1
|
Berdnikovs S, Newcomb DC, Hartert TV. How early life respiratory viral infections impact airway epithelial development and may lead to asthma. Front Pediatr 2024; 12:1441293. [PMID: 39156016 PMCID: PMC11327159 DOI: 10.3389/fped.2024.1441293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Childhood asthma is a common chronic disease of the airways that results from host and environment interactions. Most risk factor studies of asthma point to the first year of life as a susceptibility window of mucosal exposure that directly impacts the airway epithelium and airway epithelial cell development. The development of the airway epithelium, which forms a competent barrier resulting from coordinated interactions of different specialized cell subsets, occurs during a critical time frame in normal postnatal development in the first year of life. Understanding the normal and aberrant developmental trajectory of airway epithelial cells is important in identifying pathways that may contribute to barrier dysfunction and asthma pathogenesis. Respiratory viruses make first contact with and infect the airway mucosa. Human rhinovirus (HRV) and respiratory syncytial virus (RSV) are mucosal pathogens that are consistently identified as asthma risk factors. Respiratory viruses represent a unique early life exposure, different from passive irritant exposures which injure the developing airway epithelium. To replicate, respiratory viruses take over the host cell transcriptional and translational processes and exploit host cell energy metabolism. This takeover impacts the development and differentiation processes of airway epithelial cells. Therefore, delineating the mechanisms through which early life respiratory viral infections alter airway epithelial cell development will allow us to understand the maturation and heterogeneity of asthma and develop tools tailored to prevent disease in specific children. This review will summarize what is understood about the impact of early life respiratory viruses on the developing airway epithelium and define critical gaps in our knowledge.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Dawn C. Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tina V. Hartert
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
2
|
Kaneko S, Takasawa K, Asada K, Shiraishi K, Ikawa N, Machino H, Shinkai N, Matsuda M, Masuda M, Adachi S, Takahashi S, Kobayashi K, Kouno N, Bolatkan A, Komatsu M, Yamada M, Miyake M, Watanabe H, Tateishi A, Mizuno T, Okubo Y, Mukai M, Yoshida T, Yoshida Y, Horinouchi H, Watanabe SI, Ohe Y, Yatabe Y, Saloura V, Kohno T, Hamamoto R. Mechanism of ERBB2 gene overexpression by the formation of super-enhancer with genomic structural abnormalities in lung adenocarcinoma without clinically actionable genetic alterations. Mol Cancer 2024; 23:126. [PMID: 38862995 PMCID: PMC11165761 DOI: 10.1186/s12943-024-02035-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 05/30/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND In an extensive genomic analysis of lung adenocarcinomas (LUADs), driver mutations have been recognized as potential targets for molecular therapy. However, there remain cases where target genes are not identified. Super-enhancers and structural variants are frequently identified in several hundred loci per case. Despite this, most cancer research has approached the analysis of these data sets separately, without merging and comparing the data, and there are no examples of integrated analysis in LUAD. METHODS We performed an integrated analysis of super-enhancers and structural variants in a cohort of 174 LUAD cases that lacked clinically actionable genetic alterations. To achieve this, we conducted both WGS and H3K27Ac ChIP-seq analyses using samples with driver gene mutations and those without, allowing for a comprehensive investigation of the potential roles of super-enhancer in LUAD cases. RESULTS We demonstrate that most genes situated in these overlapped regions were associated with known and previously unknown driver genes and aberrant expression resulting from the formation of super-enhancers accompanied by genomic structural abnormalities. Hi-C and long-read sequencing data further corroborated this insight. When we employed CRISPR-Cas9 to induce structural abnormalities that mimicked cases with outlier ERBB2 gene expression, we observed an elevation in ERBB2 expression. These abnormalities are associated with a higher risk of recurrence after surgery, irrespective of the presence or absence of driver mutations. CONCLUSIONS Our findings suggest that aberrant gene expression linked to structural polymorphisms can significantly impact personalized cancer treatment by facilitating the identification of driver mutations and prognostic factors, contributing to a more comprehensive understanding of LUAD pathogenesis.
Collapse
Affiliation(s)
- Syuzo Kaneko
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan.
| | - Ken Takasawa
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Ken Asada
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Noriko Ikawa
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Hidenori Machino
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Norio Shinkai
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Maiko Matsuda
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Mari Masuda
- Department of Proteomics, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Shungo Adachi
- Department of Proteomics, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Satoshi Takahashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Kazuma Kobayashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Nobuji Kouno
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Amina Bolatkan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Masaaki Komatsu
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan
| | - Masayoshi Yamada
- Endoscopy Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Mototaka Miyake
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Hirokazu Watanabe
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Akiko Tateishi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Takaaki Mizuno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yu Okubo
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Masami Mukai
- Division of Medical Informatics, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yukihiro Yoshida
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Vassiliki Saloura
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, 103-0027, Japan.
| |
Collapse
|
3
|
The IRE1α-XBP1s Arm of the Unfolded Protein Response Activates N-Glycosylation to Remodel the Subepithelial Basement Membrane in Paramyxovirus Infection. Int J Mol Sci 2022; 23:ijms23169000. [PMID: 36012265 PMCID: PMC9408905 DOI: 10.3390/ijms23169000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes severe lower respiratory tract infections (LRTI) associated with decreased pulmonary function, asthma, and allergy. Recently, we demonstrated that RSV induces the hexosamine biosynthetic pathway via the unfolded protein response (UPR), which is a pathway controlling protein glycosylation and secretion of the extracellular matrix (ECM). Because the presence of matrix metalloproteinases and matricellular growth factors (TGF) is associated with severe LRTI, we studied the effect of RSV on ECM remodeling and found that RSV enhances the deposition of fibronectin-rich ECM by small airway epithelial cells in a manner highly dependent on the inositol requiring kinase (IRE1α)–XBP1 arm of the UPR. To understand this effect comprehensively, we applied pharmacoproteomics to understand the effect of the UPR on N-glycosylation and ECM secretion in RSV infection. We observe that RSV induces N-glycosylation and the secretion of proteins related to ECM organization, secretion, or proteins integral to plasma membranes, such as integrins, laminins, collagens, and ECM-modifying enzymes, in an IRE1α–XBP1 dependent manner. Using a murine paramyxovirus model that activates the UPR in vivo, we validate the IRE1α–XBP1-dependent secretion of ECM to alveolar space. This study extends understanding of the IRE1α–XBP1 pathway in regulating N-glycosylation coupled to structural remodeling of the epithelial basement membrane in RSV infection.
Collapse
|
4
|
RELA∙8-Oxoguanine DNA Glycosylase1 Is an Epigenetic Regulatory Complex Coordinating the Hexosamine Biosynthetic Pathway in RSV Infection. Cells 2022; 11:cells11142210. [PMID: 35883652 PMCID: PMC9319012 DOI: 10.3390/cells11142210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV), or human orthopneumovirus, is a negative-sense RNA virus that is the causative agent of severe lower respiratory tract infections in children and is associated with exacerbations of adult lung disease. The mechanisms how severe and/or repetitive virus infections cause declines in pulmonary capacity are not fully understood. We have recently discovered that viral replication triggers epithelial plasticity and metabolic reprogramming involving the hexosamine biosynthetic pathway (HBP). In this study, we examine the relationship between viral induced innate inflammation and the activation of hexosamine biosynthesis in small airway epithelial cells. We observe that RSV induces ~2-fold accumulation of intracellular UDP-GlcNAc, the end-product of the HBP and the obligate substrate of N glycosylation. Using two different silencing approaches, we observe that RSV replication activates the HBP pathway in a manner dependent on the RELA proto-oncogene (65 kDa subunit). To better understand the effect of RSV on the cellular N glycoproteome, and its RELA dependence, we conduct affinity enriched LC-MS profiling in wild-type and RELA-silenced cells. We find that RSV induces the accumulation of 171 N glycosylated peptides in a RELA-dependent manner; these proteins are functionally enriched in integrins and basal lamina formation. To elaborate this mechanism of HBP expression, we demonstrate that RSV infection coordinately induces the HBP pathway enzymes in a manner requiring RELA; these genes include Glutamine-Fructose-6-Phosphate Transaminase 1 (GFPT)-1/2, Glucosamine-Phosphate N-Acetyltransferase (GNPNAT)-1, phosphoglucomutase (PGM)-3 and UDP-N-Acetylglucosamine Pyrophosphorylase (UAP)-1. Using small-molecule inhibitor(s) of 8-oxoguanine DNA glycosylase1 (OGG1), we observe that OGG1 is also required for the expression of HBP pathway. In proximity ligation assays, RSV induces the formation of a nuclear and mitochondrial RELA∙OGG1 complex. In co-immunoprecipitaton (IP) experiments, we discover that RSV induces Ser 536-phosphorylated RELA to complex with OGG1. Chromatin IP experiments demonstrate a major role of OGG1 in supporting the recruitment of RELA and phosphorylated RNA Pol II to the HBP pathway genes. We conclude that the RELA∙OGG1 complex is an epigenetic regulator mediating metabolic reprogramming and N glycoprotein modifications of integrins in response to RSV. These findings have implications for viral-induced adaptive epithelial responses.
Collapse
|
5
|
Qiao D, Skibba M, Xu X, Garofalo RP, Zhao Y, Brasier AR. Paramyxovirus replication induces the hexosamine biosynthetic pathway and mesenchymal transition via the IRE1α-XBP1s arm of the unfolded protein response. Am J Physiol Lung Cell Mol Physiol 2021; 321:L576-L594. [PMID: 34318710 PMCID: PMC8461800 DOI: 10.1152/ajplung.00127.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The paramyxoviridae, respiratory syncytial virus (RSV), and murine respirovirus are enveloped, negative-sense RNA viruses that are the etiological agents of vertebrate lower respiratory tract infections (LRTIs). We observed that RSV infection in human small airway epithelial cells induced accumulation of glycosylated proteins within the endoplasmic reticulum (ER), increased glutamine-fructose-6-phosphate transaminases (GFPT1/2) and accumulation of uridine diphosphate (UDP)-N-acetylglucosamine, indicating activation of the hexosamine biosynthetic pathway (HBP). RSV infection induces rapid formation of spliced X-box binding protein 1 (XBP1s) and processing of activating transcription factor 6 (ATF6). Using pathway selective inhibitors and shRNA silencing, we find that the inositol-requiring enzyme (IRE1α)-XBP1 arm of the unfolded protein response (UPR) is required not only for activation of the HBP, but also for expression of mesenchymal transition (EMT) through the Snail family transcriptional repressor 1 (SNAI1), extracellular matrix (ECM)-remodeling proteins fibronectin (FN1), and matrix metalloproteinase 9 (MMP9). Probing RSV-induced open chromatin domains by ChIP, we find XBP1 binds and recruits RNA polymerase II to the IL6, SNAI1, and MMP9 promoters and the intragenic superenhancer of glutamine-fructose-6-phosphate transaminase 2 (GFPT2). The UPR is sustained through RSV by an autoregulatory loop where XBP1 enhances Pol II binding to its own promoter. Similarly, we investigated the effects of murine respirovirus infection on its natural host (mouse). Murine respirovirus induces mucosal growth factor response, EMT, and the indicators of ECM remodeling in an IRE1α-dependent manner, which persists after viral clearance. These data suggest that IRE1α-XBP1s arm of the UPR pathway is responsible for paramyxovirus-induced metabolic adaptation and mucosal remodeling via EMT and ECM secretion.
Collapse
Affiliation(s)
- Dianhua Qiao
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Melissa Skibba
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Xiaofang Xu
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Roberto P Garofalo
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Allan R Brasier
- Department of Internal Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.,Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
6
|
Du Y, Luan J, Jiang RP, Liu J, Ma Y. Myrcene exerts anti-asthmatic activity in neonatal rats via modulating the matrix remodeling. Int J Immunopathol Pharmacol 2021; 34:2058738420954948. [PMID: 32962470 PMCID: PMC7517990 DOI: 10.1177/2058738420954948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Myrcene (MC), an organic hydrocarbon, was found to exert anti-inflammatory, analgesic, antimutagenic and antioxidant properties. However, the protective role of MC has not been reported against neonatal asthma. Wistar rats induced with asthma were administered with MC; while asthma control and vehicle control were maintained without MC administration. At the end of the experimental period, lung histology, inflammatory cell counts, cytokine analysis, matrix protein expressions were elucidated. Rats administered with MC exerted significant (P < 0.05) defense in protecting the lung tissue with the evidenced restoration of alveolar thickening of the lung tissues. Also, the present study elicited the anti-asthmatic activity of MC, especially via modulating the extracellular matrix protein expression in the asthma-induced animals, while a significant reduction (P < 0.05) in the fibrotic markers were found in MC treated animals. Moreover, the protective effect of MC was evidenced with reduced leukocyte infiltration in BALF, hypersensitive specific IgE levels with a profound decrease in the inflammatory cytokines such as IL-2, IL-4, IL-18, and IL-21 in MC administered animals compared to the asthma-induced group. To an extent, the markers of asthmatic inflammation such as CD14, MCP-1, and TARC were also found to be attenuated in MC exposed animals. The possible application of MC is a promising drug for the treatment of asthma-mediated complications.
Collapse
Affiliation(s)
- Yanhui Du
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jie Luan
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ren Peng Jiang
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Juan Liu
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yan Ma
- Department of Pediatrics, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
7
|
Orr JC, Hynds RE. Stem Cell-derived Respiratory Epithelial Cell Cultures as Human Disease Models. Am J Respir Cell Mol Biol 2021; 64:657-668. [PMID: 33428856 PMCID: PMC8456877 DOI: 10.1165/rcmb.2020-0440tr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Advances in stem cell biology and the understanding of factors that determine lung stem cell self-renewal have enabled long-term in vitro culture of human lung cells derived from airway basal and alveolar type II cells. Improved capability to expand and study primary cells long term, including in clonal cultures that are recently derived from a single cell, will allow experiments that address fundamental questions about lung homeostasis and repair, as well as translational questions in asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and lung cancer research. Here, we provide a brief history of postnatal lung epithelial cell culture and describe recent methodological advances. We further discuss the applications of primary cultures in defining "normal" epithelium, in modeling lung disease, and in future cell therapies.
Collapse
Affiliation(s)
- Jessica C Orr
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, and
| | - Robert E Hynds
- UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
8
|
Brasier AR. Mechanisms how mucosal innate immunity affects progression of allergic airway disease. Expert Rev Respir Med 2019; 13:349-356. [PMID: 30712413 DOI: 10.1080/17476348.2019.1578211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Activation of antigen-independent inflammation (a.k.a. the 'innate' immune response (IIR)) plays a complex role in allergic asthma (AA). Although activation of the pulmonary IIR by aerosolized bacterial lipopolysaccharide early in life may be protective of AA, respiratory viral infections promote AA. The mechanisms how the mucosal IIR promotes allergic sensitization, remodeling, and altered epithelial signaling are not understood. Areas covered: This manuscript overviews: 1. Mechanistic studies identifying how allergens and viral patterns activate the mucosal IIR; 2. Research that reveals a major role played by specialized epithelial cells in the bronchiolar-alveolar junction in triggering inflammation and remodeling; 3. Reports linking the mucosal IIR with epithelial cell-state change and barrier disruption; and, 4. Observations relating mesenchymal transition with the expansion of the myofibroblast population. Expert commentary: Luminal allergens and viruses activate TLR signaling in key sentinel cells producing epithelial cell state transition, disrupting epithelial barrier function, and expanding the pulmonary myofibroblast population. These signals are transduced through a common NFκB/RelA -bromodomain containing four (BRD4) pathway, an epigenetic remodeling complex reprogramming the genome. Through this pathway, the mucosal IIR is a major modifier of adaptive immunity, AA and acute exacerbation-induced remodeling.
Collapse
Affiliation(s)
- Allan R Brasier
- a Institute for Clinical and Translational Research , University of Wisconsin-Madison School of Medicine and Public Health , Madison , WI , USA
| |
Collapse
|
9
|
Tian B, Liu Z, Litvinov J, Maroto R, Jamaluddin M, Rytting E, Patrikeev I, Ochoa L, Vargas G, Motamedi M, Ameredes BT, Zhou J, Brasier AR. Efficacy of Novel Highly Specific Bromodomain-Containing Protein 4 Inhibitors in Innate Inflammation-Driven Airway Remodeling. Am J Respir Cell Mol Biol 2019; 60:68-83. [PMID: 30153047 PMCID: PMC6348724 DOI: 10.1165/rcmb.2017-0445oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
NF-κB/RelA triggers innate inflammation by binding to bromodomain-containing protein 4 (BRD4), an atypical histone acetyltransferase (HAT). Although RelA·BRD4 HAT mediates acute neutrophilic inflammation, its role in chronic and functional airway remodeling is not known. We observed that BRD4 is required for Toll-like receptor 3 (TLR3)-mediated mesenchymal transition, a cell-state change that is characteristic of remodeling. We therefore tested two novel highly selective BRD4 inhibitors, ZL0420 and ZL0454, for their effects on chronic airway remodeling produced by repetitive TLR3 agonist challenges, and compared their efficacy with that of two nonselective bromodomain and extraterminal (BET) protein inhibitors, JQ1 and RVX208. We observed that ZL0420 and ZL0454 more potently reduced polyinosinic:polycytidylic acid-induced weight loss and fibrosis as assessed by microcomputed tomography and second harmonic generation microscopy. These measures correlated with the collagen deposition observed in histopathology. Importantly, the ZL inhibitors were more effective than the nonselective BET inhibitors at equivalent doses. The ZL inhibitors had significant effects on lung physiology, reversing TLR3-associated airway hyperresponsiveness and increasing lung compliance in vivo. At the molecular level, ZL inhibitors reduced elaboration of the transforming growth factor-β-induced growth program, thereby preventing mucosal mesenchymal transition and disrupting BRD4 HAT activity and complex formation with RelA. We also observed that ZL0454 treatment blocked polyinosinic:polycytidylic acid-associated expansion of the α-SMA1+/COL1A+ myofibroblast population and prevented myofibroblast transition in a coculture system. We conclude that 1) BRD4 is a central effector of the mesenchymal transition that results in paracrine activation of myofibroblasts, mechanistically linking innate inflammation to airway hyperresponsiveness and fibrosis, and 2) highly selective BRD4 inhibitors may be effective in reversing the effects of repetitive airway viral infections on innate inflammation-mediated remodeling.
Collapse
Affiliation(s)
- Bing Tian
- Department of Internal Medicine
- Sealy Center for Molecular Medicine
| | | | | | | | | | | | | | | | | | | | - Bill T. Ameredes
- Department of Internal Medicine
- Sealy Center for Molecular Medicine
- Department of Pharmacology and Toxicology
- Institute for Translational Sciences
- Sealy Center for Environmental Health and Medicine, University of Texas Medical Branch, Galveston, Texas; and
| | - Jia Zhou
- Sealy Center for Molecular Medicine
- Department of Pharmacology and Toxicology
| | - Allan R. Brasier
- School of Medicine and Public Health, University of Wisconsin–Madison, Madison, Wisconsin
| |
Collapse
|
10
|
Tian B, Hosoki K, Liu Z, Yang J, Zhao Y, Sun H, Zhou J, Rytting E, Kaphalia L, Calhoun WJ, Sur S, Brasier AR. Mucosal bromodomain-containing protein 4 mediates aeroallergen-induced inflammation and remodeling. J Allergy Clin Immunol 2018; 143:1380-1394.e9. [PMID: 30321559 DOI: 10.1016/j.jaci.2018.09.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 09/19/2018] [Accepted: 09/28/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Frequent exacerbations of allergic asthma lead to airway remodeling and a decrease in pulmonary function, producing morbidity. Cat dander is an aeroallergen associated with asthma risk. OBJECTIVE We sought to elucidate the mechanism of cat dander-induced inflammation-remodeling. METHODS We identified remodeling in mucosal samples from allergic asthma by using quantitative RT-PCR. We developed a model of aeroallergen-induced experimental asthma using repetitive cat dander extract exposure. We measured airway inflammation using immunofluorescence, leukocyte recruitment, and quantitative RT-PCR. Airway remodeling was measured by using histology, collagen content, myofibroblast numbers, and selected reaction monitoring. Inducible nuclear factor κB (NF-κB)-BRD4 interaction was measured by using a proximity ligation assay in situ. RESULTS Enhanced mesenchymal signatures are observed in bronchial biopsy specimens from patients with allergic asthma. Cat dander induces innate inflammation through NF-κB signaling, followed by production of a profibrogenic mesenchymal transition in primary human small airway epithelial cells. The IκB kinase-NF-κB signaling pathway is required for mucosal inflammation-coupled airway remodeling and myofibroblast expansion in the mouse model of aeroallergen exposure. Cat dander induces NF-κB/RelA to complex with and activate BRD4, resulting in modifying the chromatin environment of inflammatory and fibrogenic genes through its atypical histone acetyltransferase activity. A novel small-molecule BRD4 inhibitor (ZL0454) disrupts BRD4 binding to the NF-κB-RNA polymerase II complex and inhibits its histone acetyltransferase activity. ZL0454 prevents epithelial mesenchymal transition, myofibroblast expansion, IgE sensitization, and fibrosis in airways of naive mice exposed to cat dander. CONCLUSIONS NF-κB-inducible BRD4 activity mediates cat dander-induced inflammation and remodeling. Therapeutic modulation of the NF-κB-BRD4 pathway affects allergen-induced inflammation, epithelial cell-state changes, extracellular matrix production, and expansion of the subepithelial myofibroblast population.
Collapse
Affiliation(s)
- Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Koa Hosoki
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Zhiqing Liu
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Tex
| | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex
| | - Jia Zhou
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Tex; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex
| | - Erik Rytting
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Tex
| | - Lata Kaphalia
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex
| | - William J Calhoun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex
| | - Sanjiv Sur
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex
| | - Allan R Brasier
- Institute for Clinical and Translational Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wis.
| |
Collapse
|
11
|
Tian B, Widen SG, Yang J, Wood TG, Kudlicki A, Zhao Y, Brasier AR. The NFκB subunit RELA is a master transcriptional regulator of the committed epithelial-mesenchymal transition in airway epithelial cells. J Biol Chem 2018; 293:16528-16545. [PMID: 30166344 DOI: 10.1074/jbc.ra118.003662] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/20/2018] [Indexed: 12/14/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a multistep dedifferentiation program important in tissue repair. Here, we examined the role of the transcriptional regulator NF-κB in EMT of primary human small airway epithelial cells (hSAECs). Surprisingly, transforming growth factor β (TGFβ) activated NF-κB/RELA proto-oncogene, NF-κB subunit (RELA) translocation within 1 day of stimulation, yet induction of its downstream gene regulatory network occurred only after 3 days. A time course of TGFβ-induced EMT transition was analyzed by RNA-Seq in the absence or presence of inducible shRNA-mediated silencing of RELA. In WT cells, TGFβ stimulation significantly affected the expression of 2,441 genes. Gene set enrichment analysis identified WNT, cadherin, and NF-κB signaling as the most prominent TGFβ-inducible pathways. By comparison, RELA controlled expression of 3,138 overlapping genes mapping to WNT, cadherin, and chemokine signaling pathways. Conducting upstream regulator analysis, we found that RELA controls six clusters of upstream transcription factors, many of which overlapped with a transcription factor topology map of EMT developed earlier. RELA triggered expression of three key EMT pathways: 1) the WNT/β-catenin morphogen pathway, 2) the JUN transcription factor, and 3) the Snail family transcriptional repressor 1 (SNAI1). RELA binding to target genes was confirmed by ChIP. Experiments independently validating WNT dependence on RELA were performed by silencing RELA via genome editing and indicated that TGFβ-induced WNT5B expression and downstream activation of the WNT target AXIN2 are RELA-dependent. We conclude that RELA is a master transcriptional regulator of EMT upstream of WNT morphogen, JUN, SNAI1-ZEB1, and interleukin-6 autocrine loops.
Collapse
Affiliation(s)
- Bing Tian
- From the Departments of Internal Medicine and.,Sealy Center for Molecular Medicine, and
| | - Steven G Widen
- Sealy Center for Molecular Medicine, and.,Biochemistry and Molecular Biology
| | - Jun Yang
- From the Departments of Internal Medicine and.,Sealy Center for Molecular Medicine, and
| | - Thomas G Wood
- Sealy Center for Molecular Medicine, and.,Biochemistry and Molecular Biology
| | - Andrzej Kudlicki
- Sealy Center for Molecular Medicine, and.,Biochemistry and Molecular Biology.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas 77555 and
| | - Yingxin Zhao
- From the Departments of Internal Medicine and.,Sealy Center for Molecular Medicine, and.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas 77555 and
| | - Allan R Brasier
- Institute for Clinical and Translational Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin 53705
| |
Collapse
|
12
|
Pu Q, Zhao Y, Sun Y, Huang T, Lin P, Zhou C, Qin S, Singh BB, Wu M. TRPC1 intensifies house dust mite-induced airway remodeling by facilitating epithelial-to-mesenchymal transition and STAT3/NF-κB signaling. FASEB J 2018; 33:1074-1085. [PMID: 30067380 DOI: 10.1096/fj.201801085r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Airway remodeling with progressive epithelial alterations in the respiratory tract is a severe consequence of asthma. Although dysfunctional signaling transduction is attributed to airway inflammation, the exact mechanism of airway remodeling remains largely unknown. TRPC1, a member of the transient receptor potential canonical Ca2+ channel family, possesses versatile functions but its role in airway remodeling remains undefined. Here, we show that ablation of TRPC1 in mice alleviates airway remodeling following house dust mite (HDM) challenge with decreases in mucus production, cytokine secretion, and collagen deposition. HDM challenge induces Ca2+ influx via the TRPC1 channel, resulting in increased levels of signal transducer and activator of transcription 3 (STAT3) and proinflammatory cytokines. In contrast, STAT3 expression was significantly decreased in TRPC1-/- mouse lungs compared with wild-type controls after HDM challenge. Mechanistically, STAT3 promotes epithelial-to-mesenchymal transition and increases mucin 5AC expression. Collectively, these findings identify TRPC1 as a modulator of HDM-induced airway remodeling via STAT3-mediated increase in mucus production, which provide new insight in our understanding of the molecular basis of airway remodeling, and identify novel therapeutic targets for intervention of severe chronic asthma.-Pu, Q., Zhao, Y., Sun, Y., Huang, T., Lin, P., Zhou, C., Qin, S., Singh, B. B., Wu, M. TRPC1 intensifies house dust mite-induced airway remodeling by facilitating epithelial-to-mesenchymal transition and STAT3/NF-κB signaling.
Collapse
Affiliation(s)
- Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyu Zhao
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| | - Yuyang Sun
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| | - Ting Huang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| | - Ping Lin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| | - Chuanmin Zhou
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| | - Shugang Qin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Brij B Singh
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA; and
| |
Collapse
|
13
|
Zhu X, Li Q, Hu G, Wang J, Hu Q, Liu Z, Wu G, Zhong Y. BMS‑345541 inhibits airway inflammation and epithelial‑mesenchymal transition in airway remodeling of asthmatic mice. Int J Mol Med 2018; 42:1998-2008. [PMID: 30015827 PMCID: PMC6108878 DOI: 10.3892/ijmm.2018.3762] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/28/2018] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the possible effects and regulatory mechanism of the inhibitor of nuclear factor‑κB kinase complex β subunit (IKKβ) inhibitor BMS‑345541 on airway inflammation, airway remodeling and epithelial‑mesenchymal transition (EMT) in an ovalbumin (OVA) exposure asthma model in mice. The asthma mouse model was generated by sensitization and challenge with OVA. BMS‑345541/dimethyl sulfoxide (DMSO) was administered perorally dairy in two therapeutic groups throughout the entire OVA challenge process. At 24 h following the last challenge, airway hyperresponsiveness (AHR) and airway inflammation were examined, and serum, bronchoalveolar lavage fluid (BALF) and lung samples were collected. Lung tissue was stained and assessed for pathological changes. The total number and classification of inflammatory cells in the BALF were examined. Levels of transforming growth factor β1 (TGFβ1) in the serum and BALF were measured using an enzyme‑linked immunosorbent assay. The differential expression of EMT regulators E‑cadherin and vimentin was detected by immunohistochemical staining, reverse transcription‑quantitative polymerase chain reaction analysis and western blot analysis. The results showed that OVA successfully induced allergic asthma. The asthmatic mice had AHR, airway inflammation, airway remodeling, a high expression of TGFβ1, and evidence of EMT. Following BMS‑345541 treatment, there was significant inhibition of pathophysiological signs, including increased pulmonary eosinophilia infiltration, mucus hypersecretion and AHR. Treatment with BMS‑345541 significantly reduced levels of TGFβ1. In addition, BMS‑345541 notably downregulated the expression of vimentin and increased the expression of E‑cadherin. These data suggested that the increased secretion of TGFβ1 induced by asthmatic inflammation can lead to EMT, and the IKKβ inhibitor BMS‑345541 may alter airway remodeling by preventing EMT in an OVA asthma model. Therefore, IKKβ inhibitors require investigation as potential asthma therapies.
Collapse
Affiliation(s)
- Xiaohua Zhu
- Graduate School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiugen Li
- Respiratory Department, Jiangxi People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Guozhu Hu
- Central Laboratory, Jiangxi People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Wang
- Respiratory Department, Jiangxi People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Qinghua Hu
- Central Laboratory, Jiangxi Children's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiqiang Liu
- Clinical Laboratory, Jiangxi Children's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Gang Wu
- Graduate School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ying Zhong
- Graduate School, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Zhu XH, Li QG, Wang J, Hu GZ, Liu ZQ, Hu QH, Wu G. [Mechanism of action of BET bromodomain inhibitor JQ1 in treating airway remodeling in asthmatic mice]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:1278-1284. [PMID: 29237530 PMCID: PMC7389801 DOI: 10.7499/j.issn.1008-8830.2017.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/07/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the molecular mechanism of action of BET bromodomain inhibitor JQ1 in treating airway remodeling in asthmatic mice. METHODS A total of 24 mice were randomly divided into control group, ovalbumin (OVA)-induced asthma group (OVA group), and JQ1 intervention group (JQ1+OVA group), with 8 mice in each group. OVA sensitization/challenge was performed to establish a mouse model of asthma. At 1 hour before challenge, the mice in the JQ1+OVA group were given intraperitoneal injection of JQ1 solution (50 μg/g). Bronchoalveolar lavage fluid (BALF) and lung tissue samples were collected at 24 hours after the last challenge, and the total number of cells and percentage of eosinophils in BALF were calculated. Pathological staining was performed to observe histopathological changes in lung tissue. RT-PCR and Western blot were used to measure the mRNA and protein expression of E-cadherin and vimentin during epithelial-mesenchymal transition (EMT). RESULTS Compared with the control group, the OVA group had marked infiltration of inflammatory cells in the airway, thickening of the airway wall, increased secretion of mucus, and increases in the total number of cells and percentage of eosinophils in BALF (P<0.01). Compared with the OVA group, the JQ1+OVA group had significantly alleviated airway inflammatory response and significant reductions in the total number of cells and percentage of eosinophils in BALF (P<0.01). Compared with the control group, the OVA group had significant reductions in the mRNA and protein expression of E-cadherin and significant increases in the mRNA and protein expression of vimentin (P<0.01); compared with the OVA group, the JQ1+OVA group had significant increases in the mRNA and protein expression of E-cadherin and significant reductions in the mRNA and protein expression of vimentin (P<0.01); there were no significant differences in these indices between the JQ1+OVA group and the control group (P>0.05). CONCLUSIONS Mice with OVA-induced asthma have airway remodeling during EMT. BET bromodomain inhibitor JQ1 can reduce airway inflammation, inhibit EMT, and alleviate airway remodeling, which provides a new direction for the treatment of asthma.
Collapse
Affiliation(s)
- Xiao-Hua Zhu
- School of Medicine, Nanchang University, Nanchang 330006, China.
| | | | | | | | | | | | | |
Collapse
|
15
|
Schleimer RP, Berdnikovs S. Etiology of epithelial barrier dysfunction in patients with type 2 inflammatory diseases. J Allergy Clin Immunol 2017; 139:1752-1761. [PMID: 28583447 DOI: 10.1016/j.jaci.2017.04.010] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/11/2022]
Abstract
Epithelial barriers of the skin, gastrointestinal tract, and airway serve common critical functions, such as maintaining a physical barrier against environmental insults and allergens and providing a tissue interface balancing the communication between the internal and external environments. We now understand that in patients with allergic disease, regardless of tissue location, the homeostatic balance of the epithelial barrier is skewed toward loss of differentiation, reduced junctional integrity, and impaired innate defense. Importantly, epithelial dysfunction characterized by these traits appears to pre-date atopy and development of allergic disease. Despite our growing appreciation of the centrality of barrier dysfunction in initiation of allergic disease, many important questions remain to be answered regarding mechanisms disrupting normal barrier function. Although our external environment (proteases, allergens, and injury) is classically thought of as a principal contributor to barrier disruption associated with allergic sensitization, there is a need to better understand contributions of the internal environment (hormones, diet, and circadian clock). Systemic drivers of disease, such as alterations of the endocrine system, metabolism, and aberrant control of developmental signaling, are emerging as new players in driving epithelial dysfunction and allergic predisposition at various barrier sites. Identifying such central mediators of epithelial dysfunction using both systems biology tools and causality-driven laboratory experimentation will be essential in building new strategic interventions to prevent or reverse the process of barrier loss in allergic patients.
Collapse
Affiliation(s)
- Robert P Schleimer
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Sergejs Berdnikovs
- Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
16
|
Tian B, Patrikeev I, Ochoa L, Vargas G, Belanger KK, Litvinov J, Boldogh I, Ameredes BT, Motamedi M, Brasier AR. NF-κB Mediates Mesenchymal Transition, Remodeling, and Pulmonary Fibrosis in Response to Chronic Inflammation by Viral RNA Patterns. Am J Respir Cell Mol Biol 2017; 56:506-520. [PMID: 27911568 DOI: 10.1165/rcmb.2016-0259oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Airway remodeling is resultant of a complex multicellular response associated with a progressive decline of pulmonary function in patients with chronic airway disease. Here, repeated infections with respiratory viruses are linked with airway remodeling through largely unknown mechanisms. Although acute activation of the Toll-like receptor (TLR) 3 pathway by extracellular polyinosinic:polycytidylic acid (poly[I:C]) induces innate signaling through the NF-κB transcription factor in normal human small airway epithelial cells, prolonged (repetitive or tonic) poly(I:C) stimulation produces chronic stress fiber formation, mesenchymal transition, and activation of a fibrotic program. Chronic poly(I:C) stimulation enhanced the expression of core mesenchymal regulators Snail family zinc finger 1, zinc finger E-box binding homeobox, mesenchymal intermediate filaments (vimentin), and extracellular matrix proteins (fibronectin-1), and collagen 1A. This mesenchymal transition was prevented by silencing expression of NF-κB/RelA or administration of a small-molecule inhibitor of the IκB kinase, BMS345541. Acute poly(I:C) exposure in vivo induced profound neutrophilic airway inflammation. When administered repetitively, poly(I:C) resulted in enhanced fibrosis observed by lung micro-computed tomography, second harmonic generation microscopy of optically cleared lung tissue, and by immunohistochemistry. Epithelial flattening, expansion of the epithelial mesenchymal trophic unit, and enhanced Snail family zinc finger 1 and fibronectin 1 expression in airway epithelium were also observed. Repetitive poly(I:C)-induced airway remodeling, fibrosis, and epithelial-mesenchymal transition was inhibited by BMS345541 administration. Based on this novel model of viral inflammation-induced remodeling, we conclude that NF-κB is a major controller of epithelial-mesenchymal transition and pulmonary fibrosis, a finding that has potentially important relevance to airway remodeling produced by repetitive viral infections.
Collapse
Affiliation(s)
- Bing Tian
- Departments of 1 Internal Medicine.,2 Sealy Center for Molecular Medicine
| | | | | | | | - KarryAnne K Belanger
- Departments of 1 Internal Medicine.,4 Department of Biochemistry and Molecular Biology, and
| | - Julia Litvinov
- Departments of 1 Internal Medicine.,5 Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Istvan Boldogh
- 2 Sealy Center for Molecular Medicine.,6 Institute for Translational Sciences.,5 Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Bill T Ameredes
- Departments of 1 Internal Medicine.,2 Sealy Center for Molecular Medicine.,6 Institute for Translational Sciences
| | | | - Allan R Brasier
- Departments of 1 Internal Medicine.,2 Sealy Center for Molecular Medicine.,6 Institute for Translational Sciences
| |
Collapse
|
17
|
Epigenetic silencing of IRF1 dysregulates type III interferon responses to respiratory virus infection in epithelial to mesenchymal transition. Nat Microbiol 2017; 2:17086. [PMID: 28581456 PMCID: PMC5501188 DOI: 10.1038/nmicrobiol.2017.86] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 04/25/2017] [Indexed: 12/11/2022]
Abstract
Chronic oxidative injury produced by airway disease triggers TGFβ-mediated epigenetic reprogramming known as the epithelial-mesenchymal transition (EMT). We observe that EMT silences protective mucosal interferon (IFN)-I/-III production associated with enhanced rhinovirus (RV) and respiratory syncytial virus(RSV) replication. Mesenchymal transitioned cells are defective in inducible interferon regulatory factor (IRF)1 expression by occluding RelA and IRF3 access to the promoter. IRF1 is necessary for expression of type III IFNs (IFNLs-1 and 2/3). Induced by the EMT, Zinc Finger E-Box Binding Homeobox 1 (ZEB1) binds and silences IRF1. Ectopic ZEB1 is sufficient for IRF1 silencing, whereas ZEB1 knockdown partially restores IRF1-IFNL upregulation. ZEB1 silences IRF1 through the catalytic activity of the Enhancer of Zeste 2 Polycomb Repressive Complex 2 Subunit (EZH2), forming repressive H3K27(me3) marks. We observe that IRF1 expression is mediated by ZEB1 de-repression; our study demonstrates how airway remodeling/fibrosis is associated with a defective mucosal antiviral response through ZEB1-initiated epigenetic silencing.
Collapse
|
18
|
Tian B, Yang J, Zhao Y, Ivanciuc T, Sun H, Garofalo RP, Brasier AR. BRD4 Couples NF-κB/RelA with Airway Inflammation and the IRF-RIG-I Amplification Loop in Respiratory Syncytial Virus Infection. J Virol 2017; 91:e00007-17. [PMID: 28077651 PMCID: PMC5331805 DOI: 10.1128/jvi.00007-17] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 01/09/2023] Open
Abstract
The airway mucosa expresses protective interferon (IFN) and inflammatory cytokines in response to respiratory syncytial virus (RSV) infection. In this study, we examine the role of bromodomain containing 4 (BRD4) in mediating this innate immune response in human small airway epithelial cells. We observe that RSV induces BRD4 to complex with NF-κB/RelA. BRD4 is functionally required for expression of the NF-κB-dependent inflammatory gene regulatory network (GRN), including the IFN response factor 1 (IRF1) and IRF7, which mediate a cross talk pathway for RIG-I upregulation. Mechanistically, BRD4 is required for cyclin-dependent kinase 9 (CDK9) recruitment and phospho-Ser 2 carboxy-terminal domain (CTD) RNA polymerase (Pol) II formation on the promoters of IRF1, IRF7, and RIG-I, producing their enhanced expression by transcriptional elongation. We also find that BRD4 independently regulates CDK9/phospho-Ser 2 CTD RNA Pol II recruitment to the IRF3-dependent IFN-stimulated genes (ISGs). In vivo, poly(I·C)-induced neutrophilia and mucosal chemokine production are blocked by a small-molecule BRD4 bromodomain inhibitor. Similarly, BRD4 inhibition reduces RSV-induced neutrophilia, mucosal CXC chemokine expression, activation of the IRF7-RIG-I autoamplification loop, mucosal IFN expression, and airway obstruction. RSV infection activates BRD4 acetyltransferase activity on histone H3 Lys (K) 122, demonstrating that RSV infection activates BRD4 in vivo These data validate BRD4 as a major effector of RSV-induced inflammation and disease. BRD4 is required for coupling NF-κB to expression of inflammatory genes and the IRF-RIG-I autoamplification pathway and independently facilitates antiviral ISG expression. BRD4 inhibition may be a strategy to reduce exuberant virus-induced mucosal airway inflammation.IMPORTANCE In the United States, 2.1 million children annually require medical attention for RSV infections. A first line of defense is the expression of the innate gene network by infected epithelial cells. Expression of the innate response requires the recruitment of transcriptional elongation factors to rapidly induce innate response genes through an unknown mechanism. We discovered that RSV infection induces a complex of bromodomain containing 4 (BRD4) with NF-κB and cyclin-dependent kinase 9 (CDK9). BRD4 is required for stable CDK9 binding, phospho-Ser 2 RNA Pol II formation, and histone acetyltransferase activity. Inhibition of BRD4 blocks Toll-like receptor 3 (TLR3)-dependent neutrophilia and RSV-induced inflammation, demonstrating its importance in the mucosal innate response in vivo Our study shows that BRD4 plays a central role in inflammation and activation of the IRF7-RIG-I amplification loop vital for mucosal interferon expression. BRD4 inhibition may be a strategy for modulating exuberant mucosal airway inflammation.
Collapse
Affiliation(s)
- Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Roberto P Garofalo
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
19
|
Zhao Y, Jamaluddin M, Zhang Y, Sun H, Ivanciuc T, Garofalo RP, Brasier AR. Systematic Analysis of Cell-Type Differences in the Epithelial Secretome Reveals Insights into the Pathogenesis of Respiratory Syncytial Virus-Induced Lower Respiratory Tract Infections. THE JOURNAL OF IMMUNOLOGY 2017; 198:3345-3364. [PMID: 28258195 DOI: 10.4049/jimmunol.1601291] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/23/2017] [Indexed: 11/19/2022]
Abstract
Lower respiratory tract infections from respiratory syncytial virus (RSV) are due, in part, to secreted signals from lower airway cells that modify the immune response and trigger airway remodeling. To understand this process, we applied an unbiased quantitative proteomics analysis of the RSV-induced epithelial secretory response in cells representative of the trachea versus small airway bronchiolar cells. A workflow was established using telomerase-immortalized human epithelial cells that revealed highly reproducible cell type-specific differences in secreted proteins and nanoparticles (exosomes). Approximately one third of secretome proteins are exosomal; the remainder are from lysosomal and vacuolar compartments. We applied this workflow to three independently derived primary human cultures from trachea versus bronchioles. A total of 577 differentially expressed proteins from control supernatants and 966 differentially expressed proteins from RSV-infected cell supernatants were identified at a 1% false discovery rate. Fifteen proteins unique to RSV-infected primary human cultures from trachea were regulated by epithelial-specific ets homologous factor. A total of 106 proteins unique to RSV-infected human small airway epithelial cells was regulated by the transcription factor NF-κB. In this latter group, we validated the differential expression of CCL20/macrophage-inducible protein 3α, thymic stromal lymphopoietin, and CCL3-like 1 because of their roles in Th2 polarization. CCL20/macrophage-inducible protein 3α was the most active mucin-inducing factor in the RSV-infected human small airway epithelial cell secretome and was differentially expressed in smaller airways in a mouse model of RSV infection. These studies provide insights into the complexity of innate responses and regional differences in the epithelial secretome participating in RSV lower respiratory tract infection-induced airway remodeling.
Collapse
Affiliation(s)
- Yingxin Zhao
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Mohammad Jamaluddin
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555
| | - Roberto P Garofalo
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555; and.,Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555
| | - Allan R Brasier
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555; .,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| |
Collapse
|
20
|
Vlahopoulos SA. Aberrant control of NF-κB in cancer permits transcriptional and phenotypic plasticity, to curtail dependence on host tissue: molecular mode. Cancer Biol Med 2017; 14:254-270. [PMID: 28884042 PMCID: PMC5570602 DOI: 10.20892/j.issn.2095-3941.2017.0029] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The role of the transcription factor NF-κB in shaping the cancer microenvironment is becoming increasingly clear. Inflammation alters the activity of enzymes that modulate NF-κB function, and causes extensive changes in genomic chromatin that ultimately drastically alter cell-specific gene expression. NF-κB regulates the expression of cytokines and adhesion factors that control interactions among adjacent cells. As such, NF-κB fine tunes tissue cellular composition, as well as tissues' interactions with the immune system. Therefore, NF-κB changes the cell response to hormones and to contact with neighboring cells. Activating NF-κB confers transcriptional and phenotypic plasticity to a cell and thereby enables profound local changes in tissue function and composition. Research suggests that the regulation of NF-κB target genes is specifically altered in cancer. Such alterations occur not only due to mutations of NF-κB regulatory proteins, but also because of changes in the activity of specific proteostatic modules and metabolic pathways. This article describes the molecular mode of NF-κB regulation with a few characteristic examples of target genes.
Collapse
Affiliation(s)
- Spiros A Vlahopoulos
- The First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Athens 11527, Greece
| |
Collapse
|
21
|
Tian B, Zhao Y, Sun H, Zhang Y, Yang J, Brasier AR. BRD4 mediates NF-κB-dependent epithelial-mesenchymal transition and pulmonary fibrosis via transcriptional elongation. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1183-L1201. [PMID: 27793799 DOI: 10.1152/ajplung.00224.2016] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/25/2016] [Indexed: 02/08/2023] Open
Abstract
Chronic epithelial injury triggers a TGF-β-mediated cellular transition from normal epithelium into a mesenchymal-like state that produces subepithelial fibrosis and airway remodeling. Here we examined how TGF-β induces the mesenchymal cell state and determined its mechanism. We observed that TGF-β stimulation activates an inflammatory gene program controlled by the NF-κB/RelA signaling pathway. In the mesenchymal state, NF-κB-dependent immediate-early genes accumulate euchromatin marks and processive RNA polymerase. This program of immediate-early genes is activated by enhanced expression, nuclear translocation, and activating phosphorylation of the NF-κB/RelA transcription factor on Ser276, mediated by a paracrine signal. Phospho-Ser276 RelA binds to the BRD4/CDK9 transcriptional elongation complex, activating the paused RNA Pol II by phosphorylation on Ser2 in its carboxy-terminal domain. RelA-initiated transcriptional elongation is required for expression of the core epithelial-mesenchymal transition transcriptional regulators SNAI1, TWIST1, and ZEB1 and mesenchymal genes. Finally, we observed that pharmacological inhibition of BRD4 can attenuate experimental lung fibrosis induced by repetitive TGF-β challenge in a mouse model. These data provide a detailed mechanism for how activated NF-κB and BRD4 control epithelial-mesenchymal transition initiation and transcriptional elongation in model airway epithelial cells in vitro and in a murine pulmonary fibrosis model in vivo. Our data validate BRD4 as an in vivo target for the treatment of pulmonary fibrosis associated with inflammation-coupled remodeling in chronic lung diseases.
Collapse
Affiliation(s)
- Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas; .,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas; and.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas; and.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
22
|
Tapia D, Ross BN, Kalita A, Kalita M, Hatcher CL, Muruato LA, Torres AG. From In silico Protein Epitope Density Prediction to Testing Escherichia coli O157:H7 Vaccine Candidates in a Murine Model of Colonization. Front Cell Infect Microbiol 2016; 6:94. [PMID: 27625996 PMCID: PMC5003871 DOI: 10.3389/fcimb.2016.00094] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/17/2016] [Indexed: 11/18/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a leading cause of foodborne illnesses worldwide and is a common serotype linked to hemorrhagic colitis and an important cause of hemolytic uremic syndrome (HUS). Treatment of EHEC O157:H7 infections is complicated, as antibiotics can exacerbate Shiga toxin (Stx) production and lead to more severe symptoms including HUS. To date, no vaccines have been approved for human use, exposing a void in both treatment and prevention of EHEC O157:H7 infections. Previously, our lab has shown success in identifying novel vaccine candidates via bio- and immunoinformatics approaches, which are capable of reducing bacterial colonization in an in vivo model of intestinal colonization. In this study, we further characterized 17 of the identified vaccine candidates at the bioinformatics level and evaluated the protective capacity of the top three candidates when administered as DNA vaccines in our murine model of EHEC O157:H7 colonization. Based on further immunoinformatic predictions, these vaccine candidates were expected to induce neutralizing antibodies in a Th2-skewed immunological response. Immunization of BALB/c mice with two of these candidates resulted in reduced bacterial colonization following EHEC O157:H7 challenge. Additionally, immune sera was shown to prevent bacterial adhesion in vitro to Caco-2 cells. Together, this study provides further validation of our immunoinformatic analyses and identifies promising vaccine candidates against EHEC O157:H7.
Collapse
Affiliation(s)
- Daniel Tapia
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Brittany N Ross
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Anjana Kalita
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Mridul Kalita
- Department of Internal Medicine, University of Texas Medical Branch Galveston, TX, USA
| | - Christopher L Hatcher
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Laura A Muruato
- Department of Microbiology and Immunology, University of Texas Medical BranchGalveston, TX, USA; Institute for Translational Sciences, University of Texas Medical BranchGalveston, TX, USA
| | - Alfredo G Torres
- Department of Microbiology and Immunology, University of Texas Medical BranchGalveston, TX, USA; Institute for Translational Sciences, University of Texas Medical BranchGalveston, TX, USA; Department of Pathology and Sealy Center for Vaccine Development University of Texas Medical BranchGalveston, TX, USA
| |
Collapse
|
23
|
Zhao Y, Tian B, Sadygov RG, Zhang Y, Brasier AR. Integrative proteomic analysis reveals reprograming tumor necrosis factor signaling in epithelial mesenchymal transition. J Proteomics 2016; 148:126-38. [PMID: 27461979 DOI: 10.1016/j.jprot.2016.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/29/2022]
Abstract
UNLABELLED The airway epithelium is a semi-impermeable barrier whose disruption by growth factor reprogramming is associated with chronic airway diseases of humans. Transforming growth factor beta (TGFβ)-induced epithelial mesenchymal transition (EMT) plays important roles in airway remodeling characteristic of idiopathic lung fibrosis, asthma and chronic obstructive pulmonary disease (COPD). Inflammation of the airways leads to airway injury and tumor necrosis factor alpha (TNFα) plays an important pro-inflammatory role. Little systematic information about the effects of EMT on TNFα signaling is available. Using an in vitro model of TGFβ-induced EMT in primary human small airway epithelial cells (hSAECs), we applied quantitative proteomics and phosphoprotein profiling to understand the molecular mechanism of EMT and the impact of EMT on innate inflammatory responses. We quantified 7925 proteins and 1348 phosphorylation sites by stable isotope labeling with iTRAQ technology. We found that cellular response to TNFα is cell state dependent and the relative TNFα response in mesenchymal state is highly compressed. Combined bioinformatics analyses of proteome and phosphoproteome indicate that the EMT state is associated with reprogramming of kinome, signaling cascade of upstream transcription regulators, phosphor-networks, and NF-κB dependent cell signaling. BIOLOGICAL SIGNIFICANCE Epithelial mesenchymal transition and inflammation have important implications for clinical and physiologic manifestations of chronic airway diseases such as severe asthma, COPD, and lung fibrosis. Little systematic information on the interplay between EMT and innate inflammation is available. This study combined quantitative proteomics and phosphorproteomics approach to obtain systems-level insight into the upstream transcription regulators involved in the TGFβ-induced EMT in primary human small airway epithelial cells and to elucidate how EMT impacts on the TNFα signaling pathways. The proteomics and phosphoproteomics analysis indicates that many signaling pathways involved in TGFβ-induced EMT and EMT has profound reprogramming effects on innate inflammation response.
Collapse
Affiliation(s)
- Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, United States; Institute for Translational Sciences, UTMB, Galveston, TX, United States; Sealy Center for Molecular Medicine, UTMB, Galveston, TX, United States.
| | - Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, United States; Institute for Translational Sciences, UTMB, Galveston, TX, United States; Sealy Center for Molecular Medicine, UTMB, Galveston, TX, United States
| | - Rovshan G Sadygov
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, United States; Department of Biochemistry & Molecular Biology, UTMB, Galveston, TX, United States
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, United States; Institute for Translational Sciences, UTMB, Galveston, TX, United States; Sealy Center for Molecular Medicine, UTMB, Galveston, TX, United States.
| |
Collapse
|
24
|
Kaulmann A, Legay S, Schneider YJ, Hoffmann L, Bohn T. Inflammation related responses of intestinal cells to plum and cabbage digesta with differential carotenoid and polyphenol profiles following simulated gastrointestinal digestion. Mol Nutr Food Res 2016; 60:992-1005. [DOI: 10.1002/mnfr.201500947] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Anouk Kaulmann
- Luxembourg Institute of Science and Technology - Environmental Research and Innovation Department; Belvaux Luxembourg
| | - Sylvain Legay
- Luxembourg Institute of Science and Technology - Environmental Research and Innovation Department; Belvaux Luxembourg
| | | | - Lucien Hoffmann
- Luxembourg Institute of Science and Technology - Environmental Research and Innovation Department; Belvaux Luxembourg
| | - Torsten Bohn
- Luxembourg Institute of Science and Technology - Environmental Research and Innovation Department; Belvaux Luxembourg
- Luxembourg Institute of Health - Population Health Department; Strassen Luxembourg
| |
Collapse
|
25
|
Liu Z, Li W, Lv J, Xie R, Huang H, Li Y, He Y, Jiang J, Chen B, Guo S, Chen L. Identification of potential COPD genes based on multi-omics data at the functional level. MOLECULAR BIOSYSTEMS 2016; 12:191-204. [PMID: 26575263 DOI: 10.1039/c5mb00577a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex disease, which involves dysfunctions in multi-omics. The changes in biological processes, such as adhesion junction, signaling transduction, transcriptional regulation, and cell proliferation, will lead to the occurrence of COPD. A novel systematic approach MMMG (Methylation-MicroRNA-MRNA-GO) was proposed to identify potential COPD genes by integrating function information with a methylation profile, a microRNA expression profile and an mRNA expression profile. 8 co-functional classes and 102 potential COPD genes were identified. These genes displayed a high performance in classifying COPD patients and normal samples, revealed COPD-related pathways, and have been confirmed to be associated with COPD by Matthews correlation coefficient (MCC)-values, literature, an independent data set, and pathways. The MMMG method that analyzed multi-omics data at the functional level could effectively identify potential COPD genes. These potential COPD genes would provide in-depth insights into understanding the complexity of COPD genome landscapes, improve the early diagnostics, and guide new efforts to develop therapeutics in the future.
Collapse
Affiliation(s)
- Zhe Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
8-Oxoguanine DNA glycosylase-1-driven DNA base excision repair: role in asthma pathogenesis. Curr Opin Allergy Clin Immunol 2015; 15:89-97. [PMID: 25486379 DOI: 10.1097/aci.0000000000000135] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW To provide both an overview and evidence of the potential cause of oxidative DNA base damage and repair signaling in chronic inflammation and histological changes associated with asthma. RECENT FINDINGS Asthma is initiated/maintained by immunological, genetic/epigenetic, and environmental factors. It is a world-wide health problem, as current therapies suppress symptoms rather than prevent/reverse the disease, largely due to gaps in understanding its molecular mechanisms. Inflammation, oxidative stress, and DNA damage are inseparable phenomena, but their molecular roles in asthma pathogenesis are unclear. It was found that among oxidatively modified DNA bases, 8-oxoguanine (8-oxoG) is one of the most abundant, and its levels in DNA and body fluids are considered a biomarker of ongoing asthmatic processes. Free 8-oxoG forms a complex with 8-oxoG DNA glycosylase-1 and activates RAS-family GTPases that induce gene expression to mobilize innate and adaptive immune systems, along with genes regulating airway hyperplasia, hyper-responsiveness, and lung remodeling in atopic and nonatopic asthma. SUMMARY DNA's integrity must be maintained to prevent mutation, so its continuous repair and downstream signaling 'fuel' chronic inflammatory processes in asthma and form the basic mechanism whose elucidation will allow the development of new drug targets for the prevention/reversal of lung diseases.
Collapse
|
27
|
Tian B, Li X, Kalita M, Widen SG, Yang J, Bhavnani SK, Dang B, Kudlicki A, Sinha M, Kong F, Wood TG, Luxon BA, Brasier AR. Analysis of the TGFβ-induced program in primary airway epithelial cells shows essential role of NF-κB/RelA signaling network in type II epithelial mesenchymal transition. BMC Genomics 2015; 16:529. [PMID: 26187636 PMCID: PMC4506436 DOI: 10.1186/s12864-015-1707-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 06/17/2015] [Indexed: 12/21/2022] Open
Abstract
Background The airway epithelial cell plays a central role in coordinating the pulmonary response to injury and inflammation. Here, transforming growth factor-β (TGFβ) activates gene expression programs to induce stem cell-like properties, inhibit expression of differentiated epithelial adhesion proteins and express mesenchymal contractile proteins. This process is known as epithelial mesenchymal transition (EMT); although much is known about the role of EMT in cellular metastasis in an oncogene-transformed cell, less is known about Type II EMT, that occurring in normal epithelial cells. In this study, we applied next generation sequencing (RNA-Seq) in primary human airway epithelial cells to understand the gene program controlling Type II EMT and how cytokine-induced inflammation modifies it. Results Generalized linear modeling was performed on a two-factor RNA-Seq experiment of 6 treatments of telomerase immortalized human small airway epithelial cells (3 replicates). Using a stringent cut-off, we identified 3,478 differentially expressed genes (DEGs) in response to EMT. Unbiased transcription factor enrichment analysis identified three clusters of EMT regulators, one including SMADs/TP63 and another NF-κB/RelA. Surprisingly, we also observed 527 of the EMT DEGs were also regulated by the TNF-NF-κB/RelA pathway. This Type II EMT program was compared to Type III EMT in TGFβ stimulated A549 alveolar lung cancer cells, revealing significant functional differences. Moreover, we observe that Type II EMT modifies the outcome of the TNF program, reducing IFN signaling and enhancing integrin signaling. We confirmed experimentally that TGFβ-induced the NF-κB/RelA pathway by observing a 2-fold change in NF-κB/RelA nuclear translocation. A small molecule IKK inhibitor blocked TGFβ-induced core transcription factor (SNAIL1, ZEB1 and Twist1) and mesenchymal gene (FN1 and VIM) expression. Conclusions These data indicate that NF-κB/RelA controls a SMAD-independent gene network whose regulation is required for initiation of Type II EMT. Type II EMT dramatically affects the induction and kinetics of TNF-dependent gene networks. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1707-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA. .,Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA.
| | - Xueling Li
- Institute for Translational Sciences, UTMB, Galveston, TX, USA. .,Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA.
| | - Mridul Kalita
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA.
| | - Steven G Widen
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA.
| | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA.
| | - Suresh K Bhavnani
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA. .,Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Institute for Translational Sciences, UTMB, Galveston, TX, USA.
| | - Bryant Dang
- Institute for Translational Sciences, UTMB, Galveston, TX, USA.
| | - Andrzej Kudlicki
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Institute for Translational Sciences, UTMB, Galveston, TX, USA. .,Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA.
| | - Mala Sinha
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA. .,Bioinformatics Program, UTMB, Galveston, TX, USA.
| | - Fanping Kong
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA. .,Bioinformatics Program, UTMB, Galveston, TX, USA.
| | - Thomas G Wood
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Institute for Translational Sciences, UTMB, Galveston, TX, USA. .,Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA.
| | - Bruce A Luxon
- Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Institute for Translational Sciences, UTMB, Galveston, TX, USA. .,Department of Biochemistry and Molecular Biology, UTMB, Galveston, TX, USA. .,Bioinformatics Program, UTMB, Galveston, TX, USA.
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA. .,Sealy Center for Molecular Medicine, UTMB, Galveston, TX, USA. .,Institute for Translational Sciences, UTMB, Galveston, TX, USA.
| |
Collapse
|
28
|
Wang Q, Wang Y, Zhang Y, Zhang Y, Xiao W. Involvement of urokinase in cigarette smoke extract-induced epithelial-mesenchymal transition in human small airway epithelial cells. J Transl Med 2015; 95:469-79. [PMID: 25706093 DOI: 10.1038/labinvest.2015.33] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/23/2014] [Accepted: 12/05/2014] [Indexed: 01/04/2023] Open
Abstract
Urokinase-type plasminogen activator (uPA) augments inflammation and tissue remodeling during lung injury and repair. The uPA expression in small airway epithelium of chronic obstructive pulmonary disease (COPD) increases. Epithelial-mesenchymal transition (EMT) is important in the small airway fibrosis of COPD. This study shows the uPA regulation in cigarette smoke extract (CSE)-induced EMT in human small airway epithelial cell lines (HSAEpiCs). uPA is overexpressed in the small airway epithelium of COPD patients and CSE-treated cell lines. Furthermore, uPA expression correlated with vimentin expression in the small airway epithelium of COPD patients. uPA inhibition blocks CSE-induced EMT by reversing E-cadherin and α-catenin expression and retarding the induction of N-cadherin and vimentin, resulting in reduction in migration. uPA overexpression in HSAEpiC cells also promotes EMT and migration. EMT is partly reversed in uPA-overexpressing HSAEpiC cells through the silencing expression of uPA receptor. In conclusion, this study provides new insights into the contribution of uPA upregulation to EMT associated with small airway remodeling in COPD.
Collapse
Affiliation(s)
- Qin Wang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Yunshan Wang
- 1] Department of Anatomy, Shandong University School of Medicine, Jinan, China [2] School of Ocean, Shandong University, Weihai, China
| | - Yi Zhang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Yuke Zhang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Wei Xiao
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
29
|
Desai P, Yang J, Tian B, Sun H, Kalita M, Ju H, Paulucci-Holthauzen A, Zhao Y, Brasier AR, Sadygov RG. Mixed-effects model of epithelial-mesenchymal transition reveals rewiring of signaling networks. Cell Signal 2015; 27:1413-25. [PMID: 25862520 DOI: 10.1016/j.cellsig.2015.03.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 03/19/2015] [Accepted: 03/21/2015] [Indexed: 11/17/2022]
Abstract
The type II epithelial-mesenchymal transition (EMT) produces airway fibrosis and remodeling, contributing to the severity of asthma and chronic obstructive pulmonary disease. While numerous studies have been done on the mechanisms of the transition itself, few studies have investigated the system effects of EMT on signaling networks. Here, we use mixed effects modeling to develop a computational model of phospho-protein signaling data that compares human small airway epithelial cells (hSAECs) with their EMT-transformed counterparts across a series of perturbations with 8 ligands and 5 inhibitors, revealing previously uncharacterized changes in signaling in the EMT state. Strong couplings between menadione, TNFα and TGFβ and their known phospho-substrates were revealed after mixed effects modeling. Interestingly, the overall phospho-protein response was attenuated in EMT, with loss of Mena and TNFα coupling to heat shock protein (HSP)-27. These differences persisted after correction for EMT-induced changes in phospho-protein substrate abundance. Construction of network topology maps showed significant changes between the two cellular states, including a linkage between glycogen synthase kinase (GSK)-3α and small body size/mothers against decapentaplegic (SMAD)2. The model also predicted a loss of p38 mitogen activated protein kinase (p38MAPK)-independent HSP27 signaling, which we experimentally validated. We further characterized the relationship between HSP27 and signal transducers and activators of transcription (STAT)3 signaling, and determined that loss of HSP27 following EMT is only partially responsible for the downregulation of STAT3. These rewired connections represent therapeutic targets that could potentially reverse EMT and restore a normal phenotype to the respiratory mucosa.
Collapse
Affiliation(s)
- Poonam Desai
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Mridul Kalita
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Hyunsu Ju
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, United States; Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX 77555, United States
| | | | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Rovshan G Sadygov
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, United States; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
30
|
Ijaz T, Pazdrak K, Kalita M, Konig R, Choudhary S, Tian B, Boldogh I, Brasier AR. Systems biology approaches to understanding Epithelial Mesenchymal Transition (EMT) in mucosal remodeling and signaling in asthma. World Allergy Organ J 2014; 7:13. [PMID: 24982697 PMCID: PMC4068075 DOI: 10.1186/1939-4551-7-13] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 05/16/2014] [Indexed: 11/10/2022] Open
Abstract
A pathological hallmark of asthma is chronic injury and repair, producing dysfunction of the epithelial barrier function. In this setting, increased oxidative stress, growth factor- and cytokine stimulation, together with extracellular matrix contact produces transcriptional reprogramming of the epithelial cell. This process results in epithelial-mesenchymal transition (EMT), a cellular state associated with loss of epithelial polarity, expression of mesenchymal markers, enhanced mobility and extracellular matrix remodeling. As a result, the cellular biology of the EMT state produces characteristic changes seen in severe, refractory asthma: myofibroblast expansion, epithelial trans-differentiation and subepithelial fibrosis. EMT also induces profound changes in epithelial responsiveness that affects innate immune signaling that may have impact on the adaptive immune response and effectiveness of glucocorticoid therapy in severe asthma. We discuss how this complex phenotype is beginning to be understood using systems biology-level approaches through perturbations coupled with high throughput profiling and computational modeling. Understanding the distinct changes induced by EMT at the systems level may provide translational strategies to reverse the altered signaling and physiology of refractory asthma.
Collapse
Affiliation(s)
- Talha Ijaz
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Konrad Pazdrak
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Sealy Center for Molecular Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Institute for Translational Sciences, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Mridul Kalita
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Rolf Konig
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Department of Microbiology and Immunology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Sanjeev Choudhary
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Sealy Center for Molecular Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Department of Microbiology and Immunology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Bing Tian
- Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Istvan Boldogh
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Sealy Center for Molecular Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Department of Microbiology and Immunology, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| | - Allan R Brasier
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Institute for Translational Sciences, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA.,Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston 77555-1060, Texas, USA
| |
Collapse
|
31
|
Ahn T, Park T. Pathway-driven discovery of rare mutational impact on cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:171892. [PMID: 24883302 PMCID: PMC4026869 DOI: 10.1155/2014/171892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/14/2014] [Indexed: 12/17/2022]
Abstract
Identifying driver mutation is important in understanding disease mechanism and future application of custom tailored therapeutic decision. Functional analysis of mutational impact usually focuses on the gene expression level of the mutated gene itself. However, complex regulatory network may cause differential gene expression among functional neighbors of the mutated gene. We suggest a new approach for discovering rare mutations that have real impact in the context of pathway; the philosophy of our method is iteratively combining rare mutations until no more mutations can be added under the condition that the combined mutational event can statistically discriminate pathway level mRNA expression between groups with and without mutational events. Breast cancer patients with somatic mutation and mRNA expression were analyzed by our approach. Our approach is shown to sensitively capture mutations that change pathway level mRNA expression, concurrently discovering important mutations previously reported in breast cancer such as TP53, PIK3CA, and RB1. In addition, out of 15,819 genes considered in breast cancer, our approach identified mutational events of 32 genes showing pathway level mRNA expression differences.
Collapse
Affiliation(s)
- TaeJin Ahn
- Interdisciplinary Program in Bioinformatics, Seoul National University, San 56-1, Shilim-dong, Kwanak-gu, Seoul 151-742, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Irwon-ro 81, Seoul 136-710, Republic of Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, San 56-1, Shilim-dong, Kwanak-gu, Seoul 151-742, Republic of Korea
- Department of Statistics, Seoul National University, San 56-1, Shilim-dong, Kwanak-gu, Seoul 151-742, Republic of Korea
| |
Collapse
|
32
|
Comparative genomics and immunoinformatics approach for the identification of vaccine candidates for enterohemorrhagic Escherichia coli O157:H7. Infect Immun 2014; 82:2016-26. [PMID: 24595137 DOI: 10.1128/iai.01437-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 strains are major human food-borne pathogens, responsible for bloody diarrhea and hemolytic-uremic syndrome worldwide. Thus far, there is no vaccine for humans against EHEC infections. In this study, a comparative genomics analysis was performed to identify EHEC-specific antigens useful as potential vaccines. The genes present in both EHEC EDL933 and Sakai strains but absent in nonpathogenic E. coli K-12 and HS strains were subjected to an in silico analysis to identify secreted or surface-expressed proteins. We obtained a total of 65 gene-encoding protein candidates, which were subjected to immunoinformatics analysis. Our criteria of selection aided in categorizing the candidates as high, medium, and low priority. Three members of each group were randomly selected and cloned into pVAX-1. Candidates were pooled accordingly to their priority group and tested for immunogenicity against EHEC O157:H7 using a murine model of gastrointestinal infection. The high-priority (HP) pool, containing genes encoding a Lom-like protein (pVAX-31), a putative pilin subunit (pVAX-12), and a fragment of the type III secretion structural protein EscC (pVAX-56.2), was able to induce the production of EHEC IgG and sIgA in sera and feces. HP candidate-immunized mice displayed elevated levels of Th2 cytokines and diminished cecum colonization after wild-type challenge. Individually tested HP vaccine candidates showed that pVAX-12 and pVAX-56.2 significantly induced Th2 cytokines and production of fecal EHEC sIgA, with pVAX-56.2 reducing EHEC cecum colonization. We describe here a bioinformatics approach able to identify novel vaccine candidates potentially useful for preventing EHEC O157:H7 infections.
Collapse
|
33
|
Geisen S, Barturen G, Alganza ÁM, Hackenberg M, Oliver JL. NGSmethDB: an updated genome resource for high quality, single-cytosine resolution methylomes. Nucleic Acids Res 2013; 42:D53-9. [PMID: 24271385 PMCID: PMC3964946 DOI: 10.1093/nar/gkt1202] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The updated release of ‘NGSmethDB’ (http://bioinfo2.ugr.es/NGSmethDB) is a repository for single-base whole-genome methylome maps for the best-assembled eukaryotic genomes. Short-read data sets from NGS bisulfite-sequencing projects of cell lines, fresh and pathological tissues are first pre-processed and aligned to the corresponding reference genome, and then the cytosine methylation levels are profiled. One major improvement is the application of a unique bioinformatics protocol to all data sets, thereby assuring the comparability of all values with each other. We implemented stringent quality controls to minimize important error sources, such as sequencing errors, bisulfite failures, clonal reads or single nucleotide variants (SNVs). This leads to reliable and high-quality methylomes, all obtained under uniform settings. Another significant improvement is the detection in parallel of SNVs, which might be crucial for many downstream analyses (e.g. SNVs and differential-methylation relationships). A next-generation methylation browser allows fast and smooth scrolling and zooming, thus speeding data download/upload, at the same time requiring fewer server resources. Several data mining tools allow the comparison/retrieval of methylation levels in different tissues or genome regions. NGSmethDB methylomes are also available as native tracks through a UCSC hub, which allows comparison with a wide range of third-party annotations, in particular phenotype or disease annotations.
Collapse
Affiliation(s)
- Stefanie Geisen
- Facultad de Ciencias, Departmento de Genética, Universidad de Granada, 18071-Granada, Spain and Laboratorio de Bioinformática, Instituto de Biotecnología, Centro de Investigación Biomédica, 18100-Granada, Spain
| | | | | | | | | |
Collapse
|