Network


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

Hotspot


Dive into the research topics where William Jester is active.

Publication


Featured researches published by William Jester.


PLOS ONE | 2014

RNA-Seq Transcriptome Profiling Identifies CRISPLD2 as a Glucocorticoid Responsive Gene that Modulates Cytokine Function in Airway Smooth Muscle Cells

Blanca E. Himes; Xiaofeng Jiang; Peter J. Wagner; Ruoxi Hu; Qiyu Wang; Barbara J. Klanderman; Reid M. Whitaker; Qingling Duan; Jessica Lasky-Su; Christina Nikolos; William Jester; Martin Johnson; Reynold A. Panettieri; Kelan G. Tantisira; Scott T. Weiss; Quan Lu

Asthma is a chronic inflammatory respiratory disease that affects over 300 million people worldwide. Glucocorticoids are a mainstay therapy for asthma because they exert anti-inflammatory effects in multiple lung tissues, including the airway smooth muscle (ASM). However, the mechanism by which glucocorticoids suppress inflammation in ASM remains poorly understood. Using RNA-Seq, a high-throughput sequencing method, we characterized transcriptomic changes in four primary human ASM cell lines that were treated with dexamethasone—a potent synthetic glucocorticoid (1 µM for 18 hours). Based on a Benjamini-Hochberg corrected p-value <0.05, we identified 316 differentially expressed genes, including both well known (DUSP1, KLF15, PER1, TSC22D3) and less investigated (C7, CCDC69, CRISPLD2) glucocorticoid-responsive genes. CRISPLD2, which encodes a secreted protein previously implicated in lung development and endotoxin regulation, was found to have SNPs that were moderately associated with inhaled corticosteroid resistance and bronchodilator response among asthma patients in two previously conducted genome-wide association studies. Quantitative RT-PCR and Western blotting showed that dexamethasone treatment significantly increased CRISPLD2 mRNA and protein expression in ASM cells. CRISPLD2 expression was also induced by the inflammatory cytokine IL1β, and small interfering RNA-mediated knockdown of CRISPLD2 further increased IL1β-induced expression of IL6 and IL8. Our findings offer a comprehensive view of the effect of a glucocorticoid on the ASM transcriptome and identify CRISPLD2 as an asthma pharmacogenetics candidate gene that regulates anti-inflammatory effects of glucocorticoids in the ASM.


American Journal of Respiratory Cell and Molecular Biology | 2012

Trichostatin A Abrogates Airway Constriction, but Not Inflammation, in Murine and Human Asthma Models

Audreesh Banerjee; Chinmay M. Trivedi; Gautam Damera; Meiqi Jiang; William Jester; Toshinori Hoshi; Jonathan A. Epstein; Reynold A. Panettieri

Malignant pleural mesothelioma (MPM) is a rare cancer that is refractory to current treatments. It is characterized by a robust deposition of transitional fibrin that is in part promoted by tumor cells. MPM cells express tissue factor (TF) and the tissue factor pathway inhibitor (TFPI), but their contribution to the pathogenesis of MPM has been unclear. We found that REN MPM cells fail to express TFPI. Based on the tumor growth-promoting properties of TF, we hypothesized that the stable transfection of TFPI into REN MPM cells would decrease their aggressiveness. We tested our hypothesis using in vitro, in vivo, and ex vivo analyses. TFPI knock-in decreased the proliferation, invasion, and TF activity of REN cells in vitro. REN TFPI knock-in cells, empty vector, and naive control cells were next injected intrapleurally into nude mice. The expression of TFPI significantly decreased tissue invasion, inflammation, and the deposition of fibrin and collagen associated with tumor tissue, pleural effusions, and tumor burden. In ex vivo analyses, REN cells were cultured from harvested tumors. The overexpression of TFPI was maintained in cells propagated from TFPI knock-in tumors, and attenuated the activation of Factor X and the invasiveness of tumor cells. These analyses demonstrate that TFPI reduces the aggressiveness of MPM in vitro and in vivo, and that its effect involves the inhibition of TF procoagulant activity. These observations suggest that the interactions of TF and TFPI represent a novel therapeutic target in the treatment of MPM.Histone deacetylase (HDAC) inhibitors may offer novel approaches in the treatment of asthma. We postulate that trichostatin A (TSA), a Class 1 and 2 inhibitor of HDAC, inhibits airway hyperresponsiveness in antigen-challenged mice. Mice were sensitized and challenged with Aspergillus fumigatus antigen (AF) and treated with TSA, dexamethasone, or vehicle. Lung resistance (R(L)) and dynamic compliance were measured, and bronchial alveolar lavage fluid (BALF) was analyzed for numbers of leukocytes and concentrations of cytokines. Human precision-cut lung slices (PCLS) were treated with TSA and their agonist-induced bronchoconstriction was measured, and TSA-treated human airway smooth muscle (ASM) cells were evaluated for the agonist-induced activation of Rho and intracellular release of Ca(2+). The activity of HDAC in murine lungs was enhanced by antigen and abrogated by TSA. TSA also inhibited methacholine (Mch)-induced increases in R(L) and decreases in dynamic compliance in naive control mice and in AF-sensitized and -challenged mice. Total cell counts, concentrations of IL-4, and numbers of eosinophils in BALF were unchanged in mice treated with TSA or vehicle, whereas dexamethasone inhibited the numbers of eosinophils in BALF and concentrations of IL-4. TSA inhibited the carbachol-induced contraction of PCLS. Treatment with TSA inhibited the intracellular release of Ca(2+) in ASM cells in response to histamine, without affecting the activation of Rho. The inhibition of HDAC abrogates airway hyperresponsiveness to Mch in both naive and antigen-challenged mice. TSA inhibits the agonist-induced contraction of PCLS and mobilization of Ca(2+) in ASM cells. Thus, HDAC inhibitors demonstrate a mechanism of action distinct from that of anti-inflammatory agents such as steroids, and represent a promising therapeutic agent for airway disease.


The FASEB Journal | 2014

MicroRNA-10a controls airway smooth muscle cell proliferation via direct targeting of the PI3 kinase pathway

Ruoxi Hu; Wen-Chi Pan; Alexey V. Fedulov; William Jester; Matthew R. Jones; Scott T. Weiss; Reynold A. Panettieri; Kelan G. Tantisira; Quan Lu

Airway smooth muscle (ASM) cells play important physiological roles in the lung, and abnormal proliferation of ASM directly contributes to the airway remodeling during development of lung diseases such as asthma. MicroRNAs are small yet versatile gene tuners that regulate a variety of cellular processes, including cell growth and proliferation; however, little is known about the precise role of microRNAs in the proliferation of the ASM. Here we report that a specific microRNA (miR‐10a) controls ASM proliferation through directly inhibiting the phosphoinositide 3‐kinase (PI3K) pathway. Next‐generation sequencing identified miR‐10a as the most abundant microRNA expressed in primary human airway smooth muscle (HASM) cells, accounting for > 20% of all small RNA reads. Overexpression of miR‐10a reduced mitogen‐induced HASM proliferation by ~50%, whereas inhibition of miR‐10a increased HASM proliferation by ~40%. Microarray profiling of HASM cells expressing miR‐10a mimics identified 52 significantly down‐regulated genes as potential targets of miR‐10a, including the catalytic subunit α of PI3K (PIK3CA), the central component of the PI3K pathway. MiR‐10a directly suppresses PIK3CA expression by targeting the 3‘‐untranslated region (3‘‐UTR) of the gene. Inhibition of PIK3CA by miR‐10a reduced V‐akt murine thymoma viral oncogene homolog 1 (AKT) phosphorylation and blunted the expression of cyclins and cyclin‐dependent kinases that are required for HASM proliferation. Together, our study identifies a novel microRNA‐mediated regulatory mechanism for PI3K signaling and ASM proliferation and further suggests miR‐10a as a potential therapeutic target for lung diseases whose etiology resides in abnormal ASM proliferation.—Hu, R., Pan, W., Fedulov, A. V., Jester, W., Jones, M. R., Weiss, S. T., Panettieri, R. A., Jr., Tantisira, K., Lu, Q. MicroRNA‐10a controls airway smooth muscle cell proliferation via direct targeting of the PI3 kinase pathway. FASEB J. 28, 2347–2357 (2014). www.fasebj.org


European Journal of Pharmacology | 2010

Aclidinium bromide abrogates allergen-induced hyperresponsiveness and reduces eosinophilia in murine model of airway inflammation.

Gautam Damera; Meiqi Jiang; Hengjiang Zhao; Homer W. Fogle; William Jester; Jose Freire; Reynold A. Panettieri

Airway hyperresponsiveness and inflammation characterize the airways of individuals with asthma and chronic obstructive pulmonary disease (COPD). Hence, therapeutic approaches that attenuate such manifestations may offer promise in the management of these diseases. In the present study, we investigated whether a novel long-acting cholinergic antagonist, aclidinium bromide, modulates airway function and leukocyte trafficking in an Aspergillus fumigatus (Af)-induced murine model of asthma. Nebulized aclidinium (1 mg/ml) administration completely abrogated increases in methacholine-induced lung resistance in Af-exposed mice. Parallel assessment of dynamic compliance showed that aclidinium also completely restores methacholine-mediated decreases in naïve and Af-exposed mice. As evidenced by differential cell counts within bronchoalveolar lavage fluid, aclidinium also diminished (51±4%) Af-induced airway eosinophil numbers with no significant change in other immune cell types. Further assessment of cytokine and total protein levels in bronchoalveolar lavage fluid showed that aclidinium had little effect on IL-4 or IL-6 levels in either Af-exposed or naïve mice but markedly decreased total protein levels in bronchoalveolar lavage fluid. These data suggest that aclidinium, a selective muscarinic antagonist, not only acts as a bronchodilator but could also act as an anti-inflammatory agent with potential clinical benefits in the treatment of COPD and asthma.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2009

Ozone modulates IL-6 secretion in human airway epithelial and smooth muscle cells

Gautam Damera; Hengjiang Zhao; Miao Wang; Michael Smith; Christopher Kirby; William Jester; John A. Lawson; Reynold A. Panettieri

Although ozone enhances leukocyte function and recruitment in airways, the direct effect of ozone in modulating structural cell-derived inflammatory mediators remains unknown. Using a coculture model comprised of differentiated human airway epithelial cells (NHBE) and smooth muscle cells (ASM), we postulate that ozone regulates IL-6 secretion in basal and cytokine-primed structural cells. Air-liquid interface (ALI) cultures of NHBE cells underwent differentiation as determined by mucin secretion, transepithelial electrical resistance (TEER), and ultrastructure parameters. Whereas TNF enhanced basal secretion of IL-6 (57 +/- 3%), ozone exposure at 0.6 ppm for 6 h augmented IL-6 levels in basal (41 +/- 3%) and TNF- (50 +/- 5%) primed cocultures compared with that derived from NHBE or ASM monolayers alone. Levels of PGE(2), 6-keto-PGF(1alpha), PGF(2alpha), and thromboxane B(2) (TxB(2)) levels in basal and TNF-primed cocultures revealed that ozone selectively enhanced PGE(2) production in TNF- (6 +/- 3-fold) primed cocultures, with little effect (P > 0.05) on diluent-treated cultures. In accordance with ozone-induced increases in PGE(2) levels, cyclooxygenase inhibition with indomethacin partially abolished IL-6 secretion. Surprisingly, indomethacin had little effect on constitutive secretion of IL-6 in cocultures, whereas indomethacin completely restored ozone-mediated TEER reduction in TNF-primed cocultures. Collectively, our data for the first time suggest a dual role of ozone in modulating IL-6 secretion and TEER outcomes in a PGE(2)-dependent (in presence of TNF stimulus) and -independent manner (in absence of cytokine stimulus).


PLOS ONE | 2015

Vitamin D Modulates Expression of the Airway Smooth Muscle Transcriptome in Fatal Asthma

Blanca E. Himes; Cynthia Koziol-White; Martin Johnson; Christina Nikolos; William Jester; Barbara J. Klanderman; Augusto A. Litonjua; Kelan G. Tantisira; Kevin Truskowski; Kevin MacDonald; Reynold A. Panettieri; Scott T. Weiss

Globally, asthma is a chronic inflammatory respiratory disease affecting over 300 million people. Some asthma patients remain poorly controlled by conventional therapies and experience more life-threatening exacerbations. Vitamin D, as an adjunct therapy, may improve disease control in severe asthma patients since vitamin D enhances glucocorticoid responsiveness and mitigates airway smooth muscle (ASM) hyperplasia. We sought to characterize differences in transcriptome responsiveness to vitamin D between fatal asthma- and non-asthma-derived ASM by using RNA-Seq to measure ASM transcript expression in five donors with fatal asthma and ten non-asthma-derived donors at baseline and with vitamin D treatment. Based on a Benjamini-Hochberg corrected p-value <0.05, 838 genes were differentially expressed in fatal asthma vs. non-asthma-derived ASM at baseline, and vitamin D treatment compared to baseline conditions induced differential expression of 711 and 867 genes in fatal asthma- and non-asthma-derived ASM, respectively. Functional gene categories that were highly represented in all groups included extracellular matrix, and responses to steroid hormone stimuli and wounding. Genes differentially expressed by vitamin D also included cytokine and chemokine activity categories. Follow-up qPCR and individual analyte ELISA experiments were conducted for four cytokines (i.e. CCL2, CCL13, CXCL12, IL8) to measure TNFα-induced changes by asthma status and vitamin D treatment. Vitamin D inhibited TNFα-induced IL8 protein secretion levels to a comparable degree in fatal asthma- and non-asthma-derived ASM even though IL8 had significantly higher baseline levels in fatal asthma-derived ASM. Our findings identify vitamin D-specific gene targets and provide transcriptomic data to explore differences in the ASM of fatal asthma- and non-asthma-derived donors.


Experimental Lung Research | 2010

Inhibition of myristoylated alanine-rich C kinase substrate (MARCKS) protein inhibits ozone-induced airway neutrophilia and inflammation

Gautam Damera; William Jester; Meiqi Jiang; Hengjiang Zhao; Homer W. Fogle; Michael Mittelman; Angela Haczku; Edwin C. Murphy; Indu Parikh; Reynold A. Panettieri

ABSTRACT Evidence suggests inhibition of leukocyte trafficking mitigates, in part, ozone-induced inflammation. In the present study, the authors postulated that inhibition of myristoylated alanine-rich C kinase substrate (MARCKS), an 82-kDa protein with multiple biological roles, could inhibit ozone-induced leukocyte trafficking and cytokine secretions. BALB/c mice (n == 5/cohort) were exposed to ozone (100 ppb) or forced air (FA) for 4 hours. MARCKS-inhibiting peptides, MANS, BIO-11000, BIO-11006, or scrambled control peptide RNS, were intratracheally administered prior to ozone exposure. Ozone selectively enhanced bronchoalveolar lavage (BAL) levels of killer cells (KCs; 6 ± 0.9-fold), interleukin-6 (IL-6; 12.7 ± 1.9-fold), and tumor necrosis factor (TNF; 2.1 ± 0.5-fold) as compared to cohorts exposed to FA. Additionally, ozone increased BAL neutrophils by 21%% ± 2%% with no significant (P > .05) changes in other cell types. MANS, BIO-11000, and BIO-11006 significantly reduced ozone-induced KC secretion by 66%% ± 14%%, 47%% ± 15%%, and 71.1%% ± 14%%, and IL-6 secretion by 69%% ± 12%%, 40%% ± 7%%, and 86.1%% ± 11%%, respectively. Ozone-mediated increases in BAL neutrophils were reduced by MANS (86%% ± 7%%) and BIO-11006 (84%% ± 2.5%%), but not BIO-11000. These studies identify for the first time the novel potential of MARCKS protein inhibitors in abrogating ozone-induced increases in neutrophils, cytokines, and chemokines in BAL fluid. BIO-11006 is being developed as a treatment for chronic obstructive pulmonary disorder (COPD) and is currently being evaluated in a phase 2 clinical study.


Journal of Clinical Investigation | 2016

Epithelium-generated neuropeptide Y induces smooth muscle contraction to promote airway hyperresponsiveness

Shanru Li; Cynthia Koziol-White; Joseph A. Jude; Meiqi Jiang; Hengjiang Zhao; Gaoyuan Cao; Edwin J. Yoo; William Jester; Michael P. Morley; Su Zhou; Yi Wang; Min Min Lu; Reynold A. Panettieri; Edward E. Morrisey

Asthma is one of the most common chronic diseases globally and can be divided into presenting with or without an immune response. Current therapies have little effect on nonimmune disease, and the mechanisms that drive this type of asthma are poorly understood. Here, we have shown that loss of the transcription factors forkhead box P1 (Foxp1) and Foxp4, which are critical for lung epithelial development, in the adult airway epithelium evokes a non-Th2 asthma phenotype that is characterized by airway hyperresponsiveness (AHR) without eosinophilic inflammation. Transcriptome analysis revealed that loss of Foxp1 and Foxp4 expression induces ectopic expression of neuropeptide Y (Npy), which has been reported to be present in the airways of asthma patients, but whose importance in disease pathogenesis remains unclear. Treatment of human lung airway explants with recombinant NPY increased airway contractility. Conversely, loss of Npy in Foxp1- and Foxp4-mutant airway epithelium rescued the AHR phenotype. We determined that NPY promotes AHR through the induction of Rho kinase activity and phosphorylation of myosin light chain, which induces airway smooth muscle contraction. Together, these studies highlight the importance of paracrine signals from the airway epithelium to the underlying smooth muscle to induce AHR and suggest that therapies targeting epithelial induction of this phenotype may prove useful in treatment of noneosinophilic asthma.


Journal of Pharmacology and Experimental Therapeutics | 2010

Deletion of Microsomal Prostaglandin E Synthase-1 Does Not Alter Ozone-Induced Airway Hyper-Responsiveness

Miao Wang; Philip R. Cooper; Meiqi Jiang; Hengjiang Zhao; Yiqun Hui; Yubing Yao; Joshua C. Tate; Gautam Damera; John A. Lawson; William Jester; Angela Haczku; Reynold A. Panettieri; Garret A. FitzGerald

Nonsteroidal anti-inflammatory drugs ameliorate pain and fever by inhibiting cyclooxygenase (COX) and suppressing prostanoid formation. Microsomal prostaglandin E synthase-1 (mPGES-1) catalyzes formation of PGE2 from the COX product PGH2 and has emerged as a therapeutic target. Inhibition of mPGES-1, however, renders the PGH2 substrate available for diversion to other PG synthases. To address the possibility that substrate diversion augments formation of PGs that might modulate bronchial tone, we assessed the impact of mPGES-1 deletion in a mouse model of ozone-induced airway hyper-responsiveness. Ozone exposure increased total lung resistance to inhaled methacholine in wild-type mice. Deletion of mPGES-1 had little effect on total lung resistance in either naive or ozone-exposed animals. The carbachol-induced narrowing of luminal diameter in intrapulmonary airways of lung slices from acute ozone-exposed mice was also unaltered by mPGES-1 deletion. Likewise, although concentrations of PGE2 were reduced in bronchoalveolar lavage fluid, whereas 6-keto-PGF1α, PGD2, and PGF2α, all were increased, deletion of mPGES-1 failed to influence cell trafficking into the airways of either naive or ozone-exposed animals. Despite biochemical evidence of PGH2 substrate diversion to potential bronchomodulator PGs, deletion of mPGES-1 had little effect on ozone-induced airway inflammation or airway hyper-responsiveness. Pharmacologically targeting mPGES-1 may not predispose patients at risk to airway dysfunction.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2005

TGF-β potentiates airway smooth muscle responsiveness to bradykinin

Jenny H. Kim; Deepika Jain; Omar Tliba; Bei Yang; William Jester; Reynold A. Panettieri; Yassine Amrani; Ellen Puré

Collaboration


Dive into the William Jester's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meiqi Jiang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Gautam Damera

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Hengjiang Zhao

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Homer W. Fogle

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Kelan G. Tantisira

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Scott T. Weiss

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Angela Haczku

University of California

View shared research outputs
Top Co-Authors

Avatar

Audreesh Banerjee

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge