Network


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

Hotspot


Dive into the research topics where Carol F. Webb is active.

Publication


Featured researches published by Carol F. Webb.


Nature Genetics | 2011

Association of a functional variant downstream of TNFAIP3 with systemic lupus erythematosus

Indra Adrianto; Feng Wen; Amanda Templeton; Graham B. Wiley; Jarrod B. King; Christopher J. Lessard; Jared S. Bates; Yanqing Hu; Jennifer A. Kelly; Kenneth M. Kaufman; Joel M. Guthridge; Marta E. Alarcón-Riquelme; Juan-Manuel Anaya; Sang-Cheol Bae; So-Young Bang; Susan A. Boackle; Elizabeth E. Brown; Michelle Petri; Caroline J. Gallant; Rosalind Ramsey-Goldman; John D. Reveille; Luis M. Vilá; Lindsey A. Criswell; Jeffrey C. Edberg; Barry I. Freedman; Peter K. Gregersen; Gary S. Gilkeson; Chaim O. Jacob; Judith A. James; Diane L. Kamen

Systemic lupus erythematosus (SLE, MIM152700) is an autoimmune disease characterized by self-reactive antibodies resulting in systemic inflammation and organ failure. TNFAIP3, encoding the ubiquitin-modifying enzyme A20, is an established susceptibility locus for SLE. By fine mapping and genomic re-sequencing in ethnically diverse populations, we fully characterized the TNFAIP3 risk haplotype and identified a TT>A polymorphic dinucleotide (deletion T followed by a T to A transversion) associated with SLE in subjects of European (P = 1.58 × 10−8, odds ratio = 1.70) and Korean (P = 8.33 × 10−10, odds ratio = 2.54) ancestry. This variant, located in a region of high conservation and regulatory potential, bound a nuclear protein complex composed of NF-κB subunits with reduced avidity. Further, compared with the non-risk haplotype, the haplotype carrying this variant resulted in reduced TNFAIP3 mRNA and A20 protein expression. These results establish this TT>A variant as the most likely functional polymorphism responsible for the association between TNFAIP3 and SLE.


Journal of Immunology | 2000

The Transcription Factor Bright Associates with Bruton’s Tyrosine Kinase, the Defective Protein in Immunodeficiency Disease

Carol F. Webb; Yoshio Yamashita; Neil Ayers; Seth Evetts; Yolene Paulin; Mary Ellen Conley; Elizabeth A. Smith

Binding of the transcription factor Bright to Ig heavy chain loci after B cell activation is associated with increased heavy chain transcription. We now report that Bright coprecipitates with Bruton’s tyrosine kinase (Btk), the defective enzyme in X-linked immunodeficiency disease (xid). Furthermore, we observed Btk in the nucleus of activated murine B cells, and mobility shift assays suggest that it is a component of the Bright DNA-binding complex. While Bright protein was synthesized in activated spleen cells from xid mice, it did not bind DNA or associate stably with Btk. These data suggest that deficiencies in Bright DNA-binding activity may contribute to the defects in Ig production seen in xid mice.


Molecular and Cellular Biology | 1991

Identification of a matrix-associated region 5' of an immunoglobulin heavy chain variable region gene.

Carol F. Webb; C Das; K L Eneff; P W Tucker

In the accompanying report (C. F. Webb, C. Das, S. Eaton, K. Calame, and P. Tucker, Mol. Cell. Biol. 11:5197-5205, 1991), we characterize B-cell-specific protein-DNA interactions at -500 and -200 bp upstream of the mu immunoglobulin heavy chain promoter whose abundances were increased by interleukin-5 plus antigen. Because of the high A + T/G + C ratio of these sequences and the consistent findings by others that enhancer- and promoterlike regions are often located near matrix-associated regions, we asked whether these sequences might also be involved in binding to the nuclear matrix. Indeed, DNA fragments containing the -500 binding site were bound by nuclear matrix proteins. Furthermore, UV cross-linking studies showed that the DNA binding site for interleukin-5-plus-antigen-inducible proteins could also bind to proteins solubilized from the nuclear matrix. Nuclear matrix-associated sequences have also been demonstrated on either side of the intronic immunoglobulin heavy chain enhancer. Our data suggest a topological model by which interactions among proteins bound to the promoter and distal enhancer sequences might occur.


Molecular and Cellular Biology | 2011

The ARID Family Transcription Factor Bright Is Required for both Hematopoietic Stem Cell and B Lineage Development

Carol F. Webb; James Bryant; Melissa Popowski; Laura Allred; Dongkoon Kim; June V. Harriss; Christian Schmidt; Cathrine Miner; Kira Rose; Hwei Ling Cheng; Courtney Griffin; Philip W. Tucker

ABSTRACT Bright/Arid3a has been characterized both as an activator of immunoglobulin heavy-chain transcription and as a proto-oncogene. Although Bright expression is highly B lineage stage restricted in adult mice, its expression in the earliest identifiable hematopoietic stem cell (HSC) population suggests that Bright might have additional functions. We showed that >99% of Bright−/− embryos die at midgestation from failed hematopoiesis. Bright−/− embryonic day 12.5 (E12.5) fetal livers showed an increase in the expression of immature markers. Colony-forming assays indicated that the hematopoietic potential of Bright−/− mice is markedly reduced. Rare survivors of lethality, which were not compensated by the closely related paralogue Bright-derived protein (Bdp)/Arid3b, suffered HSC deficits in their bone marrow as well as B lineage-intrinsic developmental and functional deficiencies in their peripheries. These include a reduction in a natural antibody, B-1 responses to phosphocholine, and selective T-dependent impairment of IgG1 class switching. Our results place Bright/Arid3a on a select list of transcriptional regulators required to program both HSC and lineage-specific differentiation.


Journal of Immunology | 2008

Retinoids Accelerate B Lineage Lymphoid Differentiation

Xinrong Chen; Brandt L. Esplin; Karla P. Garrett; Robert S. Welner; Carol F. Webb; Paul W. Kincade

Retinoids are known to have potent effects on hemopoietic stem cell integrity, and our objective was to learn whether they influence cells destined to replenish the immune system. Total CD19+ B lineage cells increased substantially in the marrow and spleens of all-trans retinoic acid (ATRA)-treated C57BL6 mice, while lymphoid progenitors were reduced. All B lymphoid progenitors were targets of ATRA in culture and overall cell yields declined without reductions in proliferation. Remarkably, ATRA shortened the time required for primitive progenitors to generate CD19+ cells. PCR analysis and a panel of retinoid acid receptor (RAR)/retinoid X receptor agonist treatments suggested that RARα mediates these responses. The transcription factors EBF1 and Pax-5 were elevated during treatment and ATRA had similar effects on human B cell differentiation. That is, it inhibited the expansion of human progenitor cells and accelerated their differentiation to B lineage cells. There may be previously unsuspected side effects of ATRA therapy, and the new findings suggest retinoids can normally contribute to the lymphopoietic environment in bone marrow.


Molecular and Cellular Biology | 2006

Induction of Immunoglobulin Heavy-Chain Transcription through the Transcription Factor Bright Requires TFII-I

Jaya Rajaiya; Jamee C. Nixon; Neil Ayers; Zana P. Desgranges; Ananda L. Roy; Carol F. Webb

ABSTRACT Bright/ARID3a/Dril1, a member of the ARID family of transcription factors, is expressed in a highly regulated fashion in B lymphocytes, where it enhances immunoglobulin transcription three- to sixfold. Recent publications from our lab indicated that functional, but not kinase-inactive, Brutons tyrosine kinase (Btk) is critical for Bright activity in an in vitro model system, yet Bright itself is not appreciably tyrosine phosphorylated. These data suggested that a third protein, and Btk substrate, must contribute to Bright-enhanced immunoglobulin transcription. The ubiquitously expressed transcription factor TFII-I was identified as a substrate for Btk several years ago. In this work, we show that TFII-I directly interacts with human Bright through amino acids in Brights protein interaction domain and that specific tyrosine residues of TFII-I are essential for Bright-induced activity of an immunoglobulin reporter gene. Moreover, inhibition of TFII-I function in a B-cell line resulted in decreased heavy-chain transcript levels. These data suggest that Bright functions as a three-component protein complex in the immunoglobulin locus and tie together previous data indicating important roles for Btk and TFII-I in B lymphocytes.


Journal of Immunology | 2002

High frequency of matrix attachment regions and cut-like protein x/CCAAT-displacement protein and B cell regulator of IgH transcription binding sites flanking Ig V region genes.

Peter Goebel; Alina Montalbano; Neil Ayers; Elizabeth Kompfner; Liliane A. Dickinson; Carol F. Webb; Ann J. Feeney

A major component in controlling V(D)J recombination is differential accessibility through localized changes in chromatin structure. Attachment of DNA to the nuclear matrix via matrix attachment region (MAR) sequences, and interaction with MAR-binding proteins have been shown to alter chromatin conformation, promote histone acetylation, and influence gene transcription. In this study, the flanking regions of several human and mouse Ig VH and Ig Vκ genes were analyzed extensively for the presence of MARs by in vitro matrix-binding assay, and for interaction with the MAR-binding proteins cut-like protein x/CCAAT-displacement protein (Cux/CDP), B cell regulator of IgH transcription (Bright), and special AT-rich sequence-binding protein (SATB1) by EMSA. Cux/CDP and SATB1 are associated with repression, while Bright is an activator of Ig transcription. Binding sites were identified in the vicinity of all analyzed Ig V genes, and were also found flanking TCR Vβ genes. We also show that the binding sites of the different factors do not always occur at MAR sequences. MAR sequences were also found within the Ig V loci at a much higher frequency than throughout the rest of the genome. Overall, the frequency and location of binding sites relative to the coding regions, and the strength of DNA-protein interaction showed much heterogeneity. Thus, variations in factor binding and MAR activity could potentially influence the extent of localized accessibility to V(D)J recombination and thus could play a role in unequal rearrangement of individual V genes. These sites could also contribute to effective transcription of Ig genes in mature and/or activated B cells, bringing both the promoter as well as the enhancer regions into close proximity at the nuclear matrix.


American Journal of Human Genetics | 2014

Two Functional Lupus-Associated BLK Promoter Variants Control Cell-Type- and Developmental-Stage-Specific Transcription

Joel M. Guthridge; Rufei Lu; Harry Sun; Celi Sun; Graham B. Wiley; Nicolas Dominguez; Susan Macwana; Christopher J. Lessard; Xana Kim-Howard; Beth L. Cobb; Kenneth M. Kaufman; Jennifer A. Kelly; Carl D. Langefeld; Adam Adler; Isaac T.W. Harley; Joan T. Merrill; Gary S. Gilkeson; Diane L. Kamen; Timothy B. Niewold; Elizabeth E. Brown; Jeffery Edberg; Michelle Petri; Rosalind Ramsey-Goldman; John D. Reveille; Luis M. Vilá; Robert P. Kimberly; Barry I. Freedman; Anne M. Stevens; Susan A. Boackle; Lindsey A. Criswell

Efforts to identify lupus-associated causal variants in the FAM167A/BLK locus on 8p21 are hampered by highly associated noncausal variants. In this report, we used a trans-population mapping and sequencing strategy to identify a common variant (rs922483) in the proximal BLK promoter and a tri-allelic variant (rs1382568) in the upstream alternative BLK promoter as putative causal variants for association with systemic lupus erythematosus. The risk allele (T) at rs922483 reduced proximal promoter activity and modulated alternative promoter usage. Allelic differences at rs1382568 resulted in altered promoter activity in B progenitor cell lines. Thus, our results demonstrated that both lupus-associated functional variants contribute to the autoimmune disease association by modulating transcription of BLK in B cells and thus potentially altering immune responses.


Molecular and Cellular Biology | 2005

Bruton's Tyrosine Kinase Regulates Immunoglobulin Promoter Activation in Association with the Transcription Factor Bright

Jaya Rajaiya; Melissa Hatfield; Jamee C. Nixon; David J. Rawlings; Carol F. Webb

ABSTRACT Bright (B-cell regulator of immunoglobulin heavy chain transcription) binding to immunoglobulin heavy chain loci after B-cell activation is associated with increased heavy chain transcription. Our earlier reports demonstrated that Bright coimmunoprecipitates with Brutons tyrosine kinase (Btk) and that these proteins associate in a DNA-binding complex in primary B cells. B cells from immunodeficient mice with a mutation in Btk failed to produce stable Bright DNA-binding complexes. In order to determine if Btk is important for Bright function, a transcription activation assay was established and analyzed using real-time PCR technology. Cells lacking both Bright and Btk were transfected with Bright and/or Btk along with an immunoglobulin heavy chain reporter construct. Immunoglobulin gene transcription was enhanced when Bright and Btk were coexpressed. In contrast, neither Bright nor Btk alone led to activation of heavy chain transcription. Furthermore, Bright function required both Btk kinase activity and sequences within the pleckstrin homology domain of Btk. Bright was not appreciably phosphorylated by Btk; however, a third tyrosine-phosphorylated protein coprecipitated with Bright. Thus, the ability of Bright to enhance immunoglobulin transcription critically requires functional Btk.


Journal of Immunology | 2000

An Upstream Oct-1- and Oct-2-Binding Silencer Governs B29 (Igβ) Gene Expression

Cindy S. Malone; Lisa Patrone; Kent L. Buchanan; Carol F. Webb; Randolph Wall

The B cell-specific B29 (Igβ) gene is activated in the earliest B cell precursors and is expressed throughout B cell development. Tissue-specific expression of the murine B29 gene is controlled by a B cell-specific promoter whose activity is governed by a cassette of upstream transcriptional silencers. This study describes a potent new silencer that is located 5′ of the previously identified B29 silencer elements, FROG and TOAD. Like these known elements, the new B29 silencer is not restricted to the B29 promoter. Nuclear proteins from all cell lines tested interacted with this A+T-rich sequence, which closely resembled a noncanonical octamer binding motif and also conformed to the consensus sequence for nuclear matrix attachment regions. Interaction of Oct-1 and Oct-2 with the B29 A+T-rich sequence was confirmed using octamer-specific Abs. Oct-1/Oct-2 binding was required for the inhibitory activity of this sequence because mutations that blocked Oct-1/Oct-2 binding also eliminated inhibition of the B29 promoter. This B29 A+T-rich sequence specifically interacted with isolated nuclear matrix proteins in vitro, suggesting that it may also function as a matrix attachment region element. Maintenance of the level of B29 gene expression through the interaction of the minimal promoter and the upstream silencer elements FROG, TOAD, and the A+T-rich Oct-1/Oct-2 binding motif may be essential for normal B cell development and/or function.

Collaboration


Dive into the Carol F. Webb's collaboration.

Top Co-Authors

Avatar

Judith A. James

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Julie M. Ward

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Michelle L. Ratliff

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Jamee C. Nixon

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Cathrine Miner

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Joan T. Merrill

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Kira Rose

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Courtney G. Montgomery

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Indra Adrianto

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Joel M. Guthridge

Oklahoma Medical Research Foundation

View shared research outputs
Researchain Logo
Decentralizing Knowledge