1
|
Simmons Beck R, Liang OD, Klinger JR. Light at the ENDothelium-role of Sox17 and Runx1 in endothelial dysfunction and pulmonary arterial hypertension. Front Cardiovasc Med 2023; 10:1274033. [PMID: 38028440 PMCID: PMC10656768 DOI: 10.3389/fcvm.2023.1274033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease that is characterized by an obliterative vasculopathy of the distal pulmonary circulation. Despite significant progress in our understanding of the pathophysiology, currently approved medical therapies for PAH act primarily as pulmonary vasodilators and fail to address the underlying processes that lead to the development and progression of the disease. Endothelial dysregulation in response to stress, injury or physiologic stimuli followed by perivascular infiltration of immune cells plays a prominent role in the pulmonary vascular remodeling of PAH. Over the last few decades, our understanding of endothelial cell dysregulation has evolved and brought to light a number of transcription factors that play important roles in vascular homeostasis and angiogenesis. In this review, we examine two such factors, SOX17 and one of its downstream targets, RUNX1 and the emerging data that implicate their roles in the pathogenesis of PAH. We review their discovery and discuss their function in angiogenesis and lung vascular development including their roles in endothelial to hematopoietic transition (EHT) and their ability to drive progenitor stem cells toward an endothelial or myeloid fate. We also summarize the data from studies that link mutations in Sox17 with an increased risk of developing PAH and studies that implicate Sox17 and Runx1 in the pathogenesis of PAH. Finally, we review the results of recent studies from our lab demonstrating the efficacy of preventing and reversing pulmonary hypertension in animal models of PAH by deleting RUNX1 expression in endothelial or myeloid cells or by the use of RUNX1 inhibitors. By investigating PAH through the lens of SOX17 and RUNX1 we hope to shed light on the role of these transcription factors in vascular homeostasis and endothelial dysregulation, their contribution to pulmonary vascular remodeling in PAH, and their potential as novel therapeutic targets for treating this devastating disease.
Collapse
Affiliation(s)
- Robert Simmons Beck
- Division of Pulmonary, Sleep and Critical Care Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, United States
| | - Olin D. Liang
- Division of Hematology/Oncology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, United States
| | - James R. Klinger
- Division of Pulmonary, Sleep and Critical Care Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
2
|
Zhu Z, Li Y, Freishtat RJ, Celedón JC, Espinola JA, Harmon B, Hahn A, Camargo CA, Liang L, Hasegawa K. Epigenome-wide association analysis of infant bronchiolitis severity: a multicenter prospective cohort study. Nat Commun 2023; 14:5495. [PMID: 37679381 PMCID: PMC10485022 DOI: 10.1038/s41467-023-41300-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
Bronchiolitis is the most common lower respiratory infection in infants, yet its pathobiology remains unclear. Here we present blood DNA methylation data from 625 infants hospitalized with bronchiolitis in a 17-center prospective study, and associate them with disease severity. We investigate differentially methylated CpGs (DMCs) for disease severity. We characterize the DMCs based on their association with cell and tissues types, biological pathways, and gene expression. Lastly, we also examine the relationships of severity-related DMCs with respiratory and immune traits in independent cohorts. We identify 33 DMCs associated with severity. These DMCs are differentially methylated in blood immune cells. These DMCs are also significantly enriched in multiple tissues (e.g., lung) and cells (e.g., small airway epithelial cells), and biological pathways (e.g., interleukin-1-mediated signaling). Additionally, these DMCs are associated with respiratory and immune traits (e.g., asthma, lung function, IgE levels). Our study suggests the role of DNA methylation in bronchiolitis severity.
Collapse
Affiliation(s)
- Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yijun Li
- Department of Epidemiology, Harvard T.H.Chan School of Public Health, Boston, MA, USA
| | - Robert J Freishtat
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Juan C Celedón
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Janice A Espinola
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brennan Harmon
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Andrea Hahn
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Division of Infectious Diseases, Children's National Hospital, Washington, DC, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H.Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H.Chan School of Public Health, Boston, MA, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Rozen EJ, Ozeroff CD, Allen MA. RUN(X) out of blood: emerging RUNX1 functions beyond hematopoiesis and links to Down syndrome. Hum Genomics 2023; 17:83. [PMID: 37670378 PMCID: PMC10481493 DOI: 10.1186/s40246-023-00531-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND RUNX1 is a transcription factor and a master regulator for the specification of the hematopoietic lineage during embryogenesis and postnatal megakaryopoiesis. Mutations and rearrangements on RUNX1 are key drivers of hematological malignancies. In humans, this gene is localized to the 'Down syndrome critical region' of chromosome 21, triplication of which is necessary and sufficient for most phenotypes that characterize Trisomy 21. MAIN BODY Individuals with Down syndrome show a higher predisposition to leukemias. Hence, RUNX1 overexpression was initially proposed as a critical player on Down syndrome-associated leukemogenesis. Less is known about the functions of RUNX1 in other tissues and organs, although growing reports show important implications in development or homeostasis of neural tissues, muscle, heart, bone, ovary, or the endothelium, among others. Even less is understood about the consequences on these tissues of RUNX1 gene dosage alterations in the context of Down syndrome. In this review, we summarize the current knowledge on RUNX1 activities outside blood/leukemia, while suggesting for the first time their potential relation to specific Trisomy 21 co-occurring conditions. CONCLUSION Our concise review on the emerging RUNX1 roles in different tissues outside the hematopoietic context provides a number of well-funded hypotheses that will open new research avenues toward a better understanding of RUNX1-mediated transcription in health and disease, contributing to novel potential diagnostic and therapeutic strategies for Down syndrome-associated conditions.
Collapse
Affiliation(s)
- Esteban J Rozen
- Crnic Institute Boulder Branch, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO, 80303, USA.
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO, 80045, USA.
| | - Christopher D Ozeroff
- Crnic Institute Boulder Branch, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO, 80303, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO, 80045, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, 1945 Colorado Ave., Boulder, CO, 80309, USA
| | - Mary Ann Allen
- Crnic Institute Boulder Branch, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., Boulder, CO, 80303, USA.
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
4
|
Kurihara C, Kuniyoshi KM, Rehan VK. Preterm Birth, Developmental Smoke/Nicotine Exposure, and Life-Long Pulmonary Sequelae. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040608. [PMID: 37189857 DOI: 10.3390/children10040608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
This review delineates the main pulmonary issues related to preterm birth, perinatal tobacco/nicotine exposure, and its effects on offspring, focusing on respiratory health and its possible transmission to subsequent generations. We review the extent of the problem of preterm birth, prematurity-related pulmonary effects, and the associated increased risk of asthma later in life. We then review the impact of developmental tobacco/nicotine exposure on offspring asthma and the significance of transgenerational pulmonary effects following perinatal tobacco/nicotine exposure, possibly via its effects on germline epigenetics.
Collapse
Affiliation(s)
- Chie Kurihara
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Katherine M Kuniyoshi
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Virender K Rehan
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Allegra A, Murdaca G, Gammeri L, Ettari R, Gangemi S. Alarmins and MicroRNAs, a New Axis in the Genesis of Respiratory Diseases: Possible Therapeutic Implications. Int J Mol Sci 2023; 24:ijms24021783. [PMID: 36675299 PMCID: PMC9861898 DOI: 10.3390/ijms24021783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023] Open
Abstract
It is well ascertained that airway inflammation has a key role in the genesis of numerous respiratory pathologies, including asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome. Pulmonary tissue inflammation and anti-inflammatory responses implicate an intricate relationship between local and infiltrating immune cells and structural pulmonary cells. Alarmins are endogenic proteins discharged after cell injury in the extracellular microenvironment. The purpose of our review is to highlight the alterations in respiratory diseases involving some alarmins, such as high mobility group box 1 (HMGB1) and interleukin (IL)-33, and their inter-relationships and relationships with genetic non-coding material, such as microRNAs. The role played by these alarmins in some pathophysiological processes confirms the existence of an axis composed of HMGB1 and IL-33. These alarmins have been implicated in ferroptosis, the onset of type 2 inflammation and airway alterations. Moreover, both factors can act on non-coding genetic material capable of modifying respiratory function. Finally, we present an outline of alarmins and RNA-based therapeutics that have been proposed to treat respiratory pathologies.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Luca Gammeri
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
6
|
LIU JIA, WANG FAPING, YUAN BO, LUO FENGMING. Transcriptional factor RUNX1: A potential therapeutic target for fibrotic pulmonary disease. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
|
7
|
Agache I, Shamji MH, Kermani NZ, Vecchi G, Favaro A, Layhadi JA, Heider A, Akbas DS, Filipaviciute P, Wu LYD, Cojanu C, Laculiceanu A, Akdis CA, Adcock IM. Multidimensional endotyping using nasal proteomics predicts molecular phenotypes in the asthmatic airways. J Allergy Clin Immunol 2023; 151:128-137. [PMID: 36154846 DOI: 10.1016/j.jaci.2022.06.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 06/16/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Unsupervised clustering of biomarkers derived from noninvasive samples such as nasal fluid is less evaluated as a tool for describing asthma endotypes. OBJECTIVE We sought to evaluate whether protein expression in nasal fluid would identify distinct clusters of patients with asthma with specific lower airway molecular phenotypes. METHODS Unsupervised clustering of 168 nasal inflammatory and immune proteins and Shapley values was used to stratify 43 patients with severe asthma (endotype of noneosinophilic asthma) using a 2 "modeling blocks" machine learning approach. This algorithm was also applied to nasal brushings transcriptomics from U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes). Feature reduction and functional gene analysis were used to compare proteomic and transcriptomic clusters. Gene set variation analysis provided enrichment scores of the endotype of noneosinophilic asthma protein signature within U-BIOPRED sputum and blood. RESULTS The nasal protein machine learning model identified 2 severe asthma endotypes, which were replicated in U-BIOPRED nasal transcriptomics. Cluster 1 patients had significant airway obstruction, small airways disease, air trapping, decreased diffusing capacity, and increased oxidative stress, although only 4 of 18 were current smokers. Shapley identified 20 cluster-defining proteins. Forty-one proteins were significantly higher in cluster 1. Pathways associated with proteomic and transcriptomic clusters were linked to TH1, TH2, neutrophil, Janus kinase-signal transducer and activator of transcription, TLR, and infection activation. Gene set variation analysis of the nasal protein and gene signatures were enriched in subjects with sputum neutrophilic/mixed granulocytic asthma and in subjects with a molecular phenotype found in sputum neutrophil-high subjects. CONCLUSIONS Protein or gene analysis may indicate molecular phenotypes within the asthmatic lower airway and provide a simple, noninvasive test for non-type 2 immune response asthma that is currently unavailable.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania; Theramed Healthcare, Brasov, Romania.
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, United Kingdom; NIHR Biomedical Research Centre, London, United Kingdom.
| | - Nazanin Zounemat Kermani
- National Heart and Lung Institute, Imperial College London, United Kingdom; Data Science Institute, Imperial College London, United Kingdom
| | | | | | - Janice A Layhadi
- National Heart and Lung Institute, Imperial College London, United Kingdom; NIHR Biomedical Research Centre, London, United Kingdom
| | - Anja Heider
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Didem Sanver Akbas
- National Heart and Lung Institute, Imperial College London, United Kingdom; NIHR Biomedical Research Centre, London, United Kingdom
| | - Paulina Filipaviciute
- National Heart and Lung Institute, Imperial College London, United Kingdom; NIHR Biomedical Research Centre, London, United Kingdom
| | - Lily Y D Wu
- National Heart and Lung Institute, Imperial College London, United Kingdom; NIHR Biomedical Research Centre, London, United Kingdom
| | - Catalina Cojanu
- Faculty of Medicine, Transylvania University, Brasov, Romania; Theramed Healthcare, Brasov, Romania
| | - Alexandru Laculiceanu
- Faculty of Medicine, Transylvania University, Brasov, Romania; Theramed Healthcare, Brasov, Romania
| | - Cezmi A Akdis
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, United Kingdom; NIHR Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
8
|
Yamashiro T, Kurosaka H, Inubush T. The Association Between Runx Signaling and Craniofacial Development and Disease. Curr Osteoporos Rep 2022; 20:120-126. [PMID: 34931296 DOI: 10.1007/s11914-021-00692-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The Runx family genes (Runx1, Runx2, Runx3, and Cbfb) are important transcriptional regulators in the development of various tissues. We herein highlight the roles of the Runx family genes in morphogenesis in the craniofacial regions and in the pathogenesis of congenital morphological problems in these regions. RECENT FINDINGS A recent analysis using conditional Runx mutant animals and a human genetic study identified the novel roles of Runx genes in the development of the tooth, salivary glands, and the palate. In an animal study, Runx1/Cbfb signaling was found to regulate the Lgr5 expression and maintain the stem cells in the dental epithelium in the growing incisors. Aberrant Runx1/Cbfb signaling induced male-specific involution of the convoluted granular cell differentiation of the submandibular gland. In palatogenesis, Runx1/Cbfb signaling regulated the Tgfb3 expression in the fusing palatal epithelium through Stat3 activation. The combination of a human genetic study and a phenotype analysis of mutant animals revealed the various roles of Runx genes in the development of the tooth, palate, and salivary glands. Runx genes have functional redundancy in various tissues, which still hinder the roles of Runx genes in morphogenesis. Future studies may reveal the novel roles of Runx signaling.
Collapse
Affiliation(s)
- Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan.
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Toshihiro Inubush
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
9
|
Wu W, Gao J, Chen D, Chen G, Feng Y, Chang C, Chen S, Yi L, Zhen G. Epithelial microRNA-30a-3p targets RUNX2/HMGB1 axis to suppress airway eosinophilic inflammation in asthma. Respir Res 2022; 23:17. [PMID: 35093061 PMCID: PMC8800331 DOI: 10.1186/s12931-022-01933-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Background Type 2-high asthma is a prominent endotype of asthma which is characterized by airway eosinophilic inflammation. Airway epithelial cells play a critical role in the pathogenesis of asthma. Our previous miRNA profiling data showed that miR-30a-3p was downregulated in bronchial epithelial cells from asthma patients. We hypothesize that epithelial miR-30a-3p plays a role in asthma airway inflammation. Methods We measured miR‐30a-3p expression in bronchial brushings of asthma patients (n = 51) and healthy controls (n = 16), and analyzed the correlations between miR‐30a-3p expression and airway eosinophilia. We examined whether Runt-related transcription factor 2 (RUNX2) was a target of miR‐30a-3p and whether RUNX2 bound to the promoter of high mobility group box 1 (HMGB1) by using luciferase reporter assay and chromatin immunoprecipitation (ChIP)-PCR. The role of miR‐30a-3p was also investigated in a murine model of allergic airway inflammation. Results We found that miR-30a-3p expression were significantly decreased in bronchial brushings of asthma patients compared to control subjects. Epithelial miR-30a-3p expression was negatively correlated with parameters reflecting airway eosinophilia including eosinophils in induced sputum and bronchial biopsies, and fraction of exhaled nitric oxide in asthma patients. We verified that RUNX2 is a target of miR-30a-3p. Furthermore, RUNX2 bound to the promoter of HMGB1 and upregulated HMGB1 expression. RUNX2 and HMGB1 expression was both enhanced in airway epithelium and was correlated with each other in asthma patients. Inhibition of miR-30a-3p enhanced RUNX2 and HMGB1 expression, and RUNX2 overexpression upregulated HMGB1 in BEAS-2B cells. Intriguingly, airway overexpression of mmu-miR-30a-3p suppressed Runx2 and Hmgb1 expression, and alleviated airway eosinophilia in a mouse model of allergic airway inflammation. Conclusions Epithelial miR-30a-3p could possibly target RUNX2/HMGB1 axis to suppress airway eosinophilia in asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01933-x.
Collapse
|
10
|
Azari H, Karimi E, Shekari M, Tahmasebi A, Nikpoor AR, Negahi AA, Sanadgol N, Mousavi P. Construction of a lncRNA-miRNA-mRNA network to determine the key regulators of the Th1/Th2 imbalance in multiple sclerosis. Epigenomics 2021; 13:1797-1815. [PMID: 34726075 DOI: 10.2217/epi-2021-0296] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The exact epigenetic mechanisms that determine the balance of T helper (Th)1 and Th2 cells and autoimmune responses in multiple sclerosis (MS) remain unclear. We aim to clarify these. Methods: A combination of bioinformatics analysis and molecular evaluations was utilized to identify master hub genes. Results: A competitive endogenous RNA network containing six long noncoding RNAs (lncRNAs), 21 miRNAs and 86 mRNAs was provided through enrichment analysis and a protein-protein interaction network. NEAT1 and MALAT1 were found as differentially expressed lncRNAs using Gene Expression Omnibus (GSE21942). Quantitative real-time PCR results demonstrate dysregulation in the RUNX3 (a regulator of Th1/Th2 balance), GATA3 and TBX21, as well as miR-544a and miR-210-3p (which directly target RUNX3). ELISA also confirmed an imbalance in IFN-γ (Th1)/IL-4 (Th2) in MS patients. Conclusion: Our findings introduce novel biomarkers leading to Th1/Th2 imbalance in MS.
Collapse
Affiliation(s)
- Hanieh Azari
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 79196-93116, Iran
| | - Elham Karimi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 79196-93116, Iran
| | - Mohammad Shekari
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 79196-93116, Iran.,Hormozgan University of Medical Sciences Research Center for Molecular Medicine, Bandar Abbas, 79196-93116, Iran
| | - Ahmad Tahmasebi
- Institute of Biotechnology, Shiraz University, Shiraz, 71441-13131, Iran
| | - Amin Reza Nikpoor
- Hormozgan University of Medical Sciences Research Center for Molecular Medicine, Bandar Abbas, 79196-93116, Iran
| | - Ahmad Agha Negahi
- Department of Internal Medicine, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 79196-93116, Iran
| | - Nima Sanadgol
- Institute of Neuroanatomy, RWTH University Hospital Aachen, Aachen, 52074, Germany
| | - Pegah Mousavi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 79196-93116, Iran.,Hormozgan University of Medical Sciences Research Center for Molecular Medicine, Bandar Abbas, 79196-93116, Iran
| |
Collapse
|
11
|
Thomas AL, Marsman J, Antony J, Schierding W, O’Sullivan JM, Horsfield JA. Transcriptional Regulation of RUNX1: An Informatics Analysis. Genes (Basel) 2021; 12:1175. [PMID: 34440349 PMCID: PMC8395016 DOI: 10.3390/genes12081175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 01/04/2023] Open
Abstract
The RUNX1/AML1 gene encodes a developmental transcription factor that is an important regulator of haematopoiesis in vertebrates. Genetic disruptions to the RUNX1 gene are frequently associated with acute myeloid leukaemia. Gene regulatory elements (REs), such as enhancers located in non-coding DNA, are likely to be important for Runx1 transcription. Non-coding elements that modulate Runx1 expression have been investigated over several decades, but how and when these REs function remains poorly understood. Here we used bioinformatic methods and functional data to characterise the regulatory landscape of vertebrate Runx1. We identified REs that are conserved between human and mouse, many of which produce enhancer RNAs in diverse tissues. Genome-wide association studies detected single nucleotide polymorphisms in REs, some of which correlate with gene expression quantitative trait loci in tissues in which the RE is active. Our analyses also suggest that REs can be variant in haematological malignancies. In summary, our analysis identifies features of the RUNX1 regulatory landscape that are likely to be important for the regulation of this gene in normal and malignant haematopoiesis.
Collapse
Affiliation(s)
- Amarni L. Thomas
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (A.L.T.); (J.A.)
| | - Judith Marsman
- Department of Cardiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Jisha Antony
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (A.L.T.); (J.A.)
- The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
| | - William Schierding
- Liggins Institute, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
| | - Justin M. O’Sullivan
- The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
- Liggins Institute, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton SO17 1BJ, UK
| | - Julia A. Horsfield
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (A.L.T.); (J.A.)
- The Maurice Wilkins Centre for Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
- Genetics Otago Research Centre, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
12
|
Bermudez LG, Madariaga I, Zuñiga MI, Olaya M, Cañas A, Rodriguez LS, Moreno OM, Rojas A. RUNX1 gene expression changes in the placentas of women smokers. Exp Ther Med 2021; 22:902. [PMID: 34257715 PMCID: PMC8243315 DOI: 10.3892/etm.2021.10334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/21/2021] [Indexed: 11/28/2022] Open
Abstract
The placenta can be affected by environmental factors, such as exposure to cigarette smoke. This exposure in the fetal context is considered a risk factor for the development of short-term postnatal diseases, such as asthma. Asthma is an inflammatory disease characterized by predominant acquisition of CD4 T lymphocytes (TLs) of the Th2 type. Transcription factors such as GATA binding protein 3 (GATA3) and STAT6 actively participate in the differentiation of virgin TLs towards the Th2 profile, while transcription factors such as STAT1, T-Box transcription factor 21 (T-BET), RUNX1 and RUNX3 participate in their differentiation towards the Th1 profile. The objective of the current study was to evaluate the impact of exposure to cigarette smoke on the gene expression of STAT1, T-BET, GATA3, IL-4, RUNX1 and RUNX3 during the gestation period, and to determine whether the expression levels of these genes are associated with changes in global methylation. STAT1, GATA3, RUNX1 and RUNX3 protein and mRNA expression levels in the placental tissue of women smokers and non-smoking women were determined via immunohistochemistry and quantitative PCR (qPCR) respectively. Additionally, T-BET and IL-4 mRNA expression levels were determined by qPCR. On the other hand, global methylation was determined via ELISA. In the present study, significant increases were observed in RUNX1 transcription factor expression in placentas from women smokers when compared with placentas of non-smoking women. Similarly, significant increases in the expression of GATA3, IL-4 and RUNX3 mRNA were observed. The changes in gene expression were not associated with changes in the global methylation levels. Finally, a higher frequency of low-birth-weight infants were identified in cases of exposure to cigarette smoke during pregnancy when compared with infants not exposed to cigarette smoke during pregnancy. Thus, the data of the present study contributed to the understanding of the genetic and clinical impacts of exposure to cigarette smoke during pregnancy and its importance in maternal and fetal health.
Collapse
Affiliation(s)
- Litzy Gisella Bermudez
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Ithzayana Madariaga
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Maria Isabel Zuñiga
- Department of Pathology, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio, Bogotá 110231, Colombia
| | - Mercedes Olaya
- Department of Pathology, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio, Bogotá 110231, Colombia
| | - Alejandra Cañas
- Department of Internal Medicine, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio, Bogotá 110231, Colombia
| | - Luz-Stella Rodriguez
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Olga Maria Moreno
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Adriana Rojas
- Institute of Human Genetics, School of Medicine, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| |
Collapse
|
13
|
Zhu T, Zhang X, Chen X, Brown AP, Weirauch MT, Guilbert TW, Khurana Hershey GK, Biagini JM, Ji H. Nasal DNA methylation differentiates severe from non-severe asthma in African-American children. Allergy 2021; 76:1836-1845. [PMID: 33175399 PMCID: PMC8110596 DOI: 10.1111/all.14655] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Asthma is highly heterogeneous, and severity evaluation is key to asthma management. DNA methylation (DNAm) contributes to asthma pathogenesis. This study aimed to identify nasal epithelial DNAm differences between severe and nonsevere asthmatic children and evaluate the impact of environmental exposures. METHODS Thirty-three nonsevere and 22 severe asthmatic African American children were included in an epigenome-wide association study. Genome-wide nasal epithelial DNAm and gene expression were measured. CpG sites associated with asthma severity and environmental exposures and predictive of severe asthma were identified. DNAm was correlated with gene expression. Enrichment for transcription factor (TF) binding sites or histone modifications surrounding DNAm differences were determined. RESULTS We identified 816 differentially methylated CpG positions (DMPs) and 10 differentially methylated regions (DMRs) associated with asthma severity. Three DMPs exhibited discriminatory ability for severe asthma. Intriguingly, six DMPs were simultaneously associated with asthma, allergic asthma, total IgE, environmental IgE, and FeNO in an independent cohort of children. Twenty-seven DMPs were associated with traffic-related air pollution or secondhand smoke. DNAm at 22 DMPs was altered by diesel particles or allergen in human bronchial epithelial cells. DNAm levels at 39 DMPs were correlated with mRNA expression. Proximal to 816 DMPs, three histone marks and several TFs involved in asthma pathogenesis were enriched. CONCLUSIONS Significant differences in nasal epithelial DNAm were observed between nonsevere and severe asthma in African American children, a subset of which may be useful to predict disease severity. These CpG sites are subjected to the influences of environmental exposures and may regulate gene expression.
Collapse
Affiliation(s)
- Tao Zhu
- California National Primate Research Center, Davis, CA
| | - Xue Zhang
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | | | - Matthew T. Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Divisions of Biomedical Informatics and Developmental Biology, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Theresa W. Guilbert
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gurjit K. Khurana Hershey
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
- Divison of Asthma Research, Cincinnati Children’s Hospital Medical Center, Davis, CA
| | - Jocelyn M. Biagini
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
- Divison of Asthma Research, Cincinnati Children’s Hospital Medical Center, Davis, CA
| | - Hong Ji
- California National Primate Research Center, Davis, CA
- Department of Anatomy, Physiology and Cell biology, School of Veterinary Medicine, University of California, Davis, CA
| |
Collapse
|
14
|
Zhou X, Han X, Lyu SC, Bunning B, Kost L, Chang I, Cao S, Sampath V, Nadeau KC. Targeted DNA methylation profiling reveals epigenetic signatures in peanut allergy. JCI Insight 2021; 6:143058. [PMID: 33571165 PMCID: PMC8026193 DOI: 10.1172/jci.insight.143058] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/10/2021] [Indexed: 12/14/2022] Open
Abstract
DNA methylation (DNAm) has been shown to play a role in mediating food allergy; however, the mechanism by which it does so is poorly understood. In this study, we used targeted next-generation bisulfite sequencing to evaluate DNAm levels in 125 targeted highly informative genomic regions containing 602 CpG sites on 70 immune-related genes to understand whether DNAm can differentiate peanut allergy (PA) versus nonallergy (NA). We found PA-associated DNAm signatures associated with 12 genes (7 potentially novel to food allergy, 3 associated with Th1/Th2, and 2 associated with innate immunity), as well as DNAm signature combinations with superior diagnostic potential compared with serum peanut–specific IgE for PA versus NA. Furthermore, we found that, following peanut protein stimulation, peripheral blood mononuclear cell (PBMCs) from PA participants showed increased production of cognate cytokines compared with NA participants. The varying responses between PA and NA participants may be associated with the interaction between the modification of DNAm and the interference of environment. Using Euclidean distance analysis, we found that the distances of methylation profile comprising 12 DNAm signatures between PA and NA pairs in monozygotic (MZ) twins were smaller than those in randomly paired genetically unrelated individuals, suggesting that PA-related DNAm signatures may be associated with genetic factors.
Collapse
|
15
|
Ji Q, Hou J, Yong X, Gong G, Muddassir M, Tang T, Xie J, Fan W, Chen X. Targeted Dual Small Interfering Ribonucleic Acid Delivery via Non-Viral Polymeric Vectors for Pulmonary Fibrosis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007798. [PMID: 33604928 DOI: 10.1002/adma.202007798] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Indexed: 06/12/2023]
Abstract
Inhibiting the myofibroblast differentiation of lung-resident mesenchymal stem cells (LR-MSCs) is a promising yet challenging approach for pulmonary fibrosis (PF) therapy. Here, micelles formed by a graft copolymer of multiple PEGs modified branched polyethylenimine are used for delivering runt-related transcription factor-1 (RUNX1) small interfering RNA (siRNA) (siRUNX1) to the lung, aiming to inhibit the myofibroblast differentiation of LR-MSCs. LR-MSC targeting is achieved by functionalizing the micelle surface with an anti-stem-cell antigen-1 antibody fragment (Fab'). Consequently, therapeutic benefits are obtained by successful suppression of myofibroblast differentiation of LR-MSCs in bleomycin-induced PF model mice treated with siRUNX1-loaded micelles. Furthermore, an excellent synergistic effect of PF therapy is achieved for this micelle system loaded siRUNX1 and glioma-associated oncogene homolog-1 (Gli1) small interfering RNA (siGli1), a traditional anti-PF siRNA of glioma-associated oncogene homolog-1. Hence, this work not only provides RUNX1 as a novel PF therapeutic target, but also as a promising dual siRNA-loaded nanocarrier system for the therapy of PF.
Collapse
Affiliation(s)
- Qijian Ji
- Department of Critical Care Medicine, Xuyi People's Hospital, 28 Hongwu Road, Xuyi, Huai'an, Jiangsu, 211700, China
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Jiwei Hou
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Xueqing Yong
- Department of Nuclear Science & Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Guangming Gong
- Department of Pharmaceutics, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Mohd Muddassir
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Tianyu Tang
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Jinbing Xie
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
16
|
Li H, Wang H, Sokulsky L, Liu S, Yang R, Liu X, Zhou L, Li J, Huang C, Li F, Lei X, Jia H, Cheng J, Li F, Yang M, Zhang G. Single-cell transcriptomic analysis reveals key immune cell phenotypes in the lungs of patients with asthma exacerbation. J Allergy Clin Immunol 2021; 147:941-954. [PMID: 33039479 DOI: 10.1016/j.jaci.2020.09.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Asthma exacerbations are associated with heightened asthma symptoms, which can result in hospitalization in severe cases. However, the molecular immunologic processes that determine the course of an exacerbation remain poorly understood, impeding the progression of development of effective therapies. OBJECTIVE Our aim was to identify candidate genes that are strongly associated with asthma exacerbation at a cellular level. METHODS Subjects with asthma exacerbation and healthy control subjects were recruited, and bronchoalveolar lavage fluid was isolated from these subjects via bronchoscopy. Cells were isolated through fluorescence-activated cell sorting, and single-cell RNA sequencing was performed on enriched cell populations. RESULTS We showed that the levels of monocytes, CD8+ T cells, and macrophages are significantly elevated in the bronchoalveolar lavage fluid of patients. A set of cytokines and intracellular transduction regulators are associated with asthma exacerbations and are shared across multiple cell clusters, forming a complicated molecular framework. An additional group of core exacerbation-associated modules is activated, including eukaryotic initiation factor 2 signaling, ephrin receptor signaling, and C-X-C chemokine receptor type 4 signaling in the subpopulations of CD8+ T cells (C1-a) and monocyte clusters (C7 clusters), which are associated with infection. CONCLUSION Our study identified a significant number of severe asthma-associated genes that are differentially expressed by multiple cell clusters.
Collapse
Affiliation(s)
- Hui Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huaqi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Leon Sokulsky
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Shaoxia Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Liu
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lujia Zhou
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Juan Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chun Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Lei
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongxia Jia
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiuling Cheng
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuguang Li
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia; Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Effect of Intrauterine Smoke Exposure on microRNA-15a Expression in Human Lung Development and Subsequent Asthma Risk. Healthcare (Basel) 2020; 8:healthcare8040536. [PMID: 33291534 PMCID: PMC7761806 DOI: 10.3390/healthcare8040536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 11/16/2022] Open
Abstract
Background: In utero smoke (IUS) exposure is associated with asthma susceptibility. Objective: We sought to test the hypothesis that changes in miRNA expression by IUS exposure during human lung development is associated with asthma susceptibility. Methods: Gene expression was profiled from 53 IUS unexposed and 51 IUS exposed human fetal lung tissues. We tested for the differential expression of miRNAs across post-conception age and by IUS using linear models with covariate adjustment. We tested the IUS-associated miRNAs for association with their gene expression targets using pair-wise inverse correlation. Using our mouse model, we investigated the persistence of the IUS-associated miRNA signature using RT-PCR from the lungs of mouse pups with and without IUS at postnatal day 14. MiRNAs were then tested for association with asthma and exacerbations using whole blood gene expression profiles from Asthma BRIDGE. Results: Five miRNAs were differentially expressed across post-conception age (adjusted p < 0.0002) including two that were differentially expressed by IUS exposure in human fetal lung (p < 0.05). MiR-15a was differentially expressed by post-conception age (p = 0.00002), IUS exposure in human fetal lung (p = 0.005), and in the post-natal mouse lung (p = 0.01). MiR-15a was also associated with the in utero expression of GSDMB (adjusted p = 0.0002), a known childhood asthma gene and with asthma exacerbations (p = 0.0009) in Asthma BRIDGE. Thus, miR-15a is expressed during human lung development, is impacted by IUS exposure, regulates the intrauterine expression of asthma genes, and is associated with asthma severity. Conclusions: These results provide evidence for the role of miR-15a in the fetal origin of asthma.
Collapse
|
18
|
Portas L, Pereira M, Shaheen SO, Wyss AB, London SJ, Burney PGJ, Hind M, Dean CH, Minelli C. Lung Development Genes and Adult Lung Function. Am J Respir Crit Care Med 2020; 202:853-865. [PMID: 32392078 PMCID: PMC7491406 DOI: 10.1164/rccm.201912-2338oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 05/11/2020] [Indexed: 12/25/2022] Open
Abstract
Rationale: Poor lung health in adult life may occur partly through suboptimal growth and development, as suggested by epidemiological evidence pointing to early life risk factors.Objectives: To systematically investigate the effects of lung development genes on adult lung function.Methods: Using UK Biobank data, we tested the association of 391 genes known to influence lung development with FVC and FEV1/FVC. We split the dataset into two random subsets of 207,616 and 138,411 individuals, using the larger subset to select the most promising signals and the smaller subset for replication.Measurements and Main Results: We identified 55 genes, of which 36 (16 for FVC, 19 for FEV1/FVC, and one for both) had not been identified in the largest, most recent genome-wide study of lung function. Most of these 36 signals were intronic variants; expression data from blood and lung tissue showed that the majority affect the expression of the genes they lie within. Further testing of 34 of these 36 signals in the CHARGE and SpiroMeta consortia showed that 16 replicated after Bonferroni correction and another 12 replicated at nominal significance level. Of the 55 genes, 53 fell into four biological categories whose function is to regulate organ size and cell integrity (growth factors; transcriptional regulators; cell-to-cell adhesion; extracellular matrix), suggesting that these specific processes are important for adult lung health.Conclusions: Our study demonstrates the importance of lung development genes in regulating adult lung function and influencing both restrictive and obstructive patterns. Further investigation of these developmental pathways could lead to druggable targets.
Collapse
Affiliation(s)
- Laura Portas
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Miguel Pereira
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Congenica Ltd., Wellcome Genome Campus, Cambridge, United Kingdom
| | - Seif O Shaheen
- Institute of Population Health Sciences, Queen Mary University of London, London, United Kingdom
| | - Annah B Wyss
- Department of Health and Human Services, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| | - Stephanie J London
- Department of Health and Human Services, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| | - Peter G J Burney
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom; and
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Harwell Institute, Oxfordshire, United Kingdom
| | - Cosetta Minelli
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
19
|
Genome‑wide analysis of DNA methylation and gene expression changes in an ovalbumin‑induced asthma mouse model. Mol Med Rep 2020; 22:1709-1716. [PMID: 32705270 PMCID: PMC7411290 DOI: 10.3892/mmr.2020.11245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to establish an integrated network of DNA methylation and RNA expression in an ovalbumin (OVA)-induced asthma model, and to investigate the epigenetically-regulated genes involved in asthma development. Genome-wide CpG-DNA methylation profiling was conducted through the use of a methylated DNA immunoprecipitation microarray and RNA sequencing was performed using three lung samples from mice with OVA-induced asthma. A total of 35,401 differentially methylated regions (DMRs) were identified between mice with OVA-induced asthma and control mice. Of these, 3,060 were located in promoter regions and 370 of the genes containing these DMRs demonstrated an inverse correlation between methylation and gene expression. Kyoto Encyclopedia of Genes and Genomes pathway analysis identified that 368 genes were upregulated or downregulated in OVA-induced asthma samples, including genes involved in ‘chemokine signalling pathway’, ‘focal adhesion’, ‘leukocyte transendothelial migration’ and ‘vascular smooth muscle contraction signaling’ pathways. Integrated network analysis identified four hub genes, consisting of three upregulated genes [forkhead box O1 (FOXO1), SP1 transcription factor (SP1) and amyloid β precursor protein (APP)], and one downregulated gene [RUNX family transcription factor 1 (RUNX1)], all of which demonstrated an association between DNA methylation and gene expression. These genes were highly interconnected nodes in the Ingenuity Pathway Analysis module and were functionally significant. A total of four interconnected hub genes, FOXO1, RUNX1, SP1 and APP, were identified from the integrated DNA methylation and gene expression networks involved in asthma development. These results suggested that modulating these four genes could effectively control the development of asthma.
Collapse
|
20
|
Toppila-Salmi S, Luukkainen AT, Xu B, Lampi J, Auvinen J, Dhaygude K, Järvelin MR, Pekkanen J. Maternal smoking during pregnancy affects adult onset of asthma in offspring: a follow up from birth to age 46 years. Eur Respir J 2020; 55:13993003.01857-2019. [PMID: 32341110 DOI: 10.1183/13993003.01857-2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 03/04/2020] [Indexed: 01/17/2023]
Abstract
RATIONALE Environmental tobacco smoke (ETS) exposure increases asthma risk in children. There is limited knowledge of prenatal ETS for adult-onset asthma. OBJECTIVES To determine the association between prenatal ETS and adult onset asthma. MEASUREMENTS AND MAIN RESULTS The questionnaire and clinical data of 5200 people, free of physician-diagnosed asthma by 31 years of age, who were included in the Northern Finland Birth Cohort 1966 Study was used. The association of maternal smoking during the last 3 months of pregnancy with onset of physician-diagnosed asthma and with lung function in adult offspring was studied using adjusted multivariate regression analyses. The cumulative incidence of physician-diagnosed asthma between the ages of 31 and 46 years was 5.1% among men and 8.8% among women. Gestational smoke exposure was associated with adult-onset asthma among offspring (adjusted OR 1.54, 95% CI 1.04-2.29), namely among offspring who reported either past non-diagnosed asthma (OR 9.63, 95% CI 2.28-40.67) or past cough with wheeze (3.21, 95% CI 1.71-6.05). A significant association was detected between gestational smoke exposure and the offspring's forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) ratio at 31 years of age. In offspring with the haplotype rs11702779-AA of RUNX1, gestational smoke exposure was associated with adult-onset asthma (5.53, 95% CI 2.11-14.52, adjusted p-value for interaction 0.10). CONCLUSION Maternal smoking during pregnancy is associated with the cumulative incidence of asthma in offspring between the ages of 31 and 46 years. The association was accentuated in offspring who at age 31, reported having past respiratory problems and/or who had haplotype rs11702779-AA. A reduction in FEV1/FVC ratio was also observed at age 31 years in offspring with gestational smoke exposure. These results could reflect the early vulnerability of offspring's airways to ETS and its putative long-term effects.
Collapse
Affiliation(s)
- Sanna Toppila-Salmi
- Medicum, Haartman Institute, University of Helsinki, Helsinki, Finland .,Skin and Allergy Hospital, Helsinki University Hospital, Helsinki, Finland
| | | | - Baizhuang Xu
- Environment Health Unit, National Institute for Health and Welfare, Kuopio, Finland
| | - Jussi Lampi
- Environment Health Unit, National Institute for Health and Welfare, Kuopio, Finland
| | - Juha Auvinen
- Center for Life Course Health Research, Oulu, Finland
| | - Kishor Dhaygude
- Medicum, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Oulu, Finland.,Imperial College London, London, UK
| | - Juha Pekkanen
- Environment Health Unit, National Institute for Health and Welfare, Kuopio, Finland.,Dep of Public Health, University of Helsinki, Helsinki, Finland
| |
Collapse
|
21
|
Hammer B, Wagner C, Divac Rankov A, Reuter S, Bartel S, Hylkema MN, Krüger A, Svanes C, Krauss-Etschmann S. In utero exposure to cigarette smoke and effects across generations: A conference of animals on asthma. Clin Exp Allergy 2019; 48:1378-1390. [PMID: 30244507 DOI: 10.1111/cea.13283] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 07/24/2018] [Accepted: 09/01/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND The prevalence of asthma and chronic obstructive pulmonary disease (COPD) has risen markedly over the last decades and is reaching epidemic proportions. However, underlying molecular mechanisms are not fully understood, hampering the urgently needed development of approaches to prevent these diseases. It is well established from epidemiological studies that prenatal exposure to cigarette smoke is one of the main risk factors for aberrant lung function development or reduced fetal growth, but also for the development of asthma and possibly COPD later in life. Of note, recent evidence suggests that the disease risk can be transferred across generations, that is, from grandparents to their grandchildren. While initial studies in mouse models on in utero smoke exposure have provided important mechanistic insights, there are still knowledge gaps that need to be filled. OBJECTIVE Thus, in this review, we summarize current knowledge on this topic derived from mouse models, while also introducing two other relevant animal models: the fruit fly Drosophila melanogaster and the zebrafish Danio rerio. METHODS This review is based on an intensive review of PubMed-listed transgenerational animal studies from 1902 to 2018 and focuses in detail on selected literature due to space limitations. RESULTS This review gives a comprehensive overview of mechanistic insights obtained in studies with the three species, while highlighting the remaining knowledge gaps. We will further discuss potential (dis)advantages of all three animal models. CONCLUSION/CLINICAL RELEVANCE Many studies have already addressed transgenerational inheritance of disease risk in mouse, zebrafish or fly models. We here propose a novel strategy for how these three model organisms can be synergistically combined to achieve a more detailed understanding of in utero cigarette smoke-induced transgenerational inheritance of disease risk.
Collapse
Affiliation(s)
- Barbara Hammer
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany
| | - Christina Wagner
- Invertebrate Models, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Sabine Bartel
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany
| | - Machteld N Hylkema
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Arne Krüger
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany.,Institute for Life Science and Technology, Hanze University of Applied Sciences, Groningen, The Netherlands
| | - Cecilie Svanes
- Centre for International Health, University of Bergen, Bergen, Norway.,Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Borstel, Germany.,Institute for Experimental Medicine, Christian-Albrechts-Universitaet zu Kiel, Kiel, Germany
| |
Collapse
|
22
|
He C, Bai X, Li Y, Sun H, Kong X, Fu B, Chen L, Zhu K, Li P, Xu S. Runt-related transcription factor 1 contributes to lung cancer development by binding to tartrate-resistant acid phosphatase 5. Cell Cycle 2019; 18:3404-3419. [PMID: 31650885 DOI: 10.1080/15384101.2019.1678966] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lung cancer (LC) is one of the malignant tumors with growing morbidity and mortality. The involvement of runt-related transcription factor 1 (RUNX1) in LC patients has been elucidated. We intended to research mechanisms of RUNX1 and tartrate-resistant acid phosphatase 5 (ACP5) in LC. Firstly, ACP5 levels in LC tissues, paracancerous tissues, LC cells and tracheal epithelial cells were detected. RUNX1 overexpression plasmid and interference plasmid were constructed and transfected into 95C cells and A549 cells, respectively. The binding of RUNX1 to ACP5 promoter was tested. Additionally, the gain- and loss-of-function were performed to explore the effects of ACP5 and RUNX1 on LC biological process. The xenograft tumor in nude mice was constructed in vivo to verify in vitro results. Functional rescue experiment was performed by adding MAPK-specific activator P79350 to A549 cells with si-ACP5 to measure the effects of ERK/MAPK axis on LC progression. Consequently, we found ACP5 expression was higher in LC tissues and cells, and ACP5 silencing suppressed LC cell growth. Overexpression of ACP5 promoted malignant biological behavior of LC cells. RUNX1 could bind to ACP5 promoter, and overexpressed RUNX1 promoted ACP5 expression and LC cell growth. Moreover, ACP5 upregulated the ERK/MAPK axis and thus promoted LC progression. The results of xenograft tumor in nude mice showed that silencing ACP5 could inhibit the growth of LC cells in vivo. To conclude, silenced RUNX1 inhibits LC progression through the ERK/MAPK axis by binding to ACP5. This study may provide new approaches for LC treatment.
Collapse
Affiliation(s)
- Changjun He
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Xue Bai
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Yingbin Li
- Department of Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R.China
| | - Haobo Sun
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Xianglong Kong
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Bicheng Fu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Lantao Chen
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Kaibin Zhu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Pengju Li
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| | - Shidong Xu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
23
|
Kuniyoshi KM, Rehan VK. The impact of perinatal nicotine exposure on fetal lung development and subsequent respiratory morbidity. Birth Defects Res 2019; 111:1270-1283. [PMID: 31580538 DOI: 10.1002/bdr2.1595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/13/2019] [Accepted: 09/05/2019] [Indexed: 01/18/2023]
Abstract
Maternal smoking during pregnancy remains as a significant public health crisis as it did decades ago. Although its prevalence is decreasing in high-income countries, it has worsened globally, along with a concerning emergence of electronic-cigarette usage within the last two decades. Extensive epidemiologic and experimental evidence exists from both human and animal studies, demonstrating the detrimental long-term pulmonary outcomes in the offspring of mothers who smoke during pregnancy. Even secondhand and thirdhand smoke exposure to the developing lung might be as or even more harmful than firsthand smoke exposure. Furthermore, these effects are not limited only to the exposed progeny, but can also be transmitted transgenerationally. There is compelling evidence to support that the majority of the effects of perinatal smoke exposure on the developing lung, including the transgenerational transmission of asthma, is mediated by nicotine. Nicotine exposure induces cell-specific molecular changes in lungs, which offers a unique opportunity to prevent, halt, and/or reverse the resultant damage through targeted molecular interventions. Experimentally, the proposed interventions, such as administration of peroxisome proliferator-activated receptor gamma (PPARγ) agonists can not only block but also potentially reverse the perinatal nicotine exposure-induced respiratory morbidity in the exposed offspring. However, the development of a safe and effective intervention is still many years away. In the meantime, electropuncture at specific acupoints appears to be emerging as a more practical and safe physiologic approach to block the harmful pulmonary consequences of perinatal nicotine exposure.
Collapse
Affiliation(s)
- Katherine M Kuniyoshi
- Department of Pediatrics, David Geffen School of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor, UCLA Medical Center, Torrance, California
| | - Virender K Rehan
- Department of Pediatrics, David Geffen School of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor, UCLA Medical Center, Torrance, California
| |
Collapse
|
24
|
Ross KR, Teague WG, Gaston BM. Life Cycle of Childhood Asthma: Prenatal, Infancy and Preschool, Childhood, and Adolescence. Clin Chest Med 2018; 40:125-147. [PMID: 30691707 DOI: 10.1016/j.ccm.2018.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Asthma is a heterogeneous developmental disorder influenced by complex interactions between genetic susceptibility and exposures. Wheezing in infancy and early childhood is highly prevalent, with a substantial minority of children progressing to established asthma by school age, most of whom are atopic. Adolescence is a time of remission of symptoms with persistent lung function deficits. The transition to asthma in adulthood is not well understood.
Collapse
Affiliation(s)
- Kristie R Ross
- Division of Pediatric Pulmonology, Allergy, Immunology and Sleep Medicine, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, USA.
| | - W Gerald Teague
- Pediatric Asthma Center of Excellence, Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, Building MR4, Room 2112, PO Box 801349, Charlottesville, VA 22908, USA
| | - Benjamin M Gaston
- Division of Pediatric Pulmonology, Allergy, Immunology and Sleep Medicine, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Children's Lung Foundation, 2109 Adelbert Road, BRB 827, Cleveland, OH 44106, USA
| |
Collapse
|
25
|
Lappas M. Runt-related transcription factor 1 (RUNX1) deficiency attenuates inflammation-induced pro-inflammatory and pro-labour mediators in myometrium. Mol Cell Endocrinol 2018; 473:61-71. [PMID: 29330113 DOI: 10.1016/j.mce.2018.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/02/2018] [Accepted: 01/07/2018] [Indexed: 01/08/2023]
Abstract
Identifying new targets that regulate myometrial activation are required to develop effective treatments to stop preterm labor. Inflammation, which can be induced by sterile or infective insults, plays a role in initiating and maintaining uterine contractions. Several high throughput transcription screening studies have identified an upregulation of runt-related transcription factor 1 (RUNX1) mRNA expression in myometrium with labor. The role of RUNX1 in labor, however, is not known. We report increased RUNX1 during late gestation which was further augmented in labor, suggesting that RUNX1 may be involved in the transition of the myometrium from a quiescent into a contractile state in preparation for labor. By inhibiting the expression of RUNX1, we have established that RUNX1 induces the expression of pro-inflammatory cytokines, chemokines, adhesion molecules, contraction-associated proteins OXR and PTGFR, the uterotonic PGF2α, and the ECM remodelling enzyme MMP9. Targeting RUNX1 may be a novel approach to prevent preterm labor.
Collapse
Affiliation(s)
- Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia; Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| |
Collapse
|
26
|
Gao L, Liu X, Millstein J, Siegmund KD, Dubeau L, Maguire RL, (Jim) Zhang J, Fuemmeler BF, Kollins SH, Hoyo C, Murphy SK, Breton CV. Self-reported prenatal tobacco smoke exposure, AXL gene-body methylation, and childhood asthma phenotypes. Clin Epigenetics 2018; 10:98. [PMID: 30029617 PMCID: PMC6054742 DOI: 10.1186/s13148-018-0532-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Epigenetic modifications, including DNA methylation, act as one potential mechanism underlying the detrimental effects associated with prenatal tobacco smoke (PTS) exposure. Methylation in a gene called AXL was previously reported to differ in response to PTS. METHODS We investigated the association between PTS and epigenetic changes in AXL and how this was related to childhood asthma phenotypes. We tested the association between PTS and DNA methylation at multiple CpG loci of AXL at birth using Pyrosequencing in two separate study populations, the Children's Health Study (CHS, n = 799) and the Newborn Epigenetic Study (NEST, n = 592). Plasma cotinine concentration was used to validate findings with self-reported smoking status. The inter-relationships among AXL mRNA and miR-199a1 expression, PTS, and AXL methylation were examined. Lastly, we evaluated the joint effects of AXL methylation and PTS on the risk of asthma and related symptoms at age 10 years old. RESULTS PTS was associated with higher methylation level in the AXL gene body in both CHS and NEST subjects. In the pooled analysis, exposed subjects had a 0.51% higher methylation level in this region compared to unexposed subjects (95% CI 0.29, 0.74; p < 0.0001). PTS was also associated with 21.2% lower expression of miR-199a1 (95% CI - 37.9, - 0.1; p = 0.05), a microRNA known to regulate AXL expression. Furthermore, the combination of higher AXL methylation and PTS exposure at birth increased the risk of recent episodes of bronchitic symptoms in childhood. CONCLUSIONS PTS was associated with methylation level of AXL and the combination altered the risk of childhood bronchitic symptoms.
Collapse
Affiliation(s)
- Lu Gao
- Department of Preventive Medicine, USC Keck School of Medicine, 2001 N. Soto Street, Los Angeles, CA 90032 USA
| | - Xiaochen Liu
- Department of Preventive Medicine, USC Keck School of Medicine, 2001 N. Soto Street, Los Angeles, CA 90032 USA
| | - Joshua Millstein
- Department of Preventive Medicine, USC Keck School of Medicine, 2001 N. Soto Street, Los Angeles, CA 90032 USA
| | - Kimberly D. Siegmund
- Department of Preventive Medicine, USC Keck School of Medicine, 2001 N. Soto Street, Los Angeles, CA 90032 USA
| | - Louis Dubeau
- Department of Preventive Medicine, USC Keck School of Medicine, 2001 N. Soto Street, Los Angeles, CA 90032 USA
| | - Rachel L. Maguire
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695 USA
| | - Junfeng (Jim) Zhang
- Nicholas School of the Environment and Duke Global Health Institute, Duke University, Durham, NC 27701 USA
| | - Bernard F. Fuemmeler
- Department of Health Behavior and Policy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23219 USA
| | - Scott H. Kollins
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27705 USA
| | - Cathrine Hoyo
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695 USA
| | - Susan K. Murphy
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC 27708 USA
| | - Carrie V. Breton
- Department of Preventive Medicine, USC Keck School of Medicine, 2001 N. Soto Street, Los Angeles, CA 90032 USA
| |
Collapse
|
27
|
Chai J, Guo D, Ma W, Han D, Dong W, Guo H, Zhang Y. A feedback loop consisting of RUNX2/LncRNA-PVT1/miR-455 is involved in the progression of colorectal cancer. Am J Cancer Res 2018; 8:538-550. [PMID: 29637007 PMCID: PMC5883102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 11/14/2017] [Indexed: 06/08/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been shown to participate in cancer progression. In the present study, we explored the potential roles of lncRNA-PVT1 in the development process of colorectal cancer (CRC) via miR-455. We found that PVT1 is up-regulated in human CRC tissues compared to adjacent normal tissues. A functional study showed that the silencing of PVT1 expression by siRNAs inhibited cell proliferation, migration and invasion, whereas the overexpression of PVT1 accelerated cell proliferation, migration and invasion in vitro. A mechanistic study indicated PVT1 regulated the growth of CRC tumors by acting as a competing endogenous RNAs (ceRNA) and negatively regulated miR-455. Furthermore, we discovered that RUNX2, a functional transcription factor in CRC, up-regulated PVT1 expression. Therefore, our study suggested that the RUNX2/PVT1/miR-455 regulatory axis plays an important role in CRC tumorigenesis and may be a therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Jie Chai
- Department of General Surgery, Shandong University Affiliated Shandong Cancer Hospital and InstituteShandong Province, China
| | - Dawei Guo
- Shandong Academy of Medical SciencesShandong Province, China
| | - Wanli Ma
- Department of Orthopedics, The Second Hospital of Shandong UniversityShandong Province, China
| | - Dali Han
- Department of Radiation Oncology, Shandong University Affiliated Shandong Cancer Hospital and InstituteShandong Province, China
| | - Wei Dong
- Department of Radiation Oncology, Shandong University Affiliated Shandong Cancer Hospital and InstituteShandong Province, China
| | - Hongliang Guo
- Department of General Surgery, Shandong University Affiliated Shandong Cancer Hospital and InstituteShandong Province, China
| | - Yi Zhang
- Department of General Surgery, Shandong University Affiliated Shandong Cancer Hospital and InstituteShandong Province, China
| |
Collapse
|
28
|
Tang X, Sun L, Wang G, Chen B, Luo F. RUNX1: A Regulator of NF-kB Signaling in Pulmonary Diseases. Curr Protein Pept Sci 2018; 19:172-178. [PMID: 28990531 PMCID: PMC5876917 DOI: 10.2174/1389203718666171009111835] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 02/05/2023]
Abstract
Runt-related transcription factor 1 (RUNX1), a member of the RUNX family, is one of the key regulatory proteins in vertebrates. RUNX1 is involved in embryonic development, hematopoiesis, angiogenesis, tumorigenesis and immune response. In the past few decades, studies mainly focused on the effect of RUNX1 on acute leukemia and cancer. Only few studies about the function of RUNX1 in the pathological process of pulmonary diseases have been reported. Recent studies have demonstrated that RUNX1 is highly expressed in both mesenchymal and epithelial compartments of the developing and postnatal lung and that it plays a critical role in the lipopolysaccharide induced lung inflammation by regulating the NF-kB pathway. RUNX1 participates in the regulation of the NF-kB signaling pathway through interaction with IkB kinase complex in the cytoplasm or interaction with the NF-kB subunit P50. NF-kB is well-known signaling pathway necessary for inflammatory response in the lung. This review is to highlight the RUNX1 structure, isoforms and to present the mechanism that RUNX1 regulates NF-kB. This will illustrate the great potential role of RUNX1 in the inflammation signaling pathway in pulmonary diseases.
Collapse
Affiliation(s)
- Xiaoju Tang
- Department of Pulmonary and Critical Care Medicine
| | - Ling Sun
- Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Wang
- Department of Pulmonary and Critical Care Medicine
| | - Bojiang Chen
- Department of Pulmonary and Critical Care Medicine
| | - Fengming Luo
- Department of Pulmonary and Critical Care Medicine
| |
Collapse
|
29
|
Forno E, Wang T, Yan Q, Brehm J, Acosta-Perez E, Colon-Semidey A, Alvarez M, Boutaoui N, Cloutier MM, Alcorn JF, Canino G, Chen W, Celedón JC. A Multiomics Approach to Identify Genes Associated with Childhood Asthma Risk and Morbidity. Am J Respir Cell Mol Biol 2017; 57:439-447. [PMID: 28574721 DOI: 10.1165/rcmb.2017-0002oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Childhood asthma is a complex disease. In this study, we aim to identify genes associated with childhood asthma through a multiomics "vertical" approach that integrates multiple analytical steps using linear and logistic regression models. In a case-control study of childhood asthma in Puerto Ricans (n = 1,127), we used adjusted linear or logistic regression models to evaluate associations between several analytical steps of omics data, including genome-wide (GW) genotype data, GW methylation, GW expression profiling, cytokine levels, asthma-intermediate phenotypes, and asthma status. At each point, only the top genes/single-nucleotide polymorphisms/probes/cytokines were carried forward for subsequent analysis. In step 1, asthma modified the gene expression-protein level association for 1,645 genes; pathway analysis showed an enrichment of these genes in the cytokine signaling system (n = 269 genes). In steps 2-3, expression levels of 40 genes were associated with intermediate phenotypes (asthma onset age, forced expiratory volume in 1 second, exacerbations, eosinophil counts, and skin test reactivity); of those, methylation of seven genes was also associated with asthma. Of these seven candidate genes, IL5RA was also significant in analytical steps 4-8. We then measured plasma IL-5 receptor α levels, which were associated with asthma age of onset and moderate-severe exacerbations. In addition, in silico database analysis showed that several of our identified IL5RA single-nucleotide polymorphisms are associated with transcription factors related to asthma and atopy. This approach integrates several analytical steps and is able to identify biologically relevant asthma-related genes, such as IL5RA. It differs from other methods that rely on complex statistical models with various assumptions.
Collapse
Affiliation(s)
- Erick Forno
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ting Wang
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qi Yan
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Brehm
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Angel Colon-Semidey
- 3 Department of Pediatrics, University of Puerto Rico, San Juan, Puerto Rico; and
| | - Maria Alvarez
- 3 Department of Pediatrics, University of Puerto Rico, San Juan, Puerto Rico; and
| | - Nadia Boutaoui
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michelle M Cloutier
- 4 Department of Pediatrics, University of Connecticut Health Center, Connecticut Children's Medical Center, Farmington, Connecticut
| | - John F Alcorn
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Wei Chen
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Juan C Celedón
- 1 Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
30
|
Shorey-Kendrick LE, McEvoy CT, Ferguson B, Burchard J, Park BS, Gao L, Vuylsteke BH, Milner KF, Morris CD, Spindel ER. Vitamin C Prevents Offspring DNA Methylation Changes Associated with Maternal Smoking in Pregnancy. Am J Respir Crit Care Med 2017; 196:745-755. [PMID: 28422514 DOI: 10.1164/rccm.201610-2141oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
RATIONALE Infants whose mothers smoked during pregnancy demonstrate lifelong decreases in pulmonary function. DNA methylation changes associated with maternal smoking during pregnancy have been described in placenta and cord blood at delivery, in fetal lung, and in buccal epithelium and blood during childhood. We demonstrated in a randomized clinical trial ( ClinicalTrials.gov identifier, NCT00632476) that vitamin C supplementation to pregnant smokers can lessen the impact of maternal smoking on offspring pulmonary function and decrease the incidence of wheeze at 1 year of age. OBJECTIVES To determine whether vitamin C supplementation reduces changes in offspring methylation in response to maternal smoking and whether methylation at specific CpGs is also associated with respiratory outcomes. METHODS Targeted bisulfite sequencing was performed with a subset of placentas, cord blood samples, and buccal samples collected during the NCT00632476 trial followed by independent validation of selected cord blood differentially methylated regions, using bisulfite amplicon sequencing. MEASUREMENTS AND MAIN RESULTS The majority (69.03%) of CpGs with at least 10% methylation difference between placebo and nonsmoker groups were restored (by at least 50%) toward nonsmoker levels with vitamin C treatment. A significant proportion of restored CpGs were associated with phenotypic outcome with greater enrichment among hypomethylated CpGs. CONCLUSIONS We identified a pattern of normalization in DNA methylation by vitamin C supplementation across multiple loci. The consistency of this pattern across tissues and time suggests a systemic and persistent effect on offspring DNA methylation. Further work is necessary to determine how genome-wide changes in DNA methylation may mediate or reflect persistent effects of maternal smoking on lung function.
Collapse
Affiliation(s)
| | - Cindy T McEvoy
- 2 Oregon Health and Science University, Portland, Oregon
| | - Betsy Ferguson
- 1 Oregon National Primate Research Center, Beaverton, Oregon; and
| | - Julja Burchard
- 2 Oregon Health and Science University, Portland, Oregon
| | - Byung S Park
- 1 Oregon National Primate Research Center, Beaverton, Oregon; and.,2 Oregon Health and Science University, Portland, Oregon
| | - Lina Gao
- 1 Oregon National Primate Research Center, Beaverton, Oregon; and.,2 Oregon Health and Science University, Portland, Oregon
| | | | | | | | - Eliot R Spindel
- 1 Oregon National Primate Research Center, Beaverton, Oregon; and
| |
Collapse
|
31
|
Impact of resveratrol on bone repair in rats exposed to cigarette smoke inhalation: histomorphometric and bone-related gene expression analysis. Int J Oral Maxillofac Surg 2017; 47:541-548. [PMID: 28927744 DOI: 10.1016/j.ijom.2017.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 02/03/2023]
Abstract
This study investigated the effect of resveratrol on bone healing and its influence on the gene expression of bone-related markers in rats exposed to cigarette smoke. Two calvarial defects were created in each of 60 rats, which were assigned equally (n=20) to three groups: (1) resveratrol (10mg/kg)+smoke exposure (SMK+RESV); (2) placebo+smoke exposure (SMK+PLA); or (3) placebo+no smoke exposure (NS+PLA). Substances were administered daily for 30days following surgery. Smoke inhalation was started 7days before surgery and continued for 30days after surgery. One defect was processed for histomorphometric analysis and the other was used for mRNA quantification of bone-related gene expression by qPCR. The remaining defect was smaller in the SMK+RESV (2.27±0.61mm, P=0.0003) and NS+PLA (2.17±0.74mm, P=0.0005) groups than in the SMK+PLA group (3.12±0.47mm). Higher levels of Runx2 were observed in the NS+PLA group than in the smoke exposure groups (vs. SMK+PLA, P=0002; vs. SMK+RESV, P=0.052); levels of Lrp-5 were also higher in the no smoke exposure group (vs. SMK+RESV, P=0.009; vs. SMK+PLA, P=0.003). Resveratrol therapy decreased RANKL/OPG expression when compared to placebo (SMK+RESV vs. SMK+PLA, P=0.017). Dkk1 levels were decreased in the SMK+RESV group when compared to the SMK+PLA (P=0.006) and NS+PLA groups (P=0.005). In conclusion, resveratrol optimizes the repair of critical-sized bone defects, up-regulating the gene expression of important bone remodelling markers in rats exposed to cigarette smoke inhalation.
Collapse
|
32
|
Tang X, Sun L, Jin X, Chen Y, Zhu H, Liang Y, Wu Q, Han X, Liang J, Liu X, Liang Z, Wang G, Luo F. Runt-Related Transcription Factor 1 Regulates LPS-Induced Acute Lung Injury via NF-κB Signaling. Am J Respir Cell Mol Biol 2017; 57:174-183. [PMID: 28314106 DOI: 10.1165/rcmb.2016-0319oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Runt-related transcription factor 1 (RUNX1), a transcription factor expressed in multiple organs, plays important roles in embryonic development and hematopoiesis. Although RUNX1 is highly expressed in pulmonary tissues, its roles in lung function and homeostasis are unknown. We sought to assess the role of RUNX1 in lung development and inflammation after LPS challenge. Expression of RUNX1 was assessed in the developing and postnatal lung. RUNX1 was conditionally deleted in pulmonary epithelial cells. Pulmonary maturation was evaluated in the developing and postnatal lung, and lung inflammation was investigated in adult mice after LPS challenge. Interactions between RUNX1 and inflammatory signaling via NF-κB-IkB kinase β were assessed in vitro. RUNX1 was expressed in both mesenchymal and epithelial compartments of the developing and postnatal lung. The RUNX1 gene was efficiently deleted from respiratory epithelial cells producing Runx1∆/∆ mice. Although lung maturation was delayed, Runx1∆/∆ mice survived postnatally and subsequent growth and maturation of the lung proceeded normally. Increased respiratory distress, inflammation, and proinflammatory cytokines were observed in the Runx1-deleted mice after pulmonary LPS exposure. RUNX1 deletion was associated with the activation of NF-κB in respiratory epithelial cells. RUNX1 was required for the suppression of NF-κB signaling pathway via inhibition of IkB kinase β in in vitro studies. RUNX1 plays a critical role in the lung inflammation after LPS-induced injury.
Collapse
Affiliation(s)
- Xiaoju Tang
- 1 Department of Respiratory Medicine.,2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | - Ling Sun
- 2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | - Xiaodong Jin
- 3 Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hui Zhu
- 1 Department of Respiratory Medicine
| | - Yasha Liang
- 4 First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qingbo Wu
- 1 Department of Respiratory Medicine
| | - Xing Han
- 5 Fourth People's Hospital of Sichuan Province, Chengdu, China
| | - Jianing Liang
- 6 Department of Respiratory Medicine, Fourth Military Medical University, Xian, China
| | - Xiaojing Liu
- 2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | | | - Gang Wang
- 1 Department of Respiratory Medicine
| | | |
Collapse
|
33
|
Singh SP, Chand HS, Langley RJ, Mishra N, Barrett T, Rudolph K, Tellez C, Filipczak PT, Belinsky S, Saeed AI, Sheybani A, Exil V, Agarwal H, Sidhaye VK, Sussan T, Biswal S, Sopori M. Gestational Exposure to Sidestream (Secondhand) Cigarette Smoke Promotes Transgenerational Epigenetic Transmission of Exacerbated Allergic Asthma and Bronchopulmonary Dysplasia. THE JOURNAL OF IMMUNOLOGY 2017; 198:3815-3822. [PMID: 28381639 DOI: 10.4049/jimmunol.1700014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Embryonic development is highly sensitive to xenobiotic toxicity and in utero exposure to environmental toxins affects physiological responses of the progeny. In the United States, the prevalence of allergic asthma (AA) is inexplicably rising and in utero exposure to cigarette smoke increases the risk of AA and bronchopulmonary dysplasia (BPD) in children and animal models. We reported that gestational exposure to sidestream cigarette smoke (SS), or secondhand smoke, promoted nicotinic acetylcholine receptor-dependent exacerbation of AA and BPD in mice. Recently, perinatal nicotine injections in rats were reported to induce peroxisome proliferator-activated receptor γ-dependent transgenerational transmission of asthma. Herein, we show that first generation and second generation progeny from gestationally SS-exposed mice exhibit exacerbated AA and BPD that is not dependent on the decrease in peroxisome proliferator-activated receptor γ levels. Lungs from these mice show strong eosinophilic infiltration, excessive Th2 polarization, marked airway hyperresponsiveness, alveolar simplification, decreased lung compliance, and decreased lung angiogenesis. At the molecular level, these changes are associated with increased RUNX3 expression, alveolar cell apoptosis, and the antiangiogenic factor GAX, and decreased expression of HIF-1α and proangiogenic factors NF-κB and VEGFR2 in the 7-d first generation and second generation lungs. Moreover, the lungs from these mice exhibit lower levels of microRNA (miR)-130a and increased levels of miR-16 and miR-221. These miRs regulate HIF-1α-regulated apoptotic, angiogenic, and immune pathways. Thus the intergenerational effects of gestational SS involve epigenetic regulation of HIF-1α through specific miRs contributing to increased incidence of AA and BPD in the progenies.
Collapse
Affiliation(s)
- Shashi P Singh
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108
| | - Hitendra S Chand
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108.,Florida International University, Miami, FL 33199
| | - Raymond J Langley
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108.,University of Southern Alabama, Mobile, AL 36688
| | - Neerad Mishra
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108
| | - Ted Barrett
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108
| | - Karin Rudolph
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108
| | - Carmen Tellez
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108
| | | | - Steve Belinsky
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108
| | - Ali I Saeed
- Pulmonary and Critical Care Medicine, University of New Mexico Medical Center, Albuquerque, NM 87131
| | - Aryaz Sheybani
- Department of Pediatrics, University of New Mexico Medical Center, Albuquerque, NM 87131; and
| | - Vernat Exil
- Department of Pediatrics, University of New Mexico Medical Center, Albuquerque, NM 87131; and
| | - Hemant Agarwal
- Department of Pediatrics, University of New Mexico Medical Center, Albuquerque, NM 87131; and
| | | | - Thomas Sussan
- Environmental Health Sciences, Johns Hopkins University, Baltimore, MD 21205
| | - Shyam Biswal
- Environmental Health Sciences, Johns Hopkins University, Baltimore, MD 21205
| | - Mohan Sopori
- Lovelace Respiratory Research Institute, Albuquerque, NM 87108;
| |
Collapse
|
34
|
Delestrain C, Khen-Dunlop N, Hadchouel A, Cros P, Ducoin H, Fayon M, Gibertini I, Labbé A, Labouret G, Lebras MN, Lezmi G, Madhi F, Thouvenin G, Thumerelle C, Delacourt C. Respiratory Morbidity in Infants Born With a Congenital Lung Malformation. Pediatrics 2017; 139:peds.2016-2988. [PMID: 28202768 DOI: 10.1542/peds.2016-2988] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2016] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The actual frequency of respiratory symptoms related to congenital pulmonary malformations (CPMs) remains undetermined. The goal of this study was to prospectively evaluate the respiratory symptoms occurring in infants with prenatally diagnosed CPMs, identify factors associated with the occurrence of these symptoms, and evaluate their resolution after surgery. METHODS Infectious and noninfectious respiratory symptoms were prospectively collected in a French multicenter cohort of children with CPMs. RESULTS Eighty-five children were followed up to the mean age of 2.1 ± 0.4 years. Six children (7%) underwent surgery during the first 28 days of life. Of the 79 remaining children, 33 (42%) had respiratory symptoms during infancy before any surgery. Wheezing was the dominant symptom (24 of 79 [30%]), and only 1 infant had documented infection of the cystic lobe. Symptoms were more frequent in children with noncystic CPMs, prenatally (P = .01) or postnatally (P < .03), and with postnatally hyperlucent CPMs (P < .01). Sixty-six children underwent surgery during the follow-up period, and 40% of them displayed symptoms after the intervention. Six children had documented pneumonia during the postoperative period. At the end of the follow-up, pectus excavatum was observed in 10 children, significantly associated with thoracotomy (P < .02) or with surgery before the age of 6 months (P < .002). CONCLUSIONS CPMs are frequently associated with wheezing episodes. Surgery had no significant impact on these symptoms but was associated with a paradoxical increase in pulmonary infections, as well as an increased risk of pectus excavatum after thoracotomy.
Collapse
Affiliation(s)
| | - Naziha Khen-Dunlop
- Chirurgie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique-Hospitaux de Paris, Paris, France.,Université Paris-Descartes, Paris, France
| | - Alice Hadchouel
- Pneumologie Pédiatrique.,Université Paris-Descartes, Paris, France
| | - Pierrick Cros
- Pneumologie Pédiatrique, Centre Hospitalier Régional et Universitaire de Brest, Brest, France
| | - Héloïse Ducoin
- Pneumologie Pédiatrique, Centre Hospitalier de Lens, Lens, France
| | - Michael Fayon
- Pneumologie Pédiatrique, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Isabelle Gibertini
- Pneumologie Pédiatrique, Centre Hospitalier Universitaire de Tours, Tours, France
| | - André Labbé
- Pneumologie Pédiatrique, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Géraldine Labouret
- Pneumologie Pédiatrique, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Marie-Noëlle Lebras
- Pneumologie Pédiatrique, Hôpital Universitaire Robert Debré, Assistance Publique-Hospitaux de Paris, Paris, France
| | - Guillaume Lezmi
- Pneumologie Pédiatrique.,Université Paris-Descartes, Paris, France.,Centre de Référence des Maladies Respiratoires Rares, Paris, France
| | - Fouad Madhi
- Centre de Référence des Maladies Respiratoires Rares, Paris, France.,Pneumologie Pédiatrique, Centre Hospitalier Intercommunal Créteil,Créteil, France
| | - Guillaume Thouvenin
- Centre de Référence des Maladies Respiratoires Rares, Paris, France.,Pneumologie Pédiatrique, Hôpital Armand Trousseau, Assistance Publique Hospitaux de Paris, Paris, France; and
| | - Caroline Thumerelle
- Pneumologie Pédiatrique, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Christophe Delacourt
- Pneumologie Pédiatrique, .,Université Paris-Descartes, Paris, France.,Centre de Référence des Maladies Respiratoires Rares, Paris, France
| |
Collapse
|
35
|
England LJ, Aagaard K, Bloch M, Conway K, Cosgrove K, Grana R, Gould TJ, Hatsukami D, Jensen F, Kandel D, Lanphear B, Leslie F, Pauly JR, Neiderhiser J, Rubinstein M, Slotkin TA, Spindel E, Stroud L, Wakschlag L. Developmental toxicity of nicotine: A transdisciplinary synthesis and implications for emerging tobacco products. Neurosci Biobehav Rev 2017; 72:176-189. [PMID: 27890689 PMCID: PMC5965681 DOI: 10.1016/j.neubiorev.2016.11.013] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/18/2016] [Accepted: 11/19/2016] [Indexed: 12/24/2022]
Abstract
While the health risks associated with adult cigarette smoking have been well described, effects of nicotine exposure during periods of developmental vulnerability are often overlooked. Using MEDLINE and PubMed literature searches, books, reports and expert opinion, a transdisciplinary group of scientists reviewed human and animal research on the health effects of exposure to nicotine during pregnancy and adolescence. A synthesis of this research supports that nicotine contributes critically to adverse effects of gestational tobacco exposure, including reduced pulmonary function, auditory processing defects, impaired infant cardiorespiratory function, and may contribute to cognitive and behavioral deficits in later life. Nicotine exposure during adolescence is associated with deficits in working memory, attention, and auditory processing, as well as increased impulsivity and anxiety. Finally, recent animal studies suggest that nicotine has a priming effect that increases addiction liability for other drugs. The evidence that nicotine adversely affects fetal and adolescent development is sufficient to warrant public health measures to protect pregnant women, children, and adolescents from nicotine exposure.
Collapse
Affiliation(s)
- Lucinda J England
- Office on Smoking and Health, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Kjersti Aagaard
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Michele Bloch
- Division of Cancer Control and Population Science, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Kevin Conway
- Division of Epidemiology, Services and Prevention Research, National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, USA
| | - Kelly Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Rachel Grana
- Division of Cancer Control and Population Science, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Pennsylvania State University, PA, USA
| | | | - Frances Jensen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Denise Kandel
- Department of Psychiatry and Mailman School of Public Health, Columbia University, New York State Psychiatric Institute, New York, NY, USA
| | | | - Frances Leslie
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA, USA
| | - James R Pauly
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Jenae Neiderhiser
- Department of Psychology, Pennsylvania State University, University Park, PA, USA
| | - Mark Rubinstein
- Department of Pediatrics, School of Medicine, University of California, San Francisco, CA, USA
| | - Theodore A Slotkin
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Eliot Spindel
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Laura Stroud
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| | - Lauren Wakschlag
- Department of Medical Social Sciences Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
36
|
Shi N, Zhang J, Chen SY. Runx2, a novel regulator for goblet cell differentiation and asthma development. FASEB J 2016; 31:412-420. [PMID: 27825108 DOI: 10.1096/fj.201600954r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/28/2016] [Indexed: 01/25/2023]
Abstract
Airway goblet cell differentiation and related mucus overproduction are critical processes in the development of respiratory diseases, including asthma. The underlying mechanisms, however, are not fully understood. We identified Runt-related transcription factor 2 (Runx2) as a novel regulator for goblet cell differentiation. Runx2 was up-regulated by 6.4-fold during IL-13-induced goblet cell differentiation of human bronchial epithelial cells. Knockdown of Runx2 attenuated the IL-13-induced differentiation/mucus production by 67%. Mechanistically, Runx2 bound to the promoter of SAM-pointed domain-containing Ets-like factor (SPDEF), a known factor for goblet cell differentiation, resulting in an activation of SPDEF transcription. In vivo, Runx2 was induced by 6.2-fold in pulmonary epithelium of house dust mite-challenged mice. Blockade of Runx2 inhibited the house dust mite-induced goblet cell differentiation with a 75% reduction in mucus overproduction while improving airway responsiveness to methacholine by 41%. More importantly, a 12.3-fold increase in Runx2 expression was observed in human asthma lung epithelium, underlying the potential clinical importance of these findings.-Shi, N., Zhang, J., Chen, S.-Y. Runx2, a novel regulator for goblet cell differentiation and asthma development.
Collapse
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia, USA; and
| | - Jing Zhang
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia, USA; and.,Department of Respiratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shi-You Chen
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia, USA; and
| |
Collapse
|
37
|
Prakash YS. Emerging concepts in smooth muscle contributions to airway structure and function: implications for health and disease. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1113-L1140. [PMID: 27742732 DOI: 10.1152/ajplung.00370.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Airway structure and function are key aspects of normal lung development, growth, and aging, as well as of lung responses to the environment and the pathophysiology of important diseases such as asthma, chronic obstructive pulmonary disease, and fibrosis. In this regard, the contributions of airway smooth muscle (ASM) are both functional, in the context of airway contractility and relaxation, as well as synthetic, involving production and modulation of extracellular components, modulation of the local immune environment, cellular contribution to airway structure, and, finally, interactions with other airway cell types such as epithelium, fibroblasts, and nerves. These ASM contributions are now found to be critical in airway hyperresponsiveness and remodeling that occur in lung diseases. This review emphasizes established and recent discoveries that underline the central role of ASM and sets the stage for future research toward understanding how ASM plays a central role by being both upstream and downstream in the many interactive processes that determine airway structure and function in health and disease.
Collapse
Affiliation(s)
- Y S Prakash
- Departments of Anesthesiology, and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
38
|
Sulaiman I, Lim JCW, Soo HL, Stanslas J. Molecularly targeted therapies for asthma: Current development, challenges and potential clinical translation. Pulm Pharmacol Ther 2016; 40:52-68. [PMID: 27453494 DOI: 10.1016/j.pupt.2016.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/14/2016] [Accepted: 07/20/2016] [Indexed: 12/15/2022]
Abstract
Extensive research into the therapeutics of asthma has yielded numerous effective interventions over the past few decades. However, adverse effects and ineffectiveness of most of these medications especially in the management of steroid resistant severe asthma necessitate the development of better medications. Numerous drug targets with inherent airway smooth muscle tone modulatory role have been identified for asthma therapy. This article reviews the latest understanding of underlying molecular aetiology of asthma towards design and development of better antiasthma drugs. New drug candidates with their putative targets that have shown promising results in the preclinical and/or clinical trials are summarised. Examples of these interventions include restoration of Th1/Th2 balance by the use of newly developed immunomodulators such as toll-like receptor-9 activators (CYT003-QbG10 and QAX-935). Clinical trials revealed the safety and effectiveness of chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) antagonists such as OC0000459, BI-671800 and ARRY-502 in the restoration of Th1/Th2 balance. Regulation of cytokine activity by the use of newly developed biologics such as benralizumab, reslizumab, mepolizumab, lebrikizumab, tralokinumab, dupilumab and brodalumab are at the stage of clinical development. Transcription factors are potential targets for asthma therapy, for example SB010, a GATA-3 DNAzyme is at its early stage of clinical trial. Other candidates such as inhibitors of Rho kinases (Fasudil and Y-27632), phosphodiesterase inhibitors (GSK256066, CHF 6001, roflumilast, RPL 554) and proteinase of activated receptor-2 (ENMD-1068) are also discussed. Preclinical results of blockade of calcium sensing receptor by the use of calcilytics such as calcitriol abrogates cardinal signs of asthma. Nevertheless, successful translation of promising preclinical data into clinically viable interventions remains a major challenge to the development of novel anti-asthmatics.
Collapse
Affiliation(s)
- Ibrahim Sulaiman
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Jonathan Chee Woei Lim
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hon Liong Soo
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
39
|
Andrusaityte S, Grazuleviciene R, Kudzyte J, Bernotiene A, Dedele A, Nieuwenhuijsen MJ. Associations between neighbourhood greenness and asthma in preschool children in Kaunas, Lithuania: a case-control study. BMJ Open 2016; 6:e010341. [PMID: 27067890 PMCID: PMC4838715 DOI: 10.1136/bmjopen-2015-010341] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES The aim of this study was to investigate the associations between surrounding greenness levels and asthma among children, and to explore a possible change of this association by the distance of the residence to a city park. DESIGN A nested case-control study. SETTING Children aged 4-6 years residing at their current address since birth in Kaunas, Lithuania, whose mothers were recruited in 2007-2009 to the KANC newborns cohort study. PARTICIPANTS The participants were 1489 children whose parents in 2012-2013 filled in the questionnaires and agreed to participate in the study. PRIMARY AND SECONDARY OUTCOME MEASURES We estimated clinically diagnosed asthma risk factors. The surrounding greenness was measured as the average of the satellite-based Normalised Difference Vegetation Index (NDVI) within the buffers of 100, 300 and 500 m from each child's home address, and the distance to a city park was defined as the distance to the nearest city park. Multivariate logistic regression was performed to study the relationship between the greenness exposures and asthma adjusted for relevant covariates. RESULTS An increase in the NDVI (>median) in buffers of 100, 300 and 500 m was associated with a slightly increased risk of asthma, while an IQR increase in NDVI-100 m statistically significantly increased the risk of asthma (OR 1.43, 95% CI 1.10 to 1.85). The stratified analysis by surrounding greenness revealed indications of stronger associations for children with higher surrounding greenness (NDVI-100>median) and those living farther away from parks (>1000 m), compared to NDVI-100≤median and the distance to a city park >1000 m (OR 1.47, 95% CI 0.56 to 3.87). CONCLUSIONS A higher level of the surrounding greenness was associated with a slightly increased relative risk of asthma in children. Further investigation is needed to elucidate the influence of city parks and neighbourhood greenness levels on asthma.
Collapse
Affiliation(s)
- Sandra Andrusaityte
- Department of Environmental Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | | | - Jolanta Kudzyte
- Department of Paediatrics, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Asta Bernotiene
- Department of Paediatrics, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Audrius Dedele
- Department of Environmental Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | - Mark J Nieuwenhuijsen
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
40
|
Voon DCC, Hor YT, Ito Y. The RUNX complex: reaching beyond haematopoiesis into immunity. Immunology 2015; 146:523-36. [PMID: 26399680 DOI: 10.1111/imm.12535] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/24/2022] Open
Abstract
Among their diverse roles as transcriptional regulators during development and cell fate specification, the RUNX transcription factors are best known for the parts they play in haematopoiesis. RUNX proteins are expressed throughout all haematopoietic lineages, being necessary for the emergence of the first haematopoietic stem cells to their terminal differentiation. Although much progress has been made since their discoveries almost two decades ago, current appreciation of RUNX in haematopoiesis is largely grounded in their lineage-specifying roles. In contrast, the importance of RUNX to immunity has been mostly obscured for historic, technical and conceptual reasons. However, this paradigm is likely to shift over time, as a primary purpose of haematopoiesis is to resource the immune system. Furthermore, recent evidence suggests a role for RUNX in the innate immunity of non-haematopoietic cells. This review takes a haematopoiesis-centric approach to collate what is known of RUNX's contribution to the overall mammalian immune system and discuss their growing prominence in areas such as autoimmunity, inflammatory diseases and mucosal immunity.
Collapse
Affiliation(s)
- Dominic Chih-Cheng Voon
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan.,Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | - Yoshiaki Ito
- Cancer Biology Programme, Cancer Science Institute of Singapore, Singapore
| |
Collapse
|
41
|
Maccani JZ, Maccani MA. Altered placental DNA methylation patterns associated with maternal smoking: current perspectives. ADVANCES IN GENOMICS AND GENETICS 2015; 2015:205-214. [PMID: 26203295 PMCID: PMC4507353 DOI: 10.2147/agg.s61518] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The developmental origins of health and disease hypothesis states that adverse early life exposures can have lasting, detrimental effects on lifelong health. Exposure to maternal cigarette smoking during pregnancy is associated with morbidity and mortality in offspring, including increased risks for miscarriage, stillbirth, low birth weight, preterm birth, asthma, obesity, altered neurobehavior, and other conditions. Maternal cigarette smoking during pregnancy interferes with placental growth and functioning, and it has been proposed that this may occur through the disruption of normal and necessary placental epigenetic patterns. Epigenome-wide association studies have identified a number of differentially methylated placental genes that are associated with maternal smoking during pregnancy, including RUNX3, PURA, GTF2H2, GCA, GPR135, and HKR1. The placental methylation status of RUNX3 and NR3C1 has also been linked to adverse infant outcomes, including preterm birth and low birth weight, respectively. Candidate gene analyses have also found maternal smoking-associated placental methylation differences in the NR3C1, CYP1A1, HTR2A, and HSD11B2 genes, as well as in the repetitive elements LINE-1 and AluYb8. The differential methylation patterns of several genes have been confirmed to also exhibit altered gene expression patterns, including CYP1A1, CYP19A1, NR3C1, and HTR2A. Placental methylation patterns associated with maternal smoking during pregnancy may be largely gene-specific and tissue-specific and, to a lesser degree, involve global changes. It is important for future research to investigate the mechanistic roles that these differentially methylated genes may play in mediating the association between maternal smoking during pregnancy and disease in later life, as well as to elucidate the potential influence of emerging tobacco product use during pregnancy, including the use of electronic cigarettes, on placental epigenetics.
Collapse
Affiliation(s)
- Jennifer Zj Maccani
- Penn State Tobacco Center of Regulatory Science, College of Medicine, Department of Public Health Sciences, Hershey, PA, USA
| | - Matthew A Maccani
- Penn State Tobacco Center of Regulatory Science, College of Medicine, Department of Public Health Sciences, Hershey, PA, USA
| |
Collapse
|
42
|
Krauss-Etschmann S, Meyer KF, Dehmel S, Hylkema MN. Inter- and transgenerational epigenetic inheritance: evidence in asthma and COPD? Clin Epigenetics 2015; 7:53. [PMID: 26052354 PMCID: PMC4456695 DOI: 10.1186/s13148-015-0085-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/09/2015] [Indexed: 12/21/2022] Open
Abstract
Evidence is now emerging that early life environment can have lifelong effects on metabolic, cardiovascular, and pulmonary function in offspring, a concept also known as fetal or developmental programming. In mammals, developmental programming is thought to occur mainly via epigenetic mechanisms, which include DNA methylation, histone modifications, and expression of non-coding RNAs. The effects of developmental programming can be induced by the intrauterine environment, leading to intergenerational epigenetic effects from one generation to the next. Transgenerational epigenetic inheritance may be considered when developmental programming is transmitted across generations that were not exposed to the initial environment which triggered the change. So far, inter- and transgenerational programming has been mainly described for cardiovascular and metabolic disease risk. In this review, we discuss available evidence that epigenetic inheritance also occurs in respiratory diseases, using asthma and chronic obstructive pulmonary disease (COPD) as examples. While multiple epidemiological as well as animal studies demonstrate effects of 'toxic' intrauterine exposure on various asthma-related phenotypes in the offspring, only few studies link epigenetic marks to the observed phenotypes. As epigenetic marks may distinguish individuals most at risk of later disease at early age, it will enable early intervention strategies to reduce such risks. To achieve this goal further, well designed experimental and human studies are needed.
Collapse
Affiliation(s)
- Susanne Krauss-Etschmann
- />Comprehensive Pneumology Center, Helmholtz Center Munich and Children’s Hospital of Ludwig-Maximilians University, Max-Lebsche-Platz 31, 81377 Munich, Germany
- />Priority Area Asthma & Allergy, Leibniz Center for Medicine and Biosciences, Research Center Borstel and Christian Albrechts University Kiel, Airway Research Center North, Member of the German Center for Lung Research, Parkallee 1-40, Borstel, Germany
| | - Karolin F Meyer
- />Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
- />University of Groningen, GRIAC Research Institute, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
| | - Stefan Dehmel
- />Comprehensive Pneumology Center, Helmholtz Center Munich and Children’s Hospital of Ludwig-Maximilians University, Max-Lebsche-Platz 31, 81377 Munich, Germany
| | - Machteld N Hylkema
- />Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
- />University of Groningen, GRIAC Research Institute, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands
| |
Collapse
|
43
|
Ganguly K, Martin TM, Concel VJ, Upadhyay S, Bein K, Brant KA, George L, Mitra A, Thimraj TA, Fabisiak JP, Vuga LJ, Fattman C, Kaminski N, Schulz H, Leikauf GD. Secreted phosphoprotein 1 is a determinant of lung function development in mice. Am J Respir Cell Mol Biol 2015; 51:637-51. [PMID: 24816281 DOI: 10.1165/rcmb.2013-0471oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Secreted phosphoprotein 1 (Spp1) is located within quantitative trait loci associated with lung function that was previously identified by contrasting C3H/HeJ and JF1/Msf mouse strains that have extremely divergent lung function. JF1/Msf mice with diminished lung function had reduced lung SPP1 transcript and protein during the peak stage of alveologenesis (postnatal day [P]14-P28) as compared with C3H/HeJ mice. In addition to a previously identified genetic variant that altered runt-related transcription factor 2 (RUNX2) binding in the Spp1 promoter, we identified another promoter variant in a putative RUNX2 binding site that increased the DNA protein binding. SPP1 induced dose-dependent mouse lung epithelial-15 cell proliferation. Spp1((-/-)) mice have decreased specific total lung capacity/body weight, higher specific compliance, and increased mean airspace chord length (Lm) compared with Spp1((+/+)) mice. Microarray analysis revealed enriched gene ontogeny categories, with numerous genes associated with lung development and/or respiratory disease. Insulin-like growth factor 1, Hedgehog-interacting protein, wingless-related mouse mammary tumor virus integration site 5A, and NOTCH1 transcripts decreased in the lung of P14 Spp1((-/-)) mice as determined by quantitative RT-PCR analysis. SPP1 promotes pneumocyte growth, and mice lacking SPP1 have smaller, more compliant lungs with enlarged airspace (i.e., increased Lm). Microarray analysis suggests a dysregulation of key lung developmental transcripts in gene-targeted Spp1((-/-)) mice, particularly during the peak phase of alveologenesis. In addition to its known roles in lung disease, this study supports SPP1 as a determinant of lung development in mice.
Collapse
Affiliation(s)
- Koustav Ganguly
- 1 Department of Environmental and Occupational Health, Graduate School of Public Health
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
The impact of tobacco smoke exposure on wheezing and overweight in 4-6-year-old children. BIOMED RESEARCH INTERNATIONAL 2014; 2014:240757. [PMID: 25110663 PMCID: PMC4109218 DOI: 10.1155/2014/240757] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/08/2014] [Accepted: 06/24/2014] [Indexed: 11/17/2022]
Abstract
Aim. To investigate the association between maternal smoking during pregnancy, second-hand tobacco smoke (STS) exposure, education level, and preschool children's wheezing and overweight. Methods. This cohort study used data of the KANC cohort—1,489 4–6-year-old children from Kaunas city, Lithuania. Multivariate logistic regression was employed to study the influence of prenatal and postnatal STS exposure on the prevalence of wheezing and overweight, controlling for potential confounders. Results. Children exposed to maternal smoking during pregnancy had a slightly increased prevalence of wheezing and overweight. Postnatal exposure to STS was associated with a statistically significantly increased risk of wheezing and overweight in children born to mothers with lower education levels (OR 2.12; 95% CI 1.04–4.35 and 3.57; 95% CI 1.76–7.21, accordingly). Conclusions. The present study findings suggest that both maternal smoking during pregnancy and STS increase the risk of childhood wheezing and overweight, whereas lower maternal education might have a synergetic effect. Targeted interventions must to take this into account and address household smoking.
Collapse
|
45
|
Maccani JZJ, Koestler DC, Houseman EA, Marsit CJ, Kelsey KT. Placental DNA methylation alterations associated with maternal tobacco smoking at the RUNX3 gene are also associated with gestational age. Epigenomics 2013; 5:619-30. [PMID: 24283877 PMCID: PMC3982305 DOI: 10.2217/epi.13.63] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS The developmental origins of health and disease hypothesis states that later-life disease may be influenced by the quality of the in utero environment. Environmental toxicants can have detrimental effects on fetal development, potentially through effects on placental development and function. Maternal smoking during pregnancy is associated with low birth weight, preterm birth and other complications, and exposure to cigarette smoke in utero has been linked to gross pathologic and molecular changes to the placenta, including differential DNA methylation in placental tissue. The aim of this study was to investigate the relationship between maternal smoking during pregnancy, methylation changes in the placenta and gestational age. MATERIALS & METHODS We used Illumina(®)'s (CA, USA) Human Methylation27 BeadChip technology platform to investigate the methylation status of 21,551 autosomal, non-SNP-associated CpG loci in DNA extracted from 206 human placentas and examined loci whose variation in methylation was associated with maternal smoking during pregnancy. RESULTS We found that methylation patterns of a number of loci within the RUNX3 gene were significantly associated with smoking during pregnancy, and one of these loci was associated with decreased gestational age (p = 0.04). CONCLUSION Our findings, demonstrating maternal smoking-induced changes in DNA methylation at specific loci, suggest a mechanism by which in utero tobacco smoke exposure could exert its detrimental effects upon the health of the fetus.
Collapse
Affiliation(s)
- Jennifer ZJ Maccani
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, USA
| | - Devin C Koestler
- Section of Biostatistics & Epidemiology, Department of Community & Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | - Carmen J Marsit
- Section of Biostatistics & Epidemiology, Department of Community & Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Pharmacology & Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Karl T Kelsey
- Department of Pathology & Laboratory Medicine, Brown University, Providence, RI, USA
- Department of Epidemiology, Brown University, Providence, RI, USA
| |
Collapse
|
46
|
Prakash YS. Airway smooth muscle in airway reactivity and remodeling: what have we learned? Am J Physiol Lung Cell Mol Physiol 2013; 305:L912-33. [PMID: 24142517 PMCID: PMC3882535 DOI: 10.1152/ajplung.00259.2013] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 10/12/2013] [Indexed: 12/12/2022] Open
Abstract
It is now established that airway smooth muscle (ASM) has roles in determining airway structure and function, well beyond that as the major contractile element. Indeed, changes in ASM function are central to the manifestation of allergic, inflammatory, and fibrotic airway diseases in both children and adults, as well as to airway responses to local and environmental exposures. Emerging evidence points to novel signaling mechanisms within ASM cells of different species that serve to control diverse features, including 1) [Ca(2+)]i contractility and relaxation, 2) cell proliferation and apoptosis, 3) production and modulation of extracellular components, and 4) release of pro- vs. anti-inflammatory mediators and factors that regulate immunity as well as the function of other airway cell types, such as epithelium, fibroblasts, and nerves. These diverse effects of ASM "activity" result in modulation of bronchoconstriction vs. bronchodilation relevant to airway hyperresponsiveness, airway thickening, and fibrosis that influence compliance. This perspective highlights recent discoveries that reveal the central role of ASM in this regard and helps set the stage for future research toward understanding the pathways regulating ASM and, in turn, the influence of ASM on airway structure and function. Such exploration is key to development of novel therapeutic strategies that influence the pathophysiology of diseases such as asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis.
Collapse
Affiliation(s)
- Y S Prakash
- Dept. of Anesthesiology, Mayo Clinic, 4-184 W Jos SMH, 200 First St. SW, Rochester, MN 55905.
| |
Collapse
|
47
|
Madurga A, Mizíková I, Ruiz-Camp J, Morty RE. Recent advances in late lung development and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2013; 305:L893-905. [PMID: 24213917 DOI: 10.1152/ajplung.00267.2013] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In contrast to early lung development, a process exemplified by the branching of the developing airways, the later development of the immature lung remains very poorly understood. A key event in late lung development is secondary septation, in which secondary septa arise from primary septa, creating a greater number of alveoli of a smaller size, which dramatically expands the surface area over which gas exchange can take place. Secondary septation, together with architectural changes to the vascular structure of the lung that minimize the distance between the inspired air and the blood, are the objectives of late lung development. The process of late lung development is disturbed in bronchopulmonary dysplasia (BPD), a disease of prematurely born infants in which the structural development of the alveoli is blunted as a consequence of inflammation, volutrauma, and oxygen toxicity. This review aims to highlight notable recent developments in our understanding of late lung development and the pathogenesis of BPD.
Collapse
Affiliation(s)
- Alicia Madurga
- Dept. of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, D-61231 Bad Nauheim, Germany.
| | | | | | | |
Collapse
|
48
|
Voronov D, Gromova A, Liu D, Zoukhri D, Medvinsky A, Meech R, Makarenkova HP. Transcription factors Runx1 to 3 are expressed in the lacrimal gland epithelium and are involved in regulation of gland morphogenesis and regeneration. Invest Ophthalmol Vis Sci 2013; 54:3115-25. [PMID: 23532528 DOI: 10.1167/iovs.13-11791] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Lacrimal gland (LG) morphogenesis and repair are regulated by a complex interplay of intrinsic factors (e.g., transcription factors) and extrinsic signals (e.g., soluble growth/signaling factors). Many of these interconnections remain poorly characterized. Runt-related (Runx) factors belong to a small family of heterodimeric transcription factors known to regulate lineage-specific proliferation and differentiation of stem cells. The purpose of this study was to define the expression pattern and the role of Runx proteins in LG development and regeneration. METHODS Expression of epithelial-restricted transcription factors in murine LG was examined using immunostaining, qRT-PCR, and RT(2)Profiler PCR microarrays. The role of Runx transcription factors in LG morphogenesis was studied using siRNA and ex vivo LG cultures. Expression of Runx transcription factors during LG regeneration was assessed using in vivo model of LG regeneration. RESULTS We found that Runx factors are expressed in the epithelial compartment of the LG; in particular, Runx1 was restricted to the epithelium with highest level of expression in ductal and centroacinar cells. Downregulation of Runx1 to 3 expression using Runx-specific siRNAs abolished LG growth and branching and our data suggest that Runx1, 2, and 3 are partially redundant in LG development. In siRNA-treated LG, reduction of branching correlated with reduction of epithelial proliferation, as well as expression of cyclin D1 and the putative epithelial progenitor cell marker cytokeratin-5. Runx1, Runx3, and cytokeratin-5 expression increased significantly in regenerating LG and there was modest increase in Runx2 expression during LG differentiation. CONCLUSIONS Runx1 and 2 are new markers of the LG epithelial lineage and Runx factors are important for normal LG morphogenesis and regeneration.
Collapse
Affiliation(s)
- Dmitry Voronov
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | |
Collapse
|