Network


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

Hotspot


Dive into the research topics where Jonathan M. Woo is active.

Publication


Featured researches published by Jonathan M. Woo.


PLOS ONE | 2009

Genetic Profiling Reveals Cross-Contamination and Misidentification of 6 Adenoid Cystic Carcinoma Cell Lines: ACC2, ACC3, ACCM, ACCNS, ACCS and CAC2

Janyaporn Phuchareon; Yoshihito Ohta; Jonathan M. Woo; David W. Eisele; Osamu Tetsu

Adenoid cystic carcinoma (ACC) is the second most common malignant neoplasm of the salivary glands. Most patients survive more than 5 years after surgery and postoperative radiation therapy. The 10 year survival rate, however, drops to 40%, due to locoregional recurrences and distant metastases. Improving long-term survival in ACC requires the development of more effective systemic therapies based on a better understanding of the biologic behavior of ACC. Much preclinical research in this field involves the use of cultured cells and, to date, several ACC cell lines have been established. Authentication of these cell lines, however, has not been reported. We performed DNA fingerprint analysis on six ACC cell lines using short tandem repeat (STR) examinations and found that all six cell lines had been contaminated with other cells. ACC2, ACC3, and ACCM were determined to be cervical cancer cells (HeLa cells), whereas the ACCS cell line was composed of T24 urinary bladder cancer cells. ACCNS and CAC2 cells were contaminated with cells derived from non-human mammalian species: the cells labeled ACCNS were mouse cells and the CAC2 cells were rat cells. These observations suggest that future studies using ACC cell lines should include cell line authentication to avoid the use of contaminated or non-human cells.


BMC Genetics | 2005

Analysis of four DLX homeobox genes in autistic probands

Steven P. Hamilton; Jonathan M. Woo; Elaine J. Carlson; Noël Ghanem; Marc Ekker; John L.R. Rubenstein

BackgroundLinkage studies in autism have identified susceptibility loci on chromosomes 2q and 7q, regions containing the DLX1/2 and DLX5/6 bigene clusters. The DLX genes encode homeodomain transcription factors that control craniofacial patterning and differentiation and survival of forebrain inhibitory neurons. We investigated the role that sequence variants in DLX genes play in autism by in-depth resequencing of these genes in 161 autism probands from the AGRE collection.ResultsSequencing of exons, exon/intron boundaries and known enhancers of DLX1, 2, 5 and 6 identified several nonsynonymous variants in DLX2 and DLX5 and a variant in a DLX5/6intragenic enhancer. The nonsynonymous variants were detected in 4 of 95 families from which samples were sequenced. Two of these four SNPs were not observed in 378 undiagnosed samples from North American populations, while the remaining 2 were seen in one sample each.ConclusionSegregation of these variants in pedigrees did not generally support a contribution to autism susceptibility by these genes, although functional analyses may provide insight into the biological understanding of these important proteins.


Nature | 2017

Discovery of stimulation-responsive immune enhancers with CRISPR activation

Dimitre R. Simeonov; Benjamin G. Gowen; Mandy Boontanrart; Theodore L. Roth; John D. Gagnon; Maxwell R. Mumbach; Ansuman T. Satpathy; Youjin Lee; Nicolas Bray; Alice Y. Chan; Dmytro S. Lituiev; Michelle L. Nguyen; Rachel E. Gate; Meena Subramaniam; Zhongmei Li; Jonathan M. Woo; Therese Mitros; Graham J. Ray; Gemma L. Curie; Nicki Naddaf; Julia S. Chu; Hong Ma; Eric Boyer; Frédéric Van Gool; Hailiang Huang; Ruize Liu; Victoria R. Tobin; Kathrin Schumann; Mark J. Daly; Kyle Kai-How Farh

The majority of genetic variants associated with common human diseases map to enhancers, non-coding elements that shape cell-type-specific transcriptional programs and responses to extracellular cues. Systematic mapping of functional enhancers and their biological contexts is required to understand the mechanisms by which variation in non-coding genetic sequences contributes to disease. Functional enhancers can be mapped by genomic sequence disruption, but this approach is limited to the subset of enhancers that are necessary in the particular cellular context being studied. We hypothesized that recruitment of a strong transcriptional activator to an enhancer would be sufficient to drive target gene expression, even if that enhancer was not currently active in the assayed cells. Here we describe a discovery platform that can identify stimulus-responsive enhancers for a target gene independent of stimulus exposure. We used tiled CRISPR activation (CRISPRa) to synthetically recruit a transcriptional activator to sites across large genomic regions (more than 100 kilobases) surrounding two key autoimmunity risk loci, CD69 and IL2RA. We identified several CRISPRa-responsive elements with chromatin features of stimulus-responsive enhancers, including an IL2RA enhancer that harbours an autoimmunity risk variant. Using engineered mouse models, we found that sequence perturbation of the disease-associated Il2ra enhancer did not entirely block Il2ra expression, but rather delayed the timing of gene activation in response to specific extracellular signals. Enhancer deletion skewed polarization of naive T cells towards a pro-inflammatory T helper (TH17) cell state and away from a regulatory T cell state. This integrated approach identifies functional enhancers and reveals how non-coding variation associated with human immune dysfunction alters context-specific gene programs.


The Journal of Clinical Pharmacology | 2005

Effect of the MDR1 C3435T variant and P-glycoprotein induction on dicloxacillin pharmacokinetics.

Wendy S. Putnam; Jonathan M. Woo; Yong Huang; Leslie Z. Benet

This study investigated 2 hypotheses about genotype‐phenotype relationships for the efflux transporter, P‐glycoprotein: (1) the presence of a synonymous C3435T variant in exon 26 of the MDR1 gene correlates to higher plasma concentrations of a P‐glycoprotein substrate, dicloxacillin, and (2) the effects of genotypic differences decrease under conditions of P‐glycoprotein induction by rifampin. Eighteen healthy volunteers received two 1‐g doses of dicloxacillin, one on the 1st study day and the other on the 11th day of rifampin dosing (600 mg daily). Dicloxacillin and its 5‐hydroxymethyl metabolite were analyzed using liquid chromatography/tandem mass spectrometry. Mean dicloxacillin Cmax measurements were 30.5 ± 13.5, 33.3 ± 4.7, and 31.1 ± 12.8 μg/mL in individuals with the CC, CT, and TT genotype at position 3435 in exon 26 of the MDR1 gene. Following rifampin dosing, the mean dicloxacillin Cmax across genotypes decreased from 31.4 ± 10.8 to 22.9 ± 7.0 μg/mL (P < .05), whereas the mean oral clearance increased from 235 ± 82 to 297 ± 71 mL/min (P < .001), and the mean absorption time increased from 0.71 ± 0.55 to 1.34 ± 0.77 h (P 7< .05). Rifampin treatment increased the formation clearance, Cmax, and AUC of the 5‐hydroxymethyl metabolite by 135%, 119%, and 59%, respectively. The C3435T variant had no effect on dicloxacillin pharmacokinetics. The data suggested that rifampin induced intestinal P‐glycoprotein and increased dicloxacillin metabolism.


Pharmacogenetics and Genomics | 2015

A Pharmacogenetic Candidate Gene Study of Tenofovir-Associated Fanconi Syndrome

Amber Dahlin; Matthias B. Wittwer; Melanie De La Cruz; Jonathan M. Woo; Rujuta A. Bam; Valeska Scharen-Guivel; John F. Flaherty; Adrian S. Ray; Tomas Cihlar; Samir Gupta; Kathleen M. Giacomini

Background Tenofovir disoproxil fumarate (TDF) is a widely used antiretroviral agent with favorable efficacy, safety, and tolerability profiles. However, renal adverse events, including the rare Fanconi syndrome (FS), may occur in a small subset of patients treated for HIV infections. Objectives The aim of this study was to identify genetic variants that may be associated with TDF-associated FS (TDF-FS). Methods DNA samples collected from 19 cases with TDF-FS and 36 matched controls were sequenced, and genetic association studies were conducted on eight candidate genes: ATP-binding cassette (ABC) transporters ABCC2 (MRP2) and ABCC4 (MRP4), solute carrier family members SLC22A6 (OAT1) and SLC22A8 (OAT3), adenylate kinases 2 (AK2) and 4 (AK4), chloride transporter CIC-5 CLCN5, and Lowe syndrome protein OCRL. The functional effects of a single nucleotide polymorphism (SNP) predicted to alter the transport of tenofovir were then investigated in cells expressing an identified variant of ABCC4. Results The case group showed a trend toward a higher proportion of rare alleles. Six SNPs in ABCC2 (three SNPs), ABCC4 (one SNP), and OCRL (two SNPs) were associated with TDF-FS case status; however, this association did not remain significant after correction for multiple testing. Six SNPs, present in OCRL (four SNPs) and ABCC2 (two SNPs), were significantly associated with increased serum creatinine levels in the cases, and this association remained significant after multiple test correction (P<2×10–4). One synonymous SNP in ABCC2 (rs8187707, P=2.10×10–4, &bgr;=−73.3 ml/min/1.73 m2) was also significantly associated with the decreased estimated glomerular filtration rate of creatinine among cases. However, these results were driven by rare SNPs present in a small number of severely affected cases. Finally, a previously uncharacterized, nonsynonymous SNP, rs11568694, that was predicted to alter MRP4 function had no significant effect on tenofovir cellular accumulation in vitro. Conclusion Although no single predictive genetic marker for the development of TDF-FS was identified, the findings from our study suggest that rare variants in multiple genes involved in the renal handling of tenofovir, and/or renal cell homeostasis, may be associated with increased susceptibility to TDF-FS.


Clinical Pharmacology & Therapeutics | 2003

The effect of induction conditions and MDR1 genotypes on dicloxacillin pharmacokinetics

Wendy S. Putnam; D.G. Desai; Yong Huang; Jonathan M. Woo; Leslie Z. Benet

Clinical Pharmacology & Therapeutics (2003) 73, P57–P57; doi:


Pediatric Blood & Cancer | 2012

Evaluation of polymorphisms in EWSR1 and risk of Ewing sarcoma: A report from the childhood cancer survivor study

Steven G. DuBois; Robert E. Goldsby; Mark R. Segal; Jonathan M. Woo; Kirsten Copren; John P. Kane; Clive R. Pullinger; Katherine K. Matthay; John S. Witte; Stephen L. Lessnick; Leslie L. Robison; Smita Bhatia; Louise C. Strong

Ewing sarcoma is a malignant bone tumor characterized by a high frequency of somatic EWSR1 translocations. Ewing sarcoma is less common in people of African or African‐American ancestry, suggesting a genetic etiology.


bioRxiv | 2016

Discovery of an autoimmunity-associated IL2RA enhancer by unbiased targeting of transcriptional activation

Dimitre R. Simeonov; Benjamin G. Gowen; Mandy Boontanrart; Theo Roth; Youjin Lee; Alice Y. Chan; Michelle L. Nguyen; Rachel E. Gate; Meena Subramaniam; Jonathan M. Woo; Therese Mitros; Graham J. Ray; Nicolas Bray; Gemma L. Curie; Nicki Naddaf; Eric Boyer; Frédéric Van Gool; Kathrin Schumann; Mark J. Daly; Kyle K Fahr; Chun Ye; Jeffrey A. Bluestone; Mark S. Anderson; Jacob E. Corn; Alexander Marson

The majority of genetic variants associated with common human diseases map to enhancers, non-coding elements that shape cell type-specific transcriptional programs and responses to specific extracellular cues 1-3. In order to understand the mechanisms by which non-coding genetic variation contributes to disease, systematic mapping of functional enhancers and their biological contexts is required. Here, we develop an unbiased discovery platform that can identify enhancers for a target gene without prior knowledge of their native functional context. We used tiled CRISPR activation (CRISPRa) to synthetically recruit transcription factors to sites across large genomic regions (>100 kilobases) surrounding two key autoimmunity risk loci, CD69 and IL2RA (interleukin-2 receptor alpha; CD25). We identified several CRISPRa responsive elements (CaREs) with stimulation-dependent enhancer activity, including an IL2RA enhancer that harbors an autoimmunity risk variant. Using engineered mouse models and genome editing of human primary T cells, we found that sequence perturbation of the disease-associated IL2RA enhancer does not block IL2RA expression, but rather delays the timing of gene activation in response to specific extracellular signals. This work develops an approach to rapidly identify functional enhancers within non-coding regions, decodes a key human autoimmunity association, and suggests a general mechanism by which genetic variation can cause immune dysfunction.


bioRxiv | 2018

Genome-wide CRISPR Screens in Primary Human T Cells Reveal Key Regulators of Immune Function

Eric Shifrut; Julia Carnevale; Victoria Tobin; Theodore L. Roth; Jonathan M. Woo; Christina Bui; P. Jonathan Li; Morgan E. Diolaiti; Alan Ashworth; Alexander Marson

Human T cells are central effectors of immunity and cancer immunotherapy. CRISPR-based functional studies in T cells could prioritize novel targets for drug development and improve the design of genetically reprogrammed cell-based therapies. However, large-scale CRISPR screens have been challenging in primary human cells. We developed a new method, sgRNA lentiviral infection with Cas9 protein electroporation (SLICE), to identify regulators of stimulation responses in primary human T cells. Genome-wide loss-of-function screens identified essential T cell receptor signaling components and genes that negatively tune proliferation following stimulation. Targeted ablation of individual candidate genes validated hits and identified perturbations that enhanced cancer cell killing. SLICE coupled with single-cell RNA-Seq revealed signature stimulation-response gene programs altered by key genetic perturbations. SLICE genome-wide screening was also adaptable to identify mediators of immunosuppression, revealing genes controlling response to adenosine signaling. The SLICE platform enables unbiased discovery and characterization of functional gene targets in primary cells.


Nature | 2018

Author Correction: Discovery of stimulation-responsive immune enhancers with CRISPR activation

Dimitre R. Simeonov; Benjamin G. Gowen; Mandy Boontanrart; Theodore L. Roth; John D. Gagnon; Maxwell R. Mumbach; Ansuman T. Satpathy; Youjin Lee; Nicolas Bray; Alice Y. Chan; Dmytro S. Lituiev; Michelle L. Nguyen; Rachel E. Gate; Meena Subramaniam; Zhongmei Li; Jonathan M. Woo; Therese Mitros; Graham J. Ray; Gemma L. Curie; Nicki Naddaf; Julia S. Chu; Hong Ma; Eric Boyer; Frédéric Van Gool; Hailiang Huang; Ruize Liu; Victoria R. Tobin; Kathrin Schumann; Mark J. Daly; Kyle Kai-How Farh

In this Letter, analysis of steady-state regulatory T (Treg) cell percentages from Il2ra enhancer deletion (EDEL) and wild-type (WT) mice revealed no differences between them (Extended Data Fig. 9d). This analysis included two mice whose genotypes were incorrectly assigned. Even after correction of the genotypes, no significant differences in Treg cell percentages were seen when data across experimental cohorts were averaged (as was done in Extended Data Fig. 9d). However, if we normalize the corrected data to account for variation among experimental cohorts, a subtle decrease in EDEL Treg cell percentages is revealed and, using the corrected and normalized data, we have redrawn Extended Data Fig. 9d in Supplementary Fig. 1. The Supplementary Information to this Amendment contains the corrected and reanalysed Extended Data Fig. 9d. The sentence “This enhancer deletion (EDEL) strain also had no obvious T cell phenotypes at steady state (Extended Data Fig. 9).” should read: “This enhancer deletion (EDEL) strain had a small decrease in the percentage of Treg cells (Extended Data Fig. 9).”. This error does not affect any of the main figures in the Letter or the data from mice with the human autoimmune-associated single nucleotide polymorphism (SNP) knocked in or with a 12-base-pair deletion at the site (12DEL). In addition, we stated in the Methods that we observed consistent immunophenotypes of EDEL mice across three founders, but in fact, we observed consistent phenotypes in mice from two founders. This does not change any of our conclusions and the original Letter has not been corrected.

Collaboration


Dive into the Jonathan M. Woo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alice Y. Chan

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Boyer

University of California

View shared research outputs
Top Co-Authors

Avatar

Gemma L. Curie

University of California

View shared research outputs
Top Co-Authors

Avatar

Graham J. Ray

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge