Network


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

Hotspot


Dive into the research topics where W. Christian Wigley is active.

Publication


Featured researches published by W. Christian Wigley.


Journal of Biological Chemistry | 2000

Activity and Regulation of the Centrosome-associated Proteasome

Rosalind P. Fabunmi; W. Christian Wigley; Philip J. Thomas

Regulated proteolysis is important for maintaining appropriate cellular levels of many proteins. The bulk of intracellular protein degradation is catalyzed by the proteasome. Recently, the centrosome was identified as a novel site for concentration of the proteasome and associated regulatory proteins (Wigley, W. C., Fabunmi, R. P., Lee, M. G., Marino, C. R., Muallem, S., DeMartino, G. N., and Thomas, P. J. (1999) J. Cell Biol. 145, 481–490). Here we provide evidence that centrosomes contain the active 26 S proteasome that degrades ubiquitinated-protein and proteasome-specific peptide substrates. Moreover, the centrosomes contain an ubiquitin isopeptidase activity. The proteolytic activity is ATP-dependent and is inhibited by proteasome inhibitors. Notably, treatment of cells with inhibitors of proteasome activity promotes redistribution of the proteasome and associated regulatory proteins to the centrosome independent of an intact microtubule system. These data provide biochemical evidence for active proteasomal complexes at the centrosome, highlighting a novel function for this organizing structure.


Nature Biotechnology | 2001

Protein solubility and folding monitored in vivo by structural complementation of a genetic marker protein

W. Christian Wigley; Rhesa D. Stidham; Nathan M. Smith; John F. Hunt; Philip J. Thomas

Protein misfolding is the basis of a number of human diseases and presents an obstacle to the production of soluble recombinant proteins. We present a general method to assess the solubility and folding of proteins in vivo. The basis of this assay is structural complementation between the α- and ω- fragments of β-galactosidase (β-gal). Fusions of the α-fragment to the C terminus of target proteins with widely varying in vivo folding yield and/or solubility levels, including the Alzheimers amyloid β (Aβ) peptide and a non-amyloidogenic mutant thereof, reveal an unambiguous correlation between β-gal activity and the solubility/folding of the target. Thus, structural complementation provides a means of monitoring protein solubility/misfolding in vivo, and should find utility in the screening for compounds that influence the pathological consequences of these processes.


American Journal of Physiology-renal Physiology | 2011

Bardoxolone methyl (BARD) ameliorates ischemic AKI and increases expression of protective genes Nrf2, PPARγ, and HO-1

Qing Qing Wu; Yanxia Wang; Martin Senitko; Colin J. Meyer; W. Christian Wigley; Deborah A. Ferguson; Eric B. Grossman; Jianlin Chen; Xin J. Zhou; John R. Hartono; Pamela D. Winterberg; Bo Chen; Anapam Agarwal; Christopher Y. Lu

Ischemic acute kidney injury (AKI) triggers expression of adaptive (protective) and maladaptive genes. Agents that increase expression of protective genes should provide a therapeutic benefit. We now report that bardoxolone methyl (BARD) ameliorates ischemic murine AKI as assessed by both renal function and pathology. BARD may exert its beneficial effect by increasing expression of genes previously shown to protect against ischemic AKI, NF-E2-related factor 2 (Nrf2), peroxisome proliferator-activated receptor-γ (PPARγ), and heme oxygenase 1 (HO-1). Although we found that BARD alone or ischemia-reperfusion alone increased expression of these genes, the greatest increase occurred after the combination of both ischemia-reperfusion and BARD. BARD had a different mode of action than other agents that regulate PPARγ and Nrf2. Thus we report that BARD regulates PPARγ, not by acting as a ligand but by increasing the amount of PPARγ mRNA and protein. This should increase ligand-independent effects of PPARγ. Similarly, BARD increased Nrf2 mRNA; this increased Nrf2 protein by mechanisms in addition to the prolongation of Nrf2 protein half-life previously reported. Finally, we localized expression of these protective genes after ischemia and BARD treatment. Using double-immunofluorescence staining for CD31 and Nrf2 or PPARγ, we found increased Nrf2 and PPARγ on glomerular endothelia in the cortex; Nrf2 was also present on cortical peritubular capillaries. In contrast, HO-1 was localized to different cells, i.e., tubules and interstitial leukocytes. Although Nrf2-dependent increases in HO-1 have been described, our data suggest that BARDs effects on tubular and leukocyte HO-1 during ischemic AKI may be Nrf2 independent. We also found that BARD ameliorated cisplatin nephrotoxicity.


Recent Progress in Hormone Research | 1994

The Molecular Genetics of Steroid 5α-Reductases

David W. Russell; David M. Berman; James T. Bryant; Kristine M. Cala; Daphne L. Davis; Charles P. Landrum; James S. Prihoda; Richard I. Silver; Anice E. Thigpen; W. Christian Wigley

Publisher Summary This chapter discusses the molecular genetics of steroid 5α-reductases. 5α-reductase plays a central role in androgen action by catalyzing the conversion of testosterone into the more potent hormone dihydrotestosterone. Like other steroid and thyroid hormones, testosterone and dihydrotestosterone activate responsive genes by binding to the androgen receptor—a member of the steroid hormone receptor family of transcriptional activator proteins. Despite this mechanistic similarity, androgen action differs from other steroid hormones in that two different steroids interact with the same receptor to bring about different physiological effects. Testosterone bound to the androgen receptor is responsible for the regulation of gonadotropin production, spermatogenesis, and the formation of the internal male genitalia—epididymis, seminal vesicles, and vas deferens—from wolffian duct anlagen during phenotypic sexual differentiation in the male embryo. In contrast, dihydrotestosterone bound to the same androgen receptor brings about the formation of the male external genitalia—penis and scrotum—and prostate from urogenital sinus primordium in the male embryo and is required for sexual maturation at puberty.


Journal of Medicinal Chemistry | 2012

Synthesis, Chemical Reactivity as Michael Acceptors, and Biological Potency of Monocyclic Cyanoenones, Novel and Highly Potent Anti-inflammatory and Cytoprotective Agents(1)

Suqing Zheng; Y. R. Santosh Laxmi; Emilie David; Albena T. Dinkova-Kostova; Katherine H. Shiavoni; Yanqing Ren; Ying Zheng; Isaac Trevino; Ronald Bumeister; Iwao Ojima; W. Christian Wigley; James B. Bliska; Dale F. Mierke; Tadashi Honda

Novel monocyclic cyanoenones examined to date display unique features regarding chemical reactivity as Michael acceptors and biological potency. Remarkably, in some biological assays, the simple structure is more potent than pentacyclic triterpenoids (e.g., CDDO and bardoxolone methyl) and tricycles (e.g., TBE-31). Among monocyclic cyanoenones, 1 is a highly reactive Michael acceptor with thiol nucleophiles. Furthermore, an important feature of 1 is that its Michael addition is reversible. For the inhibition of NO production, 1 shows the highest potency. Notably, its potency is about three times higher than CDDO, whose methyl ester (bardoxolone methyl) is presently in phase III clinical trials. For the induction of NQO1, 1 also demonstrated the highest potency. These results suggest that the reactivity of these Michael acceptors is closely related to their biological potency. Interestingly, in LPS-stimulated macrophages, 1 causes apoptosis and inhibits secretion of TNF-α and IL-1β with potencies that are higher than those of bardoxolone methyl and TBE-31.


BMC Cell Biology | 2007

Hook2 contributes to aggresome formation

Györgyi Szebenyi; W. Christian Wigley; Branden Hall; Aaron Didier; Michelle Yu; Philip J. Thomas; Helmut Krämer

BackgroundAggresomes are pericentrosomal accumulations of misfolded proteins, chaperones and proteasomes. Their positioning near the centrosome, like that of other organelles, requires active, microtubule-dependent transport. Linker proteins that can associate with the motor protein dynein, organelles, and microtubules are thought to contribute to the active maintenance of the juxtanuclear localization of many membrane bound organelles and aggresomes. Hook proteins have been proposed to serve as adaptors for the association of cargos with dynein for transport on microtubules. Hook2 was shown to localize to the centrosome, bind centriolin, and contribute to centrosomal function.ResultsHere we show that overexpression of hook2 promotes the accumulation of the cystic fibrosis transmembrane regulator in aggresomes without altering its biochemical properties or its steady state level. A dominant negatively acting form of hook2 that lacks the centriolin binding C-terminal inhibits aggresome formation.ConclusionWe propose that hook2 contributes to the establishment and maintenance of the pericentrosomal localization of aggresomes by promoting the microtubule-based delivery of protein aggregates to pericentriolar aggresomes.


PLOS ONE | 2015

RTA 408, A Novel Synthetic Triterpenoid with Broad Anticancer and Anti-Inflammatory Activity

Brandon L. Probst; Isaac Trevino; Lyndsey McCauley; Ron Bumeister; Irina Dulubova; W. Christian Wigley; Deborah A. Ferguson

Semi-synthetic triterpenoids are antioxidant inflammation modulator (AIM) compounds that inhibit tumor cell growth and metastasis. Compounds in the AIM class bind to Keap1 and attenuate Nrf2 degradation. In the nucleus, Nrf2 increases antioxidant gene expression and reduces pro-inflammatory gene expression. By increasing Nrf2 activity, AIMs reduce reactive oxygen species and inflammation in the tumor microenvironment, which reverses tumor-mediated immune evasion and inhibits tumor growth and metastasis. AIMs also directly inhibit tumor cell growth by modulating oncogenic signaling pathways, such as IKKβ/NF-κB. Here, we characterized the in vitro antioxidant, anti-inflammatory, and anticancer activities of RTA 408, a novel AIM that is currently being evaluated in patients with advanced malignancies. At low concentrations (≤ 25 nM), RTA 408 activated Nrf2 and suppressed nitric oxide and pro-inflammatory cytokine levels in interferon-γ-stimulated RAW 264.7 macrophage cells. At higher concentrations, RTA 408 inhibited tumor cell growth (GI50 = 260 ± 74 nM) and increased caspase activity in tumor cell lines, but not in normal primary human cells. Consistent with the direct effect of AIMs on IKKβ, RTA 408 inhibited NF-κB signaling and decreased cyclin D1 levels at the same concentrations that inhibited cell growth and induced apoptosis. RTA 408 also increased CDKN1A (p21) levels and JNK phosphorylation. The in vitro activity profile of RTA 408 is similar to that of bardoxolone methyl, which was well-tolerated by patients at doses that demonstrated target engagement. Taken together, these data support clinical evaluation of RTA 408 for cancer treatment.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and biological evaluation of 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]-4-ethynylimidazole. A novel and highly potent anti-inflammatory and cytoprotective agent.

Tadashi Honda; Albena T. Dinkova-Kostova; Emilie David; Eric M. Padegimas; Chitra Sundararajan; Melean Visnick; Ron Bumeister; W. Christian Wigley

To explore more potent N-acylimidazole analogues of CDDO than CDDO-Im, which is one of the most potent compounds in several widely used bioassays related to protection against inflammation and carcinogenesis; we have synthesized and evaluated five new N-acyl(acetylenic)imidazole analogues. Among them, 4-ethynylimidazole 4 is nearly equivalent to CDDO-Im in potency in these bioassays. Remarkably, the solid form of 4 is more stable than that of CDDO-Im. These findings suggest that 4 is a very promising anti-inflammatory and cytoprotective agent and its further preclinical evaluation is warranted.


PLOS ONE | 2015

Cancer Cell Growth Is Differentially Affected by Constitutive Activation of NRF2 by KEAP1 Deletion and Pharmacological Activation of NRF2 by the Synthetic Triterpenoid, RTA 405

Brandon L. Probst; Lyndsey McCauley; Isaac Trevino; W. Christian Wigley; Deborah A. Ferguson

Synthetic triterpenoids are antioxidant inflammation modulators (AIMs) that exhibit broad anticancer activity. AIMs bind to KEAP1 and inhibit its ability to promote NRF2 degradation. As a result, NRF2 increases transcription of genes that restore redox balance and reduce inflammation. AIMs inhibit tumor growth and metastasis by increasing NRF2 activity in the tumor microenvironment and by modulating the activity of oncogenic signaling pathways, including NF-κB, in tumor cells. Accumulating evidence suggests that KEAP1 loss or mutation—which results in high levels of sustained NRF2 activity—may promote cancer growth and increase chemoresistance. Loss of KEAP1 also increases the levels of other oncogenic proteins, including IKKβ and BCL2. The apparent survival advantage provided to some tumor cells by loss of functional KEAP1 raises the question of whether pharmacological inhibition of KEAP1 could promote tumor growth. To address this issue, we characterized the basal levels of KEAP1 and NRF2 in a panel of human tumor cell lines and profiled the activity of an AIM, RTA 405. We found that in tumor cell lines with low or mutant KEAP1, and in Keap1 -/- murine embryonic fibroblasts, multiple KEAP1 targets including NRF2, IKKβ, and BCL2 were elevated. Keap1 -/- murine embryonic fibroblasts also had higher rates of proliferation and colony formation than their wild-type counterparts. In cells with functional KEAP1, RTA 405 increased NRF2 levels, but not IKKβ or BCL2 levels, and did not increase cell proliferation or survival. Moreover, RTA 405 inhibited growth at similar concentrations in cells with different basal NRF2 activity levels and in cells with wild-type or mutant KRAS. Finally, pre-treatment with RTA 405 did not protect tumor cells from doxorubicin- or cisplatin-mediated growth inhibition. Collectively, these data demonstrate that pharmacological activation of NRF2 by AIMs is distinct from genetic activation and does not provide a growth or survival advantage to tumor cells.


Oxidative Medicine and Cellular Longevity | 2017

NRF2 as an emerging therapeutic target

Ian M. Copple; Albena T. Dinkova-Kostova; Thomas W. Kensler; Karen T. Liby; W. Christian Wigley

1MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, UK 2Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK 3Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA 4Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA 5Reata Pharmaceuticals Inc., 2801 Gateway Drive, Irving, TX 75063, USA

Collaboration


Dive into the W. Christian Wigley's collaboration.

Top Co-Authors

Avatar

Philip J. Thomas

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rhesa D. Stidham

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen T. Liby

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Michael J. Corboy

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rosalind P. Fabunmi

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge