Network


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

Hotspot


Dive into the research topics where Francine E. Garrett-Bakelman is active.

Publication


Featured researches published by Francine E. Garrett-Bakelman.


Genome Biology | 2012

methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles

Altuna Akalin; Matthias Kormaksson; Sheng Li; Francine E. Garrett-Bakelman; Maria E. Figueroa; Ari Melnick; Christopher E. Mason

DNA methylation is a chemical modification of cytosine bases that is pivotal for gene regulation,cellular specification and cancer development. Here, we describe an R package, methylKit, thatrapidly analyzes genome-wide cytosine epigenetic profiles from high-throughput methylation andhydroxymethylation sequencing experiments. methylKit includes functions for clustering, samplequality visualization, differential methylation analysis and annotation features, thus automatingand simplifying many of the steps for discerning statistically significant bases or regions of DNAmethylation. Finally, we demonstrate methylKit on breast cancer data, in which we find statisticallysignificant regions of differential methylation and stratify tumor subtypes. methylKit is availableat http://code.google.com/p/methylkit.


PLOS Genetics | 2012

Base-pair resolution DNA methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia

Altuna Akalin; Francine E. Garrett-Bakelman; Matthias Kormaksson; Jennifer Busuttil; Lu Zhang; Irina Khrebtukova; Thomas A. Milne; Yongsheng Huang; Debabrata Biswas; Jay L. Hess; C. David Allis; Robert G. Roeder; Bob Löwenberg; Ruud Delwel; Hugo F. Fernandez; Elisabeth Paietta; Martin S. Tallman; Gary P. Schroth; Christopher E. Mason; Ari Melnick; Maria E. Figueroa

We have developed an enhanced form of reduced representation bisulfite sequencing with extended genomic coverage, which resulted in greater capture of DNA methylation information of regions lying outside of traditional CpG islands. Applying this method to primary human bone marrow specimens from patients with Acute Myelogeneous Leukemia (AML), we demonstrated that genetically distinct AML subtypes display diametrically opposed DNA methylation patterns. As compared to normal controls, we observed widespread hypermethylation in IDH mutant AMLs, preferentially targeting promoter regions and CpG islands neighboring the transcription start sites of genes. In contrast, AMLs harboring translocations affecting the MLL gene displayed extensive loss of methylation of an almost mutually exclusive set of CpGs, which instead affected introns and distal intergenic CpG islands and shores. When analyzed in conjunction with gene expression profiles, it became apparent that these specific patterns of DNA methylation result in differing roles in gene expression regulation. However, despite this subtype-specific DNA methylation patterning, a much smaller set of CpG sites are consistently affected in both AML subtypes. Most CpG sites in this common core of aberrantly methylated CpGs were hypermethylated in both AML subtypes. Therefore, aberrant DNA methylation patterns in AML do not occur in a stereotypical manner but rather are highly specific and associated with specific driving genetic lesions.


Nature Medicine | 2017

The N 6 -methyladenosine (m 6 A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells

Ly P. Vu; Brian F. Pickering; Yuanming Cheng; Sara Zaccara; Diu Nguyen; Gerard Minuesa; Timothy Chou; Arthur Chow; Yogesh Saletore; Matthew MacKay; Jessica Schulman; Christopher Famulare; Minal Patel; Virginia M. Klimek; Francine E. Garrett-Bakelman; Ari Melnick; Martin Carroll; Christopher E. Mason; Samie R. Jaffrey; Michael G. Kharas

N6-methyladenosine (m6A) is an abundant nucleotide modification in mRNA that is required for the differentiation of mouse embryonic stem cells. However, it remains unknown whether the m6A modification controls the differentiation of normal and/or malignant myeloid hematopoietic cells. Here we show that shRNA-mediated depletion of the m6A-forming enzyme METTL3 in human hematopoietic stem/progenitor cells (HSPCs) promotes cell differentiation, coupled with reduced cell proliferation. Conversely, overexpression of wild-type METTL3, but not of a catalytically inactive form of METTL3, inhibits cell differentiation and increases cell growth. METTL3 mRNA and protein are expressed more abundantly in acute myeloid leukemia (AML) cells than in healthy HSPCs or other types of tumor cells. Furthermore, METTL3 depletion in human myeloid leukemia cell lines induces cell differentiation and apoptosis and delays leukemia progression in recipient mice in vivo. Single-nucleotide-resolution mapping of m6A coupled with ribosome profiling reveals that m6A promotes the translation of c-MYC, BCL2 and PTEN mRNAs in the human acute myeloid leukemia MOLM-13 cell line. Moreover, loss of METTL3 leads to increased levels of phosphorylated AKT, which contributes to the differentiation-promoting effects of METTL3 depletion. Overall, these results provide a rationale for the therapeutic targeting of METTL3 in myeloid leukemia.


Nature Medicine | 2016

Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia

Sheng Li; Francine E. Garrett-Bakelman; Stephen S. Chung; Mathijs A. Sanders; Todd Hricik; Franck Rapaport; Jay Patel; Richard Dillon; Priyanka Vijay; Anna L. Brown; Alexander E. Perl; Joy Cannon; Lars Bullinger; Selina M. Luger; Michael W. Becker; Ian D. Lewis; L. B. To; Ruud Delwel; Bob Löwenberg; Hartmut Döhner; Konstanze Döhner; Monica L. Guzman; Duane C. Hassane; Gail J. Roboz; David Grimwade; Peter J. M. Valk; Richard J. D'Andrea; Martin Carroll; Christopher Y. Park; Donna Neuberg

Genetic heterogeneity contributes to clinical outcome and progression of most tumors, but little is known about allelic diversity for epigenetic compartments, and almost no data exist for acute myeloid leukemia (AML). We examined epigenetic heterogeneity as assessed by cytosine methylation within defined genomic loci with four CpGs (epialleles), somatic mutations, and transcriptomes of AML patient samples at serial time points. We observed that epigenetic allele burden is linked to inferior outcome and varies considerably during disease progression. Epigenetic and genetic allelic burden and patterning followed different patterns and kinetics during disease progression. We observed a subset of AMLs with high epiallele and low somatic mutation burden at diagnosis, a subset with high somatic mutation and lower epiallele burdens at diagnosis, and a subset with a mixed profile, suggesting distinct modes of tumor heterogeneity. Genes linked to promoter-associated epiallele shifts during tumor progression showed increased single-cell transcriptional variance and differential expression, suggesting functional impact on gene regulation. Thus, genetic and epigenetic heterogeneity can occur with distinct kinetics likely to affect the biological and clinical features of tumors.


Nature Immunology | 2013

Satb1 regulates the self-renewal of hematopoietic stem cells by promoting quiescence and repressing differentiation commitment

Britta Will; Thomas O. Vogler; Boris Bartholdy; Francine E. Garrett-Bakelman; Jillian Mayer; Laura Barreyro; Ashley Pandolfi; Tihomira I. Todorova; Ujunwa C. Okoye-Okafor; Robert F. Stanley; Tushar D. Bhagat; Amit Verma; Maria E. Figueroa; Ari Melnick; Michael Roth; Ulrich Steidl

How hematopoietic stem cells (HSCs) coordinate the regulation of opposing cellular mechanisms such as self-renewal and differentiation commitment remains unclear. Here we identified the transcription factor and chromatin remodeler Satb1 as a critical regulator of HSC fate. HSCs lacking Satb1 had defective self-renewal, were less quiescent and showed accelerated lineage commitment, which resulted in progressive depletion of functional HSCs. The enhanced commitment was caused by less symmetric self-renewal and more symmetric differentiation divisions of Satb1-deficient HSCs. Satb1 simultaneously repressed sets of genes encoding molecules involved in HSC activation and cellular polarity, including Numb and Myc, which encode two key factors for the specification of stem-cell fate. Thus, Satb1 is a regulator that promotes HSC quiescence and represses lineage commitment.


BMC Bioinformatics | 2013

An optimized algorithm for detecting and annotating regional differential methylation

Sheng Li; Francine E. Garrett-Bakelman; Altuna Akalin; Paul Zumbo; Ross L. Levine; Bik To; Ian D. Lewis; Anna L. Brown; Richard J. D'Andrea; Ari Melnick; Christopher E. Mason

BackgroundDNA methylation profiling reveals important differentially methylated regions (DMRs) of the genome that are altered during development or that are perturbed by disease. To date, few programs exist for regional analysis of enriched or whole-genome bisulfate conversion sequencing data, even though such data are increasingly common. Here, we describe an open-source, optimized method for determining empirically based DMRs (eDMR) from high-throughput sequence data that is applicable to enriched whole-genome methylation profiling datasets, as well as other globally enriched epigenetic modification data.ResultsHere we show that our bimodal distribution model and weighted cost function for optimized regional methylation analysis provides accurate boundaries of regions harboring significant epigenetic modifications. Our algorithm takes the spatial distribution of CpGs into account for the enrichment assay, allowing for optimization of the definition of empirical regions for differential methylation. Combined with the dependent adjustment for regional p-value combination and DMR annotation, we provide a method that may be applied to a variety of datasets for rapid DMR analysis. Our method classifies both the directionality of DMRs and their genome-wide distribution, and we have observed that shows clinical relevance through correct stratification of two Acute Myeloid Leukemia (AML) tumor sub-types.ConclusionsOur weighted optimization algorithm eDMR for calling DMRs extends an established DMR R pipeline (methylKit) and provides a needed resource in epigenomics. Our method enables an accurate and scalable way of finding DMRs in high-throughput methylation sequencing experiments. eDMR is available for download at http://code.google.com/p/edmr/.


Nature Medicine | 2016

DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling

Olga A. Guryanova; Kaitlyn Shank; Barbara Spitzer; Luisa Luciani; Richard Koche; Francine E. Garrett-Bakelman; Chezi Ganzel; Benjamin H. Durham; Abhinita Mohanty; Gregor Hoermann; Sharon A. Rivera; Alan Chramiec; Elodie Pronier; Lennart Bastian; Matthew Keller; Daniel Tovbin; Evangelia Loizou; Abby Weinstein; Adriana Rodriguez Gonzalez; Yen K. Lieu; Jacob M. Rowe; Friederike Pastore; Anna Sophia McKenney; Andrei V. Krivtsov; Wolfgang R. Sperr; Justin R. Cross; Christopher E. Mason; Martin S. Tallman; Maria E. Arcila; Omar Abdel-Wahab

Although the majority of patients with acute myeloid leukemia (AML) initially respond to chemotherapy, many of them subsequently relapse, and the mechanistic basis for AML persistence following chemotherapy has not been determined. Recurrent somatic mutations in DNA methyltransferase 3A (DNMT3A), most frequently at arginine 882 (DNMT3AR882), have been observed in AML and in individuals with clonal hematopoiesis in the absence of leukemic transformation. Patients with DNMT3AR882 AML have an inferior outcome when treated with standard-dose daunorubicin-based induction chemotherapy, suggesting that DNMT3AR882 cells persist and drive relapse. We found that Dnmt3a mutations induced hematopoietic stem cell expansion, cooperated with mutations in the FMS-like tyrosine kinase 3 gene (Flt3ITD) and the nucleophosmin gene (Npm1c) to induce AML in vivo, and promoted resistance to anthracycline chemotherapy. In patients with AML, the presence of DNMT3AR882 mutations predicts minimal residual disease, underscoring their role in AML chemoresistance. DNMT3AR882 cells showed impaired nucleosome eviction and chromatin remodeling in response to anthracycline treatment, which resulted from attenuated recruitment of histone chaperone SPT-16 following anthracycline exposure. This defect led to an inability to sense and repair DNA torsional stress, which resulted in increased mutagenesis. Our findings identify a crucial role for DNMT3AR882 mutations in driving AML chemoresistance and highlight the importance of chromatin remodeling in response to cytotoxic chemotherapy.


Genome Biology | 2014

Dynamic evolution of clonal epialleles revealed by methclone.

Sheng Li; Francine E. Garrett-Bakelman; Alexander E. Perl; Selina M. Luger; Chao Zhang; Bik To; Ian D. Lewis; Anna L. Brown; Richard J. D’Andrea; M. Elizabeth Ross; Ross L. Levine; Martin Carroll; Ari Melnick; Christopher E. Mason

We describe methclone, a novel method to identify epigenetic loci that harbor large changes in the clonality of their epialleles (epigenetic alleles). Methclone efficiently analyzes genome-wide DNA methylation sequencing data. We quantify the changes using a composition entropy difference calculation and also introduce a new measure of global clonality shift, loci with epiallele shift per million loci covered, which enables comparisons between different samples to gauge overall epiallelic dynamics. Finally, we demonstrate the utility of methclone in capturing functional epiallele shifts in leukemia patients from diagnosis to relapse. Methclone is open-source and freely available at https://code.google.com/p/methclone.


Science Translational Medicine | 2017

CD99 is a therapeutic target on disease stem cells in myeloid malignancies

Stephen S. Chung; William S. Eng; Wenhuo Hu; Mona Khalaj; Francine E. Garrett-Bakelman; Montreh Tavakkoli; Ross L. Levine; Martin Carroll; Virginia M. Klimek; Ari Melnick; Christopher Y. Park

CD99 is a potential therapeutic target enriched on disease stem cells in myeloid malignancies. Stemming the tide of leukemia development Acute myeloid leukemia and myelodysplastic syndromes are maintained by specific populations of malignant stem cells, and successful treatment requires the eradication of these disease-causing cells. Chung et al. identified CD99 as a marker expressed on the surface of leukemic stem cells but not normal hematopoietic stem cells, suggesting its potential as a therapeutic target. A monoclonal antibody against CD99 had promising preclinical effectiveness in xenograft models and was selective for malignant stem cells, paving the way for further development of this approach. Acute myeloid leukemia (AML) and the myelodysplastic syndromes (MDS) are initiated and sustained by self-renewing malignant stem cells; thus, eradication of AML and MDS stem cells is required for cure. We identified CD99 as a cell surface protein frequently overexpressed on AML and MDS stem cells. Expression of CD99 allows for prospective separation of leukemic stem cells (LSCs) from functionally normal hematopoietic stem cells in AML, and high CD99 expression on AML blasts enriches for functional LSCs as demonstrated by limiting dilution xenotransplant studies. Monoclonal antibodies (mAbs) targeting CD99 induce the death of AML and MDS cells in a SARC family kinase–dependent manner in the absence of immune effector cells or complement, and anti-CD99 mAbs exhibit antileukemic activity in AML xenografts. These data establish CD99 as a marker of AML and MDS stem cells, as well as a promising therapeutic target in these disorders.


Cancer Discovery | 2017

Combination Targeted Therapy to Disrupt Aberrant Oncogenic Signaling and Reverse Epigenetic Dysfunction in IDH2- and TET2-Mutant Acute Myeloid Leukemia.

Alan H. Shih; Cem Meydan; Kaitlyn Shank; Francine E. Garrett-Bakelman; Patrick S. Ward; Andrew M. Intlekofer; Abbas Nazir; Eytan M. Stein; Kristina M. Knapp; Jacob Glass; Jeremy Travins; Kim Straley; Camelia Gliser; Christopher E. Mason; Katharine E. Yen; Craig B. Thompson; Ari Melnick; Ross L. Levine

Genomic studies in acute myeloid leukemias (AML) have identified mutations that drive altered DNA methylation, including TET2 and IDH2 Here, we show that models of AML resulting from TET2 or IDH2 mutations combined with FLT3ITD mutations are sensitive to 5-azacytidine or to the IDH2 inhibitor AG-221, respectively. 5-azacytidine and AG-221 treatment induced an attenuation of aberrant DNA methylation and transcriptional output and resulted in a reduction in leukemic blasts consistent with antileukemic activity. These therapeutic benefits were associated with restoration of leukemic cell differentiation, and the normalization of hematopoiesis was derived from mutant cells. By contrast, combining AG-221 or 5-azacytidine with FLT3 inhibition resulted in a reduction in mutant allele burden, progressive recovery of normal hematopoiesis from non-mutant stem-progenitor cells, and reversal of dysregulated DNA methylation and transcriptional output. Together, our studies suggest combined targeting of signaling and epigenetic pathways can increase therapeutic response in AML.Significance: AMLs with mutations in TET2 or IDH2 are sensitive to epigenetic therapy through inhibition of DNA methyltransferase activity by 5-azacytidine or inhibition of mutant IDH2 through AG-221. These inhibitors induce a differentiation response and can be used to inform mechanism-based combination therapy. Cancer Discov; 7(5); 494-505. ©2017 AACR.See related commentary by Thomas and Majeti, p. 459See related article by Yen et al., p. 478This article is highlighted in the In This Issue feature, p. 443.

Collaboration


Dive into the Francine E. Garrett-Bakelman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ross L. Levine

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Martin Carroll

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Christopher Y. Park

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen S. Chung

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ian D. Lewis

Royal Adelaide Hospital

View shared research outputs
Top Co-Authors

Avatar

Franck Rapaport

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kaitlyn Shank

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge