1
|
Matera MG, Rogliani P, Novelli G, Cazzola M. The impact of genomic variants on patient response to inhaled bronchodilators: a comprehensive update. Expert Opin Drug Metab Toxicol 2023. [PMID: 37269324 DOI: 10.1080/17425255.2023.2221848] [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: 03/01/2023] [Accepted: 06/01/2023] [Indexed: 06/05/2023]
Abstract
INTRODUCTION The bronchodilator response (BDR) depends on many factors, including genetic ones. Numerous single nucleotide polymorphisms (SNPs) influencing BDR have been identified. However, despite several studies in this field, genetic variations are not currently being utilized to support the use of bronchodilators. AREAS COVERED In this narrative review, the possible impact of genetic variants on BDR is discussed. EXPERT OPINION Pharmacogenetic studies of β2-agonists have mainly focused on ADRB2 gene. Three SNPs, A46G, C79G, and C491T, have functional significance. However, other uncommon variants may contribute to individual variability in salbutamol response. SNPs haplotypes in ADRB2 may have a role. Many variants in genes coding for muscarinic ACh receptor (mAChR) have been reported, particularly in the M2 and, to a lesser degree, M3 mAChRs, but no consistent evidence for a pharmacological relevance of these SNPs has been reported. Moreover, there is a link between SNPs and ethnic and/or age profiles regarding BDR. Nevertheless, replication of pharmacogenetic results is limited and often, BDR is dissociated from what is expected based on SNP identification. Pharmacogenetic studies on bronchodilators must continue. However, they must integrate data derived from a multi-omics approach with epigenetic factors that may modify BDR.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Paola Rogliani
- Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy
| | - Mario Cazzola
- Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
2
|
Althoff MD, Peterson R, McGrath M, Jin Y, Grasemann H, Sharma S, Federman A, Wisnivesky JP, Holguin F. Phenotypic characteristics of asthma and morbidity are associated with distinct longitudinal changes in L-arginine metabolism. BMJ Open Respir Res 2023; 10:e001683. [PMID: 37270184 PMCID: PMC10254613 DOI: 10.1136/bmjresp-2023-001683] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND The L-arginine metabolome is dysregulated in asthma, though it is not understood how longitudinal changes in L-arginine metabolism differ among asthma phenotypes and relate to disease outcomes. OBJECTIVES To determine the longitudinal associations between phenotypic characteristics with L-arginine metabolites and their relationships with asthma morbidity. METHODS This is a prospective cohort study of 321 patients with asthma followed semiannually for over 18 months with assessments of plasma L-arginine metabolites, asthma control, spirometry, quality of life and exacerbations. Metabolite concentrations and ratios were transformed using the natural logarithm. RESULTS There were many differences in L-arginine metabolism among asthma phenotypes in the adjusted models. Increasing body mass index was associated with increased asymmetric dimethylarginine (ADMA) and depleted L-citrulline. Latinx was associated with increased metabolism via arginase, with higher L-ornithine, proline and L-ornithine/L-citrulline levels, and was found to have higher L-arginine availability compared with white race. With respect to asthma outcomes, increasing L-citrulline was associated with improved asthma control and increasing L-arginine and L-arginine/ADMA were associated with improved quality of life. Increased variability in L-arginine, L-arginine/ADMA, L-arginine/L-ornithine and L-arginine availability index over 12 months were associated with increased exacerbations, OR 4.70 (95% CI 1.35 to 16.37), OR 8.69 (95% CI 1.98 to 38.08), OR 4.17 (95% CI 1.40 to 12.41) and OR 4.95 (95% CI 1.42 to 17.16), respectively. CONCLUSIONS Our findings suggest that L-arginine metabolism is associated with multiple measures of asthma control and may explain, in part, the relationship between age, race/ethnicity and obesity with asthma outcomes.
Collapse
Affiliation(s)
- Meghan Dolan Althoff
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ryan Peterson
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Max McGrath
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Ying Jin
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Hartmut Grasemann
- Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alex Federman
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juan Pablo Wisnivesky
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fernando Holguin
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
3
|
Fonseca FV, Raffay TM, Xiao K, McLaughlin PJ, Qian Z, Grimmett ZW, Adachi N, Wang B, Hausladen A, Cobb BA, Zhang R, Hess DT, Gaston B, Lambert NA, Reynolds JD, Premont RT, Stamler JS. S-nitrosylation is required for β 2AR desensitization and experimental asthma. Mol Cell 2022; 82:3089-3102.e7. [PMID: 35931084 PMCID: PMC9391322 DOI: 10.1016/j.molcel.2022.06.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/18/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022]
Abstract
The β2-adrenergic receptor (β2AR), a prototypic G-protein-coupled receptor (GPCR), is a powerful driver of bronchorelaxation, but the effectiveness of β-agonist drugs in asthma is limited by desensitization and tachyphylaxis. We find that during activation, the β2AR is modified by S-nitrosylation, which is essential for both classic desensitization by PKA as well as desensitization of NO-based signaling that mediates bronchorelaxation. Strikingly, S-nitrosylation alone can drive β2AR internalization in the absence of traditional agonist. Mutant β2AR refractory to S-nitrosylation (Cys265Ser) exhibits reduced desensitization and internalization, thereby amplifying NO-based signaling, and mice with Cys265Ser mutation are resistant to bronchoconstriction, inflammation, and the development of asthma. S-nitrosylation is thus a central mechanism in β2AR signaling that may be operative widely among GPCRs and targeted for therapeutic gain.
Collapse
Affiliation(s)
- Fabio V Fonseca
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Thomas M Raffay
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kunhong Xiao
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Precious J McLaughlin
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zhaoxia Qian
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zachary W Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Naoko Adachi
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benlian Wang
- Center for Proteomics and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Alfred Hausladen
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian A Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Rongli Zhang
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Douglas T Hess
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benjamin Gaston
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - James D Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Richard T Premont
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
4
|
Watchorn D, Menzies-Gow A. Investigational approaches for unmet need in severe asthma. Expert Rev Respir Med 2022; 16:661-678. [PMID: 35786146 DOI: 10.1080/17476348.2022.2096593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/28/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Molecular antibodies (mAb) targeting inflammatory mediators are effective in T2-high asthma. The recent approval of Tezepelumab presents a novel mAb therapeutic option for those with T2-low asthma. AREAS COVERED We discuss a number of clinical problems pertinent to severe asthma that are less responsive to current therapies, such as persistent airflow obstruction and airway hyperresponsiveness. We discuss selected investigational approaches, including a number of candidate therapies under investigation in two adaptive platform trials currently in progress, with particular reference to this unmet need, as well as their potential in phenotypes such as neutrophilic asthma and obese asthma, which may or may not overlap with a T2-high phenotype. EXPERT OPINION The application of discrete targeting approaches to T2-low molecular phenotypes, including those phenotypes in which inflammation may not arise within the airway, has yielded variable results to date. Endotypes associated with T2-low asthma are likely to be diverse but await validation. Investigational therapeutic approaches must, likewise, be diverse if the goal of remission is to become attainable for all those living with asthma.
Collapse
Affiliation(s)
- David Watchorn
- Lung Division, Royal Brompton & Harefield Hospitals,London,UK
| | | |
Collapse
|
5
|
Georas SN, Wright RJ, Ivanova A, Israel E, LaVange LM, Akuthota P, Carr TF, Denlinger LC, Fajt ML, Kumar R, O'Neal WK, Phipatanakul W, Szefler SJ, Aronica MA, Bacharier LB, Burbank AJ, Castro M, Crotty Alexander L, Bamdad J, Cardet JC, Comhair SAA, Covar RA, DiMango EA, Erwin K, Erzurum SC, Fahy JV, Gaffin JM, Gaston B, Gerald LB, Hoffman EA, Holguin F, Jackson DJ, James J, Jarjour NN, Kenyon NJ, Khatri S, Kirwan JP, Kraft M, Krishnan JA, Liu AH, Liu MC, Marquis MA, Martinez F, Mey J, Moore WC, Moy JN, Ortega VE, Peden DB, Pennington E, Peters MC, Ross K, Sanchez M, Smith LJ, Sorkness RL, Wechsler ME, Wenzel SE, White SR, Zein J, Zeki AA, Noel P. The Precision Interventions for Severe and/or Exacerbation-Prone (PrecISE) Asthma Network: An overview of Network organization, procedures, and interventions. J Allergy Clin Immunol 2022; 149:488-516.e9. [PMID: 34848210 PMCID: PMC8821377 DOI: 10.1016/j.jaci.2021.10.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
Asthma is a heterogeneous disease, with multiple underlying inflammatory pathways and structural airway abnormalities that impact disease persistence and severity. Recent progress has been made in developing targeted asthma therapeutics, especially for subjects with eosinophilic asthma. However, there is an unmet need for new approaches to treat patients with severe and exacerbation-prone asthma, who contribute disproportionately to disease burden. Extensive deep phenotyping has revealed the heterogeneous nature of severe asthma and identified distinct disease subtypes. A current challenge in the field is to translate new and emerging knowledge about different pathobiologic mechanisms in asthma into patient-specific therapies, with the ultimate goal of modifying the natural history of disease. Here, we describe the Precision Interventions for Severe and/or Exacerbation-Prone Asthma (PrecISE) Network, a groundbreaking collaborative effort of asthma researchers and biostatisticians from around the United States. The PrecISE Network was designed to conduct phase II/proof-of-concept clinical trials of precision interventions in the population with severe asthma, and is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health. Using an innovative adaptive platform trial design, the PrecISE Network will evaluate up to 6 interventions simultaneously in biomarker-defined subgroups of subjects. We review the development and organizational structure of the PrecISE Network, and choice of interventions being studied. We hope that the PrecISE Network will enhance our understanding of asthma subtypes and accelerate the development of therapeutics for severe asthma.
Collapse
Affiliation(s)
- Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY.
| | | | - Anastasia Ivanova
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Elliot Israel
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Lisa M LaVange
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Praveen Akuthota
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Tara F Carr
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Merritt L Fajt
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | | | - Wanda K O'Neal
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | | | - Stanley J Szefler
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Mark A Aronica
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Allison J Burbank
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, Mo
| | - Laura Crotty Alexander
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Julie Bamdad
- Division of Lung Diseases, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Md
| | | | | | | | | | - Kim Erwin
- Institute for Healthcare Delivery Design, University of Illinois at Chicago, Chicago, Ill
| | | | - John V Fahy
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | | | - Benjamin Gaston
- Wells Center for Pediatric Research, Indiana University, Indianapolis, Ind
| | - Lynn B Gerald
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | | | - Daniel J Jackson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - John James
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Sumita Khatri
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - John P Kirwan
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, La
| | - Monica Kraft
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Ill
| | - Andrew H Liu
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Mark C Liu
- Pulmonary and Critical Care Medicine, Department of Medicine, the Johns Hopkins University, Baltimore, Md
| | - M Alison Marquis
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Fernando Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jacob Mey
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, La
| | - Wendy C Moore
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - James N Moy
- Rush University Medical Center, Chicago, Ill
| | - Victor E Ortega
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - David B Peden
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | | | - Michael C Peters
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | - Kristie Ross
- The Cleveland Clinic, Cleveland, Ohio; UH Rainbow Babies and Children's Hospitals, Cleveland, Ohio
| | - Maria Sanchez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | | | - Ronald L Sorkness
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Michael E Wechsler
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Sally E Wenzel
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Steven R White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Joe Zein
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amir A Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Patricia Noel
- Division of Lung Diseases, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Md
| |
Collapse
|
6
|
Marozkina N, Gaston B. An Update on Thiol Signaling: S-Nitrosothiols, Hydrogen Sulfide and a Putative Role for Thionitrous Acid. Antioxidants (Basel) 2020; 9:antiox9030225. [PMID: 32164188 PMCID: PMC7139563 DOI: 10.3390/antiox9030225] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/25/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022] Open
Abstract
Long considered vital to antioxidant defenses, thiol chemistry has more recently been recognized to be of fundamental importance to cell signaling. S-nitrosothiols—such as S-nitrosoglutathione (GSNO)—and hydrogen sulfide (H2S) are physiologic signaling thiols that are regulated enzymatically. Current evidence suggests that they modify target protein function primarily through post-translational modifications. GSNO is made by NOS and other metalloproteins; H2S by metabolism of cysteine, homocysteine and cystathionine precursors. GSNO generally acts independently of NO generation and has a variety of gene regulatory, immune modulator, vascular, respiratory and neuronal effects. Some of this physiology is shared with H2S, though the mechanisms differ. Recent evidence also suggests that molecules resulting from reactions between GSNO and H2S, such as thionitrous acid (HSNO), could also have a role in physiology. Taken together, these data suggest important new potential targets for thiol-based drug development.
Collapse
Affiliation(s)
- Nadzeya Marozkina
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
- Indiana University, School of Medicine, 1044 W. Walnut Street, R4-474 Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +317-274-7427
| | - Benjamin Gaston
- Herman Wells Center for Pediatric Research, Riley Hospital for Children, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| |
Collapse
|
7
|
Einisman HJ, Gaston B, Wijers C, Smith LA, Lewis TH, Lewis SJ, Raffay TM. Tracheomalacia in bronchopulmonary dysplasia: Trachealis hyper-relaxant responses to S-nitrosoglutathione in a hyperoxic murine model. Pediatr Pulmonol 2019; 54:1989-1996. [PMID: 31486289 PMCID: PMC7329187 DOI: 10.1002/ppul.24513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) with airway hyperreactivity is a long-term pulmonary complication of prematurity. The endogenous nonadrenergic, noncholinergic signaling molecule, S-nitrosoglutathione (GSNO) and its catabolism by GSNO reductase (GSNOR) modulate airway reactivity. Tracheomalacia is a major, underinvestigated complication of BPD. We studied trachealis, left main bronchus (LB), and intrapulmonary bronchiolar (IPB) relaxant responses to GSNO in a murine hyperoxic BPD model. METHODS Wild-type (WT) or GSNOR knockout (KO) newborn mice were raised in 60% (BPD) or 21% (control) oxygen during the first 3 weeks of life. After room air recovery, adult trachealis, LB, and IPB smooth muscle relaxant responses to GSNO (after methacholine preconstriction) were studied using wire myographs. Studies were repeated after GSNOR inhibitor (GSNORi) pretreatment and in KO mice. RESULTS GSNO relaxed all airway preparations. GSNO relaxed WT BPD trachealis substantially more than WT controls (P < .05). Pharmacologic or genetic ablation of GSNOR abolished the exaggerated BPD tracheal relaxation to GSNO and also augmented BPD IPB relaxation to GSNO. LB ring contractility was not significantly different between groups or conditions. Additionally, GSNORi treatment induced relaxation of WT IPBs but not trachealis or LB. CONCLUSION GSNO dramatically relaxed the trachealis in our BPD model, an effect paradoxically reversed by loss of GSNOR. Conversely, GSNOR inhibition augmented IBP relaxation. These data suggest that GSNOR inhibition could benefit both the BPD trachealis and distal airways, restoring relaxant responses to those of room air controls. Because therapeutic options are limited in this high-risk population, future studies of GSNOR inhibition are needed.
Collapse
Affiliation(s)
- Helly J Einisman
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Pediatric Pulmonology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Benjamin Gaston
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Pediatric Pulmonology, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Christiaan Wijers
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Laura A Smith
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Tristan H Lewis
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Stephen J Lewis
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Pediatric Pulmonology, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Thomas M Raffay
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Division of Neonatology, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| |
Collapse
|
8
|
Zhang E, Levin AM, Williams LK. How does race and ethnicity effect the precision treatment of asthma? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019; 4:337-356. [PMID: 33015363 DOI: 10.1080/23808993.2019.1690396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction Asthma is a common condition that affects large numbers of children and adults, yet the burden of disease is not equally distributed amongst groups. In the United States, African Americans and Puerto Ricans have higher rates of asthma and its complications when compared with European Americans. However, clinical trials and genetic studies have largely focused on the latter group. Areas covered Here we examine what is known regarding differences in asthma treatment response by race-ethnicity. We also review existing genetic studies related to the use of asthma medications, paying special attention to studies that included substantial numbers of non-white population groups. Publicly accessible search engines of the medical literature were queried using combinations of the terms asthma, race, ethnicity, pharmacogenomics, and pharmacogenetics, as well as the names of individual asthma medication classes. The list of articles reviewed was supplemented by bibliographies and expert knowledge. Expert opinion A substantial and coordinated effort is still needed to both identify and validate genetic biomarkers of asthma medication response, as currently there are no clinically actionable genetic markers available for this purpose. The path to identifying such markers in non-white populations is even more formidable, since these groups are underrepresented in existing data.
Collapse
Affiliation(s)
- Ellen Zhang
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - L Keoki Williams
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|
9
|
Almomani BA, Al-Eitan LN, Al-Sawalha NA, Samrah SM, Al-Quasmi MN. Association of genetic variants with level of asthma control in the Arab population. J Asthma Allergy 2019; 12:35-42. [PMID: 30774389 PMCID: PMC6350652 DOI: 10.2147/jaa.s186252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Rates of asthma in Jordan have been doubled in the past decade, but this increased prevalence was not met with improved asthma control protocols. The aim of the present study was to assess whether there was any significant association between the level of asthma control and certain single-nucleotide polymorphisms (SNPs) in five genes: (ADRB2; rs1042713 and rs1042714), (CRHR1; rs1876828, rs242939, and rs242941), (STIP1; rs2236647), (ADH5, rs1154400), and (ARG1; rs2781659). These SNPs were selected based on their involvement in enzymes and receptors that are related to asthma pathways and subsequent response to medication and based on a high degree of linkage disequilibrium. Patients and methods A cross-sectional genetic association study was conducted from June 2016 to June 2017 in the two major hospitals in Jordan. The present study involved sampling from adult asthmatic patients of Arab descent who were selected from two phenotypic groups, ie, controlled and uncontrolled asthma. The blood samples and medical data were collected from the participants. DNA samples were extracted, quantified, and genotyped according to standard operating procedure. Allelic and haplotypic analyses were performed using the Haploview®. Results A total of 245 Arab asthmatic patients were enrolled in this study. Genotyping analysis revealed that the two SNPs (rs1042713 and rs1042714) in ADRB2 gene, along with their related haplotypes, were nominally significantly associated with asthma control in the Jordanian population. The A-allele of rs1042713 and the C-allele of rs1042714 were more common in the uncontrolled asthma group than in the controlled asthma group (P=0.048 and P=0.017, respectively). Conclusion This was the first study that identified the nominal significant association between the level of asthma control and genetic variants in ADRB2 gene in Arab population. Further studies in other Arab region with larger sample size are recommended to confirm the relationship.
Collapse
Affiliation(s)
- Basima A Almomani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan,
| | - Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, Jordan.,Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour A Al-Sawalha
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan,
| | - Shaher M Samrah
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan.,Department of Internal Medicine, King Abdullah University Hospital, Irbid, Jordan
| | - Mohammed N Al-Quasmi
- Hematology Laboratory, Department of Medical Laboratory, King Abdullah University Hospital, Irbid, Jordan
| |
Collapse
|
10
|
Que LG, Yang Z, Lugogo NL, Katial RK, Shoemaker SA, Troha JM, Rodman DM, Tighe RM, Kraft M. Effect of the S-nitrosoglutathione reductase inhibitor N6022 on bronchial hyperreactivity in asthma. IMMUNITY INFLAMMATION AND DISEASE 2018; 6:322-331. [PMID: 29642282 PMCID: PMC5946144 DOI: 10.1002/iid3.220] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 12/20/2022]
Abstract
RATIONALE Patients with asthma demonstrate depletion of the endogenous bronchodilator GSNO and upregulation of GSNOR. OBJECTIVES An exploratory proof of concept clinical study of N6022 in mild asthma to determine the potential bronchoprotective effects of GSNOR inhibition. Mechanistic studies aimed to provide translational evidence of effect. METHODS Fourteen mild asthma patients were treated with intravenous N6022 (5 mg) or placebo and observed for 7 days, with repeated assessments of the provocative dose of methacholine causing a 20% fall in FEV1 (methacholine PC20 FEV1), followed by a washout period and crossover treatment and observation. In vitro studies in isolated eosinophils investigated the effect of GSNO and N6022 on apoptosis. MEASUREMENTS AND MAIN RESULTS This was a negative trial as it failed to reach its primary endpoint, which was change from baseline in methacholine PC20 FEV1 at 24 h. However, our exploratory analysis demonstrated significantly more two dose-doubling increases in PC20 FEV1 for N6022 compared with placebo (21% vs 6%, P < 0.05) over the 7-day observation period. Furthermore, a significant treatment effect was observed in the change in PC20 FEV1 from baseline averaged over the 7-day observation period (mean change: +0.82 mg/ml [N6022] from 1.34 mg/ml [baseline] vs -0.18 mg/ml [placebo] from 1.16 mg/ml [baseline], P = 0.023). N6022 was well tolerated in mild asthmatics. In vitro studies demonstrated enhanced eosinophilic apoptosis with N6022. CONCLUSIONS In this early phase exploratory proof of concept trial in asthma, N6022 did not significantly alter methacholine PC20 FEV1 at 24 h, but did have a treatment effect at 7 days compared to baseline. Further investigation of the efficacy of S-nitrosoglutathione reductase inhibition in a patient population with eosinophilic asthma is warranted.
Collapse
Affiliation(s)
- Loretta G Que
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Zhonghui Yang
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Njira L Lugogo
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Rohit K Katial
- Division of Allergy & Clinical Immunology, National Jewish Health, Denver, Colorado, USA
| | | | | | | | - Robert M Tighe
- Departmentof Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Monica Kraft
- Department of Medicine, College of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| |
Collapse
|
11
|
Sánchez-Martín A, García-Sánchez A, Isidoro-García M. Review on Pharmacogenetics and Pharmacogenomics Applied to the Study of Asthma. Methods Mol Biol 2017; 1434:255-72. [PMID: 27300544 DOI: 10.1007/978-1-4939-3652-6_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nearly one-half of asthmatic patients do not respond to the most common therapies. Evidence suggests that genetic factors may be involved in the heterogeneity in therapeutic response and adverse events to asthma therapies. We focus on the three major classes of asthma medication: β-adrenergic receptor agonist, inhaled corticosteroids, and leukotriene modifiers. Pharmacogenetics and pharmacogenomics studies have identified several candidate genes associated with drug response.In this chapter, the main pharmacogenetic and pharmacogenomic studies in addition to the future perspectives in personalized medicine will be reviewed. The ideal treatment of asthma would be a tailored approach to health care in which adverse effects are minimized and the therapeutic benefit for an individual asthmatic is maximized leading to a more cost-effective care.
Collapse
Affiliation(s)
- Almudena Sánchez-Martín
- Department of Pharmacy, University Hospital of Salamanca, Salamanca, Spain.,Salamanca Institute for Biomedical Research (IBSAL), Salamanca, Spain
| | - Asunción García-Sánchez
- Salamanca Institute for Biomedical Research (IBSAL), Salamanca, Spain.,Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - María Isidoro-García
- Salamanca Institute for Biomedical Research (IBSAL), Salamanca, Spain. .,Department of Clinical Biochemistry, University Hospital of Salamanca, Salamanca, Spain. .,Department of Medicine, University of Salamanca, Salamanca, Spain.
| |
Collapse
|
12
|
Isidoro-García M, Sánchez-Martín A, García-Sánchez A, Sanz C, García-Berrocal B, Dávila I. Pharmacogenetics and the treatment of asthma. Pharmacogenomics 2017; 18:1271-1280. [PMID: 28776467 DOI: 10.2217/pgs-2017-0024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heterogeneity defines both the natural history of asthma as well as patient's response to treatment. Pharmacogenomics contribute to understand the genetic basis of drug response and thus to define new therapeutic targets or molecular biomarkers to evaluate treatment effectiveness. This review is initially focused on different genes so far involved in the pharmacological response to asthma treatment. Specific considerations regarding allergic asthma, the pharmacogenetics aspects of polypharmacy and the application of pharmacogenomics in new drugs in asthma will also be addressed. Finally, future perspectives related to epigenetic regulatory elements and the potential impact of systems biology in pharmacogenetics of asthma will be considered.
Collapse
Affiliation(s)
- María Isidoro-García
- Department of Clinical Biochemistry, Pharmacogenetics Unit, University Hospital of Salamanca, Salamanca, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), Allergy Department, Salamanca, Spain.,Department of Medicine, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Almudena Sánchez-Martín
- Institute for Biomedical Research of Salamanca (IBSAL), Allergy Department, Salamanca, Spain.,Department of Pharmacy, Faculty of Medicine, University Hospital of Salamanca, Salamanca, Spain
| | - Asunción García-Sánchez
- Institute for Biomedical Research of Salamanca (IBSAL), Allergy Department, Salamanca, Spain.,Department of Biomedical & Diagnostic Sciences, Faculty of Medicine, University of Salamanca, Spain
| | - Catalina Sanz
- Institute for Biomedical Research of Salamanca (IBSAL), Allergy Department, Salamanca, Spain.,Department of Microbiology & Genetics, Faculty of Biology, University of Salamanca, Salamanca, Spain
| | - Belén García-Berrocal
- Department of Clinical Biochemistry, Pharmacogenetics Unit, University Hospital of Salamanca, Salamanca, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), Allergy Department, Salamanca, Spain
| | - Ignacio Dávila
- Institute for Biomedical Research of Salamanca (IBSAL), Allergy Department, Salamanca, Spain.,Department of Biomedical & Diagnostic Sciences, Faculty of Medicine, University of Salamanca, Spain.,Department of Allergy, Faculty of Medicine, University Hospital of Salamanca, Salmanaca, Spain
| |
Collapse
|
13
|
Sun BL, Palmer L, Alam SR, Adekoya I, Brown-Steinke K, Periasamy A, Mutus B. O-Aminobenzoyl-S-nitrosoglutathione: A fluorogenic, cell permeable, pseudo-substrate for S-nitrosoglutathione reductase. Free Radic Biol Med 2017; 108:445-451. [PMID: 28419866 DOI: 10.1016/j.freeradbiomed.2017.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 10/19/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR) is a multifunctional enzyme. It can catalyze NADH-dependent reduction of S-nitrosoglutathione (GSNO); as well as NAD+-dependent oxidation of hydroxymethylglutathione (HMGSH; an adduct formed by the spontaneous reaction between formaldehyde and glutathione). While initially recognized as the enzyme that is involved in formaldehyde detoxification, increasing amount of evidence has shown that GSNOR also plays a significant role in nitric oxide mediated signaling through its modulation of protein S-nitrosothiol signaling. In humans, GSNOR/S-nitrosothiols have been implicated in the etiology of several diseases including lung cancer, cystic fibrosis, asthma, pulmonary hypertension, and neuronal dysfunction. Currently, it is not possible to monitor the activity of GSNOR in live cells. In this article, we present a new compound, O-aminobenzoyl-S-nitrosoglutathione (OAbz-GSNO), which acts as a fluorogenic pseudo-substrate for GSNOR with an estimated Km value of 320µM. The weak OAbz-GSNO fluorescence increases by approximately 14 fold upon reduction of its S-NO moiety. In live cell imaging studies, OAbz-GSNO is readily taken up by primary pulmonary endothelial cells and localizes to the same perinuclear region as GSNOR. The perinuclear OAbz-GSNO fluorescence increases in a time dependent manner and this increase in fluorescence is abolished by siRNA knockdown of GSNOR or by treatment with GSNOR-specific inhibitors N6022 and C3. Taken together, these data demonstrate that OAbz-GSNO can be used as a tool to monitor the activity of GSNOR in live cells.
Collapse
Affiliation(s)
- Bei Lei Sun
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Lisa Palmer
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | | | - Itunuoluwa Adekoya
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | | | - Ammasi Periasamy
- W. M. Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Bulent Mutus
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada.
| |
Collapse
|
14
|
Barnett SD, Buxton ILO. The role of S-nitrosoglutathione reductase (GSNOR) in human disease and therapy. Crit Rev Biochem Mol Biol 2017; 52:340-354. [PMID: 28393572 PMCID: PMC5597050 DOI: 10.1080/10409238.2017.1304353] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR), or ADH5, is an enzyme in the alcohol dehydrogenase (ADH) family. It is unique when compared to other ADH enzymes in that primary short-chain alcohols are not its principle substrate. GSNOR metabolizes S-nitrosoglutathione (GSNO), S-hydroxymethylglutathione (the spontaneous adduct of formaldehyde and glutathione), and some alcohols. GSNOR modulates reactive nitric oxide (•NO) availability in the cell by catalyzing the breakdown of GSNO, and indirectly regulates S-nitrosothiols (RSNOs) through GSNO-mediated protein S-nitrosation. The dysregulation of GSNOR can significantly alter cellular homeostasis, leading to disease. GSNOR plays an important regulatory role in smooth muscle relaxation, immune function, inflammation, neuronal development and cancer progression, among many other processes. In recent years, the therapeutic inhibition of GSNOR has been investigated to treat asthma, cystic fibrosis and interstitial lung disease (ILD). The direct action of •NO on cellular pathways, as well as the important regulatory role of protein S-nitrosation, is closely tied to GSNOR regulation and defines this enzyme as an important therapeutic target.
Collapse
Affiliation(s)
- Scott D Barnett
- a Department of Pharmacology , University of Nevada, Reno School of Medicine , Reno , NV , USA
| | - Iain L O Buxton
- a Department of Pharmacology , University of Nevada, Reno School of Medicine , Reno , NV , USA
| |
Collapse
|
15
|
Personalized Medicine. Respir Med 2017. [DOI: 10.1007/978-3-319-43447-6_13] [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: 10/20/2022]
|
16
|
Almomani BA, Al-Eitan LN, Samrah SM, Al-Quasmi MN, McKnight AJ. Candidate gene analysis of asthma in a population of Arab descent: a case-control study in Jordan. Per Med 2016; 14:51-61. [PMID: 29749828 DOI: 10.2217/pme-2016-0059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AIM To evaluate whether SNPs (n = 15) in ten candidate genes (ADRB2, ADH5, ARGI, CRHR1, STIP1, LTA4H, LTC4S, ALOX5, ABCC1 and OATP2B1) are associated with asthma in Jordanian population of Arab descent. METHODS A case-control study included 245 adult asthmatics and 249 controls. RESULTS Significant genetic association was identified at the rs2236647 (T/C) SNP in STIP1 and risk of asthma (p < 0.001). The C allele and CC genotype of this SNP were significantly higher in asthmatics compared with controls. The rs1141370 SNP (Val34Met) in ADRB2 is not polymorphic in our cohort. CONCLUSION The rs2236647 SNP could act as a reliable tool to identify individuals at risk of developing asthma and provision of early intervention in population of Arab descent.
Collapse
Affiliation(s)
- Basima A Almomani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science & Technology, Irbid, Jordan
| | - Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science & Technology, Irbid, Jordan.,Department of Biotechnology & Genetic Engineering, Jordan University of Science & Technology, Irbid, Jordan
| | - Shaher M Samrah
- Faculty of Medicine, Jordan University of Science & Technology, Irbid, Jordan.,Department of Internal Medicine, King Abdullah University Hospital, Irbid, Jordan
| | - Mohammed N Al-Quasmi
- Department of Medical Laboratory, King Abdullah University Hospital, Irbid, Jordan
| | - Amy J McKnight
- Centre for Public Health, Queen's University of Belfast, Northern Ireland, UK
| |
Collapse
|
17
|
Carmichael SL, Yang W, Ma C, Roberts E, Kegley S, English P, Lammer EJ, Witte JS, Shaw GM. Joint effects of genetic variants and residential proximity to pesticide applications on hypospadias risk. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2016; 106:653-8. [PMID: 27098078 PMCID: PMC4983249 DOI: 10.1002/bdra.23508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 01/24/2023]
Abstract
BACKGROUND We examined risks associated with joint exposure of gene variants and pesticides. METHODS Analyses included 189 cases and 390 male controls born from 1991 to 2003 in California's San Joaquin Valley. We used logistic regression to examine risks associated with joint exposures of gene variants and pesticides that our previous work identified as associated with hypospadias. Genetic variables were based on variants in DGKK, genes involved in sex steroid synthesis/metabolism, and genes involved in genital tubercle development. Pesticide exposure was based on residential proximity to commercial agricultural pesticide applications. RESULTS Odds ratios (ORs) were highest among babies with joint exposures, who had two- to fourfold increased risks; for example, the OR was 3.7 (95% confidence interval [CI], 0.8-16.5) among subjects with the risk-associated DGKK haplotype and pesticide exposure; OR, 1.5 (95% CI, 0.7-3.1) among subjects with the haplotype and no pesticide exposure; and OR, 0.9 (95% CI, 0.5-1.6) among subjects without the haplotype but with pesticide exposure, relative to subjects with neither. However, results did not provide statistical evidence that these risks were significantly greater than expected on an additive scale, relative to risks associated with one exposure at a time. CONCLUSION We observed elevated risks associated with joint exposures to selected pesticides and genetic variants but no statistical evidence for interaction. Birth Defects Research (Part A) 106:653-658, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Suzan L Carmichael
- Department of Pediatrics, Division of Neonatology and Developmental Medicine, Stanford University School of Medicine, Stanford, California
| | - Wei Yang
- Department of Pediatrics, Division of Neonatology and Developmental Medicine, Stanford University School of Medicine, Stanford, California
| | - Chen Ma
- Department of Pediatrics, Division of Neonatology and Developmental Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Susan Kegley
- Pesticide Research Institute, Berkeley, California
| | - Paul English
- California Department of Public Health, Richmond, California
| | - Edward J Lammer
- UCSF Benioff Children's Hospital Oakland, Oakland, California
| | - John S Witte
- Department of Epidemiology and Biostatistics and Institute of Human Genetics, University of California, San Francisco, California
| | - Gary M Shaw
- Department of Pediatrics, Division of Neonatology and Developmental Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
18
|
Ortega VE, Meyers DA, Bleecker ER. Asthma pharmacogenetics and the development of genetic profiles for personalized medicine. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2015; 8:9-22. [PMID: 25691813 PMCID: PMC4325626 DOI: 10.2147/pgpm.s52846] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human genetics research will be critical to the development of genetic profiles for personalized or precision medicine in asthma. Genetic profiles will consist of gene variants that predict individual disease susceptibility and risk for progression, predict which pharmacologic therapies will result in a maximal therapeutic benefit, and predict whether a therapy will result in an adverse response and should be avoided in a given individual. Pharmacogenetic studies of the glucocorticoid, leukotriene, and β2-adrenergic receptor pathways have focused on candidate genes within these pathways and, in addition to a small number of genome-wide association studies, have identified genetic loci associated with therapeutic responsiveness. This review summarizes these pharmacogenetic discoveries and the future of genetic profiles for personalized medicine in asthma. The benefit of a personalized, tailored approach to health care delivery is needed in the development of expensive biologic drugs directed at a specific biologic pathway. Prior pharmacogenetic discoveries, in combination with additional variants identified in future studies, will form the basis for future genetic profiles for personalized tailored approaches to maximize therapeutic benefit for an individual asthmatic while minimizing the risk for adverse events.
Collapse
Affiliation(s)
- Victor E Ortega
- Center for Genomics and Personalized Medicine Research, Pulmonary Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Deborah A Meyers
- Center for Genomics and Personalized Medicine Research, Pulmonary Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Eugene R Bleecker
- Center for Genomics and Personalized Medicine Research, Pulmonary Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
19
|
Marozkina NV, Wang XQ, Stsiapura V, Fitzpatrick A, Carraro S, Hawkins GA, Bleecker E, Meyers D, Jarjour N, Fain SB, Wenzel S, Busse W, Castro M, Panettieri RA, Moore W, Lewis SJ, Palmer LA, Altes T, de Lange EE, Erzurum S, Teague WG, Gaston B. Phenotype of asthmatics with increased airway S-nitrosoglutathione reductase activity. Eur Respir J 2015; 45:87-97. [PMID: 25359343 PMCID: PMC4283933 DOI: 10.1183/09031936.00042414] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
S-Nitrosoglutathione is an endogenous airway smooth muscle relaxant. Increased airway S-nitrosoglutathione breakdown occurs in some asthma patients. We asked whether patients with increased airway catabolism of this molecule had clinical features that distinguished them from other asthma patients. We measured S-nitrosoglutathione reductase expression and activity in bronchoscopy samples taken from 66 subjects in the Severe Asthma Research Program. We also analysed phenotype and genotype data taken from the program as a whole. Airway S-nitrosoglutathione reductase activity was increased in asthma patients (p=0.032). However, only a subpopulation was affected and this subpopulation was not defined by a "severe asthma" diagnosis. Subjects with increased activity were younger, had higher IgE and an earlier onset of symptoms. Consistent with a link between S-nitrosoglutathione biochemistry and atopy: 1) interleukin 13 increased S-nitrosoglutathione reductase expression and 2) subjects with an S-nitrosoglutathione reductase single nucleotide polymorphism previously associated with asthma had higher IgE than those without this single nucleotide polymorphism. Expression was higher in airway epithelium than in smooth muscle and was increased in regions of the asthmatic lung with decreased airflow. An early-onset, allergic phenotype characterises the asthma population with increased S-nitrosoglutathione reductase activity.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- Dept of Paediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Xin-Qun Wang
- Dept of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Vitali Stsiapura
- Dept of Chemistry, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - Eugene Bleecker
- Dept of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Deborah Meyers
- Dept of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Nizar Jarjour
- Dept of Medicine, University of Wisconsin, Madison, WI, USA
| | - Sean B Fain
- Dept of Medical Physics, University of Wisconsin, Madison, WI, USA
| | | | - William Busse
- Dept of Medicine, University of Wisconsin, Madison, WI, USA
| | - Mario Castro
- Dept of Medicine, Washington University, St. Louis, MO, USA
| | - Reynold A Panettieri
- Pulmonary, Allergy and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Wendy Moore
- Dept of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Stephen J Lewis
- Dept of Paediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Lisa A Palmer
- Dept of Paediatrics, University of Virginia, Charlottesville, VA, USA
| | - Talissa Altes
- Dept of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Eduard E de Lange
- Dept of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Serpil Erzurum
- Dept of Pathobiology, Cleveland Clinic, Cleveland, OH, USA Dept of Pulmonary, Allergy, and Critical Care Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - W Gerald Teague
- Dept of Paediatrics, University of Virginia, Charlottesville, VA, USA
| | - Benjamin Gaston
- Dept of Paediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
20
|
Huffaker MF, Phipatanakul W. Pediatric asthma: guidelines-based care, omalizumab, and other potential biologic agents. Immunol Allergy Clin North Am 2014; 35:129-44. [PMID: 25459581 DOI: 10.1016/j.iac.2014.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Over the past several decades, the evidence supporting rational pediatric asthma management has grown considerably. As more is learned about the various phenotypes of asthma, the complexity of management will continue to grow. This article focuses on the evidence supporting the current guidelines-based pediatric asthma management and explores the future of asthma management with respect to phenotypic heterogeneity and biologics.
Collapse
Affiliation(s)
- Michelle Fox Huffaker
- Division of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Wanda Phipatanakul
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Lima JJ. Do genetic polymorphisms alter patient response to inhaled bronchodilators? Expert Opin Drug Metab Toxicol 2014; 10:1231-40. [PMID: 25102170 DOI: 10.1517/17425255.2014.939956] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Short- and long-acting β agonists (SABA and LABA) are bronchodilators for treating asthma. Bronchodilator response (BDR) is quantified by measuring air expired in the first second during a forced expiratory maneuver, prior to and following inhalation of SABA. BDR has been associated with a significant degree of heterogeneity, in part attributable to genetic variation. Heritability, the proportion of phenotypic variability accounted for by genetic variation is estimated to account for 50% of pulmonary function and 28.5% for BDR. AREAS COVERED A MEDLINE search for English articles published from January 1990 to June 2014 was completed using the terms: bronchodilator, bronchodilator response, short-acting bronchodilator, long-acting bronchodilator, β2 adrenergic receptor gene (ADRB2), asthma and pharmacogenomics. The effects of ADRB2 variants on BDR and the safety of SABA and LABA + inhaled corticosteroids have been studied with equivocal results. Single and candidate gene studies have identified variants in other genes that alter response to bronchodilators. Associations were recently observed between hospital admission rates and two rare ADRB2 polymorphisms: Thr164Ile and a 25 base pair insertion-deletion at nucleotide -376. This was the first report of life-threatening events associated with LABA being linked to rare ADRB2 variants. EXPERT OPINION Pharmacogenomic studies over the last two decades clearly demonstrate that polymorphisms alter patient response to bronchodilators in patients with asthma.
Collapse
Affiliation(s)
- John J Lima
- Center for Pharmacogenomics and Translational Research, Nemours Children's Clinic , 807 Children's Way, Jacksonville, FL 32207 , USA , +1 904 697 3683 ; +1 904 687 7988 ;
| |
Collapse
|
22
|
Ortega VE. Pharmacogenetics of beta2 adrenergic receptor agonists in asthma management. Clin Genet 2014; 86:12-20. [PMID: 24641588 DOI: 10.1111/cge.12377] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/10/2014] [Accepted: 03/10/2014] [Indexed: 12/25/2022]
Abstract
Beta2 (β2) adrenergic receptor agonists (beta agonists) are a commonly prescribed treatment for asthma despite the small increase in risk for life-threatening adverse responses associated with long-acting beta agonist (LABA). The concern for life-threatening adverse effects associated with LABA and the inter-individual variability of therapeutic responsiveness to LABA-containing combination therapies provide the rationale for pharmacogenetic studies of beta agonists. These studies primarily evaluated genes within the β2-adrenergic receptor and related pathways; however, recent genome-wide studies have identified novel loci for beta agonist response. Recent studies have identified a role for rare genetic variants in determining beta agonist response and, potentially, the risk for rare, adverse responses to LABA. Before genomics research can be applied to the development of genetic profiles for personalized medicine, it will be necessary to continue adapting to the analysis of an increasing volume of genetic data in larger cohorts with a combination of analytical methods and in vitro studies.
Collapse
Affiliation(s)
- V E Ortega
- Center for Genomics and Personalized Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
23
|
Ortega VE, Meyers DA. Pharmacogenetics: implications of race and ethnicity on defining genetic profiles for personalized medicine. J Allergy Clin Immunol 2014; 133:16-26. [PMID: 24369795 DOI: 10.1016/j.jaci.2013.10.040] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/22/2013] [Accepted: 10/23/2013] [Indexed: 01/06/2023]
Abstract
Pharmacogenetics is being used to develop personalized therapies specific to subjects from different ethnic or racial groups. To date, pharmacogenetic studies have been primarily performed in trial cohorts consisting of non-Hispanic white subjects of European descent. A "bottleneck" or collapse of genetic diversity associated with the first human colonization of Europe during the Upper Paleolithic period, followed by the recent mixing of African, European, and Native American ancestries, has resulted in different ethnic groups with varying degrees of genetic diversity. Differences in genetic ancestry might introduce genetic variation, which has the potential to alter the therapeutic efficacy of commonly used asthma therapies, such as β2-adrenergic receptor agonists (β-agonists). Pharmacogenetic studies of admixed ethnic groups have been limited to small candidate gene association studies, of which the best example is the gene coding for the receptor target of β-agonist therapy, the β2-adrenergic receptor (ADRB2). Large consortium-based sequencing studies are using next-generation whole-genome sequencing to provide a diverse genome map of different admixed populations, which can be used for future pharmacogenetic studies. These studies will include candidate gene studies, genome-wide association studies, and whole-genome admixture-based approaches that account for ancestral genetic structure, complex haplotypes, gene-gene interactions, and rare variants to detect and replicate novel pharmacogenetic loci.
Collapse
Affiliation(s)
- Victor E Ortega
- Center for Genomics and Personalized Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Deborah A Meyers
- Center for Genomics and Personalized Medicine, Wake Forest School of Medicine, Winston-Salem, NC.
| |
Collapse
|
24
|
Pharmacogenetics and the development of personalized approaches for combination therapy in asthma. Curr Allergy Asthma Rep 2014; 13:443-52. [PMID: 23912588 DOI: 10.1007/s11882-013-0372-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Asthma is a common, chronic disease of the airways that is treated with a combination of different therapies. The combination of LABA and ICS therapy results in a synergistic interaction that is efficacious in improving asthma symptom control; however, genetic variation has the potential to alter therapeutic efficacy. Both agents mediate complex molecular pathways consisting of gene variation that has been investigated with the analysis of candidate genes in the β2-adrenergic receptor and glucocorticoid pathway. These pharmacogenetic studies have been limited to retrospective analyses of clinical trial cohorts and a small number of prospective, genotype-stratified trials. More recently, genome-wide association studies in combination with replication in additional cohorts and in vitro cell-based models have been used to identify novel pathway-related pharmacogenetic variations. This review of the pharmacogenetics of the β2-adrenergic receptor and glucocorticoid pathways highlights the genotypic effects of variation in multiple genes from interacting pathways which may contribute to differential responses to inhaled beta agonists and glucocorticoids. As our understanding of these genetic mechanisms improves, panels of biomarkers may be developed to determine which combination therapies are the most effective with the least risk to an individual asthma patient. Before we can usher in an era of personalized medicine for asthma, it is first important to improve our ability to analyze large volumes of genetic data in large clinical trial cohorts using a combination of study designs, analytical methods, and in vitro functional studies.
Collapse
|
25
|
Blonder JP, Mutka SC, Sun X, Qiu J, Green LH, Mehra NK, Boyanapalli R, Suniga M, Look K, Delany C, Richards JP, Looker D, Scoggin C, Rosenthal GJ. Pharmacologic inhibition of S-nitrosoglutathione reductase protects against experimental asthma in BALB/c mice through attenuation of both bronchoconstriction and inflammation. BMC Pulm Med 2014; 14:3. [PMID: 24405692 PMCID: PMC3893392 DOI: 10.1186/1471-2466-14-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 01/03/2014] [Indexed: 12/16/2022] Open
Abstract
Background S-nitrosoglutathione (GSNO) serves as a reservoir for nitric oxide (NO) and thus is a key homeostatic regulator of airway smooth muscle tone and inflammation. Decreased levels of GSNO in the lungs of asthmatics have been attributed to increased GSNO catabolism via GSNO reductase (GSNOR) leading to loss of GSNO- and NO- mediated bronchodilatory and anti-inflammatory actions. GSNOR inhibition with the novel small molecule, N6022, was explored as a therapeutic approach in an experimental model of asthma. Methods Female BALB/c mice were sensitized and subsequently challenged with ovalbumin (OVA). Efficacy was determined by measuring both airway hyper-responsiveness (AHR) upon methacholine (MCh) challenge using whole body plethysmography and pulmonary eosinophilia by quantifying the numbers of these cells in the bronchoalveolar lavage fluid (BALF). Several other potential biomarkers of GSNOR inhibition were measured including levels of nitrite, cyclic guanosine monophosphate (cGMP), and inflammatory cytokines, as well as DNA binding activity of nuclear factor kappa B (NFκB). The dose response, onset of action, and duration of action of a single intravenous dose of N6022 given from 30 min to 48 h prior to MCh challenge were determined and compared to effects in mice not sensitized to OVA. The direct effect of N6022 on airway smooth muscle tone also was assessed in isolated rat tracheal rings. Results N6022 attenuated AHR (ED50 of 0.015 ± 0.002 mg/kg; Mean ± SEM) and eosinophilia. Effects were observed from 30 min to 48 h after treatment and were comparable to those achieved with three inhaled doses of ipratropium plus albuterol used as the positive control. N6022 increased BALF nitrite and plasma cGMP, while restoring BALF and plasma inflammatory markers toward baseline values. N6022 treatment also attenuated the OVA-induced increase in NFκB activation. In rat tracheal rings, N6022 decreased contractile responses to MCh. Conclusions The significant bronchodilatory and anti-inflammatory actions of N6022 in the airways are consistent with restoration of GSNO levels through GSNOR inhibition. GSNOR inhibition may offer a therapeutic approach for the treatment of asthma and other inflammatory lung diseases. N6022 is currently being evaluated in clinical trials for the treatment of inflammatory lung disease.
Collapse
Affiliation(s)
- Joan P Blonder
- N30 Pharmaceuticals, Inc, 3122 Sterling Circle, Suite 200, Boulder, CO 80301, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Carmichael SL, Witte JS, Ma C, Lammer EJ, Shaw GM. Hypospadias and variants in genes related to sex hormone biosynthesis and metabolism. Andrology 2013; 2:130-7. [PMID: 24281767 DOI: 10.1111/j.2047-2927.2013.00165.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/24/2013] [Accepted: 10/29/2013] [Indexed: 11/29/2022]
Abstract
We examined whether variants in genes related to sex hormone biosynthesis and metabolism were associated with hypospadias in humans. We examined 332 relatively common tag single-nucleotide polymorphisms (tagSNPs) in 20 genes. Analyses included 633 cases (84 mild, 322 moderate, 212 severe and 15 undetermined severity) and 855 population-based non-malformed male controls born in California from 1990 to 2003. We used logistic regression models to estimate odds ratios (OR) and 95% confidence intervals (CI) for each SNP. Several of the 332 studied SNPs had p < 0.01: one in CYP3A4, four in HSD17B3, one in HSD3B1, two in STARD3, 10 in SRD5A2 and seven in STS. In addition, haplotype analyses gave several associations with p < 0.01. For HSD17B3, 14-SNP and 5-SNP blocks had ORs of 1.5 (95% CI 1.1, 2.0, p < 0.001) and 2.8 (95% CI 1.6, 4.8, p < 0.001) respectively. For SRD5A2, 9-SNP, 3-SNP and 8-SNP blocks had ORs of 1.7 (95% CI 1.3, 2.2, p < 0.001), 1.4 (95% CI 1.1, 1.8, p = 0.008) and 1.5 (95% CI 1.2, 1.9, p = 0.002) respectively. Our study indicates that several genes that contribute to sex hormone biosynthesis and metabolism are associated with hypospadias risk.
Collapse
Affiliation(s)
- S L Carmichael
- Division of Neonatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | |
Collapse
|
27
|
Increased susceptibility to Klebsiella pneumonia and mortality in GSNOR-deficient mice. Biochem Biophys Res Commun 2013; 442:122-6. [PMID: 24239886 DOI: 10.1016/j.bbrc.2013.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/06/2013] [Indexed: 12/16/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR) is a key denitrosylase and critically important for protecting immune and other cells from nitrosative stress. Pharmacological inhibition of GSNOR is being actively pursued as a therapeutic approach to increase S-nitrosoglutathione levels for the treatment of asthma and cystic fibrosis. In the present study, we employed GSNOR-deficient (GSNOR(-/-)) mice to investigate whether inactivation of GSNOR may increase susceptibility to pulmonary infection by Klebsiella pneumoniae, a common cause of nosocomial pneumonia. We found that compared to wild-type mice, bacterial colony forming units 48 h after intranasal infection with K. pneumoniae were increased over 4-folds in lung and spleen and strikingly, over a 1000-folds in blood of GSNOR(-/-) mice. Lung injury was comparable between infected wild-type and GSNOR(-/-) mice, but inflammation and injury was significantly elevated in spleen of GSNOR(-/-) mice. Whereas all wild-type mice survived 48 h after infection, 10 of 23 GSNOR(-/-) mice died. Thus, GSNOR appears to play a crucial role in controlling pulmonary and systemic infection by K. pneumoniae. Our results suggest that patients treated in clinical trials with inhibitors of GSNOR should be carefully monitored for signs of infection.
Collapse
|
28
|
Carmichael SL, Ma C, Choudhry S, Lammer EJ, Witte JS, Shaw GM. Hypospadias and genes related to genital tubercle and early urethral development. J Urol 2013; 190:1884-92. [PMID: 23727413 PMCID: PMC4103581 DOI: 10.1016/j.juro.2013.05.061] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2013] [Indexed: 11/19/2022]
Abstract
PURPOSE We determined whether variants in genes associated with genital tubercle (the anlage for the penis) and early urethral development were associated with hypospadias in humans. MATERIALS AND METHODS We examined 293 relatively common tag single nucleotide polymorphisms in BMP4, BMP7, FGF8, FGF10, FGFR2, HOXA13, HOXD13, HOXA4, HOXB6, SRY, WT1, WTAP, SHH, GLI1, GLI2 and GLI3. The analysis included 624 cases (81 mild, 319 moderate, 209 severe, 15 undetermined severity) and 844 population based nonmalformed male controls born in California from 1990 to 2003. RESULTS There were 28 single nucleotide polymorphisms for which any of the comparisons (ie overall or for a specific severity) had a p value of less than 0.01. The homozygous variant genotypes for 4 single nucleotide polymorphisms in BMP7 were associated with at least a twofold increased risk of hypospadias regardless of severity. Five single nucleotide polymorphisms for FGF10 were associated with threefold to fourfold increased risks, regardless of severity. For 4 of them the results were restricted to whites. For GLI1, GLI2 and GLI3 there were 12 associated single nucleotide polymorphisms but results were inconsistent by severity and race/ethnicity. For SHH 1 single nucleotide polymorphism was associated with a 2.4-fold increased risk of moderate hypospadias. For WT1 6 single nucleotide polymorphisms were associated with approximately a twofold increased risk, primarily for severe hypospadias. CONCLUSIONS This study provides evidence that single nucleotide polymorphisms in several genes that contribute to genital tubercle and early urethral development are associated with hypospadias risk.
Collapse
Affiliation(s)
- Suzan L Carmichael
- Department of Pediatrics, Division of Neonatology, Stanford University School of Medicine, Stanford, California.
| | | | | | | | | | | |
Collapse
|
29
|
Tse SM, Gold DR, Sordillo JE, Hoffman EB, Gillman MW, Rifas-Shiman SL, Fuhlbrigge AL, Tantisira KG, Weiss ST, Litonjua AA. Diagnostic accuracy of the bronchodilator response in children. J Allergy Clin Immunol 2013; 132:554-559.e5. [PMID: 23683464 PMCID: PMC3759549 DOI: 10.1016/j.jaci.2013.03.031] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/09/2013] [Accepted: 03/20/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND The bronchodilator response (BDR) reflects the reversibility of airflow obstruction and is recommended as an adjunctive test to diagnose asthma. The validity of the commonly used definition of BDR, a 12% or greater change in FEV1 from baseline, has been questioned in childhood. OBJECTIVES We sought to examine the diagnostic accuracy of the BDR test by using 3 large pediatric cohorts. METHODS Cases include 1041 children with mild-to-moderate asthma from the Childhood Asthma Management Program. Control subjects (nonasthmatic and nonwheezing) were chosen from Project Viva and Home Allergens, 2 population-based pediatric cohorts. Receiver operating characteristic curves were constructed, and areas under the curve were calculated for different BDR cutoffs. RESULTS A total of 1041 cases (59.7% male; mean age, 8.9 ± 2.1 years) and 250 control subjects (46.8% male; mean age, 8.7 ± 1.7 years) were analyzed, with mean BDRs of 10.7% ± 10.2% and 2.7% ± 8.4%, respectively. The BDR test differentiated asthmatic patients from nonasthmatic patients with a moderate accuracy (area under the curve, 73.3%). Despite good specificity, a cutoff of 12% was associated with poor sensitivity (35.6%). A cutoff of less than 8% performed significantly better than a cutoff of 12% (P = .03, 8% vs 12%). CONCLUSIONS Our findings highlight the poor sensitivity associated with the commonly used 12% cutoff for BDR. Although our data show that a threshold of less than 8% performs better than 12%, given the variability of this test in children, we conclude that it might be not be appropriate to choose a specific BDR cutoff as a criterion for the diagnosis of asthma.
Collapse
Affiliation(s)
- Sze Man Tse
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Massachusetts General Hospital, Department of Pediatrics, Division of Pediatric Pulmonary Medicine, Boston, Mass.
| | - Diane R Gold
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Joanne E Sordillo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Elaine B Hoffman
- Harvard School of Public Health, Department of Biostatistics, Boston, Mass
| | - Matthew W Gillman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Mass
| | - Sheryl L Rifas-Shiman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Mass
| | - Anne L Fuhlbrigge
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Kelan G Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Augusto A Litonjua
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| |
Collapse
|
30
|
Dahlin A, Tantisira KG. Integrative systems biology approaches in asthma pharmacogenomics. Pharmacogenomics 2013; 13:1387-404. [PMID: 22966888 DOI: 10.2217/pgs.12.126] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In order to improve therapeutic outcomes, there is a tremendous need to identify patients who are likely to respond to a given asthma treatment. Pharmacogenomic studies have explained a portion of the variability in drug response and provided an increasing list of candidate genes and SNPs. However, as phenotypic variation arises from a network of complex interactions among genetic and environmental factors, rather than individual genes or SNPs, a multidisciplinary, systems-level approach is required in order to understand the inter-relationships among these factors. Systems biology, which seeks to capture interactions between genetic factors and other variables, offers a promising approach to improved therapeutic outcomes in asthma. This aritcle will review and update progress in the pharmacogenomics of asthma and then discuss the application of systems biology approaches to asthma pharmacogenomics.
Collapse
Affiliation(s)
- Amber Dahlin
- Channing Laboratory, Brigham & Women's Hospital & Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | | |
Collapse
|
31
|
Sengupta R, Holmgren A. Thioredoxin and thioredoxin reductase in relation to reversible S-nitrosylation. Antioxid Redox Signal 2013; 18:259-69. [PMID: 22702224 DOI: 10.1089/ars.2012.4716] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) regulates a diverse range of cellular processes, including vasodilation, neurotransmission, and antimicrobial and anti-tumor activities. S-nitrosylation with the formation of S-nitrosothiols (RSNOs) is an important feature of NO signaling regulating protein function. In mammalian cells, glutathione (GSH), S-nitrosoglutathione reductase (GSNOR), and thioredoxin (Trx) have been identified as the major protein denitrosylases. RECENT ADVANCES Human cytosolic/nuclear Trx1 in the disulfide form can be nitrosylated at Cys73 and transnitrosylate target proteins, including caspase 3. Thus, similar to GSH, which by forming S-nitrosoglutathione (GSNO) can transnitrosylate proteins, Trx can either denitrosylate or nitrosylate proteins depending on its oxidation state. CRITICAL ISSUES In this review, we discuss the regulation of cellular processes by reversible S-nitrosylation and Trx-mediated cellular homeostasis of RSNOs and S-nitrosoproteins. FUTURE DIRECTIONS Functions of RSNOs in vivo and their pharmacological uses have not yet been fully studied. Further investigations on the role of Trx systems in relation to biologically relevant RSNOs, their functions, and the mechanisms of denitrosylation will facilitate the development of drugs and therapies. Antioxid. Redox Signal. 18, 259-269.
Collapse
Affiliation(s)
- Rajib Sengupta
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
32
|
Szefler SJ, Chinchilli VM, Israel E, Denlinger LC, Lemanske RF, Calhoun W, Peters SP. Key observations from the NHLBI Asthma Clinical Research Network. Thorax 2012; 67:450-5. [PMID: 22514237 DOI: 10.1136/thoraxjnl-2012-201876] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The National Heart, Lung and Blood Institute (NHLBI) Asthma Clinical Research Network (ACRN) recently completed its work after 20 years of collaboration as a multicentre clinical trial network. When formed, its stated mission was to perform multiple controlled clinical trials for treating patients with asthma by dispassionately examining new and existing therapies, and to rapidly communicate its findings to the medical community. The ACRN conducted 15 major clinical trials. In addition, clinical data, manual of operations, protocols and template informed consents from all ACRN trials are available via NHLBI BioLINCC (https://biolincc.nhlbi.nih.gov/studies/). This network contributed major insights into the use of inhaled corticosteroids, short-acting and long-acting ß-adrenergic agonists, leukotriene receptor antagonists, and novel agents (tiotropium, colchicine and macrolide antibiotics). They also pioneered studies of the variability in drug response, predictors of treatment response and pharmacogenetics. This review highlights the major research observations from the ACRN that have impacted the current management of asthma.
Collapse
Affiliation(s)
- Stanley J Szefler
- Department of Pediatrics, National Jewish Health and University of Colorado Denver, School of Medicine, Denver, Colorado 80206, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Lane HY, Tsai GE, Lin E. Assessing Gene-Gene Interactions in Pharmacogenomics. Mol Diagn Ther 2012; 16:15-27. [DOI: 10.1007/bf03256426] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
34
|
Portelli M, Sayers I. Genetic basis for personalized medicine in asthma. Expert Rev Respir Med 2012; 6:223-36. [PMID: 22455494 DOI: 10.1586/ers.12.9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There is heterogeneity in patient responses to current asthma medications. Significant progress has been made identifying genetic polymorphisms that influence the efficacy and potential for adverse effects to asthma drugs, including; β(2)-adrenergic receptor agonists, corticosteroids and leukotriene modifiers. Pharmacogenetics holds great promise to maximise clinical outcomes and minimize adverse effects. Asthma is heterogeneous with respect to clinical presentation and inflammatory mechanisms underlying the disease, which is likely to contribute to variable results in clinical trials targeting specific inflammatory mediators. Genome-wide association studies have begun to identify genes underlying asthma (e.g., IL1RL1), which represent future therapeutic targets. In this article, we review and update the pharmacogenetics of current asthma therapies and discuss the genetics underlying selected Phase II and future targets.
Collapse
Affiliation(s)
- Michael Portelli
- Division of Therapeutics and Molecular Medicine, Nottingham Respiratory Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
35
|
Fitzpatrick AM, Jones DP, Brown LAS. Glutathione redox control of asthma: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2012; 17:375-408. [PMID: 22304503 PMCID: PMC3353819 DOI: 10.1089/ars.2011.4198] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/22/2012] [Accepted: 01/22/2012] [Indexed: 12/11/2022]
Abstract
Asthma is a chronic inflammatory disorder of the airways associated with airway hyper-responsiveness and airflow limitation in response to specific triggers. Whereas inflammation is important for tissue regeneration and wound healing, the profound and sustained inflammatory response associated with asthma may result in airway remodeling that involves smooth muscle hypertrophy, epithelial goblet-cell hyperplasia, and permanent deposition of airway extracellular matrix proteins. Although the specific mechanisms responsible for asthma are still being unraveled, free radicals such as reactive oxygen species and reactive nitrogen species are important mediators of airway tissue damage that are increased in subjects with asthma. There is also a growing body of literature implicating disturbances in oxidation/reduction (redox) reactions and impaired antioxidant defenses as a risk factor for asthma development and asthma severity. Ultimately, these redox-related perturbations result in a vicious cycle of airway inflammation and injury that is not always amenable to current asthma therapy, particularly in cases of severe asthma. This review will discuss disruptions of redox signaling and control in asthma with a focus on the thiol, glutathione, and reduced (thiol) form (GSH). First, GSH synthesis, GSH distribution, and GSH function and homeostasis are discussed. We then review the literature related to GSH redox balance in health and asthma, with an emphasis on human studies. Finally, therapeutic opportunities to restore the GSH redox balance in subjects with asthma are discussed.
Collapse
Affiliation(s)
- Anne M Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta, Georgia 30322, USA.
| | | | | |
Collapse
|
36
|
van der Baan FH, Knol MJ, Maitland-van der Zee AH, Regieli JJ, van Iperen EPA, Egberts ACG, Klungel OH, Grobbee DE, Jukema JW. Added value of pharmacogenetic testing in predicting statin response: results from the REGRESS trial. THE PHARMACOGENOMICS JOURNAL 2012; 13:318-24. [DOI: 10.1038/tpj.2012.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/24/2012] [Accepted: 04/02/2012] [Indexed: 12/16/2022]
|
37
|
Weiss ST. New approaches to personalized medicine for asthma: where are we? J Allergy Clin Immunol 2012; 129:327-34. [PMID: 22284929 DOI: 10.1016/j.jaci.2011.12.971] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 12/19/2011] [Indexed: 12/24/2022]
Abstract
Access to an electronic medical record is essential for personalized medicine. Currently, only 40% of US physicians have such access, but this is rapidly changing. It is expected that 100,000 Americans will have their whole genome sequenced in 2012. The cost of such sequencing is rapidly dropping, and is estimated to be $1000 by 2013. These technological advances will make interpretation of whole genome sequence data a major clinical challenge for the foreseeable future. At present, a relatively small number of genes have been identified to determine drug treatment response phenotypes for asthma. It is anticipated that this will dramatically increase over the next 10 years as personalized medicine becomes more of a reality for asthma patients.
Collapse
Affiliation(s)
- Scott T Weiss
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Boston, Mass 02115, USA.
| |
Collapse
|
38
|
Foster MW, Yang Z, Gooden DM, Thompson JW, Ball CH, Turner ME, Hou Y, Pi J, Moseley MA, Que LG. Proteomic characterization of the cellular response to nitrosative stress mediated by s-nitrosoglutathione reductase inhibition. J Proteome Res 2012; 11:2480-91. [PMID: 22390303 DOI: 10.1021/pr201180m] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The S-nitrosoglutathione-metabolizing enzyme, GSNO reductase (GSNOR), has emerged as an important regulator of protein S-nitrosylation. GSNOR ablation is protective in models of asthma and heart failure, raising the idea that GSNOR inhibitors might hold therapeutic value. Here, we investigated the effects of a small molecule inhibitor of GSNOR (GSNORi) in mouse RAW 264.7 macrophages. We found that GSNORi increased protein S-nitrosylation in cytokine-stimulated cells, and we utilized stable isotope labeling of amino acids in cell culture (SILAC) to quantify the cellular response to this "nitrosative stress". The expression of several cytokine-inducible immunomodulators, including osteopontin, cyclooxygenase-2, and nitric oxide synthase isoform 2 (NOS2), were decreased by GSNORi. In addition, selective targets of the redox-regulated transcription factor, nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-including heme oxygenase 1 (HO-1) and glutamate cysteine ligase modulatory subunit-were induced by GSNORi in a NOS2- and Nrf2-dependent manner. In cytokine-stimulated cells, Nrf2 protected from GSNORi-induced glutathione depletion and cytotoxicity and HO-1 activity was required for down-regulation of NOS2. Interestingly, GSNORi also affected a marked increase in NOS2 protein stability. Collectively, these data provide the most complete description of the global effects of GSNOR inhibition and demonstrate several important mechanisms for inducible response to GSNORi-mediated nitrosative stress.
Collapse
Affiliation(s)
- Matthew W Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Small Molecule Synthesis Facility and Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina 27710, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Identifying that health inequalities exist is not enough; nor does the knowledge that a patient has a high-risk genotype or comes from a higher risk socioeconomic background does not, by itself, help the patient. To thoroughly examine the origins of health disparities, a broad view of environmental and molecular influences must be included. As these factors are identified, it is important to focus on interventions that can change outcomes for patients. Tools for education, community involvement, literacy, and environmental safety need to be developed, tested and disseminated. The basic science of health disparities must move forward in a coordinated fashion by structuring research that is an integrated effort between basic sciences, clinical medicine and include all traditionally underserved communities. Only through these collaborations can we hope to eliminate health inequalities in the future.
Collapse
Affiliation(s)
- Kimberly Danieli Watts
- Children's Memorial Hospital, Northwestern University Feinberg School of Medicine, 2300 Children's Plaza Box # 43, Chicago, IL 60614, USA.
| |
Collapse
|
40
|
Perinatal gene-gene and gene-environment interactions on IgE production and asthma development. Clin Dev Immunol 2012; 2012:270869. [PMID: 22481967 PMCID: PMC3299317 DOI: 10.1155/2012/270869] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 11/01/2011] [Accepted: 11/17/2011] [Indexed: 12/23/2022]
Abstract
Atopic asthma is a complex disease associated with IgE-mediated immune reactions. Numerous genome-wide studies identified more than 100 genes in 22 chromosomes associated with atopic asthma, and different genetic backgrounds in different environments could modulate susceptibility to atopic asthma. Current knowledge emphasizes the effect of tobacco smoke on the development of childhood asthma. This suggests that asthma, although heritable, is significantly affected by gene-gene and gene-environment interactions. Evidence has recently shown that molecular mechanism of a complex disease may be limited to not only DNA sequence differences, but also gene-environmental interactions for epigenetic difference. This paper reviews and summarizes how gene-gene and gene-environment interactions affect IgE production and the development of atopic asthma in prenatal and childhood stages. Based on the mechanisms responsible for perinatal gene-environment interactions on IgE production and development of asthma, we formulate several potential strategies to prevent the development of asthma in the perinatal stage.
Collapse
|
41
|
Enzymatic mechanisms regulating protein S-nitrosylation: implications in health and disease. J Mol Med (Berl) 2012; 90:233-44. [PMID: 22361849 DOI: 10.1007/s00109-012-0878-z] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/02/2012] [Accepted: 02/06/2012] [Indexed: 10/28/2022]
Abstract
Nitric oxide participates in cellular signal transduction largely through S-nitrosylation of allosteric and active-site cysteine thiols within proteins, forming S-nitroso-proteins (SNO-proteins). S-nitrosylation of proteins has been demonstrated to affect a broad range of functional parameters including enzymatic activity, subcellular localization, protein-protein interactions, and protein stability. Analogous to other ubiquitous posttranslational modifications that are regulated enzymatically, including phosphorylation and ubiquitinylation, accumulating evidence suggests the existence of enzymatic mechanisms for regulating protein S-nitrosylation. In particular, studies have led to the identification of multiple enzymes (nitrosylases and denitrosylases) that participate in targeted S-nitrosylation or denitrosylation of proteins in physiological settings. Nitrosylases are best characterized in the context of transnitrosylation in which a SNO-protein transfers an NO group to an acceptor protein (Cys-to-Cys transfer), but examples of transnitrosylation catalyzed by metalloproteins (Metal-to-Cys transfer) also exist. By contrast, denitrosylases remove the NO group from SNO-proteins, ultimately using reducing equivalents derived from NADH or NADPH. Here, we focus on the recent discoveries of nitrosylases and denitrosylases and the notion that their aberrant activities may play roles in health and disease.
Collapse
|
42
|
Lane HY, Tsai GE, Lin E. Assessing gene-gene interactions in pharmacogenomics. Mol Diagn Ther 2012; 16:15-27. [PMID: 22352452 DOI: 10.2165/11597270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
In pharmacogenomics studies, gene-gene interactions play an important role in characterizing a trait that involves complex pharmacokinetic and pharmacodynamic mechanisms, particularly when each involved feature only demonstrates a minor effect. In addition to the candidate gene approach, genome-wide association studies (GWAS) are widely utilized to identify common variants that are associated with treatment response. In the wake of recent advances in scientific research, a paradigm shift from GWAS to whole-genome sequencing is expected, because of the reduced cost and the increased throughput of next-generation sequencing technologies. This review first outlines several promising methods for addressing gene-gene interactions in pharmacogenomics studies. We then summarize some candidate gene studies for various treatments with consideration of gene-gene interactions. Furthermore, we give a brief overview for the pharmacogenomics studies with the GWAS approach and describe the limitations of these GWAS in terms of gene-gene interactions. Future research in translational medicine promises to lead to mechanistic findings related to drug responsiveness in light of complex gene-gene interactions and will probably make major contributions to individualized medicine and therapeutic decision-making.
Collapse
Affiliation(s)
- Hsien-Yuan Lane
- Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan
| | | | | |
Collapse
|
43
|
Tse SM, Tantisira K, Weiss ST. The pharmacogenetics and pharmacogenomics of asthma therapy. THE PHARMACOGENOMICS JOURNAL 2011; 11:383-92. [PMID: 21987090 DOI: 10.1038/tpj.2011.46] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the availability of several classes of asthma medications and their overall effectiveness, a significant portion of patients fail to respond to these therapeutic agents. Evidence suggests that genetic factors may partly mediate the heterogeneity in asthma treatment response. This review discusses important findings in asthma pharmacogenetic and pharmacogenomic studies conducted to date, examines limitations of these studies and, finally, proposes future research directions in this field. The focus will be on the three major classes of asthma medications: β-adrenergic receptor agonists, inhaled corticosteroids and leukotriene modifiers. Although many studies are limited by small sample sizes and replication of the findings is needed, several candidate genes have been identified. High-throughput technologies are also allowing for large-scale genetic investigations. Thus, the future is promising for a personalized treatment of asthma, which will improve therapeutic outcomes, minimize side effects and lead to a more cost-effective care.
Collapse
Affiliation(s)
- S M Tse
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
44
|
Gaston B. The biochemistry of asthma. Biochim Biophys Acta Gen Subj 2011; 1810:1017-24. [PMID: 21718756 DOI: 10.1016/j.bbagen.2011.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 05/18/2011] [Accepted: 06/15/2011] [Indexed: 01/27/2023]
Abstract
BACKGROUND Asthma is not one disease. Different patients have biochemically distinct phenotypes. SCOPE OF REVIEW Biomarker analysis was developed to identify inflammation in the asthmatic airway. It has led to a renewed interest in biochemical abnormalities in the asthmatic airway. The biochemical determinants of asthma heterogeneity are many. Examples include decreased activity of superoxide dismutases; increased activity of eosinophil peroxidase, S-nitrosoglutathione reductase, and arginases; decreased airway pH; and increased levels of asymmetric dimethyl arginine. MAJOR CONCLUSIONS New discoveries suggest that biomarkers such as exhaled nitric oxide reflect complex airway biochemistry. This biochemistry can be informative and therapeutically relevant. GENERAL SIGNIFICANCE Improved understanding of airway biochemistry will lead to new tests to identify biochemically unique subpopulations of patients with asthma. It will also likely lead to new, targeted treatments for these specific asthma subpopulations. This article is part of a Special Issue entitled Biochemistry of Asthma.
Collapse
Affiliation(s)
- Benjamin Gaston
- Universit of Virginia School of Medicine, Pediatric Respiratory Medicine, Charlottes, VA 22908-0386, USA.
| |
Collapse
|
45
|
Hwang R, Souza RP, Tiwari AK, Zai CC, Müller DJ, Potkin SG, Lieberman JA, Meltze HY, Kennedy JL. Gene-gene interaction analyses between NMDA receptor subunit and dopamine receptor gene variants and clozapine response. Pharmacogenomics 2011; 12:277-91. [PMID: 21332319 DOI: 10.2217/pgs.10.182] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS To investigate the possible association and gene-gene interaction effects of polymorphisms in NMDA receptor subunit (GRIN1, GRIN2A and GRIN2B) and dopamine receptor (DRD1, DRD2 and DRD3) genes with clozapine response. MATERIALS & METHODS GRIN1 rs11146020 (G1001C), GRIN2A GT-repeat and GRIN2B rs10193895 (G-200T) polymorphisms were tested for association in a Caucasian (n = 183) and an African-American (n = 49) sample using χ(2) and ANOVA tests. Logistic regression and two-way ANOVA were used to explore gene-gene interaction effects with dopamine receptor gene variants. RESULTS & CONCLUSION This study does not support the involvement of the NMDA receptor subunit gene polymorphisms in clozapine response. All tests for an association were negative. Gene-gene interaction analyses however yielded promising leads, including an observed effect between DRD1 rs686 and DRD3 Ser9Gly polymorphisms on clozapine response (p = 0.002).
Collapse
Affiliation(s)
- Rudi Hwang
- Neurogenetics Section, Centre for Addiction & Mental Health, 250 College Street, Toronto, Ontario, 5T1R8, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Moore PE. Influence of gene-gene interactions on response to albuterol therapy. Pharmacogenomics 2011; 12:1-3. [PMID: 21174616 DOI: 10.2217/pgs.10.192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
47
|
|
48
|
Thompson CM, Grafström RC. Considerations for the Implausibility of Leukemia Induction by Formaldehyde. Toxicol Sci 2010; 120:230-2; author reply 233. [DOI: 10.1093/toxsci/kfq340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
49
|
Kevil CG, Patel RP. S-Nitrosothiol biology and therapeutic potential in metabolic disease. CURRENT OPINION IN INVESTIGATIONAL DRUGS (LONDON, ENGLAND : 2000) 2010; 11:1127-1134. [PMID: 20872315 PMCID: PMC3677214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
S-Nitrosothiols (RSNOs) have been used widely as experimental nitric oxide (NO) donors, but the clinical use of these agents remains limited. Recent data support a role for endogenous RSNOs as mediators of NO signaling via the post-translational modification of proteins. This review discusses the increased understanding of the role of RSNOs in NO signaling, as well as emerging insights into NO donor-dependent and -independent mechanisms of action of RSNOs, in the context of emerging and potential therapeutics that target endogenous RSNOs or use synthetic RSNOs to stimulate NO signaling. The focus of this review is the treatment of diabetes and metabolic disease, pathologies in which dysfunction in NO signaling is clearly implicated.
Collapse
Affiliation(s)
- Christopher G Kevil
- Departments of Pathology and Molecular and Cellular Physiology, 1501 Kings Hwy, Louisiana State University Health Sciences Center, Shreveport, LA 71130
| | - Rakesh P Patel
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham
| |
Collapse
|