Network


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

Hotspot


Dive into the research topics where Joel V. Weinstock is active.

Publication


Featured researches published by Joel V. Weinstock.


European Journal of Immunology | 2004

Heligmosomoides polygyrus inhibits established colitis in IL-10-deficient mice

David E. Elliott; Tommy Setiawan; Ahmed Metwali; Arthur M. Blum; Joseph F. Urban; Joel V. Weinstock

Inflammatory bowel disease (IBD) is prevalent in industrialized countries, but rare in less‐developed countries. Helminths, common in less‐developed countries, may induce immunoregulatory circuits protective against IBD. IL‐10–/– mice given piroxicam develop severe and persistent colitis. Lamina propria mononuclear cells from colitic IL‐10–/– mice released IFN‐γ and IL‐12. The ongoing piroxicam‐induced colitis could be partially blocked with anti‐IL‐12 monoclonal antibody suggesting that the inflammation was at least partly IL‐12 dependent. Colonization of piroxicam‐treated colitic IL‐10–/– mice with Heligmosomoides polygyrus (an intestinal helminth) suppressed established inflammation and inhibited mucosal IL‐12 and IFN‐γ production. H. polygyrus augmented mucosal IL‐13, but not IL‐4 or IL‐5 production. Transfer of mesenteric lymph node (MLN) T cells from IL‐10–/– animals harboring H. polygyrus into colitic IL‐10–/– recipients inhibited colitis. MLN T cells from worm‐free mice did not. Foxp3 (scurfin) drives regulatory T cell function. H. polygyrus enhanced Foxp3 mRNA expression in MLN T cells that had regulatory activity. This suggests that H. polygyrus inhibits ongoing IL‐10–/– colitis in part through blocking mucosal Th1 cytokine production. Resolution of inflammation is associated with increased IL‐13 production and can be adoptively transferred by MLN T cells.


The FASEB Journal | 2000

Does the failure to acquire helminthic parasites predispose to Crohn’s disease?

David E. Elliott; Joe F. Urban; Curtis K. Argo; Joel V. Weinstock

Two polarized patterns (Th1 and Th2) of cytokines regulate inflammatory responses. Each cytokine pattern inhibits production of the opposing pattern. Lymphocytes from inflamed intestine due to Crohns disease secrete a Th1 pattern of cytokines. Crohns disease is most prevalent in highly industrialized countries with temperate climates. It occurs rarely in tropical third world countries with poor sanitation. We propose that exposure to an environmental agent predisposes individuals to Crohns disease. Parasitic worms (helminths) are common in tropical climates and in populations subject to crowding and poor sanitation. Children are most subject to helminthic colonization. Many helminths live within or migrate through the human gut where they interact with the mucosal immune system. The host mounts a mucosal response that includes Th2 cytokine production limiting helminthic colonization. Helminths and their eggs probably are the most potent stimulators of mucosal Th2 responses. The Th2 response provoked by parasitic worms can modulate immune reactions to unrelated parasitic, bacterial, and viral infections. Many people in developed countries now live in increasingly hygienic environments, avoiding exposure to helminths. Perhaps failure to acquire these parasites and experience mucosal Th2 conditioning predisposes to Crohns disease, which is an overly active Th1 inflammation.— Elliott, D. E., Urban, J. F., Jr., Argo, C. K., Weinstock, J. V. FASEB J. 14, 1848–1855 (2000)


Inflammatory Bowel Diseases | 2010

Alteration of the murine gut microbiota during infection with the parasitic helminth, Heligmosomoides polygyrus

Seth T. Walk; Arthur M. Blum; Sarah A. Ewing; Joel V. Weinstock; Vincent B. Young

Background: In a murine model of inflammatory bowel disease (IBD), treatment of colitis in IL‐10 gene‐deficient mice with the parasitic helminth Heligmosomoides polygyrus ameliorates colonic inflammation. The cellular and molecular mechanisms driving this therapeutic host response are being studied vigorously. One proposed mechanism is that H. polygyrus infection favors the outgrowth or suppression of certain bacteria, which in turn help modulate host immunity. Methods: To quantify the effect of H. polygyrus infection on the composition of the gastrointestinal (GI) tract microbiota, we conducted two independent microbial ecology analyses of C57BL/6 mice. We obtained and analyzed 3,353 bacterial 16S rRNA encoding gene sequences from the ileum and cecum of infected and uninfected mice as well as incective H. polygyrus larvae at the outset of the second experiment and adult worms taken directly from the mouse duodenum at the end of the second experiment. Results: We found that a significant shift in the abundance and relative distribution of bacterial species in the ileum of mice is associated with H. polygyrus infection. Members of the bacterial family Lactobacillaceae significantly increased in abundance in the ileum of infected mice reproducibly in two independent experiments despite having different microbiotas present at the outset of each experiment. Conclusions: These data support the concept that helminth infection shifts the composition of intestinal bacteria. The clinical consequences of these shifts in intestinal flora are yet to be explored. (Inflamm Bowel Dis 2010)


Science | 2016

Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut

Michael R. Howitt; Sydney Lavoie; Monia Michaud; Arthur M. Blum; Sara V. Tran; Joel V. Weinstock; Carey Ann Gallini; Kevin Redding; Robert F. Margolskee; Lisa C. Osborne; David Artis; Wendy S. Garrett

Tuft cells help contain parasites Trillions of microbes inhabit our guts, including worms and other parasites. Epithelial cells that line the gut orchestrate parasite-targeted immune responses. Howitt et al. now identify a key cellular player in immunity to parasites: tuft cells (see the Perspective by Harris). Tuft cells make up a small fraction of gut epithelial cells but expand when parasites colonize or infect the gut. Parasites cause tuft cells to secrete large amounts of interleukin-25, a key cytokine for parasite clearance that also indirectly feeds back on tuft cells to expand their numbers. Tuft cells express chemosensory signaling machinery: disrupting this blocked parasite-triggered tuft cell expansion and weakened the ability of mice to control a parasitic infection. Science, this issue p. 1329; see also p. 1264 Gut epithelial tuft cells are key players in mucosal immune responses against parasites. [Also see Perspective by Harris] The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.


Inflammatory Bowel Diseases | 2009

Helminths and the IBD hygiene hypothesis

Joel V. Weinstock; David E. Elliott

&NA; Helminths are parasitic animals that have evolved over 100,000,000 years to live in the intestinal track or other locations of their hosts. Colonization of humans with these organisms was nearly universal until the early 20th century. More than 1,000,000,000 people in less developed countries carry helminths even today. Helminths must quell their hosts immune system to successfully colonize. It is likely that helminths sense hostile changes in the local host environment and take action to control such responses. Inflammatory bowel disease (IBD) probably results from an inappropriately vigorous immune response to contents of the intestinal lumen. Environmental factors strongly affect the risk for IBD. People living in less developed countries are protected from IBD. The “IBD hygiene hypothesis” states that raising children in extremely hygienic environments negatively affects immune development, which predisposes them to immunological diseases like IBD later in life. Modern day absence of exposure to intestinal helminths appears to be an important environmental factor contributing to development of these illnesses. Helminths interact with both host innate and adoptive immunity to stimulate immune regulatory circuitry and to dampen effector pathways that drive aberrant inflammation. The first prototype worm therapies directed against immunological diseases are now under study in the United States and various countries around the world. Additional studies are in the advanced planning stage.


Journal of Immunology | 2006

Intestinal helminths protect in a murine model of asthma.

Kunihiko Kitagaki; Thomas R. Businga; Doina Racila; David E. Elliott; Joel V. Weinstock; Joel N. Kline

Underdeveloped nations are relatively protected from the worldwide asthma epidemic; the hygiene hypothesis suggests this is due to suppression of Th2-mediated inflammation by increased exposure to pathogens and their products. Although microbial exposures can promote Th2-suppressing Th1 responses, even Th2-skewing infections, such as helminths, appear to suppress atopy, suggesting an alternate explanation for these observations. To investigate whether induction of regulatory responses by helminths may counter allergic inflammation, we examined the effects of helminth infection in a murine model of atopic asthma. We chose Heligosomoides polygyrus, a gastrointestinal nematode, as the experimental helminth; this worm does not enter the lung in its life cycle. We found that H. polygyrus infection suppressed allergen-induced airway eosinophilia, bronchial hyperreactivity, and in vitro allergen-recall Th2 responses in an IL-10-dependent manner; total and OVA-specific IgE, however, were increased by worm infection. Finally, helminth-infected mice were protected against eosinophilic inflammation induced by adoptive transfer of OVA-stimulated CD4+ cells, and transfer of cells from helminth-infected/OVA-exposed mice suppressed OVA-induced eosinophilic inflammation, suggesting a role for regulatory cells. Increased CD4+CD25+Foxp3+ cells were found in thoracic lymph nodes of helminth-infected/OVA-exposed mice. Helminthic colonization appears to protect against asthma and atopic disorders; the regulatory cytokine, IL-10, may be a critical player.


Journal of Immunology | 2001

Inducible Costimulator Regulates Th2-Mediated Inflammation, but Not Th2 Differentiation, in a Model of Allergic Airway Disease

Amanda G. Tesciuba; Sumit K. Subudhi; Russell P. Rother; Susan J. Faas; Aric M. Frantz; David Elliot; Joel V. Weinstock; Louis A. Matis; Jeffrey A. Bluestone; Anne I. Sperling

A novel costimulatory molecule expressed on activated T cells, inducible costimulator (ICOS), and its ligand, B7-related protein-1 (B7RP-1), were recently identified. ICOS costimulation leads to the induction of Th2 cytokines without augmentation of IL-2 production, suggesting a role for ICOS in Th2 cell differentiation and expansion. In the present study, a soluble form of murine ICOS, ICOS-Ig, was used to block ICOS/B7RP-1 interactions in a Th2 model of allergic airway disease. In this model, mice are sensitized with inactivated Schistosoma mansoni eggs and are subsequently challenged with soluble S. mansoni egg Ag directly in the airways. Treatment of C57BL/6 mice with ICOS-Ig during sensitization and challenge attenuated airway inflammation, as demonstrated by a decrease in cellular infiltration into the lung tissue and airways, as well as by a decrease in local IL-5 production. These inhibitory effects were not due to a lack of T cell priming nor to a defect in Th2 differentiation. In addition, blockade of ICOS/B7RP-1 interactions during ex vivo restimulation of lung Th2 effector cells prevented cytokine production. Thus, blockade of ICOS signaling can significantly reduce airway inflammation without affecting Th2 differentiation in this model of allergic airway disease.


Annals of the New York Academy of Sciences | 2012

Helminth–host immunological interactions: prevention and control of immune-mediated diseases

David E. Elliott; Joel V. Weinstock

Exposure to commensal and pathogenic organisms strongly influences our immune system. Exposure to helminths was frequent before humans constructed their current highly hygienic environment. Today, in highly industrialized countries, contact between humans and helminths is rare. Congruent with the decline in helminth infections is an increase in the prevalence of autoimmune and inflammatory disease. It is possible that exclusion of helminths from the environment has permitted the emergence of immune‐mediated disease. We review the protective effects of helminths on expression of inflammatory bowel disease, multiple sclerosis, and animal models of these and other inflammatory diseases. We also review the immune pathways altered by helminths that may afford protection from these illnesses. Helminth exposure tends to inhibit IFN‐γ and IL‐17 production, promote IL‐4, IL‐10, and TGF‐β release, induce CD4+ T cell Foxp3 expression, and generate regulatory macrophages, dendritic cells, and B cells. Helminths enable protective pathways that may vary by specific species and disease model. Helminths or their products likely have therapeutic potential to control or prevent immune‐mediated illness.


Inflammatory Bowel Diseases | 2009

Therapeutic potential of helminth soluble proteins in TNBS‐induced colitis in mice

Nathalie E. Ruyssers; Benedicte Y. De Winter; Joris G. De Man; Alex Loukas; Mark S. Pearson; Joel V. Weinstock; Rita M. Van den Bossche; Wim Martinet; Paul A. Pelckmans; Tom G. Moreels

Background: The hygiene hypothesis suggests an inverse relationship between the incidence of parasitic infections and chronic inflammatory bowel diseases (IBD). We investigated the therapeutic potential of Schistosoma mansoni and Ancylostoma caninum soluble proteins on experimental colitis in mice. Methods: Colitis was induced by intrarectal administration of 10 mg trinitrobenzene sulfonic acid (TNBS) in 30% ethanol. Six hours after TNBS injection, mice were treated intraperitoneally with helminth proteins. Three days later, colonic inflammation was scored based on 5 inflammatory parameters: clinical disease activity, macroscopic and microscopic inflammation score, extent of inflammation, and myeloperoxidase (MPO) activity. To determine immunological pathways induced by S. mansoni proteins we measured cytokine profiles of T‐lymphocytes from colon, mesenteric lymph nodes (MLN), and spleen by real‐time reverse‐transcriptase polymerase chain reaction (RT‐PCR). Results: Control mice showed no signs of inflammation, whereas all inflammatory parameters were significantly increased in mice with colitis. Treatment of mice with colitis with S. mansoni or A. caninum proteins decreased the macroscopic inflammation score, extent of inflammation, and MPO activity. Immunologically, induction of colitis significantly increased expression of IFN‐γ mRNA in the inflamed colon. Treatment with S. mansoni proteins caused a decrease of proinflammatory cytokines (IFN‐γ, IL‐17) in colon and MLN, whereas the production of regulatory cytokines (IL‐10, TGF‐β) increased significantly in colon tissue. Conclusions: Treatment with proteins of S. mansoni and A. caninum ameliorated TNBS‐induced colitis in mice. S. mansoni proteins increased mRNA expression of regulatory cytokines while suppressing expression of proinflammatory cytokines. Therefore, we suggest a therapeutic potential for helminth proteins in the treatment of IBD. (Inflamm Bowel Dis 2009)


Journal of Neuroimmunology | 1994

Eosinophils within the healthy or inflamed human intestine produce substance P and vasoactive intestinal peptide

Ahmed Metwali; Arthur M. Blum; Luca Ferraris; John S. Klein; Claudio Fiocchi; Joel V. Weinstock

The purpose of this study was to show if inflammatory cells within healthy or diseased human intestinal mucosa produce some regulatory neuropeptides. First, inflammatory cells were isolated from the intestinal lamina propria of 11 patients with ulcerative colitis or Crohns disease. Also collected were cells from anatomically normal intestine derived from five patients requiring bowel resection for diseases not related to inflammatory bowel disease. Extracts of these isolated cells contained authentic substance P (SP) and vasoactive intestinal peptide (VIP) as shown by RIA and their elution profiles on HPLC. Immunostaining of cells from nine of 13 additional patients localized immunoreactive SP and VIP to secretory granules within most mucosal eosinophils. No other cell types stained positive. Messenger RNA encoding SP and VIP was localized to lamina propria eosinophils by in situ hybridization. Mucosa inflammatory cells, from eight of nine more patients, cultured in vitro, released detectable amounts of VIP, but not SP. It is concluded that intestinal eosinophils produce SP and VIP. Since the eosinophils store and release more VIP than SP, it is possible that VIP is the preferred secretory product.

Collaboration


Dive into the Joel V. Weinstock's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matyas Sandor

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph F. Urban

United States Department of Agriculture

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge