentation being used for the discovery of biomarkers in research and translation into clinic as diagnostic aids for the choice of patient-specific therapies.
Collapse
Affiliation(s)
- B Fatemeh Nobakht M Gh
- Faculty of Paramedical Sciences, Proteomics Research Center, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | | | | | | | | |
Collapse
|
36
|
Jeanson L, Guerrera IC, Papon JF, Chhuon C, Zadigue P, Prulière-Escabasse V, Amselem S, Escudier E, Coste A, Edelman A. Proteomic analysis of nasal epithelial cells from cystic fibrosis patients. PLoS One 2014; 9:e108671. [PMID: 25268127 PMCID: PMC4182543 DOI: 10.1371/journal.pone.0108671] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/24/2014] [Indexed: 11/21/2022] Open
Abstract
The pathophysiology of cystic fibrosis (CF) lung disease remains incompletely understood. New explanations for the pathogenesis of CF lung disease may be discovered by studying the patterns of protein expression in cultured human nasal epithelial cells (HNEC). To that aim, we compared the level of protein expressions in primary cultures of HNEC from nasal polyps secondary to CF (CFNP, n = 4), primary nasal polyps (NP, n = 8) and control mucosa (CTRL, n = 4) using isobaric tag for relative and absolute quantification (iTRAQ) labeling coupled with liquid chromatography (LC)-MS-MS. The analysis of the data revealed 42 deregulated protein expressions in CFNP compared to NP and CTRL, suggesting that these alterations are related to CF. Overall, AmiGo analysis highlighted six major pathways important for cell functions that seem to be impaired: metabolism, G protein process, inflammation and oxidative stress response, protein folding, proteolysis and structural proteins. Among them, glucose and fatty acid metabolic pathways could be impaired in CF with nine deregulated proteins. Our proteomic study provides a reproducible set of differentially expressed proteins in airway epithelial cells from CF patients and reveals many novel deregulated proteins that could lead to further studies aiming to clarify the involvement of such proteins in CF pathophysiology.
Collapse
Affiliation(s)
- Ludovic Jeanson
- Service de Génétique et Embryologie Médicales, Unité Mixte de Recherche_Scientifique 933, Institut National de la Santé et de la Recherche Médicale, Université Pierre et Marie Curie – Paris 6, and Assistance Publique – Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
- Unité_1151, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes, Paris, France
| | - Ida Chiara Guerrera
- Unité_1151, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes, Paris, France
- Plateau Proteome Necker, Structure Fédérative de Recherche de Necker, Université Paris Descartes, Paris, France
| | - Jean-François Papon
- Unité Mixte de Recherche_Scientifique 855, Institut National de la Santé et de la Recherche Médicale, Université Paris 12, Faculté de Médecine, Créteil, France
- Service d’Otorhinolaryngologie et de chirurgie cervico-faciale, Assistance Publique – Hôpitaux de Paris, Hôpital inter-communal et Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| | - Cerina Chhuon
- Unité_1151, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes, Paris, France
- Plateau Proteome Necker, Structure Fédérative de Recherche de Necker, Université Paris Descartes, Paris, France
| | - Patricia Zadigue
- Unité Mixte de Recherche_Scientifique 855, Institut National de la Santé et de la Recherche Médicale, Université Paris 12, Faculté de Médecine, Créteil, France
| | - Virginie Prulière-Escabasse
- Service d’Otorhinolaryngologie et de chirurgie cervico-faciale, Assistance Publique – Hôpitaux de Paris, Hôpital inter-communal et Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| | - Serge Amselem
- Service de Génétique et Embryologie Médicales, Unité Mixte de Recherche_Scientifique 933, Institut National de la Santé et de la Recherche Médicale, Université Pierre et Marie Curie – Paris 6, and Assistance Publique – Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Estelle Escudier
- Service de Génétique et Embryologie Médicales, Unité Mixte de Recherche_Scientifique 933, Institut National de la Santé et de la Recherche Médicale, Université Pierre et Marie Curie – Paris 6, and Assistance Publique – Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - André Coste
- Unité Mixte de Recherche_Scientifique 855, Institut National de la Santé et de la Recherche Médicale, Université Paris 12, Faculté de Médecine, Créteil, France
- Service d’Otorhinolaryngologie et de chirurgie cervico-faciale, Assistance Publique – Hôpitaux de Paris, Hôpital inter-communal et Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| | - Aleksander Edelman
- Unité_1151, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes, Paris, France
- Plateau Proteome Necker, Structure Fédérative de Recherche de Necker, Université Paris Descartes, Paris, France
| |
Collapse
|
37
|
Serum metabonomic analysis of protective effects of Curcuma aromatica oil on renal fibrosis rats. PLoS One 2014; 9:e108678. [PMID: 25265289 PMCID: PMC4181651 DOI: 10.1371/journal.pone.0108678] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 08/24/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Curcuma aromatica oil is a traditional herbal medicine demonstrating protective and anti-fibrosis activities in renal fibrosis patients. However, study of its mechanism of action is challenged by its multiple components and multiple targets that its active agent acts on. METHODOLOGY/PRINCIPAL FINDINGS Nuclear magnetic resonance (NMR)-based metabonomics combined with clinical chemistry and histopathology examination were performed to evaluate intervening effects of Curcuma aromatica oil on renal interstitial fibrosis rats induced by unilateral ureteral obstruction. The metabolite levels were compared based on integral values of serum 1H NMR spectra from rats on 3, 7, 14, and 28 days after the medicine administration. Time trajectory analysis demonstrated that metabolic profiles of the agent-treated rats were restored to control levels after 7 days of dosage. The results confirmed that the agent would be an effective anti-fibrosis medicine in a time-dependent manner, especially in early renal fibrosis stage. Targeted metabolite analysis showed that the medicine could lower levels of lipid, acetoacetate, glucose, phosphorylcholine/choline, trimethylamine oxide and raise levels of pyruvate, glycine in the serum of the rats. Serum clinical chemistry and kidney histopathology examination dovetailed well with the metabonomics data. CONCLUSIONS/SIGNIFICANCES The results substantiated that Curcuma aromatica oil administration can ameliorate renal fibrosis symptoms by inhibiting some metabolic pathways, including lipids metabolism, glycolysis and methylamine metabolism, which are dominating targets of the agent working in vivo. This study further strengthens the novel analytical approach for evaluating the effect of traditional herbal medicine and elucidating its molecular mechanism.
Collapse
|
38
|
Abstract
Pentose phosphate (PP) pathway, which is ubiquitously present in all living organisms, is one of the major metabolic pathways associated with glucose metabolism. The most important functions of this pathway includes the generation of reducing equivalents in the form of NADPH for reductive biosynthesis, and production of ribose sugars for the biosynthesis of nucleotides, amino acids, and other macromolecules required by all living cells. Under normal conditions of growth, PP pathway is important for cell cycle progression, myelin formation, and the maintenance of the structure and function of brain, liver, cortex and other organs. Under diseased conditions, such as in cases of many metabolic, neurological or malignant diseases, pathological mechanisms augment due to defects in the PP pathway genes. Adoption of alternative metabolic pathways by cells that are metabolically abnormal, or malignant cells that are resistant to chemotherapeutic drugs often plays important roles in disease progression and severity. Accordingly, the PP pathway has been suggested to play critical roles in protecting cancer or abnormal cells by providing reduced environment, to protect cells from oxidative damage and generating structural components for nucleic acids biosynthesis. Novel drugs that targets one or more components of the PP pathway could potentially serve to overcome challenges associated with currently available therapeutic options for many metabolic and non-metabolic diseases. However, careful designing of drugs is critical that takes into the accounts of cell’s broader genomic, proteomic and metabolic contexts under consideration, in order to avoid undesirable side-effects. In this review, we discuss the role of PP pathway under normal and abnormal physiological conditions and the potential of the PP pathway as a target for new drug development to treat metabolic and non-metabolic diseases.
Collapse
|
39
|
Joseloff E, Sha W, Bell SC, Wetmore DR, Lawton KA, Milburn MV, Ryals JA, Guo L, Muhlebach MS. Serum metabolomics indicate altered cellular energy metabolism in children with cystic fibrosis. Pediatr Pulmonol 2014; 49:463-72. [PMID: 23847148 DOI: 10.1002/ppul.22859] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 04/21/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is a multi-system disease affecting multiple organs and cells besides the respiratory system. Metabolomic profiling allows simultaneous detection of biochemicals originating from cells, organs, or exogenous origin that may be valuable for monitoring of disease severity or in diagnosis. AIM We hypothesized that metabolomics using serum from children would differentiate CF from non-CF lung disease subjects and would provide insight into metabolism in CF. METHODS Serum collected from children with CF (n = 31) and 31 age and gender matched children with other lung diseases was used for metabolomic profiling by gas- and liquid-chromatography. Relative concentration of metabolites was compared between the groups using partial least square discriminant analyses (PLS-DA) and linear modeling. RESULTS A clear separation of the two groups was seen in PLS-DA. Linear model found that among the 459 detected metabolites 92 differed between CF and non-CF. These included known biochemicals in lipid metabolism, oxidants, and markers consistent with abnormalities in bile acid processing. Bacterial metabolites were identified and differed between the groups indicating intestinal dysbiosis in CF. As a novel finding several pathways were markedly different in CF, which jointly point towards decreased activity in the β-oxidation of fatty acids. These pathways include low ketone bodies, low medium chain carnitines, elevated di-carboxylic acids and decreased 2-hydroxybutyrate from amino acid metabolism in CF compared to non-CF. CONCLUSION Serum metabolomics discriminated CF from non-CF and show altered cellular energy metabolism in CF potentially reflecting mitochondrial dysfunction. Future studies are indicated to examine their relation to the underlying CF defect and their use as biomarkers for disease severity or for cystic fibrosis transmembrane regulator (CFTR) function in an era of CFTR modifying drugs.
Collapse
Affiliation(s)
- Elizabeth Joseloff
- Cystic Fibrosis Foundation Therapeutics (CFFT), Inc., Bethesda, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ziady AG, Hansen J. Redox balance in cystic fibrosis. Int J Biochem Cell Biol 2014; 52:113-23. [PMID: 24657650 DOI: 10.1016/j.biocel.2014.03.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/28/2014] [Accepted: 03/11/2014] [Indexed: 11/30/2022]
Abstract
The homeostatic balance between oxidants and antioxidants in biological systems is known as redox balance, and is regulated by complex processes. Redox balance regulates many of the known cellular pathways and disease processes. The dysregulation of redox balance can lead to acute or long-term oxidative or reductive stresses that are associated with many of the abnormalities observed in cystic fibrosis (CF). Over the past 5 decades researchers have examined contributors to redox dysregulation, their molecular products, and their impact on ion transport, cell proliferation, inflammation, bacterial killing, and the metabolism of nucleic acids, proteins, and lipids in CF. CF patients exhibit elevated markers of oxidative stress when compared to non-CF healthy controls; however, whether the reported redox imbalance is sufficient to produce pathology has been controversial. In addition, comparisons between CF and non-CF disease controls have been lacking. To better understand the mechanisms which mediate the generation of oxidants and antioxidants in CF and the importance of their balance in effecting oxidative or reductive stress, we will review the determinants of redox balance in the blood, lumen, and cellular compartments. From the perspective of methodological application, we will focus on the approaches most often used to study oxidant and antioxidants in CF, including biochemical, proteomic, metabolomic, and lipidomic studies, with a discussion of the few transcriptomic analyses that predict changes in the expression of regulators of redox. Finally, we will discuss the utility of oxidants and antioxidants as biomarkers of disease and the use of antioxidant therapy in CF.
Collapse
Affiliation(s)
- Assem G Ziady
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Jason Hansen
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
41
|
Microbiota and metabolite profiling reveal specific alterations in bacterial community structure and environment in the cystic fibrosis airway during exacerbation. PLoS One 2013; 8:e82432. [PMID: 24358183 PMCID: PMC3866110 DOI: 10.1371/journal.pone.0082432] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/23/2013] [Indexed: 01/04/2023] Open
Abstract
Chronic polymicrobial infections of the lung are the foremost cause of morbidity and mortality in cystic fibrosis (CF) patients. The composition of the microbial flora of the airway alters considerably during infection, particularly during patient exacerbation. An understanding of which organisms are growing, their environment and their behaviour in the airway is of importance for designing antibiotic treatment regimes and for patient prognosis. To this end, we have analysed sputum samples taken from separate cohorts of CF and non-CF subjects for metabolites and in parallel, and we have examined both isolated DNA and RNA for the presence of 16S rRNA genes and transcripts by high-throughput sequencing of amplicon or cDNA libraries. This analysis revealed that although the population size of all dominant orders of bacteria as measured by DNA- and RNA- based methods are similar, greater discrepancies are seen with less prevalent organisms, some of which we associated with CF for the first time. Additionally, we identified a strong relationship between the abundance of specific anaerobes and fluctuations in several metabolites including lactate and putrescine during patient exacerbation. This study has hence identified organisms whose occurrence within the CF microbiome has been hitherto unreported and has revealed potential metabolic biomarkers for exacerbation.
Collapse
|
42
|
Brown MV, Compton SA, Milburn MV, Lawton KA, Cheatham B. Metabolomic signatures in lipid-loaded HepaRGs reveal pathways involved in steatotic progression. Obesity (Silver Spring) 2013; 21:E561-70. [PMID: 23512965 PMCID: PMC3689848 DOI: 10.1002/oby.20440] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/21/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVES A spectrum of disorders including simple steatosis, nonalcoholic steatohepatitis, fibrosis, and cirrhosis is described by nonalcoholic fatty liver disease (NAFLD). With the increased prevalence of obesity, and consequently NAFLD, there is a need for novel therapeutics in this area. To facilitate this effort, a cellular model of hepatic steatosis was developed using HepaRG cells and the resulting biochemical alterations were determined. DESIGN AND METHODS Using global metabolomic profiling, by means of a novel metabolite extraction procedure, the metabolic profiles in response to the saturated fatty acid palmitate, and a mixture of saturated and unsaturated fatty acids, palmitate and oleate (1:2) were examined. RESULTS We observed elevated levels of the branched chain amino acids, tricarboxylic acid cycle intermediates, sphingosine and acylcarnitines, and reduced levels of carnitine in the steatotic HepaRG model with both palmitate and palmitate:oleate treatments. In addition, elevated levels of diacylglycerols and monoacylglycerols as well as altered bile acid metabolism were selectively displayed by palmitate-induced steatotic cells. CONCLUSIONS Biochemical changes in pathways important in the transition to hepatic steatosis including insulin resistance, altered mitochondrial metabolism, and oxidative stress are revealed by this global metabolomic approach. Moreover, the utility of this in vitro model for investigating the mechanisms of steatotic progression, insulin resistance, and lipotoxicity in NAFLD was demonstrated.
Collapse
|
43
|
Abstract
Metabolomics, the global interrogation of the biochemical components in a biological sample, has become an important complement to genomics and proteomics to aid in the understanding of pathophysiology. Major advantages of metabolomics are the size of the metabolome relative to the genome or proteome and the fact that it provides a view of the existing biochemical phenotype. As such, metabolomics is fast becoming an important discovery tool for new diagnostic and prognostic biomarkers. Although many methods exist for performing metabolomics, relatively few have led to successful development of new diagnostic tests. This review will aid the reader in understanding various metabolomic methods and their applications, as well as some of their inherent advantages and disadvantages. In addition, we present one example of the application of metabolomics to the identification of new fasting blood biomarkers for the diagnosis and monitoring of insulin resistance.
Collapse
|
44
|
Dutta M, Joshi M, Srivastava S, Lodh I, Chakravarty B, Chaudhury K. A metabonomics approach as a means for identification of potential biomarkers for early diagnosis of endometriosis. MOLECULAR BIOSYSTEMS 2013; 8:3281-7. [PMID: 23079773 DOI: 10.1039/c2mb25353d] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our present study focuses on the identification of predictive biomarkers in serum for the early diagnosis of endometriosis in a minimally invasive manner using (1)H-NMR based metabonomics. PLS-DA modeling of bins obtained from CPMG spectra of serum samples discriminated endometriosis patients from controls with sensitivity and specificity levels of about 80% and 90%, respectively. Compared with those from controls, serum samples from endometriosis patients showed increased levels of lactate, 3-hydroxybutyrate, alanine, leucine, valine, threonine, lysine, glycerophosphatidylcholine, succinic acid and 2-hydroxybutyrate as well as decreased levels of lipids, glucose, isoleucine and arginine. Our work offers valuable information for non-invasive diagnosis of endometriosis and may be of potential benefit to understand pathogenesis of the disease.
Collapse
Affiliation(s)
- Mainak Dutta
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, India
| | | | | | | | | | | |
Collapse
|
45
|
Cicek AE, Bederman I, Henderson L, Drumm ML, Ozsoyoglu G. ADEMA: an algorithm to determine expected metabolite level alterations using mutual information. PLoS Comput Biol 2013; 9:e1002859. [PMID: 23341761 PMCID: PMC3547803 DOI: 10.1371/journal.pcbi.1002859] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 10/23/2012] [Indexed: 01/07/2023] Open
Abstract
Metabolomics is a relatively new “omics” platform, which analyzes a discrete set of metabolites detected in bio-fluids or tissue samples of organisms. It has been used in a diverse array of studies to detect biomarkers and to determine activity rates for pathways based on changes due to disease or drugs. Recent improvements in analytical methodology and large sample throughput allow for creation of large datasets of metabolites that reflect changes in metabolic dynamics due to disease or a perturbation in the metabolic network. However, current methods of comprehensive analyses of large metabolic datasets (metabolomics) are limited, unlike other “omics” approaches where complex techniques for analyzing coexpression/coregulation of multiple variables are applied. This paper discusses the shortcomings of current metabolomics data analysis techniques, and proposes a new multivariate technique (ADEMA) based on mutual information to identify expected metabolite level changes with respect to a specific condition. We show that ADEMA better predicts De Novo Lipogenesis pathway metabolite level changes in samples with Cystic Fibrosis (CF) than prediction based on the significance of individual metabolite level changes. We also applied ADEMA's classification scheme on three different cohorts of CF and wildtype mice. ADEMA was able to predict whether an unknown mouse has a CF or a wildtype genotype with 1.0, 0.84, and 0.9 accuracy for each respective dataset. ADEMA results had up to 31% higher accuracy as compared to other classification algorithms. In conclusion, ADEMA advances the state-of-the-art in metabolomics analysis, by providing accurate and interpretable classification results. Metabolomics is an experimental approach that analyzes differences in metabolite levels detected in experimental samples. It has been used in the literature to understand the changes in metabolism with respect to diseases or drugs. Unlike transcriptomics or proteomics, which analyze gene and protein expression levels respectively, the techniques that consider co-regulation of multiple metabolites are quite limited. In this paper, we propose a novel technique, called ADEMA, which computes the expected level changes for each metabolite with respect to a given condition. ADEMA considers multiple metabolites at the same time and is mutual information (MI)-based. We show that ADEMA predicts metabolite level changes for young mice with Cystic Fibrosis (CF) better than significance testing that considers one metabolite at a time. Using three different datasets that contain CF and wild-type (WT) mice, we show that ADEMA can classify an individual as being CF or WT based on the metabolic profiles (with 1.0, 0.84, and 0.9 accuracy, respectively). Compared to other well-known classification algorithms, ADEMA's accuracy is higher by up to 31%.
Collapse
Affiliation(s)
- A Ercument Cicek
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, Ohio, USA.
| | | | | | | | | |
Collapse
|
46
|
Application of metabolomics approaches to the study of respiratory diseases. Bioanalysis 2013; 4:2265-90. [PMID: 23046268 DOI: 10.4155/bio.12.218] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Metabolomics is the global unbiased analysis of all the small-molecule metabolites within a biological system, under a given set of conditions. These methods offer the potential for a holistic approach to clinical medicine, as well as improving disease diagnosis and understanding of pathological mechanisms. Respiratory diseases including asthma and chronic obstructive pulmonary disorder are increasing globally, with the latter predicted to become the third leading cause of global mortality by 2020. The root causes for disease onset remain poorly understood and no cures are available. This review presents an overview of metabolomics followed by in-depth discussion of its application to the study of respiratory diseases, including the design of metabolomics experiments, choice of clinical material collected and potentially confounding experimental factors. Particular challenges in the field are presented and placed within the context of the future of the applications of metabolomics approaches to the study of respiratory diseases.
Collapse
|
47
|
Ciavardelli D, D'Orazio M, Pieroni L, Consalvo A, Rossi C, Sacchetta P, Di Ilio C, Battistoni A, Urbani A. Proteomic and ionomic profiling reveals significant alterations of protein expression and calcium homeostasis in cystic fibrosis cells. MOLECULAR BIOSYSTEMS 2013; 9:1117-26. [DOI: 10.1039/c3mb25594h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
48
|
Eiserich JP, Yang J, Morrissey BM, Hammock BD, Cross CE. Omics approaches in cystic fibrosis research: a focus on oxylipin profiling in airway secretions. Ann N Y Acad Sci 2012; 1259:1-9. [PMID: 22758630 DOI: 10.1111/j.1749-6632.2012.06580.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cystic fibrosis (CF) is associated with abnormal lipid metabolism, intense respiratory tract (RT) infection, and inflammation, eventually resulting in lung tissue destruction and respiratory failure. The CF RT inflammatory milieu, as reflected by airway secretions, includes a complex array of inflammatory mediators, bacterial products, and host secretions. It is dominated by neutrophils and their proteolytic and oxidative products and includes a wide spectrum of bioactive lipids produced by both host and presumably microbial metabolic pathways. The fairly recent advent of "omics" technologies has greatly increased capabilities of further interrogating this easily obtainable RT compartment that represents the apical culture media of the underlying RT epithelial cells. This paper discusses issues related to the study of CF omics with a focus on the profiling of CF RT oxylipins. Challenges in their identification/quantitation in RT fluids, their pathways of origin, and their potential utility for understanding CF RT inflammatory and oxidative processes are highlighted. Finally, the utility of oxylipin metabolic profiling in directing optimal therapeutic approaches and determining the efficacy of various interventions is discussed.
Collapse
Affiliation(s)
- Jason P Eiserich
- Department of Internal Medicine, University of California, Davis, California, USA
| | | | | | | | | |
Collapse
|
49
|
Anjos SM, Robert R, Waller D, Zhang DL, Balghi H, Sampson HM, Ciciriello F, Lesimple P, Carlile GW, Goepp J, Liao J, Ferraro P, Phillipe R, Dantzer F, Hanrahan JW, Thomas DY. Decreasing Poly(ADP-Ribose) Polymerase Activity Restores ΔF508 CFTR Trafficking. Front Pharmacol 2012; 3:165. [PMID: 22988441 PMCID: PMC3439826 DOI: 10.3389/fphar.2012.00165] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/21/2012] [Indexed: 12/04/2022] Open
Abstract
Most cystic fibrosis is caused by mutations in CFTR that prevent its trafficking from the ER to the plasma membrane and is associated with exaggerated inflammation, altered metabolism, and diminished responses to oxidative stress. PARP-1 is activated by oxidative stress and causes energy depletion and cell dysfunction. Inhibition of this enzyme protects against excessive inflammation and recent studies have also implicated it in intracellular protein trafficking. We hypothesized that PARP-1 activity is altered in CF and affects trafficking and function of the most common CF mutant ΔF508 CFTR. Indeed, PARP-1 activity was 2.9-fold higher in CF (ΔF508/ΔF508) human bronchial epithelial primary cells than in non-CF cells, and similar results were obtained by comparing CF vs. non-CF bronchial epithelial cell lines (2.5-fold higher in CFBE41o− vs. 16HBE14o−, P < 0.002). A PARP-1 inhibitor (ABT-888, Veliparib) partially restored CFTR channel activity in CFBE41o− cells overexpressing ΔF508 CFTR. Similarly, reducing PARP-1 activity by 85% in ileum from transgenic CF mice (Cftrtm1Eur) partially rescued ΔF508 CFTR activity to 7% of wild type mouse levels, and similar correction (7.8%) was observed in vivo by measuring salivary secretion. Inhibiting PARP-1 with ABT-888 or siRNA partially restored ΔF508 CFTR trafficking in cell lines, and most ΔF508 CFTR was complex glycosylated when heterologously expressed in PARP-1−/− mouse embryonic fibroblasts. Finally, levels of the mature glycoform of CFTR were reduced by peroxynitrite, a strong activator of PARP-1. These results demonstrate that PARP-1 activity is increased in CF, and identify a novel pathway that could be targeted by proteostatic correctors of CFTR trafficking.
Collapse
Affiliation(s)
- Suzana M Anjos
- Cystic Fibrosis Translational Research Center, Department of Biochemistry, McGill University Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
A Study of Human Bladder Cancer by Serum and Urine Metabonomics. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2012. [DOI: 10.1016/s1872-2040(11)60570-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|