1
|
Wang H, Liu J, Gao J, Yan W, Rehan VK. Perinatal Exposure to Nicotine Alters Sperm RNA Profiles in Rats. Front Endocrinol (Lausanne) 2022; 13:893863. [PMID: 35600600 PMCID: PMC9114732 DOI: 10.3389/fendo.2022.893863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/07/2022] [Indexed: 01/31/2023] Open
Abstract
Perinatal exposure to smoking has been associated with childhood asthma, one of the most common pediatric conditions affecting millions of children globally. Of great interest, this disease phenotype appears heritable as it can persist across multiple generations even in the absence of persistent exposure to smoking in subsequent generations. Although the molecular mechanisms underlying childhood asthma induced by perinatal exposure to smoking or nicotine remain elusive, an epigenetic mechanism has been proposed, which is supported by the data from our earlier analyses on germline DNA methylation (5mC) and histone marks (H3 and H4 acetylation). To further investigate the potential epigenetic inheritance of childhood asthma induced by perinatal nicotine exposure, we profiled both large and small RNAs in the sperm of F1 male rats. Our data revealed that perinatal exposure to nicotine leads to alterations in the profiles of sperm-borne RNAs, including mRNAs and small RNAs, and that rosiglitazone, a PPARγ agonist, can attenuate the effect of nicotine and reverse the sperm-borne RNA profiles of F1 male rats to close to placebo control levels.
Collapse
Affiliation(s)
- Hetan Wang
- Department of Medical Genetics, China Medical University, Shenyang, China
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jie Liu
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jianjun Gao
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Wei Yan, ; Virender K. Rehan,
| | - Virender K. Rehan
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Wei Yan, ; Virender K. Rehan,
| |
Collapse
|
2
|
Liu H, Huang Q, Lv Y, Dong Y, Song D. CEBPB knockdown sensitizes nasopharyngeal carcinoma cells to cisplatin by promoting the expression of serine protease inhibitor Kazal-type 5. Anticancer Drugs 2022; 33:e327-e335. [PMID: 34387602 DOI: 10.1097/cad.0000000000001208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Serine protease inhibitor Kazal-type 5 (SPINK5) has been indicated to act as a prognostic predictor for patients with head and neck squamous cell carcinoma. However, its specific role in nasopharyngeal carcinoma (NPC), a malignancy that has a high propensity for chemoresistance, remains largely obscure. We, thus, sought to investigate the importance of SPINK5 expression in regulating chemoresistance in NPC. Differentially expressed genes in NPC were screened using the cancer genome atlas-head and neck squamous cell carcinoma database and microarray analysis. SPINK5 was downregulated in NPC tissues and cells. After SPINK5 upregulation, the cells treated with cisplatin showed reduced cell survival and the ability to migrate, invade and metastasize. Mechanistically, the transcription factors regulating SPINK5 were queried through the JASPAR website, followed by dual-luciferase and Chromatin immunoprecipitation assay validation. CCAAT enhancer-binding protein (CEBP) beta (CEBPB) bound to the SPINK5 promoter region in NPC cells. The silencing of CEBPB enhanced the expression of SPINK5. CEBPB overexpression reversed the inhibitory effects of cisplatin on NPC cell malignant phenotype in the presence of SPINK5 overexpression. In conclusion, CEBPB silencing promoted chemoresistance of NPC cells via activating SPINK5, signifying that targeting CEBPB was a new approach to enhance the chemotherapy efficacy in NPC.
Collapse
Affiliation(s)
- Hong Liu
- Department of Oncology, Binzhou Central Hospital, Binzhou
| | - Qingli Huang
- Department of Otolaryngology, Liaocheng People's Hospital
| | - Yunxia Lv
- Department of Otolaryngology, Yanggu People's Hospital, Liaocheng
| | - Youwei Dong
- Department of Otolaryngology, Juxian people's Hospital, Rizhao
| | - Daoliang Song
- Department of Otorhinolargology Head and Neck Surgery, Zibo Central Hospital, Zibo, Shandong, People's Republic of China
| |
Collapse
|
3
|
Luger T, Amagai M, Dreno B, Dagnelie MA, Liao W, Kabashima K, Schikowski T, Proksch E, Elias PM, Simon M, Simpson E, Grinich E, Schmuth M. Atopic dermatitis: Role of the skin barrier, environment, microbiome, and therapeutic agents. J Dermatol Sci 2021; 102:142-157. [PMID: 34116898 DOI: 10.1016/j.jdermsci.2021.04.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
Atopic dermatitis (AD) is a chronic, inflammatory skin disorder characterized by eczematous and pruritic skin lesions. In recent decades, the prevalence of AD has increased worldwide, most notably in developing countries. The enormous progress in our understanding of the complex composition and functions of the epidermal barrier allows for a deeper appreciation of the active role that the skin barrier plays in the initiation and maintenance of skin inflammation. The epidermis forms a physical, chemical, immunological, neuro-sensory, and microbial barrier between the internal and external environment. Not only lesional, but also non-lesional areas of AD skin display many morphological, biochemical and functional differences compared with healthy skin. Supporting this notion, genetic defects affecting structural proteins of the skin barrier, including filaggrin, contribute to an increased risk of AD. There is evidence to suggest that natural environmental allergens and man-made pollutants are associated with an increased likelihood of developing AD. A compromised epidermal barrier predisposes the skin to increased permeability of these compounds. Numerous topical and systemic therapies for AD are currently available or in development; while anti-inflammatory therapy is central to the treatment of AD, some existing and novel therapies also appear to exert beneficial effects on skin barrier function. Further research on the skin barrier, particularly addressing epidermal differentiation and inflammation, lipid metabolism, and the role of bacterial communities for skin barrier function, will likely expand our understanding of the complex etiology of AD and lead to identification of novel targets and the development of new therapies.
Collapse
Affiliation(s)
- Thomas Luger
- Department of Dermatology, University of Münster, Münster, Germany.
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Brigitte Dreno
- Dermatology Department, Nantes University, CHU Nantes, CIC 1413, CRCINA, Nantes, France
| | - Marie-Ange Dagnelie
- Dermatology Department, Nantes University, CHU Nantes, CIC 1413, CRCINA, Nantes, France
| | - Wilson Liao
- Department of Dermatology, University of California, San Francisco, CA, United States
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tamara Schikowski
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | | | - Peter M Elias
- San Francisco VA Medical Center, University of California, San Francisco, CA, United States
| | - Michel Simon
- UDEAR, Inserm, University of Toulouse, U1056, Toulouse, France
| | - Eric Simpson
- Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
| | - Erin Grinich
- Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
| | - Matthias Schmuth
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Lv Z, Wu K, Qin X, Yuan J, Yan M, Zhang J, Wang L, Ji T, Cao W, Chen W. A Novel Tumor Suppressor SPINK5 Serves as an Independent Prognostic Predictor for Patients with Head and Neck Squamous Cell Carcinoma. Cancer Manag Res 2020; 12:4855-4869. [PMID: 32606974 PMCID: PMC7320891 DOI: 10.2147/cmar.s236266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 04/25/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In our previous study, serine protease inhibitor Kazal-type 5 (SPINK5), which encodes the product of serine protease inhibitor lymphoepithelial Kazal-type-related inhibitor (LEKTI) was found to be down-regulated in head and neck squamous cell carcinoma (HNSCC) using oligonucleotide microarrays. However, the function and clinical implications of SPINK5/LEKTI remain obscure in HNSCC. METHODS The endogenous expression level of SPINK5/LEKTI was further verified in 9 HNSCC cell lines and HNSCCs by means of reverse transcription-polymerase chain reaction, real-time PCR, Western blotting and immunohistochemistry. The biological function of SPINK5/LEKTI was investigated in vitro and in vivo experiments. Kaplan-Meier survival analysis and Cox proportional hazards regression model were used to determine the correlation between SPINK5/LEKTI expression and clinical outcome. RESULTS Down-regulation expression of SPINK5/LEKTI was found in six out of nine HNSCC cell lines and in 85.7% HNSCC specimens (P<0.0001). Upon silencing of SPINK5/LEKTI, the cell proliferation, plate colony formation and cell invasion of WU-HN6 cells were significantly increased, while exogenous overexpression of SPINK5/LEKTI, the proliferation, plate colony and invasion of WU-HN13 and HN30 cells were remarkably inhibited with the arrest of G1 cell cycle (P=0.0001, P=0.003, respectively). HNSCC patients with lower LEKTI levels had significantly inferior overall survival compared to those patients with higher LEKTI (P=0.0017) by Kaplan-Meier survival analysis. Univariate and multivariate Cox proportional hazards regression model analysis revealed that LEKTI expression was an independent prognostic predictor for HNSCC patients (HR=0.114, 95% CI:0.044-0.292, P<0.001). CONCLUSION Our results demonstrate that SPINK5/LEKTI might be a tumor suppressor in HNSCCs and serve as an independent prognostic predictor for HNSCC patients.
Collapse
Affiliation(s)
- Zhongjing Lv
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Department of Stomatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, People’s Republic of China
| | - Kun Wu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
| | - Xing Qin
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
| | - Jian Yuan
- Department of Stomatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, People’s Republic of China
| | - Ming Yan
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
| | - Lizhen Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Department of Oral Pathology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Tong Ji
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
| | - Wei Cao
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, People’s Republic of China
- Correspondence: Wantao Chen; Wei Cao Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China Email ;
| |
Collapse
|
5
|
Christou EAA, Giardino G, Stefanaki E, Ladomenou F. Asthma: An Undermined State of Immunodeficiency. Int Rev Immunol 2019; 38:70-78. [PMID: 30939053 DOI: 10.1080/08830185.2019.1588267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Asthma is a heterogeneous chronic respiratory disease characterized by an increased burden of infections. Respiratory tract infections associated with an increased risk for asthma especially when occurring in the first months of life, also represent the most common cause of asthma exacerbations. The association between asthma and the increased frequency of infections and microbiota dysbiosis might be explained by a common mechanism, such as an underlying immune system defect. Apart from the well-established association between primary immunodeficiencies and asthma, several alterations in the immune response following infection have also been observed in asthmatic patients. An impairment in lung epithelial barrier integrity exists and is associated with both an increased susceptibility to infections and the development of asthma. Asthmatic patients are also found to have a deficient interferon (IFN) response upon infection. Additionally, defects in Toll-like receptor (TLR) signaling are observed in asthma and are correlated with both recurrent infections and asthma development. In this review, we summarize the common pathophysiological background of asthma and infections, highlighting the importance of an underlying immune system defect that predispose individuals to recurrent infections and asthma.
Collapse
Affiliation(s)
| | - Giuliana Giardino
- b Department of Translational Medical Sciences , University of Naples Federico II , Naples , Italy
| | - Evangelia Stefanaki
- c Department of Pediatrics , Venizeleion General Hospital , Heraklion , Greece
| | - Fani Ladomenou
- c Department of Pediatrics , Venizeleion General Hospital , Heraklion , Greece
| |
Collapse
|
6
|
Tripathi P, Deng F, Scruggs AM, Chen Y, Huang SK. Variation in doses and duration of particulate matter exposure in bronchial epithelial cells results in upregulation of different genes associated with airway disorders. Toxicol In Vitro 2018; 51:95-105. [PMID: 29753051 PMCID: PMC6464127 DOI: 10.1016/j.tiv.2018.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/04/2018] [Accepted: 05/05/2018] [Indexed: 11/25/2022]
Abstract
Exposure to particulate matter < 2.5 μm (PM2.5) is associated with a variety of airway diseases. Although studies have demonstrated that high doses of PM2.5 cause cytotoxicity and changes to gene expression in bronchial epithelial cells, the effect of lower doses and repeated exposure to PM2.5 are less well studied. Here, we treated BEAS-2B cells with varying doses of PM2.5 for 1-7 days and examined the expression of a variety of genes implicated in airway disorders. At high doses, PM2.5 increased the expression of IL6, TNF, TSLP, CSF2, PTGS2, IL4R, and SPINK5. Other genes such as ADAM33, ORMDL3, DPP10 and CYP1A1, however, were increased by PM2.5 at much lower doses (≤1 μg/cm2). Repeated exposure to PM2.5 at 1 or 5 μg/cm2 every day for 7 days increased the sensitivity and magnitude of change for all of the aforementioned genes. Genes such as IL13 and TGFB1, increased only when cells were repeatedly exposed to PM2.5. Treatment with an antioxidant, or inhibitors to aryl hydrocarbon receptor or NF-κB attenuated the effect of PM2.5. These data demonstrate that PM2.5 exerts pleiotropic actions that differ by dose and duration that affect a variety of genes important to the development of airway disease.
Collapse
Affiliation(s)
- Priya Tripathi
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Furong Deng
- School of Public Health, Peking University, Beijing, China
| | - Anne M Scruggs
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yahong Chen
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Heymann PW, Nguyen HT, Steinke JW, Turner RB, Woodfolk JA, Platts-Mills TAE, Martin L, He H, Biagini Myers J, Lindsey M, Sivaprasad U, Medvedovic M, Mahi N, Carper H, Murphy DD, Patrie J, Khurana Hershey GK. Rhinovirus infection results in stronger and more persistent genomic dysregulation: Evidence for altered innate immune response in asthmatics at baseline, early in infection, and during convalescence. PLoS One 2017; 12:e0178096. [PMID: 28552993 PMCID: PMC5446117 DOI: 10.1371/journal.pone.0178096] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/07/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Rhinovirus (HRV) is associated with the large majority of virus-induced asthma exacerbations in children and young adults, but the mechanisms remain poorly defined. METHODS Asthmatics and non-asthmatic controls were inoculated with HRV-A16, and nasal epithelial samples were obtained 7 days before, 36 hours after, and 7 days after viral inoculation. RNA was extracted and subjected to RNA-seq analysis. RESULTS At baseline, 57 genes were differentially expressed between asthmatics and controls, and the asthmatics had decreased expression of viral replication inhibitors and increased expression of genes involved in inflammation. At 36 hours (before the emergence of peak symptoms), 1329 genes were significantly altered from baseline in the asthmatics compared to 62 genes in the controls. At this time point, asthmatics lacked an increase in IL-10 signaling observed in the controls. At 7 days following HRV inoculation, 222 genes were significantly dysregulated in the asthmatics, whereas only 4 genes were dysregulated among controls. At this time point, the controls but not asthmatics demonstrated upregulation of SPINK5. CONCLUSIONS As judged by the magnitude and persistence of dysregulated genes, asthmatics have a substantially different host response to HRV-A16 infection compared with non-asthmatic controls. Gene expression differences illuminate biologically plausible mechanisms that contribute to a better understanding of the pathogenesis of HRV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Peter W. Heymann
- Division of Allergy, Immunology and Respiratory Medicine, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Huyen-Tran Nguyen
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - John W. Steinke
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ronald B. Turner
- Division of Infectious Diseases, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Judith A. Woodfolk
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas A. E. Platts-Mills
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lisa Martin
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Hua He
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jocelyn Biagini Myers
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Mark Lindsey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Umasundari Sivaprasad
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Mario Medvedovic
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Naim Mahi
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Holliday Carper
- Division of Allergy, Immunology and Respiratory Medicine, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Deborah D. Murphy
- Division of Allergy, Immunology and Respiratory Medicine, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - James Patrie
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
8
|
Kim SH. Airway epithelial cells in airway inflammation and remodeling in asthma. ALLERGY ASTHMA & RESPIRATORY DISEASE 2016. [DOI: 10.4168/aard.2016.4.2.82] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
9
|
Martínez-Aguilar NE, Del Río-Navarro BE, Navarro-Olivos E, García-Ortíz H, Orozco L, Jiménez-Morales S. SPINK5 and ADRB2 haplotypes are risk factors for asthma in Mexican pediatric patients. J Asthma 2014; 52:232-9. [PMID: 25233048 DOI: 10.3109/02770903.2014.966913] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Asthma is one of the most common respiratory diseases worldwide, and the complexity of its etiology has been widely documented. Chromosome 5q31-33 is one of the main loci implicated in asthma and asthma-related traits. IL13, CD14 and ADRB2, which are located in this risk locus, are among the genes most strongly associated with asthma susceptibility. OBJECTIVES This study evaluated whether single-nucleotide polymorphisms or haplotypes at 5q31-33 conferred risk for asthma in Mexican-Mestizo pediatric patients. METHODS We performed a case-controlled study including 851 individuals, 421 of them affected with childhood-onset asthma and 430 ethnically matched unaffected subjects. We used the TaqMan Allelic Discrimination Assay to genotype 20 single-nucleotide polymorphisms within IL5, RAD50, IL13, IL4, CD14, SPINK5, HTR4, ADRB2 and IL12B. RESULTS Although no association was detected for any risk allele, three SPINK5 haplotypes (GGCT: p = 6 × 10(-6); AATC: p = 0.0001; AGTT: p = 0.0001) and five ADRB2 haplotypes (AGGACC: p = 0.0014; AGGAAG: p = 0.0002; TGAGAG: p = 0.0001; AGGAAC: p = 0.0002; AAGGAG: p = 0.003) were associated with asthma. Notably, the AGTT SPINK5 haplotype exhibited a male gender-dependent association (p = 7.6 × 10(-5)). CONCLUSION Our results suggest that SPINK5 and ADRB2 haplotypes might play a role in the susceptibility to childhood-onset asthma.
Collapse
Affiliation(s)
- N E Martínez-Aguilar
- Escuela Superior de Medicina, Instituto Politecnico Nacional , México City , Mexico
| | | | | | | | | | | |
Collapse
|
10
|
Biagini Myers JM, Martin LJ, Kovacic MB, Mersha TB, He H, Pilipenko V, Lindsey MA, Ericksen MB, Bernstein DI, LeMasters GK, Lockey JE, Khurana Hershey GK. Epistasis between serine protease inhibitor Kazal-type 5 (SPINK5) and thymic stromal lymphopoietin (TSLP) genes contributes to childhood asthma. J Allergy Clin Immunol 2014; 134:891-899.e3. [PMID: 24831437 DOI: 10.1016/j.jaci.2014.03.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Epithelial genes have previously been associated with asthma but only explain a small fraction of heritability. In part, this might be due to epistasis, which is often not considered. OBJECTIVE We sought to determine independent and epistatic associations between filaggrin (FLG), serine protease inhibitor Kazal-type 5 (SPINK5), and thymic stromal lymphopoietin (TSLP) gene variants and childhood asthma. METHODS Using a candidate gene approach, we genotyped 29 variants in FLG, SPINK5, and TSLP in asthmatic, allergic, and nonallergic nonasthmatic white and black children participating in the well-phenotyped Greater Cincinnati Pediatric Clinic Repository. Associations with asthma were also assessed in 6 replication populations. RESULTS We observed independent associations of variants in SPINK5 (P = .003) and TSLP (P = .006) with childhood asthma; a SPINK5 single nucleotide polymorphism was replicated. In subjects with 1 or more SPINK5 risk alleles, the absence of the TSLP protective minor alleles was associated with a significant increase in asthma (67% vs 53%, P = .0017). In contrast, the presence or absence of TSLP minor alleles did not affect asthma risk in subjects without the SPINK5 risk alleles. The SPINK5 and TSLP epistasis was replicated in a black population (P = .036) who did not display independent association with variants in these genes. CONCLUSIONS Our results support epistasis between SPINK5 and TSLP, which contributes to childhood asthma. These findings emphasize the importance of using biology to inform analyses to identify genetic susceptibility to complex diseases. The results from our study have clinical relevance and support that the therapeutic effects of anti-TSLP therapy in asthmatic patients might be dependent on SPINK5 genotype.
Collapse
Affiliation(s)
- Jocelyn M Biagini Myers
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Lisa J Martin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Melinda Butsch Kovacic
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Tesfaye B Mersha
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Hua He
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Valentina Pilipenko
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Mark A Lindsey
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Mark B Ericksen
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - David I Bernstein
- Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Grace K LeMasters
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio
| | - James E Lockey
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio
| | - Gurjit K Khurana Hershey
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
11
|
Wagener AH, Zwinderman AH, Luiten S, Fokkens WJ, Bel EH, Sterk PJ, van Drunen CM. The impact of allergic rhinitis and asthma on human nasal and bronchial epithelial gene expression. PLoS One 2013; 8:e80257. [PMID: 24282527 PMCID: PMC3839950 DOI: 10.1371/journal.pone.0080257] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 10/02/2013] [Indexed: 01/30/2023] Open
Abstract
Background The link between upper and lower airways in patients with both asthma and allergic rhinitis is still poorly understood. As the biological complexity of these disorders can be captured by gene expression profiling we hypothesized that the clinical expression of rhinitis and/or asthma is related to differential gene expression between upper and lower airways epithelium. Objective Defining gene expression profiles of primary nasal and bronchial epithelial cells from the same individuals and examining the impact of allergic rhinitis with and without concomitant allergic asthma on expression profiles. Methods This cross-sectional study included 18 subjects (6 allergic asthma and allergic rhinitis; 6 allergic rhinitis; 6 healthy controls). The estimated false discovery rate comparing 6 subjects per group was approximately 5%. RNA was extracted from isolated and cultured epithelial cells from bronchial brushings and nasal biopsies, and analyzed by microarray (Affymetrix U133+ PM Genechip Array). Data were analysed using R and Bioconductor Limma package. For gene ontology GeneSpring GX12 was used. Results The study was successfully completed by 17 subjects (6 allergic asthma and allergic rhinitis; 5 allergic rhinitis; 6 healthy controls). Using correction for multiple testing, 1988 genes were differentially expressed between healthy lower and upper airway epithelium, whereas in allergic rhinitis with or without asthma this was only 40 and 301 genes, respectively. Genes influenced by allergic rhinitis with or without asthma were linked to lung development, remodeling, regulation of peptidases and normal epithelial barrier functions. Conclusions Differences in epithelial gene expression between the upper and lower airway epithelium, as observed in healthy subjects, largely disappear in patients with allergic rhinitis with or without asthma, whilst new differences emerge. The present data identify several pathways and genes that might be potential targets for future drug development.
Collapse
Affiliation(s)
- Ariane H. Wagener
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands
- * E-mail:
| | - Aeilko H. Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Silvia Luiten
- Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Elisabeth H. Bel
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Peter J. Sterk
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Cornelis M. van Drunen
- Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
12
|
Yavuz ST, Buyuktiryaki B, Sahiner UM, Birben E, Tuncer A, Yakarisik S, Karabulut E, Kalayci O, Sackesen C. Factors that predict the clinical reactivity and tolerance in children with cow's milk allergy. Ann Allergy Asthma Immunol 2013; 110:284-9. [PMID: 23535094 DOI: 10.1016/j.anai.2013.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 01/13/2013] [Accepted: 01/22/2013] [Indexed: 11/16/2022]
Abstract
BACKGROUND Specific IgE (sIgE) may be used for the diagnosis of cow's milk allergy (CMA) and as a guide to perform food challenge tests in patients with CMA. The effect of genetic variants on the prognosis of food allergy is largely unknown. OBJECTIVE To examine the performance of sIgE analysis and the utility of the genetic variants of CD14, STAT6, IL13, IL10, SPINK5, and TSLP in predicting the clinical course in children with CMA. METHODS Serum sIgE levels of 94 children who underwent open food challenges and 54 children with anaphylaxis due to cow's milk (CM) were retrospectively analyzed between January 2002 and May 2009. The genetic polymorphisms were determined in 72 children. RESULTS A total of 148 children were followed up for a median of 3.5 years, and 42 of the 94 challenge results were positive. The probability curves with 95% decision points were 2.8 kU/L for younger than 1 year, 11.1 for younger than 2 years, 11.7 for younger than 4 years, and 13.7 for younger than 6 years. Sixty-six children outgrew CMA during follow-up. Children with initial an CM sIgE level less than 6 kU/L outgrew CMA earlier than children with an initial CM sIgE level of 6 kU/L or higher (P < .001). The age of tolerance development for CM was significantly higher in children with the GG genotype at rs324015 of the STAT6 gene compared with those with the AA+AG genotype (2 years [range, 1.5-3.9 years] vs 1.2 years [range, 1.0-2.2 years]) (P = .02). CONCLUSION The decision points of sIgE obtained in different age groups may help to determine the likelihood of clinical reactivity more precisely. The results suggest that sIgE levels and STAT6 gene variants may be important determinants to predict longer persistence of CMA.
Collapse
Affiliation(s)
- S Tolga Yavuz
- Faculty of Medicine, Department of Pediatric Allergy and Asthma, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
The role of SCCA1 in asthma related physiological events in the airway epithelium and the effect of promoter variants on asthma and gene function. Respir Med 2012. [PMID: 23199842 DOI: 10.1016/j.rmed.2012.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Even though the systemic level of SCCA1, a serine protease inhibitor, was shown to be elevated in asthma, its physiological role is unknown. OBJECTIVE We sought to determine the effect of SCCA1 on apoptosis, cytokine expression and mucus production by A549 cells and define the effect of promoter variants on gene expression and association with asthma. METHODS SCCA levels were measured by ELISA. Promoter variants were determined by direct sequencing. 442 asthmatic children and 191 controls were genotyped by RFLP. The functional effect of the polymorphisms was assessed in transient transfection experiments using reporter constructs. A transcription factor ELISA was used for differential binding of GATA proteins to the variant region. The effects of SCCA1 on cytokine synthesis, mucus production and apoptosis were determined in A549 cells transfected with SCCA1 pcDNA vector. MUC5AC expression in A549 cells was determined with RT-PCR. RESULTS SCCA1 protein level was significantly higher in asthmatic children compared to healthy controls. Four polymorphisms SCCA1 promoter that were in linkage disequilibrium were associated with skin test positivity in asthmatic children and showed higher promoter activity and higher binding of GATA-2 and GATA-3 after IL-4 + IL-13 stimulation. IL-6, IL-8 levels were significantly higher in cells transfected with SCCA1 whereas RANTES increased only after IL-4 stimulation. Transfection of A549 cells with SCCA1 resulted in decreased MUC5AC expression and conferred protection against apoptosis. CONCLUSION Our results showed that SCCA1 has diverse effects on many of the cellular events that characterize asthma and its role extends beyond protease inhibition.
Collapse
|