Network


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

Hotspot


Dive into the research topics where Anthony Thomas is active.

Publication


Featured researches published by Anthony Thomas.


Proceedings of the National Academy of Sciences of the United States of America | 2011

R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/β-catenin signaling

Kendra S. Carmon; Xing Gong; Qiushi Lin; Anthony Thomas; Qingyun Liu

The Wnt/β-catenin signaling system plays essential roles in embryonic development and in the self-renewal and maintenance of adult stem cells. R-spondins (RSPOs) are a group of secreted proteins that enhance Wnt/β-catenin signaling and have pleiotropic functions in development and stem cell growth. LGR5, an orphan receptor of the G protein-coupled receptor (GPCR) superfamily, is specifically expressed in stem cells of the intestinal crypt and hair follicle. Knockout of LGR5 in the mouse results in neonatal lethality. LGR4, a receptor closely related to LGR5, also has essential roles in development, as its knockout leads to reduced viability and retarded growth. Overexpression of both receptors has been reported in several types of cancer. Here we demonstrate that LGR4 and LGR5 bind the R-spondins with high affinity and mediate the potentiation of Wnt/β-catenin signaling by enhancing Wnt-induced LRP6 phosphorylation. Interestingly, neither receptor is coupled to heterotrimeric G proteins or to β-arrestin when stimulated by the R-spondins, indicating a unique mechanism of action. The findings provide a basis for stem cell-specific effects of Wnt/β-catenin signaling and for the broad range of functions LGR4, LGR5, and the R-spondins have in normal and malignant growth.


Molecular and Cellular Biology | 2012

LGR5 interacts and cointernalizes with Wnt receptors to modulate Wnt/β-catenin signaling.

Kendra S. Carmon; Qiushi Lin; Xing Gong; Anthony Thomas; Qingyun Liu

ABSTRACT LGR5, a seven-transmembrane domain receptor of the rhodopsin family, is a Wnt target gene and a bona fide marker of adult stem cells in the gastrointestinal tract and hair follicle bulge. Recently, we and others demonstrated that LGR5 and its homologues function as receptors of the R-spondin family of stem cell factors to potentiate Wnt/β-catenin signaling. However, the mechanism of how LGR5 enhances the signaling output remains unclear. Here we report that following costimulation with the ligands R-spondin1 and Wnt3a, LGR5 interacts and forms a supercomplex with the Wnt coreceptors LRP6 and Fzd5 which is rapidly internalized and then degraded. Internalization of LGR5 is mediated through a dynamin- and clathrin-dependent pathway. Inhibition of this endocytic process has no effect on LGR5 signaling. Deletion of the C-terminal tail of LGR5 maintains its ability to interact with LRP6, yet this LGR5 mutant exhibits increased signaling activity and a decreased rate of endocytosis in response to R-spondin1 compared to the wild-type receptor. This study provides direct evidence that LGR5 becomes part of the Wnt signaling complex at the membrane level to enhance Wnt/β-catenin signaling. However, internalization of LGR5 does not appear to be essential for potentiating the canonical Wnt signaling pathway.


PLOS ONE | 2012

LGR6 Is a High Affinity Receptor of R-Spondins and Potentially Functions as a Tumor Suppressor

Xing Guo Gong; Kendra S. Carmon; Qiushi Lin; Anthony Thomas; Jing He Zhangfei Li Yu Yi; Qingyun Liu

Background LGR6 (leucine-rich repeat containing, G protein-coupled receptor 6) is a member of the rhodopsin-like seven transmembrane domain receptor superfamily with the highest homology to LGR4 and LGR5. LGR6 was found as one of the novel genes mutated in colon cancer through total exon sequencing and its promoter region is hypermethylated in 20–50% of colon cancer cases. In the skin, LGR6 marks a population of stem cells that can give rise to all cell lineages. Recently, we and others demonstrated that LGR4 and LGR5 function as receptors of R-spondins to potentiate Wnt/β-catenin signaling. However, the binding affinity and functional response of LGR6 to R-spondins, and the activity of colon cancer mutants of LGR6 have not been determined. Principal Findings We found that LGR6 also binds and responds to R-spondins 1–3 with high affinity to enhance Wnt/β-catenin signaling through increased LRP6 phosphorylation. Similar to LGR4 and LGR5, LGR6 is not coupled to heterotrimeric G proteins or to β-arrestin following R-spondin stimulation. Functional and expression analysis of three somatic mutations identified in colon cancer samples indicates that one mutant fails to bind and respond to R-spondin (loss-of-function), but the other two have no significant effect on receptor function. Overexpression of wild-type LGR6 in HeLa cells leads to increased cell migration following co-treatment with R-spondin1 and Wnt3a when compared to vector control cells or cells overexpressing the loss-of-function mutant. Conclusions LGR6 is a high affinity receptor for R-spondins 1–3 and potentially functions as a tumor suppressor despite its positive effect on Wnt/β-catenin signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2014

RSPO–LGR4 functions via IQGAP1 to potentiate Wnt signaling

Kendra S. Carmon; Xing Gong; Jing Yi; Anthony Thomas; Qingyun Liu

Significance R-spondins (RSPOs) and LGR4 emerged as a major ligand–receptor system in the regulation of Wnt signaling as manifested in their pleotropic roles in development and survival of adult stem cells. The mechanism of how RSPO–LGR4 interacts with the Wnt signaling system remains poorly understood. In this work, we describe the identification of IQGAP1 as the first intracellular signaling partner of RSPO–LGR4 and the delineation of IQGAP1’s roles and mechanism in mediating RSPO–LGR4-induced potentiation of Wnt signaling. We also elucidate the relationship between the RSPO–LGR4–IQGAP1 pathway and the function of RSPO–LGR4 in inhibiting RNF43/ZNRF3. The findings uncovered a unique mechanism of RSPO–LGR4 signaling and provide a mechanistic basis for the pleiotropic functions of RSPO–LGR4 signaling in normal and pathological processes. R-spondins (RSPOs) and their receptor leucine-rich repeat-containing G-protein coupled receptor 4 (LGR4) play pleiotropic roles in normal and cancer development as well as the survival of adult stem cells through potentiation of Wnt signaling. Current evidence indicates that RSPO–LGR4 functions to elevate levels of Wnt receptors through direct inhibition of two membrane-bound E3 ligases (RNF43 and ZNRF3), which otherwise ubiquitinate Wnt receptors for degradation. Whether RSPO–LGR4 is coupled to intracellular signaling proteins to regulate Wnt pathways remains unknown. We identified the intracellular scaffold protein IQ motif containing GTPase-activating protein 1 (IQGAP1) as an LGR4-interacting protein that mediates RSPO–LGR4’s interaction with the Wnt signalosome. IQGAP1 binds to and modulates the activities of a plethora of signaling molecules, including MAP kinases, Rho GTPases, and components of the Wnt signaling pathways. Interaction of LGR4 with IQGAP1 brings RSPO–LGR4 to the Wnt signaling complex through enhanced IQGAP1–DVL interaction following RSPO stimulation. In this configuration, RSPO–LGR4–IQGAP1 potentiates β-catenin–dependent signaling by promoting MEK1/2-medidated phosphorylation of LRP5/6 as well as β-catenin–independent signaling through regulation of actin dynamics. Overall, these findings reveal that RSPO–LGR4 not only induces the clearance of RNF43/ZNRF3 to increase Wnt receptor levels but also recruits IQGAP1 into the Wnt signaling complex, leading to potent and robust potentiation of both the canonical and noncanonical pathways of Wnt signaling.


Journal of Biological Chemistry | 2008

α1 Soluble Guanylyl Cyclase (sGC) Splice Forms as Potential Regulators of Human sGC Activity

Iraida Sharina; Filip Jelen; Elena Bogatenkova; Anthony Thomas; Emil Martin; Ferid Murad

Soluble guanylyl cyclase (sGC), a key protein in the NO/cGMP signaling pathway, is an obligatory heterodimeric protein composed of one α- and one β-subunit. The α1/β1 sGC heterodimer is the predominant form expressed in various tissues and is regarded as the major isoform mediating NO-dependent effects such as vasodilation. We have identified three new α1 sGC protein variants generated by alternative splicing. The 363 residue N1-α1 sGC splice variant contains the regulatory domain, but lacks the catalytic domain. The shorter N2-α1 sGC maintains 126 N-terminal residues and gains an additional 17 unique residues. The C-α1 sGC variant lacks 240 N-terminal amino acids, but maintains a part of the regulatory domain and the entire catalytic domain. Q-PCR of N1-α1, N2-α1 sGC mRNA levels together with RT-PCR analysis for C-α1 sGC demonstrated that the expression of the α1 sGC splice forms vary in different human tissues indicative of tissue-specific regulation. Functional analysis of the N1-α1 sGC demonstrated that this protein has a dominant-negative effect on the activity of sGC when coexpressed with the α1/β1 heterodimer. The C-α1 sGC variant heterodimerizes with the β1 subunit and produces a fully functional NO- and BAY41-2272-sensitive enzyme. We also found that despite identical susceptibility to inhibition by ODQ, intracellular levels of the 54-kDa C-α1 band did not change in response to ODQ treatments, while the level of 83 kDa α1 band was significantly affected by ODQ. These studies suggest that modulation of the level and diversity of splice forms may represent novel mechanisms modulating the function of sGC in different human tissues.


Proceedings of the National Academy of Sciences of the United States of America | 2003

CCAAT-binding factor regulates expression of the β1 subunit of soluble guanylyl cyclase gene in the BE2 human neuroblastoma cell line

Iraida Sharina; Emil Martin; Anthony Thomas; Karen L. Uray; Ferid Murad

Soluble guanylyl cyclase (sGC) is a cytosolic enzyme producing the intracellular messenger cyclic guanosine monophosphate (cGMP) on activation with nitric oxide (NO). sGC is an obligatory heterodimer composed of α and β subunits. We investigated human β1 sGC transcriptional regulation in BE2 human neuroblastoma cells. The 5′ upstream region of the β1 sGC gene was isolated and analyzed for promoter activity by using luciferase reporter constructs. The transcriptional start site of the β1 sGC gene in BE2 cells was identified. The functional significance of consensus transcriptional factor binding sites proximal to the transcriptional start site was investigated by site deletions in the 800-bp promoter fragment. The elimination of CCAAT-binding factor (CBF) and growth factor independence 1 (GFI1) binding cores significantly diminished whereas deletion of the NF1 core elevated the transcription. Electrophoretic mobility-shift assay (EMSA) and Western analysis of proteins bound to biotinated EMSA probes confirmed the interaction of GFI1, CBF, and NF1 factors with the β1 sGC promoter. Treatment of BE2 cells with genistein, known to inhibit the CBF binding to DNA, significantly reduced protein levels of β1 sGC by inhibiting transcription. In summary, our study represents an analysis of the human β1 sGC promoter regulation in human neuroblastoma BE2 cells and identifies CBF as a critically important factor in β1 sGC expression.


Oncogene | 2015

Aberrant RSPO3-LGR4 signaling in Keap1-deficient lung adenocarcinomas promotes tumor aggressiveness

Xing Gong; Jing Yi; Kendra S. Carmon; Christine A. Crumbley; Wei Xiong; Anthony Thomas; Xuejun Fan; Shan Guo; Zhiqiang An; Jeffrey T. Chang; Qingyun Liu

The four R-spondins (RSPO1–4) and their three related receptors LGR4, 5 and 6 (LGR4–6) have emerged as a major ligand-receptor system with critical roles in development and stem cell survival through modulation of Wnt signaling. Recurrent, gain-of-expression gene fusions of RSPO2 (to EIF3E) and RSPO3 (to PTPRK) occur in a subset of human colorectal cancer. However, the exact roles and mechanisms of the RSPO-LGR system in oncogenesis remain largely unknown. We found that RSPO3 is aberrantly expressed at high levels in approximately half of Keap1-mutated lung adenocarcinomas (ADs). This high RSPO3 expression is driven by a combination of demethylation of its own promoter region and deficiency in Keap1 instead of gene fusion as in colon cancer. Patients with RSPO3-high tumors (~9%, 36/412) displayed much poorer survival than the rest of the cohort (median survival of 28 vs 163 months, log-rank test P<0.0001). Knockdown (KD) of RSPO3, LGR4 or their signaling mediator IQGAP1 in lung cancer cell lines with Keap1 deficiency and high RSPO3-LGR4 expression led to reduction in cell proliferation and migration in vitro, and KD of LGR4 or IQGAP1 resulted in decrease in tumor growth and metastasis in vivo. These findings suggest that aberrant RSPO3-LGR4 signaling potentially acts as a driving mechanism in the aggressiveness of Keap1-deficient lung ADs.


PLOS ONE | 2012

Hydrogen Peroxide alters splicing of soluble Guanylyl cyclase and selectively modulates expression of splicing regulators in human cancer cells

Gilbert J. Cote; Wen Zhu; Anthony Thomas; Emil Martin; Ferid Murad; Iraida Sharina

Background Soluble guanylyl cyclase (sGC) plays a central role in nitric oxide (NO)-mediated signal transduction in the cardiovascular, nervous and gastrointestinal systems. Alternative RNA splicing has emerged as a potential mechanism to modulate sGC expression and activity. C-α1 sGC is an alternative splice form that is resistant to oxidation-induced protein degradation and demonstrates preferential subcellular distribution to the oxidized environment of endoplasmic reticulum (ER). Methodology/Principal Findings Here we report that splicing of C-α1 sGC can be modulated by H2O2 treatment in BE2 neuroblastoma and MDA-MD-468 adenocarcinoma human cells. In addition, we show that the H2O2 treatment of MDA-MD-468 cells selectively decreases protein levels of PTBP1 and hnRNP A2/B1 splice factors identified as potential α1 gene splicing regulators by in silico analysis. We further demonstrate that down-regulation of PTBP1 by H2O2 occurs at the protein level with variable regulation observed in different breast cancer cells. Conclusions/Significance Our data demonstrate that H2O2 regulates RNA splicing to induce expression of the oxidation-resistant C-α1 sGC subunit. We also report that H2O2 treatment selectively alters the expression of key splicing regulators. This process might play an important role in regulation of cellular adaptation to conditions of oxidative stress.


Journal of Biological Chemistry | 2017

LGR5 receptor promotes cell-cell adhesion in stem cells and colon cancer cells via the IQGAP1 -Rac1 pathway

Kendra S. Carmon; Xing Gong; Jing Yi; Ling Wu; Anthony Thomas; Catherine M. Moore; Ikuo Masuho; David J. Timson; Kirill A. Martemyanov; Qingyun Liu

Leucine-rich repeat-containing G protein–coupled receptor 5 (LGR5) is a bona fide marker of adult stem cells in several epithelial tissues, most notably in the intestinal crypts, and is highly up-regulated in many colorectal, hepatocellular, and ovarian cancers. LGR5 activation by R-spondin (RSPO) ligands potentiates Wnt/β-catenin signaling in vitro; however, deletion of LGR5 in stem cells has little or no effect on Wnt/β-catenin signaling or cell proliferation in vivo. Remarkably, modulation of LGR5 expression has a major impact on the actin cytoskeletal structure and cell adhesion in the absence of RSPO stimulation, but the molecular mechanism is unclear. Here, we show that LGR5 interacts with IQ motif-containing GTPase-activating protein 1 (IQGAP1), an effector of Rac1/CDC42 GTPases, in the regulation of actin cytoskeleton dynamics and cell–cell adhesion. Specifically, LGR5 decreased levels of IQGAP1 phosphorylation at Ser-1441/1443, leading to increased binding of Rac1 to IQGAP1 and thus higher levels of cortical F-actin and enhanced cell–cell adhesion. LGR5 ablation in colon cancer cells and crypt stem cells resulted in loss of cortical F-actin, reduced cell–cell adhesion, and disrupted localization of adhesion-associated proteins. No evidence of LGR5 coupling to any of the four major subtypes of heterotrimeric G proteins was found. These findings suggest that LGR5 primarily functions via the IQGAP1–Rac1 pathway to strengthen cell–cell adhesion in normal adult crypt stem cells and colon cancer cells.


Molecular and Cellular Biology | 2017

Correction: "LGR5 interacts and cointernalizes with Wnt receptors to modulate Wnt/ß-catenin signaling [32, 11, (2054-2064), (2012)] https://doi.org/10.1128/MCB.00272-12

Kendra S. Carmon; Qiushi Lin; Xing Gong; Anthony Thomas; Qingyun Liu

Volume 32, no. 11, p. 2054 –2064, 2012, https://doi.org/10.1128/MCB.00272-12. Page 2061, Fig. 6A legend, line 2: The following sentence should be added. “Zero-minute images in the bottom two rows are duplicates of the 0-min images in the top two rows to make it easier to visualize the different extents of internalization in the presence and absence of ligands, especially at the 15-min time point.” The duplication in the figure does not affect any conclusions of the study, and we apologize for any confusion. Citation Carmon KS, Lin Q, Gong X, Thomas A, Liu Q. 2017. Correction for Carmon et al., “LGR5 interacts and cointernalizes with Wnt receptors to modulate Wnt/β-catenin signaling.” Mol Cell Biol 37:e00203-17. https://doi.org/10.1128/MCB .00203-17. Copyright

Collaboration


Dive into the Anthony Thomas's collaboration.

Top Co-Authors

Avatar

Kendra S. Carmon

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Qingyun Liu

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Xing Gong

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Qiushi Lin

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Emil Martin

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Ferid Murad

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Iraida Sharina

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Jing Yi

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Christine A. Crumbley

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Elena Bogatenkova

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge