Network


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

Hotspot


Dive into the research topics where L. Wayne Duck is active.

Publication


Featured researches published by L. Wayne Duck.


Inflammatory Bowel Diseases | 2007

Isolation of Flagellated Bacteria Implicated in Crohn's Disease

L. Wayne Duck; Mark R. Walter; Jan Novak; Denise Kelly; Maurizio Tomasi; Yingzi Cong; Charles O. Elson

Background: Serologic expression cloning has identified flagellins of the intestinal microbiota as immunodominant antigens in experimental colitis in mice and in individuals with Crohns disease (CD). The present study was done to identify the microbial source of such flagellins. Methods: Using a variety of isolation and culture approaches, a number of previously unknown flagellated bacteria were isolated. Based on 16S ribosomal DNA sequences, these bacteria fall into the family Lachnospiraceae of the phylum Firmicutes. Results: Serum IgG from patients with CD and from mice with colitis reacted to the flagellins of these bacteria, and only their flagellins, whereas serum IgG from controls did not. The sequence of these flagellins demonstrate conserved amino‐ and carboxy‐terminal domains that cluster phylogenetically and have a predicted 3D structure similar to Salmonella fliC, including an intact TLR5 binding site. The flagellin of 1 of these bacteria was likely O‐glycosylated. Conclusions: The conserved immune response in both mouse and human to these previously unknown flagellins of the microbiota indicate that they play an important role in host–microbe interactions in the intestine. (Inflamm Bowel Dis 2007)


Endocrinology | 1998

Potential Regulatory Roles for G Protein-Coupled Receptor Kinases and β-Arrestins in Gonadotropin-Releasing Hormone Receptor Signaling1

Jimmy D. Neill; L. Wayne Duck; Lois C. Musgrove; Jeffrey C. Sellers

GnRH stimulates gonadotropin secretion, which desensitizes unless the releasing hormone is secreted or administered in a pulsatile fashion. The mechanism of desensitization is unknown, but as the GnRH receptor is G protein coupled, it might involve G protein-coupled receptor kinases (GRKs). Such kinases phosphorylate the intracellular regions of seven-transmembrane receptors, permitting β-arrestin to bind, which prevents the receptor from activating G proteins. Here, we tested the effect of GRKs and β-arrestins on GnRH-induced inositol trisphosphate (IP3) production in COS cells transfected with the GnRH receptor complementary DNA. GRK2, -3, and -6 overexpression inhibited IP3 production by 50–75% during the 30 sec of GnRH treatment. Coexpression of GRK2 and β-arrestin-2 suppressed GnRH-induced IP3 production more than that of either alone. Immunocytochemical staining of rat anterior pituitary revealed that all cells expressed GRK2, -3, and -6; all cells also expressed theβ -arrestins. Western blots on cy...


European Journal of Immunology | 2014

Downregulation of microRNA-107 in intestinal CD11c(+) myeloid cells in response to microbiota and proinflammatory cytokines increases IL-23p19 expression.

Xiaochang Xue; Anthony T. Cao; Xiaocang Cao; Suxia Yao; Eric D. Carlsen; Lynn Soong; Chang Gong Liu; Xiuping Liu; Zhanju Liu; L. Wayne Duck; Charles O. Elson; Yingzi Cong

Commensal flora plays an important role in the development of the mucosal immune system and in maintaining intestinal homeostasis. However, the mechanisms involved in regulation of host‐microbiota interaction are still not completely understood. In this study, we examined how microbiota and intestinal inflammatory conditions regulate host microRNA expression and observed lower microRNA‐107 (miR‐107) expression in the inflamed intestines of colitic mice, compared with that in normal control mice. miR‐107 was predominantly reduced in epithelial cells and CD11c+ myeloid cells including dendritic cells and macrophages in the inflamed intestines. We demonstrate that IL‐6, IFN‐γ, and TNF‐α downregulated, whereas TGF‐β promoted, miR‐107 expression. In addition, miR‐107 expression was higher in the intestines of germ‐free mice than in mice housed under specific pathogen‐free conditions, and the presence of microbiota downregulated miR‐107 expression in DCs and macrophages in a MyD88‐ and NF‐κB‐dependent manner. We determined that the ectopic expression of miR‐107 specifically repressed the expression of IL‐23p19, a key molecule in innate immune responses to commensal bacteria. We concluded that regulation of miR‐107 by intestinal microbiota and proinflammatory cytokine serve as an important pathway for maintaining intestinal homeostasis.


Molecular and Cellular Endocrinology | 1997

Epitope-tagged gonadotropin-releasing hormone receptors heterologously-expressed in mammalian (COS-1) and insect (Sf9) cells.

Jimmy D. Neill; Jeffrey C. Sellers; Lois C. Musgrove; L. Wayne Duck

The molecular cloning and nucleotide sequencing of the gonadotropin-releasing hormone (GnRH) receptor represented an enhanced step in the experimental effort to understand this key molecule in the reproductive process at a cell and molecular level. A subsequent step in this broad effort is heterologous expression of the receptor in model cell systems for studies of signal transduction and desensitization, processes that may require immunologic detection of the receptor. Therefore, the GnRH receptor was tagged at its N-terminus using recombinant DNA procedures with the HA-1 epitope that is bound by a monoclonal antibody (12CA5). COS-1 cells expressing this receptor bound [(125)I]D-Ala6-desGly10-GnRH ethylamide (GnRH-A) with the expected high affinity (IC(50) = 0.47 nM), and were immunocytochemically stained by the 12CA5 antibody. Signal transduction was demonstrated by GnRH-induced [(3)H]inositol phosphate accumulation in receptor-expressing COS-1 cells. Western blotting of COS-1 cell membranes expressing the receptor revealed protein bands at 67, 57, and 32 kDa. Immunoprecipitation occurred when the solubilized receptor from COS-1 cell membranes was reacted with 12CA5 antibody and anti-mouse IgG Sepharose, and the presence of the receptor demonstrated either by its binding of [(125)I]GnRH-A or by its detection on Western blots. Desensitization of inositol 1,4,5-trisphosphate (IP(3)) production by N-epitope-tagged GnRH receptor expressing COS-1 cells was evoked by a five min GnRH pretreatment; [(32)P]i labeling of such cells during desensitization followed by immunoprecipitation of the N-epitope-tagged receptor was not associated with receptor phosphorylation. Finally, the epitope tagged receptor was expressed in the high-yield baculovirus/insect Sf9 cell system: the membrane receptor bound [(125)I]GnRH-A with slightly lowered affinity (IC(50) = 1.4 nM), and in Western blots yielded protein bands of 32, 56/57, 69, and 120/140 kDa. The development and validation of these heterologous systems will permit the study of several GnRH receptor-mediated processes that are poorly understood.


The Journal of Allergy and Clinical Immunology | 2015

Human seroreactivity to gut microbiota antigens

Benjamin S. Christmann; Thomas R. Abrahamsson; Charles N. Bernstein; L. Wayne Duck; Peter J. Mannon; Göran Berg; Bengt Björkstén; Maria C. Jenmalm; Charles O. Elson

BACKGROUND Although immune responses directed against antigens from the intestinal microbiota are observed in certain diseases, the normal human adaptive immune response to intestinal microbiota is poorly defined. OBJECTIVE Our goal was to assess the adaptive immune response to the intestinal microbiota present in 143 healthy adults and compare this response with the response observed in 52 children and their mothers at risk of having allergic disease. METHODS Human serum was collected from adults and children followed from birth to 7 years of age, and the serum IgG response to a panel of intestinal microbiota antigens was assessed by using a novel protein microarray. RESULTS Nearly every subject tested, regardless of health status, had serum IgG that recognized a common set of antigens. Seroreactivity to the panel of antigens was significantly lower in atopic adults. Healthy infants expressed the highest level of IgG seroreactivity to intestinal microbiota antigens. This adaptive response developed between 6 and 12 months of age and peaked around 2 years of age. Low IgG responses to certain clusters of microbiota antigens during infancy were associated with allergy development during childhood. CONCLUSIONS There is an observed perturbation of the adaptive response to antigens from the microbiota in allergic subjects. These perturbations are observable even in childhood, suggesting that optimal stimulation of the adaptive immune system by the microbiota might be needed to prevent certain immune-mediated diseases.


European Journal of Immunology | 2016

Commensal A4 bacteria inhibit intestinal Th2-cell responses through induction of dendritic cell TGF-β production

Wei Wu; Hou Pu Liu; Feidi Chen; Han Liu; Anthony T. Cao; Suxia Yao; Mingming Sun; Heather L. Evans-Marin; Ye Zhao; Qing Zhao; L. Wayne Duck; Charles O. Elson; Zhanju Liu; Yingzi Cong

It has been shown that while commensal bacteria promote Th1, Th17 and Treg cells in lamina propria (LP) in steady‐state conditions, they suppress mucosal Th2 cells. However, it is still unclear whether there are specific commensal organisms down‐regulating Th2 responses, and the mechanism involved. Here we demonstrate that commensal A4 bacteria, a member of the Lachnospiraceae family, which produce an immunodominant microbiota CBir1 antigen, inhibits LP Th2‐cell development. When transferred into the intestines of RAG−/− mice, CBir1‐specific T cells developed predominately towards Th1 cells and Th17 cells, but to a lesser extent into Th2 cells. The addition of A4 bacterial lysates to CD4+ T‐cell cultures inhibited production of IL‐4. A4 bacteria stimulated dendritic cell production of TGF‐β, and blockade of TGF‐β abrogated A4 bacteria inhibition of Th2‐cell development in vitro and in vivo. Collectively, our data show that A4 bacteria inhibit Th2‐cell differentiation by inducing dendritic cell production of TGF‐β.


Life Sciences | 1999

Dual intracellular pathways in gonadotropin releasing hormone (GNRH) induced desensitization of luteinizing hormone (LH) secretion.

M.Patricia Cassina; Lois C. Musgrove; L. Wayne Duck; Jeffrey C. Sellers; Jimmy D. Neill

The mechanisms of GnRH-induced desensitization of LH secretion are poorly understood. Protein kinase C (PKC) and protein kinase A (PKA) desensitize some receptors of the 7-membrane type, and the GnRH receptor has consensus phosphorylation sites for PKC in the first and third intracellular loops, and a site for PKA in the first intracellular loop. In the first set of experiments we determined whether synthetic peptides representing the three intracellular loops of the receptor could be phosphorylated in vitro by purified PKC and PKA. As compared with a model substrate peptide for PKC, the third intracellular loop was phosphorylated 74% and the first intracellular loop 21%; PKA-phosphorylated the first intracellular loop peptide 17% as well as a model peptide substrate. In the second set of experiments, we used phorbol 12-myristate 13 acetate (PMA), an established PKC stimulator, and cholera toxin (CTX), established to activate the Gs protein and presumed to activate PKA, to treat cultured rat pituitary cells followed by LH measurements. Treatment with both drugs severely impaired GnRH-stimulated LH secretion whereas neither drug alone reduced LH secretion. Dibutyryl cAMP did not duplicate the effects of cholera toxin suggesting that the CTX action could not be explained by an increase in cAMP. These results suggest that more than one intracellular signaling pathway requires activation in order to induce desensitization; one pathway involves PKC and the other involves a pathway stimulated by cholera toxin, presumably Gs protein, which does not involve PKA.


Nature Genetics | 1998

Mutations in gonadotropin-releasing hormone receptor gene cause hypogonadotropic hypogonadism

Lawrence C. Layman; David P. Cohen; Mei Jin; Jun Xie; Zhu Li; Richard H. Reindollar; Shahla Bolbolan; David Bick; Richard R. Sherins; L. Wayne Duck; Lois C. Musgrove; Jeffrey C. Sellers; Jimmy D. Neill


Endocrinology | 1997

Potential Role for a Regulator of G Protein Signaling (RGS3) in Gonadotropin-Releasing Hormone (GnRH) Stimulated Desensitization

Jimmy D. Neill; L. Wayne Duck; Jeffrey C. Sellers; Lois C. Musgrove; Astrid Scheschonka; Kirk M. Druey; John H. Kehrl


Endocrinology | 1999

High Efficiency Method for Gene Transfer in Normal Pituitary Gonadotropes: Adenoviral-Mediated Expression of G Protein-Coupled Receptor Kinase 2 Suppresses Luteinizing Hormone Secretion1

Jimmy D. Neill; Lois C. Musgrove; L. Wayne Duck; Jeffrey C. Sellers

Collaboration


Dive into the L. Wayne Duck's collaboration.

Top Co-Authors

Avatar

Jeffrey C. Sellers

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jimmy D. Neill

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Lois C. Musgrove

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Charles O. Elson

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Yingzi Cong

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Anthony T. Cao

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Benjamin S. Christmann

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Peter J. Mannon

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Suxia Yao

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge