Richard J. von Furstenberg
University of North Carolina at Chapel Hill
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Richard J. von Furstenberg.
Gut | 2014
Michael T. Shanahan; Ian M. Carroll; Emily Grossniklaus; Andrew M. White; Richard J. von Furstenberg; Roshonda Barner; Anthony A. Fodor; Susan J. Henning; R. Balfour Sartor; Ajay S. Gulati
Objective Although polymorphisms of the NOD2 gene predispose to the development of ileal Crohns disease, the precise mechanisms of this increased susceptibility remain unclear. Previous work has shown that transcript expression of the Paneth cell (PC) antimicrobial peptides (AMPs) α-defensin 4 and α-defensin-related sequence 10 are selectively decreased in Nod2−/− mice. However, the specific mouse background used in this previous study is unclear. In light of recent evidence suggesting that mouse strain strongly influences PC antimicrobial activity, we sought to characterise PC AMP function in commercially available Nod2−/− mice on a C57BL/6 (B6) background. Specifically, we hypothesised that Nod2−/− B6 mice would display reduced AMP expression and activity. Design Wild-type (WT) and Nod2−/− B6 ileal AMP expression was assessed via real-time PCR, acid urea polyacrylamide gel electrophoresis and mass spectrometry. PCs were enumerated using flow cytometry. Functionally, α-defensin bactericidal activity was evaluated using a gel-overlay antimicrobial assay. Faecal microbial composition was determined using 454-sequencing of the bacterial 16S gene in cohoused WT and Nod2−/− littermates. Results WT and Nod2−/− B6 mice displayed similar PC AMP expression patterns, equivalent α-defensin profiles, and identical antimicrobial activity against commensal and pathogenic bacterial strains. Furthermore, minimal differences in gut microbial composition were detected between the two cohoused, littermate mouse groups. Conclusions Our data reveal that Nod2 does not directly regulate PC antimicrobial activity in B6 mice. Moreover, we demonstrate that previously reported Nod2-dependent influences on gut microbial composition may be overcome by environmental factors, such as cohousing with WT littermates.
Nature | 2017
Kelley S. Yan; Claudia Y. Janda; Junlei Chang; Grace X. Y. Zheng; Kathryn A. Larkin; Vincent C. Luca; Luis A. Chia; Amanda T. Mah; Arnold Han; Jessica M. Terry; Akifumi Ootani; Kelly Roelf; Mark Lee; Jenny Yuan; Xiao Li; Christopher R. Bolen; Julie Wilhelmy; Paige S. Davies; Hiroo Ueno; Richard J. von Furstenberg; Phillip Belgrader; Solongo B. Ziraldo; Heather Ordonez; Susan J. Henning; Melissa H. Wong; Michael Snyder; Irving L. Weissman; Aaron J. W. Hsueh; Tarjei S. Mikkelsen; K. Christopher Garcia
The canonical Wnt/β-catenin signalling pathway governs diverse developmental, homeostatic and pathological processes. Palmitoylated Wnt ligands engage cell-surface frizzled (FZD) receptors and LRP5 and LRP6 co-receptors, enabling β-catenin nuclear translocation and TCF/LEF-dependent gene transactivation. Mutations in Wnt downstream signalling components have revealed diverse functions thought to be carried out by Wnt ligands themselves. However, redundancy between the 19 mammalian Wnt proteins and 10 FZD receptors and Wnt hydrophobicity have made it difficult to attribute these functions directly to Wnt ligands. For example, individual mutations in Wnt ligands have not revealed homeostatic phenotypes in the intestinal epithelium—an archetypal canonical, Wnt pathway-dependent, rapidly self-renewing tissue, the regeneration of which is fueled by proliferative crypt Lgr5+ intestinal stem cells (ISCs). R-spondin ligands (RSPO1–RSPO4) engage distinct LGR4–LGR6, RNF43 and ZNRF3 receptor classes, markedly potentiate canonical Wnt/β-catenin signalling, and induce intestinal organoid growth in vitro and Lgr5+ ISCs in vivo. However, the interchangeability, functional cooperation and relative contributions of Wnt versus RSPO ligands to in vivo canonical Wnt signalling and ISC biology remain unknown. Here we identify the functional roles of Wnt and RSPO ligands in the intestinal crypt stem-cell niche. We show that the default fate of Lgr5+ ISCs is to differentiate, unless both RSPO and Wnt ligands are present. However, gain-of-function studies using RSPO ligands and a new non-lipidated Wnt analogue reveal that these ligands have qualitatively distinct, non-interchangeable roles in ISCs. Wnt proteins are unable to induce Lgr5+ ISC self-renewal, but instead confer a basal competency by maintaining RSPO receptor expression that enables RSPO ligands to actively drive and specify the extent of stem-cell expansion. This functionally non-equivalent yet cooperative interaction between Wnt and RSPO ligands establishes a molecular precedent for regulation of mammalian stem cells by distinct priming and self-renewal factors, with broad implications for precise control of tissue regeneration.
PLOS ONE | 2012
Ajay S. Gulati; Michael T. Shanahan; Janelle C. Arthur; Emily Grossniklaus; Richard J. von Furstenberg; Lieselotte Kreuk; Susan J. Henning; Christian Jobin; R. Balfour Sartor
Background Increasing evidence supports the central role of Paneth cells in maintaining intestinal host-microbial homeostasis. However, the direct impact of host genotype on Paneth cell function remains unclear. Here, we characterize key differences in Paneth cell function and intestinal microbial composition in two widely utilized, genetically distinct mouse strains (C57BL/6 and 129/SvEv). In doing so, we demonstrate critical influences of host genotype on Paneth cell activity and the enteric microbiota. Methodology and Principal Findings Paneth cell numbers were determined by flow cytometry. Antimicrobial peptide (AMP) expression was evaluated using quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR), acid urea-polyacrylamide gel electrophoresis, and mass spectrometry. Effects of mouse background on microbial composition were assessed by reciprocal colonization of germ-free mice from both background strains, followed by compositional analysis of resultant gut bacterial communities using terminal restriction fragment length polymorphism analysis and 16 S qPCR. Our results revealed that 129/SvEv mice possessed fewer Paneth cells and a divergent AMP profile relative to C57BL/6 counterparts. Novel 129/SvEv á-defensin peptides were identified, including Defa2/18v, Defa11, Defa16, and Defa18. Host genotype profoundly affected the global profile of the intestinal microbiota, while both source and host factors were found to influence specific bacterial groups. Interestingly, ileal α-defensins from 129/SvEv mice displayed attenuated antimicrobial activity against pro-inflammatory E. coli strains, a bacterial species found to be expanded in these animals. Conclusions and Significance This work establishes the important impact of host genotype on Paneth cell function and the composition of the intestinal microbiota. It further identifies specific AMP and microbial alterations in two commonly used inbred mouse strains that have varying susceptibilities to a variety of disorders, ranging from obesity to intestinal inflammation. This will be critical for future studies utilizing these murine backgrounds to study the effects of Paneth cells and the intestinal microbiota on host health and disease.
American Journal of Physiology-gastrointestinal and Liver Physiology | 2012
Jeffrey B. King; Richard J. von Furstenberg; Brian J. Smith; Kirk K. McNaughton; Joseph A. Galanko; Susan J. Henning
A growing body of evidence has implicated CD24, a cell-surface protein, as a marker of colorectal cancer stem cells and target for antitumor therapy, although its presence in normal colonic epithelium has not been fully characterized. Previously, our group showed that CD24-based cell sorting can be used to isolate a fraction of murine small intestinal epithelial cells enriched in actively cycling stem cells. Similarly, we hypothesized that CD24-based isolation of colonic epithelial cells would generate a fraction enriched in actively cycling colonic epithelial stem cells (CESCs). Immunohistochemistry performed on mouse colonic tissue showed CD24 expression in the bottom half of proximal colon crypts and the crypt base in the distal colon. This pattern of distribution was similar to enhanced green fluorescent protein (EGFP) expression in Lgr5-EGFP mice. Areas expressing CD24 contained actively proliferating cells as determined by ethynyl deoxyuridine (EdU) incorporation, with a distinct difference between the proximal colon, where EdU-labeled cells were most frequent in the midcrypt, and the distal colon, where they were primarily at the crypt base. Flow cytometric analyses of single epithelial cells, identified by epithelial cell adhesion molecule (EpCAM) positivity, from mouse colon revealed an actively cycling CD24(+) fraction that contained the majority of Lgr5-EGFP(+) putative CESCs. Transcript analysis by quantitative RT-PCR confirmed enrichment of active CESC markers [leucine-rich-repeat-containing G protein-coupled receptor 5 (Lgr5), ephrin type B receptor 2 (EphB2), and CD166] in the CD24(+)EpCAM(+) fraction but also showed enrichment of quiescent CESC markers [leucine-rich repeats and immunoglobin domains (Lrig), doublecortin and calmodulin kinase-like 1 (DCAMKL-1), and murine telomerase reverse transcriptase (mTert)]. We conclude that CD24-based sorting in wild-type mice isolates a colonic epithelial fraction highly enriched in actively cycling and quiescent putative CESCs. Furthermore, the presence of CD24 expression in normal colonic epithelium may have important implications for the use of anti-CD24-based colorectal cancer therapies.
PLOS ONE | 2015
Julie M. Davies; Rebeca Santaolalla; Richard J. von Furstenberg; Susan J. Henning; Maria T. Abreu
Background & Aims The intestinal epithelium is the first line of defense against enteric pathogens. We investigated the response of small intestinal and colonic crypt cultures to a panel of toll-like receptor ligands to assess the impact of microbial pattern recognition on epithelial growth. Methods Primary murine jejunal enteroids and colonoids were cultured with lipopeptide Pam3CSK4, lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (Poly I:C) for 4 to 6 days. Surface area, budding and survival were assessed. Proliferation and numbers of lysozyme positive cells were quantified by flow cytometry. Gene expression was assessed by Nanostring and qRT-PCR. Results Exposure to Pam3CSK4 and LPS had minimal impact on either enteroids or colonoids. In contrast, Poly I:C increased the surface area of enteroids, while colonoids demonstrated decreased budding. Survival was decreased by Poly I:C in enteroids but not in colonoids. Both enteroids and colonoids exhibited upregulated gene expression of chemokines, but these were increased in magnitude in enteroids. Decreases in gene expression associated with epithelial differentiation and lysozyme positive cells were more apparent in enteroids than in colonoids. Baseline gene expression between enteroids and colonoids differed markedly in levels of stem cell and inflammatory markers. The changes in morphology induced by Poly I:C were mediated by the toll-like receptor adaptor molecule 1 (Ticam1) in enteroids but not in colonoids. Conclusions Poly I:C alters the molecular program of epithelial cells and shifts from absorption and digestion towards defense and inflammation. Diversity of responses to microbial patterns in enteroids and colonoids may underlie differences in susceptibility to infection along the intestinal tract.
Gut microbes | 2018
Alexi A. Schoenborn; Richard J. von Furstenberg; Smrithi Valsaraj; Farah S. Hussain; Molly Stein; Michael T. Shanahan; Susan J. Henning; Ajay S. Gulati
ABSTRACT Paneth cells (PCs) are epithelial cells found in the small intestine, next to intestinal stem cells (ISCs) at the base of the crypts. PCs secrete antimicrobial peptides (AMPs) that regulate the commensal gut microbiota. In contrast, little is known regarding how the enteric microbiota reciprocally influences PC function. In this study, we sought to characterize the impact of the enteric microbiota on PC biology in the mouse small intestine. This was done by first enumerating jejunal PCs in germ-free (GF) versus conventionally raised (CR) mice. We next evaluated the possible functional consequences of altered PC biology in these experimental groups by assessing epithelial proliferation, ISC numbers, and the production of AMPs. We found that PC numbers were significantly increased in CR versus GF mice; however, there were no differences in ISC numbers or cycling activity between groups. Of the AMPs assessed, only Reg3γ transcript expression was significantly increased in CR mice. Intriguingly, this increase was abrogated in cultured CR versus GF enteroids, and could not be re-induced with various bacterial ligands. Our findings demonstrate the enteric microbiota regulates PC function by increasing PC numbers and inducing Reg3γ expression, though the latter effect may not involve direct interactions between bacteria and the intestinal epithelium. In contrast, the enteric microbiota does not appear to regulate jejunal ISC census and proliferation. These are critical findings for investigators using GF mice and the enteroid system to study PC and ISC biology.
Gastroenterology | 2015
Julie M. Davies; Rebeca Santaolalla; Richard J. von Furstenberg; Susan J. Henning; Maria T. Abreu
The small intestine and colon differ greatly in both function and exposure to microbial products. Specific studies are needed to assess the response of microbial signaling in the small intestine versus the colon. Double-stranded RNAs (ds-RNA) are primarily viral products, but can also be derived from bacteria. Acute enteric viruses usually impact the small intestine, but viruses have also been investigated in inflammatory bowel disease and in post-infective irritable bowel syndrome and viruses are frequently found in colon cancer tissue. As such, understanding differences in the response of epithelial cells from the small intestine and colon to exposure to viral products will be important for dissecting the impact of infection at the different anatomical locations. Using newly developed culture techniques for growing primary enteroids and colonoids, we sought to compare the responses of enteroids versus colonoids to stimulation with the synthetic ds-RNA polyinosinic-polycytidylic acid (poly I:C). Murine crypts from jejunum or colon were plated in Matrigel for two days prior to the addition of poly I:C for the duration of culture. Stimulation of enteroids with poly I:C significantly altered the surface area but decreased the number of surviving enteroids compared to unstimulated. In colonoids, stimulation with poly I:C resulted in a significant decrease in bud count, but did not impact the area or survival of the organoids. Gene expression measured by the probe-based Nanostring technology in enteroids following poly I:C stimulation showed significantly decreased expression of stem cell markers including: Sox9, Lgr5, Dclk1, Tert and Lrig1. Additionally, differentiation markers Sis and Muc2 were significantly decreased. In contrast, fewer genes were significantly impacted by poly I:C stimulation in colonoids. Stem cell markers were not altered by poly I:C stimulation in colonoids. As expected, in both enteroids and colonoids inflammatory markers were significantly increased in response to poly I:C stimulation. Additionally, pro-apoptotic genes in both enteroids and colonoids were decreased following poly I:C stimulation. Comparing the response of enteroids and colonoids to poly I:C stimulation we found differences in the magnitude or direction of change in several classes of genes. These distinctions may derive frombaseline differences in expression levels. Enteroids had significantly increased expression of stem cell markers at baseline compared to colonoids, while colonoids had increased levels of inflammatory markers and toll-like receptors. Together, these data indicate that important functional differences exist between enteroids and colonoids at baseline and after viral product stimulation providing evidence which may help explain the different anatomical responses to viral infection throughout the intestinal tract.
American Journal of Physiology-gastrointestinal and Liver Physiology | 2011
Richard J. von Furstenberg; Ajay S. Gulati; Anand Baxi; Jason M. Doherty; Thaddeus S. Stappenbeck; Adam D. Gracz; Scott T. Magness; Susan J. Henning
Cell Stem Cell | 2017
Kelley S. Yan; Olivier Gevaert; Grace X. Y. Zheng; Benedict Anchang; Christopher S. Probert; Kathryn A. Larkin; Paige S. Davies; Zhuan fen Cheng; John S. Kaddis; Arnold Han; Kelly Roelf; Ruben I. Calderon; Esther Cynn; Xiaoyi Hu; Komal Mandleywala; Julie Wilhelmy; Susan M. Grimes; David C Corney; Stéphane C. Boutet; Jessica M. Terry; Phillip Belgrader; Solongo B. Ziraldo; Tarjei S. Mikkelsen; Fengchao Wang; Richard J. von Furstenberg; Nicholas R. Smith; Parthasarathy Chandrakesan; Randal May; Mary Ann S. Chrissy; Rajan Jain
Stem Cell Research | 2014
Richard J. von Furstenberg; Simon J.A. Buczacki; Brian J. Smith; Kristen Seiler; Douglas J. Winton; Susan J. Henning