Network


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

Hotspot


Dive into the research topics where Wendy A. Goodman is active.

Publication


Featured researches published by Wendy A. Goodman.


Journal of Immunology | 2009

IL-6 Signaling in Psoriasis Prevents Immune Suppression by Regulatory T Cells

Wendy A. Goodman; Alan D. Levine; Jessica V. Massari; Hideaki Sugiyama; Thomas S. McCormick; Kevin D. Cooper

T memory/effector cells (Tmem/eff) isolated from psoriatic patients are chronically activated and poorly suppressed by regulatory T cells (Treg). The proinflammatory cytokine IL-6, which signals through Stat3, allows escape of Tmem/eff cells from Treg-mediated suppression in a murine system. We show here that IL-6 protein is markedly elevated and most highly expressed by CD31+ endothelial cells and CD11c+ dermal dendritic cells (DCs) in lesional psoriatic skin. We hypothesized that exposure to high IL-6 in lesional tissue may lead to the dampened Treg function observed in psoriasis patients. Indeed, we found that IL-6, but not other Stat3-activating cytokines, was necessary and sufficient to reverse human T cell suppression by Treg in an in vitro model using activated DCs as a source of IL-6. IL-6Rα and gp130 expression was significantly elevated in psoriatic effector T cells compared with normal controls. Overall, IL-6Rα expression on Treg exceeded that of effector T cells, and both populations phosphorylated Stat3 in response to IL-6. Phosphorylation of Stat3 in T cells contributes to Th17 differentiation and we identify cells within lesional tissue that coexpress CD3, IL-17, and IL-6, indicating that Th17 cells are present in vivo within the psoriatic Tmem/eff population and contribute to IL-6-mediated resistance to Treg suppression. Taken together, T lymphocytes trafficking into lesional psoriatic skin encounter high IL-6 from endothelial cells, DCs, and Th17 cells, enabling cutaneous T cell escape from Treg suppression and Th17 participation in inflammation. Targeting IL-6 signaling pathways in psoriasis may rebalance Treg/T effector activity and ameliorate disease.


Journal of Immunology | 2011

Stat3 Phosphorylation Mediates Resistance of Primary Human T Cells to Regulatory T Cell Suppression

Wendy A. Goodman; Andrew Young; Thomas S. McCormick; Kevin D. Cooper; Alan D. Levine

Human autoimmune diseases are characterized by systemic T cell dysfunction, resulting in chronically activated Th1 and Th17 cells that are inadequately suppressed by regulatory T cells (Tregs). IL-6, which is overexpressed in tissue and serum of patients with autoimmune diseases, inhibits human Treg function. We sought to determine the mechanism for the antitolerogenic properties of IL-6 by examining the signaling pathways downstream of IL-6R in primary human T cells. Inhibition of Stat3 signaling in MLCs containing IL-6 restores Treg-mediated suppression, demonstrating that IL-6–mediated loss of Treg suppression requires phosphorylation of Stat3. Cultures in which either effector T cells (Teffs) or Tregs were pretreated with Stat3 inhibitors indicate that phosphorylated (p)Stat3 is required in both T cell populations for IL-6–mediated reversal of Treg function. IL-21, which signals preferentially through pStat3, also reverses Treg suppression, in contrast to IL-27 and IFN-γ, which signal preferentially through Stat1 and do not inhibit Treg function. Interestingly, both Teffs and Tregs respond to IL-6 stimulation through strong Stat3 phosphorylation with minimal MAPK/Erk activation and moderate Stat1 phosphorylation. Finally, Teffs stimulated strongly through the TCR are also resistant to suppression by Tregs and show concurrent Stat3 phosphorylation. In these cultures, inhibition of pStat3 restores functional suppression by Tregs. Taken together, our findings suggest that an early dominance of Stat3 signaling, prior to subsequent T cell activation, is required for the loss of functional Treg suppression and that kinase-specific inhibitors may hold therapeutic promise in the treatment of autoimmune and chronic inflammatory diseases.


American Journal of Pathology | 2016

IL-33 drives eosinophil infiltration and pathogenic type 2 helper T-cell immune responses leading to chronic experimental ileitis

Carlo De Salvo; X.M. Wang; L. Pastorelli; Benedetta Mattioli; Sara Omenetti; Kristine Ann G Buela; Saleem Chowdhry; Rekha R. Garg; Wendy A. Goodman; Alex Rodriguez-Palacios; Dirk E. Smith; Fabio Cominelli; Giorgos Bamias; Wei Xin; James J. Lee; Maurizio Vecchi; Theresa T. Pizarro

Although a clear association has been established between IL-33 and inflammatory bowel disease, mechanistic studies to date, primarily using acute murine models of colitis, have yielded contradicting results, demonstrating both pathogenic and protective roles. We used a well-characterized, spontaneous model of inflammatory bowel disease [ie, SAMP1/YitFc (SAMP) mice] to investigate the role of IL-33 during chronic intestinal inflammation. Our results showed marked eosinophil infiltration into the gut mucosa with increased levels of eotaxins and type 2 helper T-cell (Th2) cytokines as disease progressed and became more severe, which could be reversed upon either eosinophil depletion or blockade of IL-33 signaling. Exogenous IL-33 administration recapitulated these effects in ilea of uninflamed (parental) control AKR/J mice. Human data supported these findings, showing colocalization and up-regulation of IL-33 and eosinophils in the colonic mucosa of inflammatory bowel disease patients versus noninflamed controls. Finally, colonization of commensal flora by fecal material transplantation into germ-free SAMP and the presence of the gut microbiome induced IL-33, subsequent eosinophil infiltration, and mounting of Th2 immune responses, leading to exacerbation of chronic intestinal inflammation characteristic of SAMP mice. These data demonstrate a pathogenic role for IL-33-mediated eosinophilia and activation of Th2 immunity in chronic intestinal inflammation that is dependent on the gut microbiome. Targeting IL-33 may represent a novel therapeutic approach to treat patients with inflammatory bowel disease.


Journal of Leukocyte Biology | 2014

Immunosuppressive monocytes: possible homeostatic mechanism to restrain chronic intestinal inflammation

Elvira Kurmaeva; Dhruva Bhattacharya; Wendy A. Goodman; Sara Omenetti; Amber Merendino; Seth Berney; Theresa T. Pizarro; Dmitry V. Ostanin

Chronic colitis is accompanied by extensive myelopoiesis and accumulation of CD11b+Gr‐1+ cells in spleens and secondary lymphoid tissues. Although cells with similar phenotype have been described in cancer, chronic infection, or autoimmunity, where they were associated with suppression of T cell responses, little is known regarding how these cells affect CD4 T cell responses in the context of chronic intestinal inflammation. Therefore, we undertook this study to characterize the interplay between colitis‐induced myeloid cells and CD4 T cell. Within the CD11b+Gr‐1+ population, only monocytes (Ly6GnegLy6Chigh) but not other myeloid cell subsets suppressed proliferation and production of cytokines by CD4 T cells. Suppression was mediated by cell‐contact, NO and partially by IFN‐γ and PGs. Interestingly, Ly6Chigh MDCs, isolated from colitic colons, showed up‐regulation of iNOS and arginase‐1 and were more potent suppressors than those isolated from spleen. On a single‐cell level, MDCs inhibited Th1 responses but enhanced generation of foxp3+ T cells. MDCs, cocultured with activated/Teffs, isolated from inflamed colons under hypoxic (1% O2) conditions typical for the inflamed intestine, suppressed proliferation but not their production of proinflammatory cytokines and chemokines. Taken together, expansion of monocytes and MDCs and activation of their suppressive properties may represent a homeostatic mechanism aimed at restraining excessive T cell activation during chronic inflammatory settings. The contribution of immunosuppressive monocytes/MDCs to chronic colitis and their role in shaping T cell responses in vivo require further investigation.


Mucosal Immunology | 2014

Loss of estrogen-mediated immunoprotection underlies female gender bias in experimental Crohn’s-like ileitis

Wendy A. Goodman; Rekha R. Garg; Brian K. Reuter; Benedetta Mattioli; Emilie F. Rissman; Theresa T. Pizarro

The incidence and severity of Crohn’s disease (CD) are increased in female patients. Using SAMP1/YitFc (SAMP) mice, a spontaneous model of chronic intestinal inflammation that displays histologic and pathogenic similarities to human CD, we investigated the potential mechanism(s) contributing to sex differences observed in CD. Similar to gender differences observed in CD patients, SAMP female (SAMP-F) mice displayed an earlier onset and more severe ileitis compared with SAMP male (SAMP-M) mice. Furthermore, T-regulatory cells (Tregs) from gut-associated lymphoid tissue (GALT) of SAMP-F mice were reduced in frequency and impaired in their in vitro and in vivo suppressive functions compared with that of SAMP-M mice. Given the interaction between sex hormones and Treg function, we investigated the possible role of estrogen (E2) in SAMP ileitis. SAMP-M mice responded to exogenous E2 administration by expanding Treg frequency and reducing ileal inflammation, whereas SAMP-F mice were resistant. Conventional T cells and Tregs responded differentially to estrogen signaling, leading to distinct immunoprotective effects mediated by distinct estrogen receptor (ER) isoforms. These mechanisms were impaired in T cells from SAMP-F mice. Thus, hormone signaling influences the expansion and function of GALT Tregs in an ER-dependent manner and contributes to gender-based differences in experimental CD.


Mucosal Immunology | 2016

KLF6 contributes to myeloid cell plasticity in the pathogenesis of intestinal inflammation

Wendy A. Goodman; Sara Omenetti; D. Date; L. Di Martino; C. De Salvo; G. D. Kim; Shakeel A. Chowdhry; Giorgos Bamias; Fabio Cominelli; Theresa T. Pizarro; G. H. Mahabeleshwar

Inflammatory bowel disease (IBD) is associated with dysregulated macrophage responses, such that quiescent macrophages acquire a pro-inflammatory activation state and contribute to chronic intestinal inflammation. The transcriptional events governing macrophage activation and gene expression in the context of chronic inflammation such as IBD remain incompletely understood. Here, we identify Kruppel-like transcription factor-6 (KLF6) as a critical regulator of pathogenic myeloid cell activation in human and experimental IBD. We found that KLF6 was significantly upregulated in myeloid cells and intestinal tissue from IBD patients and experimental models of IBD, particularly in actively inflamed regions of the colon. Using complementary gain- and loss-of-function studies, we observed that KLF6 promotes pro-inflammatory gene expression through enhancement of nuclear factor κB (NFκB) signaling, while simultaneously suppressing anti-inflammatory gene expression through repression of signal transducer and activator of transcription 3 (STAT3) signaling. To study the in vivo role of myeloid KLF6, we treated myeloid-specific KLF6-knockout mice (Mac-KLF6-KO) with dextran sulfate sodium (DSS) and found that Mac-KLF6-KO mice were protected against chemically-induced colitis; this highlights the central role of myeloid KLF6 in promoting intestinal inflammation. Collectively, our results point to a novel gene regulatory program underlying pathogenic, pro-inflammatory macrophage activation in the setting of chronic intestinal inflammation.


Gastroenterology | 2015

SAMP1/YitFc Mice Develop Ileitis via Loss of CCL21 and Defects in Dendritic Cell Migration

Zbigniew Mikulski; Rebecca Johnson; Iftach Shaked; Gisen Kim; Heba Nowyhed; Wendy A. Goodman; Grzegorz Chodaczek; Theresa T. Pizarro; Fabio Cominelli; Klaus Ley

BACKGROUND & AIMS The lymphatic chemokine CCL21 is required for dendritic cell (DC) migration from tissues to lymph nodes, which helps establish tolerance to foreign yet harmless antigens. We demonstrate that CCL21 is almost completely absent from SAMP1/YitFc (SAMP) mice, which spontaneously develop chronic ileitis that resembles Crohns disease, and that DC migration is severely impaired in these mice compared with AKR mice (controls). Toll-like receptor agonists like the Toll-like receptor 7 agonist R848 induce DC maturation and mobilization. METHODS We collected intestinal and other tissues and mesenteric lymph nodes (MLN) from SAMP mice. Expression of CCL21 was measured by quantitative reverse transcription polymerase chain reaction and immunofluorescence analyses; spontaneous and induced migration of DCs were assessed by flow cytometry. We analyzed production of retinoic acid by DCs and their ability to induce development of regulatory T cells. Mice were fed R848 to determine its effects on migration of DCs and development of ileitis in SAMP mice. RESULTS SAMP mice expressed almost no CCL21 in any tissue tested. Their CD11b(+)CD103(+) DCs were defective in migration from the ileal lamina propria to the MLN. DCs from SAMP mice also had a greatly reduced ability to produce retinoic acid and induce development of regulatory T cells compared with control mice. Young SAMP mice had reduced CCL21 expression and decreased DC migration before developing ileitis. Administration of R848 to adult SAMP mice increased migration of DC to the MLN and development of regulatory T cells there, and reduced the severity of ileitis. CONCLUSIONS Loss of CCL21 signaling and DC migration is required for development of ileitis in SAMP mice. Reagents such as R848, which activate DC migration to the MLN, may be developed as treatments for patients with Crohns disease.


Current Opinion in Gastroenterology | 2013

Regulatory cell populations in the intestinal mucosa.

Wendy A. Goodman; Theresa T. Pizarro

Purpose of review The purpose of this review is to summarize recent advances in the fields of intestinal T-regulatory cell (Treg) and tolerogenic dendritic cell subsets. Under homeostatic conditions, CD4+ Tregs and tolerogenic dendritic cells function to maintain mucosal tolerance. Loss of immune homeostasis is the primary cause of intestinal abnormalities, including inflammatory bowel disease. Thus, an improved understanding of cellular mechanisms promoting tolerance will be critical for the development of more efficacious therapies to treat chronic intestinal inflammation. Recent findings Significant progress has been made in the past year in the study of mucosal Treg and dendritic cell populations. In particular, efforts have focused on the migration and differentiation of these cells in the intestinal mucosa, the functional consequences of cross-talk with the intestinal microbiome, mechanisms by which tolerogenic dendritic cells take up antigen, and regulation of retinoic acid synthesis. Summary Recent studies examining tolerogenic cell populations of the intestinal mucosa highlight the progress in understanding the function, regulation, and cross-talk of Treg and dendritic cell populations, and their interactions with the gut microbiota. Scientific advances in these areas will undoubtedly lead to the development of more effective therapeutic strategies for intestinal abnormalities such as inflammatory bowel disease.


Cellular and molecular gastroenterology and hepatology | 2015

Dysregulated Intrahepatic CD4+ T-Cell Activation Drives Liver Inflammation in Ileitis-Prone SAMP1/YitFc Mice

Sara Omenetti; Marco Brogi; Wendy A. Goodman; Colleen M. Croniger; Saada Eid; Alex Y. Huang; Giacomo Laffi; Tania Roskams; Fabio Cominelli; Massimo Pinzani; Theresa T. Pizarro

Background & Aims Liver inflammation is a common extraintestinal manifestation of inflammatory bowel disease (IBD), but whether liver involvement is a consequence of a primary intestinal defect or results from alternative pathogenic processes remains unclear. Therefore, we sought to determine the potential pathogenic mechanism(s) of concomitant liver inflammation in an established murine model of IBD. Methods Liver inflammation and immune cell subsets were characterized in ileitis-prone SAMP1/YitFc (SAMP) and AKR/J (AKR) control mice, lymphocyte-depleted SAMP (SAMPxRag-1−/−), and immunodeficient SCID recipient mice receiving SAMP or AKR donor CD4+ T cells. Proliferation and suppressive capacity of CD4+ T-effector (Teff) and T-regulatory (Treg) cells from gut-associated lymphoid tissue (GALT) and livers of SAMP and AKR mice were measured. Results Surprisingly, prominent inflammation was detected in 4-week-old SAMP livers before histologic evidence of ileitis, whereas both disease phenotypes were absent in age-matched AKR mice. SAMP liver disease was characterized by abundant infiltration of lymphocytes, required for hepatic inflammation to occur, a TH1-skewed environment, and phenotypically activated CD4+ T cells. SAMP intrahepatic CD4+ T cells also had the ability to induce liver and ileal inflammation when adoptively transferred into SCID recipients, whereas GALT-derived CD4+ T cells produced milder ileitis but not liver inflammation. Interestingly, SAMP intrahepatic CD4+ Teff cells showed increased proliferation compared with both SAMP GALT- and AKR liver-derived CD4+ Teff cells, and SAMP intrahepatic Tregs were decreased among CD4+ T cells and impaired in in vitro suppressive function compared with AKR. Conclusions Activated intrahepatic CD4+ T cells induce liver inflammation and contribute to experimental ileitis via locally impaired hepatic immunosuppressive function.


Inflammatory Bowel Diseases | 2017

Epithelial-specific Toll-like Receptor (TLR)5 Activation Mediates Barrier Dysfunction in Experimental Ileitis.

Loris Riccardo Lopetuso; Ruo Jia; X.M. Wang; Li Guo Jia; Valentina Petito; Wendy A. Goodman; Jon Meddings; Fabio Cominelli; Brian K. Reuter; Theresa T. Pizarro

Background: A large body of evidence supports a central role of TLR5 and its natural ligand, flagellin, in Crohns disease (CD), with the precise mechanism(s) still unresolved. Methods: We investigated the role of flagellin/TLR5 in SAMP1/YitFc (SAMP) mice, a spontaneous model of Crohns disease-like ileitis. Results: Ileal Tlr5 and serum antiflagellin IgG antibodies were increased in SAMP before the onset of inflammation and during established disease; these trends were abrogated in the absence of colonizing commensal bacteria. Irradiated SAMP receiving either wild-type (AKR) or SAMP bone marrow (BM) developed severe ileitis and displayed increased ileal Tlr5 compared with AKR recipients of either SAMP or AKR bone marrow, neither of which conferred ileitis, suggesting that elevated TLR5 in native SAMP is derived primarily from a nonhematopoietic (e.g., epithelial) source. Indeed, ileal epithelial TLR5 in preinflamed SAMP was increased compared with age-matched AKR and germ-free SAMP. TLR5-specific ex vivo activation of SAMP ileal tissues decreased epithelial barrier resistance, indicative of increased permeability, and was accompanied by altered expression of the tight junction proteins, claudin-3, occludin, and zonula occludens-1. Conclusions: Our results provide evidence that aberrant, elevated TLR5 expression is present in the ileal epithelium of SAMP mice, is augmented in the presence of the gut microbiome, and that TLR5 activation in response to bacterial flagellin results in a deficiency to maintain appropriate epithelial barrier integrity. Together, these findings represent a potential mechanistic pathway leading to the exacerbation and perpetuation of chronic gut inflammation in experimental ileitis and possibly, in patients with Crohns disease.

Collaboration


Dive into the Wendy A. Goodman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fabio Cominelli

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Loris Riccardo Lopetuso

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Antonio Gasbarrini

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Franco Scaldaferri

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

C. De Salvo

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

L. Di Martino

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin D. Cooper

University Hospitals of Cleveland

View shared research outputs
Top Co-Authors

Avatar

Rekha R. Garg

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge