Network


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

Hotspot


Dive into the research topics where Joseph D. Sherrill is active.

Publication


Featured researches published by Joseph D. Sherrill.


Nature Genetics | 2010

Common variants at 5q22 associate with pediatric eosinophilic esophagitis

Marc E. Rothenberg; Jonathan M. Spergel; Joseph D. Sherrill; Kiran Annaiah; Lisa J. Martin; Antonella Cianferoni; Laura M. Gober; Cecilia Kim; Joseph T. Glessner; Edward C. Frackelton; Kelly Thomas; Carine Blanchard; Chris A. Liacouras; Ritu Verma; Seema S. Aceves; Margaret H. Collins; Terri F. Brown-Whitehorn; Phil E. Putnam; James P. Franciosi; Rosetta M. Chiavacci; J. Struan F.A. Grant; J. Pablo Abonia; Patrick Sleiman; Hakon Hakonarson

Eosinophilic esophagitis (EoE) is an allergic disorder characterized by the accumulation of eosinophils in the esophagus. We report association of EoE with variants at chromosome 5q22 encompassing TSLP and WDR36 (rs3806932, combined P = 3.19 × 10−9). TSLP is overexpressed in esophageal biopsies from individuals with EoE compared with unaffected individuals, whereas WDR36 expression is unaltered between the two groups. These data implicate the 5q22 locus in the pathogenesis of EoE and identify TSLP as the most likely candidate gene in the region.


The Journal of Allergy and Clinical Immunology | 2010

Variants of thymic stromal lymphopoietin and its receptor associate with eosinophilic esophagitis

Joseph D. Sherrill; Peisong Gao; Emily M. Stucke; Carine Blanchard; Margaret H. Collins; Phil E. Putnam; James P. Franciosi; Jonathan P. Kushner; J. Pablo Abonia; Amal H. Assa'ad; Melinda Butsch Kovacic; Jocelyn M. Biagini Myers; Bruce S. Bochner; Hua He; Gurjit K. Khurana Hershey; Lisa J. Martin; Marc E. Rothenberg

BACKGROUND The genetic cause of eosinophilic esophagitis (EE) has been largely unexplored until a recent genome-wide association study identified a disease susceptibility locus on 5q22, a region that harbors the thymic stromal lymphopoietin (TSLP) gene. However, it is unclear whether the observed genetic associations with EE are disease-specific or confounded by the high rate of allergy in patients with EE. In addition, the genetic contributions of other allergy-associated genes to EE risk have not been explored. OBJECTIVE We aimed to delineate single nucleotide polymorphisms (SNPs) that associated with EE apart from allergy. METHODS We used a custom array containing 738 SNPs in 53 genes implicated in allergic responses, immune responses, or both to genotype 220 allergic or 246 nonallergic control subjects and a discovery cohort of 170 patients with EE. We replicated a statistically significant SNP association in an independent case-control cohort and examined the induction of the candidate gene in primary esophageal epithelial cells. RESULTS A single SNP residing in the TSLP gene reached Bonferroni linkage disequilibrium-adjusted significance but only when patients with EE were compared with allergic control subjects (rs10062929; P = 4.11 x 10(-5); odds ratio, 0.35). A nonsynonymous polymorphism in the thymic stromal lymphopoietin receptor (TSLPR) gene on Xp22.3 and Yp11.3 was significantly associated with disease only in male patients with EE. Primary esophageal epithelial cells expressed TSLP mRNA after Toll-like receptor 3 stimulation. CONCLUSION These data collectively identify TSLP as a candidate gene critically involved in EE susceptibility beyond its role in promoting T(H)2 responses.


The Journal of Allergy and Clinical Immunology | 2012

MicroRNA signature in patients with eosinophilic esophagitis, reversibility with glucocorticoids, and assessment as disease biomarkers.

Thomas X. Lu; Joseph D. Sherrill; Ting Wen; Andrew J. Plassard; John A. Besse; J.P. Abonia; James P. Franciosi; Philip E. Putnam; Michael Eby; Lisa J. Martin; Bruce J. Aronow; Marc E. Rothenberg

BACKGROUND The role of microRNAs (miRNAs), a key class of regulators of mRNA expression and translation, in patients with eosinophilic esophagitis (EoE) has not been explored. OBJECTIVE We aimed to identify miRNAs dysregulated in patients with EoE and assess the potential of these miRNAs as disease biomarkers. METHODS Esophageal miRNA expression was profiled in patients with active EoE and those with glucocorticoid-induced disease remission. Expression profiles were compared with those of healthy control subjects and patients with chronic (noneosinophilic) esophagitis. Expression levels of the top differentially expressed miRNAs from the plasma of patients with active EoE and patients with EoE remission were compared with those of healthy control subjects. RESULTS EoE was associated with 32 differentially regulated miRNAs and was distinguished from noneosinophilic forms of esophagitis. The expression levels of the most upregulated miRNAs (miR-21 and miR-223) and the most downregulated miRNA (miR-375) strongly correlated with esophageal eosinophil levels. Bioinformatic analysis predicted interplay of miR-21 and miR-223 with key roles in the polarization of adaptive immunity and regulation of eosinophilia, and indeed, these miRNAs correlated with key elements of the EoE transcriptome. The differentially expressed miRNAs were largely reversible in patients who responded to glucocorticoid treatment. EoE remission induced a single miRNA (miR-675) likely to be involved in DNA methylation. Plasma analysis of the most upregulated esophageal miRNAs identified miR-146a, miR-146b, and miR-223 as the most differentially expressed miRNAs in the plasma. CONCLUSIONS We have identified a marked dysregulated expression of a select group of miRNAs in patients with EoE and defined their reversibility with glucocorticoid treatment and their potential value as invasive and noninvasive biomarkers.


Genes and Immunity | 2014

Analysis and expansion of the eosinophilic esophagitis transcriptome by RNA sequencing

Joseph D. Sherrill; Kiran Kc; Carine Blanchard; Emily M. Stucke; Katherine A. Kemme; Margaret H. Collins; J.P. Abonia; Philip E. Putnam; Vincent A. Mukkada; Ajay Kaul; Samuel Kocoshis; Jonathan P. Kushner; Andrew J. Plassard; Rebekah Karns; Phillip Dexheimer; Bruce J. Aronow; Marc E. Rothenberg

Eosinophilic esophagitis (EoE) is an allergic inflammatory disorder of the esophagus that is compounded by genetic predisposition and hypersensitivity to environmental antigens. Using high-density oligonucleotide expression chips, a disease-specific esophageal transcript signature was identified and was shown to be largely reversible with therapy. In an effort to expand the molecular signature of EoE, we performed RNA sequencing on esophageal biopsies from healthy controls and patients with active EoE and identified a total of 1607 significantly dysregulated transcripts (1096 upregulated, 511 downregulated). When clustered by raw expression levels, an abundance of immune cell-specific transcripts are highly induced in EoE but expressed at low (or undetectable) levels in healthy controls. Moreover, 66% of the gene signature identified by RNA sequencing was previously unrecognized in the EoE transcript signature by microarray-based expression profiling and included several long non-coding RNAs (lncRNA), an emerging class of transcriptional regulators. The lncRNA BRAF-activated non-protein coding RNA (BANCR) was upregulated in EoE and induced in interleukin-13 (IL-13)–treated primary esophageal epithelial cells. Repression of BANCR significantly altered the expression of IL-13–induced proinflammatory genes. Together, these data comprise new potential biomarkers of EoE and demonstrate a novel role for lncRNAs in EoE and IL-13–associated responses.


Journal of Immunology | 2013

Intestinal CCL11 and Eosinophilic Inflammation Is Regulated by Myeloid Cell–Specific RelA/p65 in Mice

Amanda Waddell; Richard Ahrens; Yi Ting Tsai; Joseph D. Sherrill; Lee A. Denson; Kris A. Steinbrecher; Simon P. Hogan

In inflammatory bowel diseases (IBDs), particularly ulcerative colitis, intestinal macrophages (MΦs), eosinophils, and the eosinophil-selective chemokine CCL11, have been associated with disease pathogenesis. MΦs, a source of CCL11, have been reported to be of a mixed classical (NF-κB–mediated) and alternatively activated (STAT-6–mediated) phenotype. The importance of NF-κB and STAT-6 pathways to the intestinal MΦ/CCL11 response and eosinophilic inflammation in the histopathology of experimental colitis is not yet understood. Our gene array analyses demonstrated elevated STAT-6– and NF-κB–dependent genes in pediatric ulcerative colitis colonic biopsies. Dextran sodium sulfate (DSS) exposure induced STAT-6 and NF-κB activation in mouse intestinal F4/80+CD11b+Ly6Chi (inflammatory) MΦs. DSS-induced CCL11 expression, eosinophilic inflammation, and histopathology were attenuated in RelA/p65Δmye mice, but not in the absence of STAT-6. Deletion of p65 in myeloid cells did not affect inflammatory MΦ recruitment or alter apoptosis, but did attenuate LPS-induced cytokine production (IL-6) and Ccl11 expression in purified F4/80+CD11b+Ly6Chi inflammatory MΦs. Molecular and cellular analyses revealed a link between expression of calprotectin (S100a8/S100a9), Ccl11 expression, and eosinophil numbers in the DSS-treated colon. In vitro studies of bone marrow–derived MΦs showed calprotectin-induced CCL11 production via a p65-dependent mechanism. Our results indicate that myeloid cell–specific NF-κB–dependent pathways play an unexpected role in CCL11 expression and maintenance of eosinophilic inflammation in experimental colitis. These data indicate that targeting myeloid cells and NF-κB–dependent pathways may be of therapeutic benefit for the treatment of eosinophilic inflammation and histopathology in IBD.


Journal of Biological Chemistry | 2006

G Protein-coupled Receptor (GPCR) Kinase 2 Regulates Agonist-independent Gq/11 Signaling from the Mouse Cytomegalovirus GPCR M33

Joseph D. Sherrill; William E. Miller

The mouse cytomegalovirus M33 protein is highly homologous to mammalian G protein-coupled receptors (GPCRs) yet functions in an agonist-independent manner to activate a number of classical GPCR signal transduction pathways. M33 is functionally similar to the human cytomegalovirus-encoded US28 GPCR in its ability to induce inositol phosphate accumulation, activate NF-κB, and promote smooth muscle cell migration. This ability to promote cellular migration suggests a role for viral GPCRs like M33 in viral dissemination in vivo, and accordingly, M33 is required for efficient murine cytomegalovirus replication in the mouse. Although previous studies have identified several M33-induced signaling pathways, little is known regarding the membrane-proximal events involved in signaling and regulation of this receptor. In this study, we used recombinant retroviruses to express M33 in wild-type and Gαq/11–/– mouse embryonic fibroblasts and show that M33 couples directly to the Gq/11 signaling pathway to induce high levels of total inositol phosphates in an agonist-independent manner. Our data also show that GRK2 is a potent regulator of M33-induced Gq/11 signaling through its ability to phosphorylate M33 and sequester Gαq/11 proteins. Taken together, the results from this study provide the first genetic evidence of a viral GPCR coupling to a specific G protein signaling pathway as well as identify the first viral GPCR to be regulated specifically by both the catalytic activity of the GRK2 kinase domain and the Gαq/11 binding activity of the GRK2 RH domain.


Mucosal Immunology | 2015

Neurotrophic tyrosine kinase receptor 1 is a direct transcriptional and epigenetic target of IL-13 involved in allergic inflammation

Mark Rochman; Andrey V. Kartashov; Julie M. Caldwell; Margaret H. Collins; Emily M. Stucke; K Kc; Joseph D. Sherrill; J Herren; Artem Barski; Marc E. Rothenberg

Although interleukin (IL)-13 and neurotrophins are functionally important for the pathogenesis of immune responses, the interaction of these pathways has not been explored. Herein, by interrogating IL-13-induced responses in human epithelial cells we show that neurotrophic tyrosine kinase receptor, type 1 (NTRK1), a cognate, high-affinity receptor for nerve growth factor (NGF), is an early transcriptional IL-13 target. Induction of NTRK1 was accompanied by accumulation of activating epigenetic marks in the promoter; transcriptional and epigenetic changes were signal transducer and activator of transcription 6 dependent. Using eosinophilic esophagitis as a model for human allergic inflammation, we found that NTRK1 was increased in inflamed tissue and dynamically expressed as a function of disease activity and that the downstream mediator of NTRK1 signaling early growth response 1 protein was elevated in allergic inflammatory tissue compared with control tissue. Unlike NTRK1, its ligand NGF was constitutively expressed in control and disease states, indicating that IL-13-stimulated NTRK1 induction is a limiting factor in pathway activation. In epithelial cells, NGF and IL-13 synergistically induced several target genes, including chemokine (C-C motif) ligand 26 (eotaxin-3). In summary, we have demonstrated that IL-13 confers epithelial cell responsiveness to NGF by regulating NTRK1 levels by a transcriptional and epigenetic mechanism and that this process likely contributes to allergic inflammation.


PLOS ONE | 2015

In vitro model for studying esophageal epithelial differentiation and allergic inflammatory responses identifies keratin involvement in eosinophilic esophagitis.

Kiran Kc; Marc E. Rothenberg; Joseph D. Sherrill

Epithelial differentiation is an essential physiological process that imparts mechanical strength and barrier function to squamous epithelia. Perturbation of this process can give rise to numerous human diseases, such as atopic dermatitis, in which antigenic stimuli can penetrate the weakened epithelial barrier to initiate the allergic inflammatory cascade. We recently described a simplified air-liquid interface (ALI) culture system that facilitates the study of differentiated squamous epithelia in vitro. Herein, we use RNA sequencing to define the genome-wide transcriptional changes that occur within the ALI system during epithelial differentiation and in response to allergic inflammation. We identified 2,191 and 781 genes that were significantly altered upon epithelial differentiation or dysregulated in the presence of interleukin 13 (IL-13), respectively. Notably, 286 genes that were modified by IL-13 in the ALI system overlapped with the gene signature present within the inflamed esophageal tissue from patients with eosinophilic esophagitis (EoE), an allergic inflammatory disorder of the esophagus that is characterized by elevated IL-13 levels, altered epithelial differentiation, and pro-inflammatory gene expression. Pathway analysis of these overlapping genes indicated enrichment in keratin genes; for example, the gene encoding keratin 78, an uncharacterized type II keratin, was upregulated during epithelial differentiation (45-fold) yet downregulated in response to IL-13 and in inflamed esophageal tissue from patients. Thus, our findings delineate an in vitro experimental system that models epithelial differentiation that is dynamically regulated by IL-13. Using this system and analyses of patient tissues, we identify an altered expression profile of novel keratin differentiation markers in response to IL-13 and disease activity, substantiating the potential of this combined approach to identify relevant molecular processes that contribute to human allergic inflammatory disease.


The Journal of Allergy and Clinical Immunology | 2017

Profound loss of esophageal tissue differentiation in patients with eosinophilic esophagitis

Mark Rochman; Jared Travers; Cora E. Miracle; Mary C. Bedard; Ting Wen; Nurit P. Azouz; Julie M. Caldwell; Kiran Kc; Joseph D. Sherrill; Benjamin P. Davis; Jeffrey K. Rymer; Kenneth M. Kaufman; Bruce J. Aronow; Marc E. Rothenberg

Background A key question in the allergy field is to understand how tissue‐specific disease is manifested. Eosinophilic esophagitis (EoE) is an emerging tissue‐specific allergic disease with an unclear pathogenesis. Objective Herein we tested the hypothesis that a defect in tissue‐specific esophageal genes is an integral part of EoE pathogenesis. Methods We interrogated the pattern of expression of esophagus‐specific signature genes derived from the Human Protein Atlas in the EoE transcriptome and in EPC2 esophageal epithelial cells. Western blotting and immunofluorescence were used for evaluating expression of esophageal proteins in biopsy specimens from control subjects and patients with active EoE. Whole‐exome sequencing was performed to identify mutations in esophagus‐specific genes. Results We found that approximately 39% of the esophagus‐specific transcripts were altered in patients with EoE, with approximately 90% being downregulated. The majority of transcriptional changes observed in esophagus‐specific genes were reproduced in vitro in esophageal epithelial cells differentiated in the presence of IL‐13. Functional enrichment analysis revealed keratinization and differentiation as the most affected biological processes and identified IL‐1 cytokines and serine peptidase inhibitors as the most dysregulated esophagus‐specific protein families in patients with EoE. Accordingly, biopsy specimens from patients with EoE evidenced a profound loss of tissue differentiation, decreased expression of keratin 4 (KRT4) and cornulin (CRNN), and increased expression of KRT5 and KRT14. Whole‐exome sequencing of 33 unrelated patients with EoE revealed 39 rare mutations in 18 esophagus‐specific differentially expressed genes. Conclusions A tissue‐centered analysis has revealed a profound loss of esophageal tissue differentiation (identity) as an integral and specific part of the pathophysiology of EoE and implicated protease‐ and IL‐1–related activities as putative central pathways in disease pathogenesis.


Gastroenterology Clinics of North America | 2014

Genetic and Epigenetic Underpinnings of Eosinophilic Esophagitis

Joseph D. Sherrill; Marc E. Rothenberg

Eosinophilic esophagitis (EoE) is a complex, polygenic disorder caused by genetic predisposition and environmental exposures. Because of the recent emergence of EoE as a bona fide global health concern, a paucity of available therapeutic and diagnostic options exists. However, rapid progress has been made in an effort to rectify this lack and to improve understanding of the factors that cause EoE. This article highlights key advances in elucidating the genetic (and epigenetic) components involved in EoE.

Collaboration


Dive into the Joseph D. Sherrill's collaboration.

Top Co-Authors

Avatar

Marc E. Rothenberg

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Emily M. Stucke

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Margaret H. Collins

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mark Rochman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ting Wen

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

J. Pablo Abonia

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Julie M. Caldwell

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa J. Martin

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Simon P. Hogan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge