Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Burton F. Dickey is active.

Publication


Featured researches published by Burton F. Dickey.


The New England Journal of Medicine | 2010

Airway Mucus Function and Dysfunction

John V. Fahy; Burton F. Dickey

Pulmonary defense against environmental injury depends on airway mucus, which traps inhaled toxins that are then cleared from the lungs by ciliary beating and cough. This review examines the normal and abnormal formation and clearance of mucus, as well as the therapy of mucus dysfunction.


The New England Journal of Medicine | 2011

A common MUC5B promoter polymorphism and pulmonary fibrosis.

Max A. Seibold; Anastasia L. Wise; Marcy C. Speer; Mark P. Steele; Kevin K. Brown; James E. Loyd; Tasha E. Fingerlin; Weiming Zhang; Gunnar Gudmundsson; Steve D. Groshong; Christopher M. Evans; Stavros Garantziotis; Kenneth B. Adler; Burton F. Dickey; Roland M. du Bois; Ivana V. Yang; Aretha Herron; Dolly Kervitsky; Janet Talbert; Cheryl Markin; Joungjoa Park; Anne L. Crews; Susan Slifer; Scott S. Auerbach; Michelle G. Roy; Jia Lin; Corinne E. Hennessy; Marvin I. Schwarz; David A. Schwartz

BACKGROUND The mutations that have been implicated in pulmonary fibrosis account for only a small proportion of the population risk. METHODS Using a genomewide linkage scan, we detected linkage between idiopathic interstitial pneumonia and a 3.4-Mb region of chromosome 11p15 in 82 families. We then evaluated genetic variation in this region in gel-forming mucin genes expressed in the lung among 83 subjects with familial interstitial pneumonia, 492 subjects with idiopathic pulmonary fibrosis, and 322 controls. MUC5B expression was assessed in lung tissue. RESULTS Linkage and fine mapping were used to identify a region of interest on the p-terminus of chromosome 11 that included gel-forming mucin genes. The minor-allele of the single-nucleotide polymorphism (SNP) rs35705950, located 3 kb upstream of the MUC5B transcription start site, was present at a frequency of 34% among subjects with familial interstitial pneumonia, 38% among subjects with idiopathic pulmonary fibrosis, and 9% among controls (allelic association with familial interstitial pneumonia, P=1.2×10(-15); allelic association with idiopathic pulmonary fibrosis, P=2.5×10(-37)). The odds ratios for disease among subjects who were heterozygous and those who were homozygous for the minor allele of this SNP were 6.8 (95% confidence interval [CI], 3.9 to 12.0) and 20.8 (95% CI, 3.8 to 113.7), respectively, for familial interstitial pneumonia and 9.0 (95% CI, 6.2 to 13.1) and 21.8 (95% CI, 5.1 to 93.5), respectively, for idiopathic pulmonary fibrosis. MUC5B expression in the lung was 14.1 times as high in subjects who had idiopathic pulmonary fibrosis as in those who did not (P<0.001). The variant allele of rs35705950 was associated with up-regulation in MUC5B expression in the lung in unaffected subjects (expression was 37.4 times as high as in unaffected subjects homozygous for the wild-type allele, P<0.001). MUC5B protein was expressed in lesions of idiopathic pulmonary fibrosis. CONCLUSIONS A common polymorphism in the promoter of MUC5B is associated with familial interstitial pneumonia and idiopathic pulmonary fibrosis. Our findings suggest that dysregulated MUC5B expression in the lung may be involved in the pathogenesis of pulmonary fibrosis. (Funded by the National Heart, Lung, and Blood Institute and others.).


Nature | 2014

Muc5b is required for airway defence

Michelle G. Roy; Alessandra Livraghi-Butrico; Ashley A. Fletcher; Melissa M. McElwee; Scott E. Evans; Ryan M. Boerner; Samantha N. Alexander; Lindsey K. Bellinghausen; Alfred S. Song; Youlia Petrova; Michael J. Tuvim; Roberto Adachi; Irlanda Romo; Andrea S. Bordt; M. Gabriela Bowden; Joseph H. Sisson; Prescott G. Woodruff; David J. Thornton; Karine Rousseau; Maria Miguelina De La Garza; Seyed Javad Moghaddam; Harry Karmouty-Quintana; Michael R. Blackburn; Scott M. Drouin; C. William Davis; Kristy A. Terrell; Barbara R. Grubb; Wanda K. O'Neal; Sonia C. Flores; Adela Cota-Gomez

Respiratory surfaces are exposed to billions of particulates and pathogens daily. A protective mucus barrier traps and eliminates them through mucociliary clearance (MCC). However, excessive mucus contributes to transient respiratory infections and to the pathogenesis of numerous respiratory diseases. MUC5AC and MUC5B are evolutionarily conserved genes that encode structurally related mucin glycoproteins, the principal macromolecules in airway mucus. Genetic variants are linked to diverse lung diseases, but specific roles for MUC5AC and MUC5B in MCC, and the lasting effects of their inhibition, are unknown. Here we show that mouse Muc5b (but not Muc5ac) is required for MCC, for controlling infections in the airways and middle ear, and for maintaining immune homeostasis in mouse lungs, whereas Muc5ac is dispensable. Muc5b deficiency caused materials to accumulate in upper and lower airways. This defect led to chronic infection by multiple bacterial species, including Staphylococcus aureus, and to inflammation that failed to resolve normally. Apoptotic macrophages accumulated, phagocytosis was impaired, and interleukin-23 (IL-23) production was reduced in Muc5b−/− mice. By contrast, in mice that transgenically overexpress Muc5b, macrophage functions improved. Existing dogma defines mucous phenotypes in asthma and chronic obstructive pulmonary disease (COPD) as driven by increased MUC5AC, with MUC5B levels either unaffected or increased in expectorated sputum. However, in many patients, MUC5B production at airway surfaces decreases by as much as 90%. By distinguishing a specific role for Muc5b in MCC, and by determining its impact on bacterial infections and inflammation in mice, our results provide a refined framework for designing targeted therapies to control mucin secretion and restore MCC.


Journal of Experimental Medicine | 2011

Muc5ac: a critical component mediating the rejection of enteric nematodes.

Sumaira Z. Hasnain; Christopher M. Evans; Michelle G. Roy; Amanda L. Gallagher; Kristen N. Kindrachuk; Luke Barron; Burton F. Dickey; Mark S. Wilson; Thomas A. Wynn; Richard K. Grencis; David J. Thornton

The mucin Muc5ac is essential for the expulsion of Trichuris muris and other gut-dwelling nematodes.


Current Opinion in Pulmonary Medicine | 2009

Mucus hypersecretion in asthma: causes and effects

Christopher M. Evans; Kyubo Kim; Michael J. Tuvim; Burton F. Dickey

Purpose of review Airway mucus plugging has long been recognized as a principal cause of death in asthma. However, molecular mechanisms of mucin overproduction and secretion have not been understood until recently. These mechanisms are reviewed together with ongoing investigations relating them to lung pathophysiology. Recent findings Of the five secreted gel-forming mucins in mammals, only MUC5AC and MUC5B are produced in significant quantities in intrapulmonary airways. MUC5B is the principal gel-forming mucin at baseline in small airways of humans and mice, and therefore likely performs most homeostatic clearance functions. MUC5AC is the principal gel-forming mucin upregulated in airway inflammation and is under negative control by forkhead box a2 (Foxa2) and positive control by hypoxia inducible factor-1 (HIF-1). Mucin secretion is regulated separately from production, principally by extracellular triphosphate nucleotides that bind P2Y2 receptors on the lumenal surface of airway secretory cells, generating intracellular second messengers that activate the exocytic proteins, Munc13-2 and synaptotagmin-2. Summary Markedly upregulated production of MUC5AC together with stimulated secretion leads to airflow obstruction in asthma. As MUC5B appears to mediate homeostatic functions, it may be possible to selectively inhibit MUC5AC production without impairing airway function. The precise roles of mucin hypersecretion in asthma symptoms such as dyspnea and cough and in physiologic phenomena such as airway hyperresponsiveness remain to be defined.


The Journal of Neuroscience | 2006

Synaptotagmin-2 is essential for survival and contributes to Ca2+ triggering of neurotransmitter release in central and neuromuscular synapses.

Zhiping P. Pang; Ernestina Melicoff; Daniel Padgett; Yun Liu; Andrew Franklin Teich; Burton F. Dickey; Weichun Lin; Roberto Adachi; Thomas C. Südhof

Biochemical and genetic data suggest that synaptotagmin-2 functions as a Ca2+ sensor for fast neurotransmitter release in caudal brain regions, but animals and/or synapses lacking synaptotagmin-2 have not been examined. We have now generated mice in which the 5′ end of the synaptotagmin-2 gene was replaced by lacZ. Using β-galactosidase as a marker, we show that, consistent with previous studies, synaptotagmin-2 is widely expressed in spinal cord, brainstem, and cerebellum, but is additionally present in selected forebrain neurons, including most striatal neurons and some hypothalamic, cortical, and hippocampal neurons. Synaptotagmin-2-deficient mice were indistinguishable from wild-type littermates at birth, but subsequently developed severe motor dysfunction, and perished at ∼3 weeks of age. Electrophysiological studies in cultured striatal neurons revealed that the synaptotagmin-2 deletion slowed the kinetics of evoked neurotransmitter release without altering the total amount of release. In contrast, synaptotagmin-2-deficient neuromuscular junctions (NMJs) suffered from a large reduction in evoked release and changes in short-term synaptic plasticity. Furthermore, in mutant NMJs, the frequency of spontaneous miniature release events was increased both at rest and during stimulus trains. Viewed together, our results demonstrate that the synaptotagmin-2 deficiency causes a lethal impairment in synaptic transmission in selected synapses. This impairment, however, is less severe than that produced in forebrain neurons by deletion of synaptotagmin-1, presumably because at least in NMJs, synaptotagmin-1 is coexpressed with synaptotagmin-2, and both together mediate fast Ca2+-triggered release. Thus, synaptotagmin-2 is an essential synaptotagmin isoform that functions in concert with other synaptotagmins in the Ca2+ triggering of neurotransmitter release.


Annual Review of Physiology | 2010

Inducible Innate Resistance of Lung Epithelium to Infection

Scott E. Evans; Yi Xu; Michael J. Tuvim; Burton F. Dickey

Most studies of innate immunity have focused on leukocytes such as neutrophils, macrophages, and natural killer cells. However, epithelial cells play key roles in innate defenses that include providing a mechanical barrier to microbial entry, signaling to leukocytes, and directly killing pathogens. Importantly, all these defenses are highly inducible in response to the sensing of microbial and host products. In healthy lungs, the level of innate immune epithelial function is low at baseline. This is indicated by low levels of spontaneous microbial killing and cytokine release, reflecting low constitutive stimulation in the nearly sterile lower respiratory tract when mucociliary clearance mechanisms are functioning effectively. This contrasts with the colon, where bacteria are continuously present and epithelial cells are constitutively activated. Although the surface area of the lungs presents a large target for microbial invasion, activated lung epithelial cells that are closely apposed to deposited pathogens are ideally positioned for microbial killing.


Proceedings of the National Academy of Sciences of the United States of America | 2009

β2-Adrenoceptor signaling is required for the development of an asthma phenotype in a murine model

Long P. Nguyen; Rui Lin; Sergio Parra; Ozozoma Omoluabi; Nicola A. Hanania; Michael J. Tuvim; Brian J. Knoll; Burton F. Dickey; Richard A. Bond

Chronic regular use of β2-adrenoceptor (β2-AR) agonists in asthma is associated with a loss of disease control and increased risk of death. Conversely, we have found that administration of β2-AR inverse agonists results in attenuation of the asthma phenotype in an allergen-driven murine model. Besides antagonizing agonist-induced signaling and reducing signaling by empty receptors, β-AR inverse agonists can also activate signaling by novel pathways. To determine the mechanism of the β-AR inverse agonists, we compared the asthma phenotype in β2-AR-null and wild-type mice. Antigen challenge of β2-AR-null mice produced results similar to what was observed with chronic β2-AR inverse agonist treatment, namely, reductions in mucous metaplasia, airway hyperresponsiveness (AHR), and inflammatory cells in the lungs. These results indicate that the effects of β2-AR inverse agonists are caused by inhibition of β2-AR signaling rather than by the induction of novel signaling pathways. Chronic administration of alprenolol, a β-blocker without inverse agonist properties, did not attenuate the asthma phenotype, suggesting that it is signaling by empty receptors, rather than agonist-induced β2-AR signaling, that supports the asthma phenotype. In conclusion, our results demonstrate that, in a murine model of asthma, β2-AR signaling is required for the full development of three cardinal features of asthma: mucous metaplasia, AHR, and the presence of inflammatory cells in the lungs.


American Journal of Respiratory and Critical Care Medicine | 2008

Stimulation of Lung Innate Immunity Protects against Lethal Pneumococcal Pneumonia in Mice

Cecilia G. Clement; Scott E. Evans; Christopher M. Evans; David H. Hawke; Ryuji Kobayashi; Paul R. Reynolds; Seyed Javad Moghaddam; Brenton L. Scott; Ernestina Melicoff; Roberto Adachi; Burton F. Dickey; Michael J. Tuvim

RATIONALE The lungs are a common site of serious infection in both healthy and immunocompromised subjects, and the most likely route of delivery of a bioterror agent. Since the airway epithelium shows great structural plasticity in response to inflammatory stimuli, we hypothesized it might also show functional plasticity. OBJECTIVES To test the inducibility of lung defenses against bacterial challenge. METHODS Mice were treated with an aerosolized lysate of ultraviolet-killed nontypeable (unencapsulated) Haemophilus influenzae (NTHi), then challenged with a lethal dose of live Streptococcus pneumoniae (Spn) delivered by aerosol. MEASUREMENTS AND MAIN RESULTS Treatment with the NTHi lysate induced complete protection against challenge with a lethal dose of Spn if treatment preceded challenge by 4 to 24 hours. Lesser levels of protection occurred at shorter (83% at 2 h) and longer (83% at 48-72 h) intervals between treatment and challenge. There was also some protection when treatment was given 2 hours after challenge (survival increased from 14 to 57%), but not 24 hours after challenge. Protection did not depend on recruited neutrophils or resident mast cells and alveolar macrophages. Protection was specific to the airway route of infection, correlated in magnitude and time with rapid bacterial killing within the lungs, and was associated with increases of multiple antimicrobial polypeptides in lung lining fluid. CONCLUSIONS We infer that protection derives from stimulation of local innate immune mechanisms, and that activated lung epithelium is the most likely cellular effector of this response. Augmentation of innate antimicrobial defenses of the lungs might have therapeutic value.


American Journal of Respiratory Cell and Molecular Biology | 2009

Promotion of Lung Carcinogenesis by Chronic Obstructive Pulmonary Disease–Like Airway Inflammation in a K-ras–Induced Mouse Model

Seyed Javad Moghaddam; Huaiguang Li; Sung Nam Cho; Megan K. Dishop; Ignacio I. Wistuba; Lin Ji; Jonathan M. Kurie; Burton F. Dickey; Francesco J. DeMayo

Lung cancer is the leading cause of cancer deaths in the United States. In addition to genetic abnormalities induced by cigarette smoke, several epidemiologic studies have found that smokers with chronic obstructive pulmonary disease (COPD), an inflammatory disease of the lungs, have an increased risk of lung cancer (1.3- to 4.9-fold) compared to smokers without COPD. This suggests a link between chronic airway inflammation and lung carcinogenesis, independent of tobacco smoke exposure. We studied this association by assaying the inflammatory impact of products of nontypeable Haemophilus influenzae, which colonizes the airways of patients with COPD, on lung cancer promotion in mice with an activated K-ras mutation in their airway epithelium. Two new mouse models of lung cancer were generated by crossing mice harboring the LSL-K-ras(G12D) allele with mice containing Cre recombinase inserted into the Clara cell secretory protein (CCSP) locus, with or without the neomycin cassette excised (CCSP(Cre) and CCSP(Cre-Neo), respectively). Lung lesions in CCSP(Cre-Neo)/LSL-K-ras(G12D) and CCSP(Cre)/LSL-K-ras(G12D) mice appeared at 4 and 1 month of age, respectively, and were classified as epithelial hyperplasia of the bronchioles, adenoma, and adenocarcinoma. Weekly exposure of CCSP(Cre)/LSL-K-ras(G12D) mice to aerosolized nontypeable Haemophilus influenzae lysate from age 6-14 weeks resulted in neutrophil/macrophage/CD8 T-cell-associated COPD-like airway inflammation, a 3.2-fold increase in lung surface tumor number (156 +/- 9 versus 45 +/- 7), and an increase in total lung tumor burden. We conclude that COPD-like airway inflammation promotes lung carcinogenesis in a background of a G12D-activated K-ras allele in airway secretory cells.

Collaboration


Dive into the Burton F. Dickey's collaboration.

Top Co-Authors

Avatar

Michael J. Tuvim

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher M. Evans

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Scott E. Evans

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Seyed Javad Moghaddam

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Roberto Adachi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicola A. Hanania

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Seyedeh Golsar Mirabolfathinejad

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amir Sharafkhaneh

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge