Network


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

Hotspot


Dive into the research topics where W. Jason Cummings is active.

Publication


Featured researches published by W. Jason Cummings.


Current Biology | 2005

MutSα Binds to and Promotes Synapsis of Transcriptionally Activated Immunoglobulin Switch Regions

Erik D. Larson; Michelle L. Duquette; W. Jason Cummings; Raphael J. Streiff; Nancy Maizels

Immunoglobulin class switch recombination joins a new constant (C) region to the rearranged and expressed heavy chain variable (VDJ) region in antigen-activated B cells (Figure 1A) (reviewed in [1, 2]). Switch recombination is activated by transcription of intronic, G-rich and repetitive switch (S) regions and produces junctions that are heterogeneous in sequence and position in the S regions. Switch recombination depends upon the B cell-specific cytidine deaminase, AID, and conserved DNA repair factors, including the mismatch repair heterodimer, MutSalpha (MSH2/MSH6). In mice, ablation of Msh2 or Msh6, but not Msh3, decreases levels of switch recombination and diminishes heterogeneity of switch junctions [3-7]. Here, we demonstrate that MSH2 associates with transcribed S regions in primary murine B cells activated for switch recombination. Electron microscopic imaging reveals that MutSalpha binds in vitro to DNA structures formed within transcribed S regions and mediates their synapsis. MutSalpha binds with high affinity to G4 DNA formed upon transcription of the S regions and also binds to U.G mismatches, initial products of DNA deamination by AID. These results suggest that MutSalpha interacts with the S regions in switching B cells to promote DNA synapsis and recombination.


PLOS Biology | 2007

Chromatin structure regulates gene conversion.

W. Jason Cummings; Munehisa Yabuki; Ellen C. Ordinario; David W. Bednarski; Simon Quay; Nancy Maizels

Homology-directed repair is a powerful mechanism for maintaining and altering genomic structure. We asked how chromatin structure contributes to the use of homologous sequences as donors for repair using the chicken B cell line DT40 as a model. In DT40, immunoglobulin genes undergo regulated sequence diversification by gene conversion templated by pseudogene donors. We found that the immunoglobulin Vλ pseudogene array is characterized by histone modifications associated with active chromatin. We directly demonstrated the importance of chromatin structure for gene conversion, using a regulatable experimental system in which the heterochromatin protein HP1 (Drosophila melanogaster Su[var]205), expressed as a fusion to Escherichia coli lactose repressor, is tethered to polymerized lactose operators integrated within the pseudo-Vλ donor array. Tethered HP1 diminished histone acetylation within the pseudo-Vλ array, and altered the outcome of Vλ diversification, so that nontemplated mutations rather than templated mutations predominated. Thus, chromatin structure regulates homology-directed repair. These results suggest that histone modifications may contribute to maintaining genomic stability by preventing recombination between repetitive sequences.


Journal of Immunology | 2009

E2A Acts in cis in G1 Phase of Cell Cycle to Promote Ig Gene Diversification

Munehisa Yabuki; Ellen C. Ordinario; W. Jason Cummings; Monica M. Fujii; Nancy Maizels

Rearranged Ig genes undergo diversification in sequence and structure initiated by the DNA deaminase, activation-induced deaminase. Ig genes must be transcribed for diversification to occur, but whether there are additional requirements for cis activation has not been established. Here we show, by chromatin immunoprecipitation, that the regulatory factor E2A associates with the rearranged IgλR gene in the chicken DT40 B cell line, which performs constitutive Ig gene diversification. By analysis of a DT40 derivative in which polymerized lactose operator tags the rearranged λR gene, we show that E2A must function in cis to promote diversification and that stimulation of diversification in cis depends on the E2A activation domains. By direct imaging, we show that λR/E2A colocalizations are most prominent in G1. We further show that expression of the E2A antagonist Id1 prevents λR/E2A colocalizations in G1 and impairs diversification but not transcription of λR. Thus, E2A acts in cis to promote Ig gene diversification, and G1 phase is the critical window for E2A action.


PLOS ONE | 2008

Genetic Variation Stimulated by Epigenetic Modification

W. Jason Cummings; David W. Bednarski; Nancy Maizels

Homologous recombination is essential for maintaining genomic integrity. A common repair mechanism, it uses a homologous or homeologous donor as a template for repair of a damaged target gene. Such repair must be regulated, both to identify appropriate donors for repair, and to avoid excess or inappropriate recombination. We show that modifications of donor chromatin structure can promote homology-directed repair. These experiments demonstrate that either the activator VP16 or the histone chaperone, HIRA, accelerated gene conversion approximately 10-fold when tethered within the donor array for Ig gene conversion in the chicken B cell line DT40. VP16 greatly increased levels of acetylated histones H3 and H4, while tethered HIRA did not affect histone acetylation, but caused an increase in local nucleosome density and levels of histone H3.3. Thus, epigenetic modification can stimulate genetic variation. The evidence that distinct activating modifications can promote similar functional outcomes suggests that a variety of chromatin changes may regulate homologous recombination, and that disregulation of epigenetic marks may have deleterious genetic consequences.


The FASEB Journal | 2017

Lectin pathway effector enzyme mannan-binding lectin-associated serine protease-2 can activate native complement C3 in absence of C4 and/or C2.

Sadam Yaseen; Gregory A. Demopulos; Thomas Dudler; Munehisa Yabuki; Christi L. Wood; W. Jason Cummings; Larry W. Tjoelker; Teizo Fujita; Steven H. Sacks; Peter Garred; Peter W. Andrew; Robert B. Sim; Peter J. Lachmann; Russell Wallis; Nicholas J. Lynch; Wilhelm J. Schwaeble

All 3 activation pathways of complement—the classic pathway (CP), the alternative pathway, and the lectin pathway (LP)— converge into a common central event: the cleavage and activation of the abundant third complement component, C3, via formation of C3‐activating enzymes (C3 convertases). The fourth complement component, C4, and the second component, C2, are indispensable constituents of the C3 convertase complex, C4bC2a, which is formed by both the CP and the LP. Whereas in the absence of C4, CP can no longer activate C3, LP retains a residual but physiologically critical capacity to convert native C3 into its activation fragments, C3a and C3b. This residual C4 and/or C2 bypass route is dependent on LP‐specific mannan‐binding lectin‐associated serine protease‐2. By using various serum sources with defined complement deficiencies, we demonstrate that, under physiologic conditions LP‐specific C4 and/or C2 bypass activation of C3 is mediated by direct cleavage of native C3 by mannan‐binding lectin‐associated serine protease‐2 bound to LP‐activation complexes captured on ligand‐coated surfaces.—Yaseen, S., Demopulos, G., Dudler, T., Yabuki, M., Wood, C. L., Cummings, W. J., Tjoelker, L. W., Fujita, T., Sacks, S., Garred, P., Andrew, P., Sim, R. B., Lachmann, P. J., Wallis, R., Lynch, N., Schwaeble, W. J. Lectin pathway effector enzyme mannan‐binding lectin‐associated serine protease‐2 can activate native complement C3 in absence of C4 and/or C2. FASEB J. 31, 2210–2219 (2017). www.fasebj.org


BMC Molecular Biology | 2009

RAD51 paralogs promote homology-directed repair at diversifying immunoglobulin V regions

Ellen C. Ordinario; Munehisa Yabuki; Priya Handa; W. Jason Cummings; Nancy Maizels

BackgroundGene conversion depends upon the same factors that carry out more general process of homologous recombination, including homologous gene targeting and recombinational repair. Among these are the RAD51 paralogs, conserved factors related to the key recombination factor, RAD51. In chicken and other fowl, gene conversion (templated mutation) diversifies immunoglobulin variable region sequences. This allows gene conversion and recombinational repair to be studied using the chicken DT40 B cell line, which carries out constitutive gene conversion and provides a robust and physiological model for homology-directed repair in vertebrate cells.ResultsWe show that DT40 contains constitutive nuclear foci of the repair factors RAD51D and XRCC2, consistent with activated homologous recombination. Single-cell imaging of a DT40 derivative in which the rearranged and diversifying immunoglobulin λR light chain gene is tagged with polymerized lactose operator, DT40 PolyLacO-λR, showed that RAD51D and XRCC2 localize to the diversifying λR gene. Colocalizations correlate both functionally and physically with active immunoglobulin gene conversion. Ectopic expression of either RAD51D or XRCC2 accelerated the clonal rate of gene conversion, and conversion tracts were significantly longer in RAD51D than XRCC2 transfectants.ConclusionThese results demonstrate direct functions of RAD51D and XRCC2 in immunoglobulin gene conversion, and also suggest that modulation of levels of repair factors may be a useful strategy to promote gene correction in other cell types.


PLOS ONE | 2012

Antibody discovery ex vivo accelerated by the LacO/LacI regulatory network.

Munehisa Yabuki; W. Jason Cummings; John B. Leppard; Robert M. Immormino; Christi L. Wood; Daniel S. Allison; Patrick W. Gray; Larry W. Tjoelker; Nancy Maizels

Monoclonal antibodies (mAbs) can be potent and highly specific therapeutics, diagnostics and research reagents. Nonetheless, mAb discovery using current in vivo or in vitro approaches can be costly and time-consuming, with no guarantee of success. We have established a platform for rapid discovery and optimization of mAbs ex vivo. This DTLacO platform derives from a chicken B cell line that has been engineered to enable rapid selection and seamless maturation of high affinity mAbs. We have validated the DTLacO platform by generation of high affinity and specific mAbs to five cell surface targets, the receptor tyrosine kinases VEGFR2 and TIE2, the glycoprotein TROP2, the small TNF receptor family member FN14, and the G protein-coupled receptor FZD10. mAb discovery is rapid and humanization is straightforward, establishing the utility of the DTLacO platform for identification of mAbs for therapeutic and other applications.


Molecular Cell | 2005

MRE11/RAD50 cleaves DNA in the AID/UNG-dependent pathway of immunoglobulin gene diversification.

Erik D. Larson; W. Jason Cummings; David W. Bednarski; Nancy Maizels


Archive | 2008

Inducible mutagenesis of target genes

Nancy Maizels; W. Jason Cummings; Munehisa Yabuki


Archive | 2012

Anti-FZD10 monoclonal antibodies and methods for their use

W. Jason Cummings; Munehisa Yabuki; John B. Leppard; Christi L. Wood; Nancy Maizels; Daniel S. Allison; Larry W. Tjoelker

Collaboration


Dive into the W. Jason Cummings's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nancy Maizels

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erik D. Larson

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge