Network


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

Hotspot


Dive into the research topics where Dimitre R. Simeonov is active.

Publication


Featured researches published by Dimitre R. Simeonov.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Generation of knock-in primary human T cells using Cas9 ribonucleoproteins

Kathrin Schumann; Steven Lin; Eric Boyer; Dimitre R. Simeonov; Meena Subramaniam; Rachel E. Gate; Genevieve E. Haliburton; Chun Ye; Jeffrey A. Bluestone; Jennifer A. Doudna; Alexander Marson

Significance T-cell genome engineering holds great promise for cancer immunotherapies and cell-based therapies for HIV, primary immune deficiencies, and autoimmune diseases, but genetic manipulation of human T cells has been inefficient. We achieved efficient genome editing by delivering Cas9 protein pre-assembled with guide RNAs. These active Cas9 ribonucleoproteins (RNPs) enabled successful Cas9-mediated homology-directed repair in primary human T cells. Cas9 RNPs provide a programmable tool to replace specific nucleotide sequences in the genome of mature immune cells—a longstanding goal in the field. These studies establish Cas9 RNP technology for diverse experimental and therapeutic genome engineering applications in primary human T cells. T-cell genome engineering holds great promise for cell-based therapies for cancer, HIV, primary immune deficiencies, and autoimmune diseases, but genetic manipulation of human T cells has been challenging. Improved tools are needed to efficiently “knock out” genes and “knock in” targeted genome modifications to modulate T-cell function and correct disease-associated mutations. CRISPR/Cas9 technology is facilitating genome engineering in many cell types, but in human T cells its efficiency has been limited and it has not yet proven useful for targeted nucleotide replacements. Here we report efficient genome engineering in human CD4+ T cells using Cas9:single-guide RNA ribonucleoproteins (Cas9 RNPs). Cas9 RNPs allowed ablation of CXCR4, a coreceptor for HIV entry. Cas9 RNP electroporation caused up to ∼40% of cells to lose high-level cell-surface expression of CXCR4, and edited cells could be enriched by sorting based on low CXCR4 expression. Importantly, Cas9 RNPs paired with homology-directed repair template oligonucleotides generated a high frequency of targeted genome modifications in primary T cells. Targeted nucleotide replacement was achieved in CXCR4 and PD-1 (PDCD1), a regulator of T-cell exhaustion that is a validated target for tumor immunotherapy. Deep sequencing of a target site confirmed that Cas9 RNPs generated knock-in genome modifications with up to ∼20% efficiency, which accounted for up to approximately one-third of total editing events. These results establish Cas9 RNP technology for diverse experimental and therapeutic genome engineering applications in primary human T cells.


Annals of clinical and translational neurology | 2014

GRIN2A mutation and early-onset epileptic encephalopathy: personalized therapy with memantine.

Tyler Mark Pierson; Hongjie Yuan; Eric D. Marsh; Karin Fuentes-Fajardo; David Adams; Thomas C. Markello; Gretchen Golas; Dimitre R. Simeonov; Conisha Holloman; Anel Tankovic; Manish M. Karamchandani; John M. Schreiber; James C. Mullikin; Cynthia J. Tifft; Camilo Toro; Cornelius F. Boerkoel; Stephen F. Traynelis; William A Gahl

Early‐onset epileptic encephalopathies have been associated with de novo mutations of numerous ion channel genes. We employed techniques of modern translational medicine to identify a disease‐causing mutation, analyze its altered behavior, and screen for therapeutic compounds to treat the proband.


European Journal of Human Genetics | 2012

Exome sequencing and SNP analysis detect novel compound heterozygosity in fatty acid hydroxylase-associated neurodegeneration

Tyler Mark Pierson; Dimitre R. Simeonov; Murat Sincan; David A Adams; Thomas C. Markello; Gretchen Golas; Karin Fuentes-Fajardo; Nancy F. Hansen; Praveen F. Cherukuri; Pedro Cruz; Craig Blackstone; Cynthia J. Tifft; Cornelius F. Boerkoel; William A. Gahl

Fatty acid hydroxylase-associated neurodegeneration due to fatty acid 2-hydroxylase deficiency presents with a wide range of phenotypes including spastic paraplegia, leukodystrophy, and/or brain iron deposition. All previously described families with this disorder were consanguineous, with homozygous mutations in the probands. We describe a 10-year-old male, from a non-consanguineous family, with progressive spastic paraplegia, dystonia, ataxia, and cognitive decline associated with a sural axonal neuropathy. The use of high-throughput sequencing techniques combined with SNP array analyses revealed a novel paternally derived missense mutation and an overlapping novel maternally derived ∼28-kb genomic deletion in FA2H. This patient provides further insight into the consistent features of this disorder and expands our understanding of its phenotypic presentation. The presence of a sural nerve axonal neuropathy had not been previously associated with this disorder and so may extend the phenotype.


Nature Genetics | 2017

Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements

Maxwell R. Mumbach; Ansuman T. Satpathy; Evan A. Boyle; Chao Dai; Benjamin G. Gowen; Seung Woo Cho; Michelle L. Nguyen; Adam J Rubin; Jeffrey M. Granja; Katelynn R. Kazane; Yuning Wei; Trieu Nguyen; Peyton Greenside; M. Ryan Corces; Josh Tycko; Dimitre R. Simeonov; Nabeela Suliman; Rui Li; Jin Xu; Ryan A. Flynn; Anshul Kundaje; Paul A. Khavari; Alexander Marson; Jacob E. Corn; Thomas Quertermous; William J. Greenleaf; Howard Y. Chang

The challenge of linking intergenic mutations to target genes has limited molecular understanding of human diseases. Here we show that H3K27ac HiChIP generates high-resolution contact maps of active enhancers and target genes in rare primary human T cell subtypes and coronary artery smooth muscle cells. Differentiation of naive T cells into T helper 17 cells or regulatory T cells creates subtype-specific enhancer–promoter interactions, specifically at regions of shared DNA accessibility. These data provide a principled means of assigning molecular functions to autoimmune and cardiovascular disease risk variants, linking hundreds of noncoding variants to putative gene targets. Target genes identified with HiChIP are further supported by CRISPR interference and activation at linked enhancers, by the presence of expression quantitative trait loci, and by allele-specific enhancer loops in patient-derived primary cells. The majority of disease-associated enhancers contact genes beyond the nearest gene in the linear genome, leading to a fourfold increase in the number of potential target genes for autoimmune and cardiovascular diseases.


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.


Human Mutation | 2012

VAR‐MD: A tool to analyze whole exome–genome variants in small human pedigrees with mendelian inheritance

Murat Sincan; Dimitre R. Simeonov; David Adams; Thomas C. Markello; Tyler Mark Pierson; Camilo Toro; William A. Gahl; Cornelius F. Boerkoel

The analysis of variants generated by exome sequencing (ES) of families with rare Mendelian diseases is a time‐consuming, manual process that represents one barrier to applying the technology routinely. To address this issue, we have developed a software tool, VAR‐MD (http://research.nhgri.nih.gov/software/var‐md/), for analyzing the DNA sequence variants produced by human ES. VAR‐MD generates a ranked list of variants using predicted pathogenicity, Mendelian inheritance models, genotype quality, and population variant frequency data. VAR‐MD was tested using two previously solved data sets and one unsolved data set. In the solved cases, the correct variant was listed at the top of VAR‐MDs variant ranking. In the unsolved case, the correct variant was highly ranked, allowing for subsequent identification and validation. We conclude that VAR‐MD has the potential to enhance mutation identification using family based, annotated next generation sequencing data. Moreover, we predict an incremental advancement in software performance as the reference databases, such as Single Nucleotide Polymorphism Database and Human Gene Mutation Database, continue to improve. Hum Mutat 33:593–598, 2012.


Neurology | 2012

Exome sequencing as a diagnostic tool in a case of undiagnosed juvenile-onset GM1-gangliosidosis.

Tyler Mark Pierson; David Adams; Thomas C. Markello; Gretchen Golas; Sandra Yang; Murat Sincan; Dimitre R. Simeonov; Karin Fuentes Fajardo; Nancy F. Hansen; Praveen F. Cherukuri; Pedro Cruz; Jamie K. Teer; James C. Mullikin; Cornelius F. Boerkoel; William A. Gahl; Cynthia J. Tifft

Objective: To utilize high-throughput sequencing to determine the etiology of juvenile-onset neurodegeneration in a 19-year-old woman with progressive motor and cognitive decline. Methods: Exome sequencing identified an initial list of 133,555 variants in the probands family, which were filtered using segregation analysis, presence in dbSNP, and an empirically derived gene exclusion list. The filtered list comprised 52 genes: 21 homozygous variants and 31 compound heterozygous variants. These variants were subsequently scrutinized with predicted pathogenicity programs and for association with appropriate clinical syndromes. Results: Exome sequencing data identified 2 GLB1 variants (c.602G>A, p.R201H; c.785G>T, p.G262V). β-Galactosidase enzyme analysis prior to our evaluation was reported as normal; however, subsequent testing was consistent with juvenile-onset GM1-gangliosidosis. Urine oligosaccharide analysis was positive for multiple oligosaccharides with terminal galactose residues. Conclusions: We describe a patient with juvenile-onset neurodegeneration that had eluded diagnosis for over a decade. GM1-gangliosidosis had previously been excluded from consideration, but was subsequently identified as the correct diagnosis using exome sequencing. Exome sequencing can evaluate genes not previously associated with neurodegeneration, as well as most known neurodegeneration-associated genes. Our results demonstrate the utility of “agnostic” exome sequencing to evaluate patients with undiagnosed disorders, without prejudice from prior testing results.


Journal of Clinical Investigation | 2011

Nitisinone improves eye and skin pigmentation defects in a mouse model of oculocutaneous albinism

Ighovie F. Onojafe; David Adams; Dimitre R. Simeonov; Jun Zhang; Chi-Chao Chan; Isa Bernardini; Yuri V. Sergeev; Monika B. Dolinska; Ramakrishna P. Alur; Murray H. Brilliant; William A. Gahl; Brian P. Brooks

Mutation of the tyrosinase gene (TYR) causes oculocutaneous albinism, type 1 (OCA1), a condition characterized by reduced skin and eye melanin pigmentation and by vision loss. The retinal pigment epithelium influences postnatal visual development. Therefore, increasing ocular pigmentation in patients with OCA1 might enhance visual function. There are 2 forms of OCA1, OCA-1A and OCA-1B. Individuals with the former lack functional tyrosinase and therefore lack melanin, while individuals with the latter produce some melanin. We hypothesized that increasing plasma tyrosine concentrations using nitisinone, an FDA-approved inhibitor of tyrosine degradation, could stabilize tyrosinase and improve pigmentation in individuals with OCA1. Here, we tested this hypothesis in mice homozygous for either the Tyrc-2J null allele or the Tyrc-h allele, which model OCA-1A and OCA-1B, respectively. Only nitisinone-treated Tyrc-h/c-h mice manifested increased pigmentation in their fur and irides and had more pigmented melanosomes. High levels of tyrosine improved the stability and enzymatic function of the Tyrc-h protein and also increased overall melanin levels in melanocytes from a human with OCA-1B. These results suggest that the use of nitisinone in OCA-1B patients could improve their pigmentation and potentially ameliorate vision loss.


Science immunology | 2018

Revisiting IL-2: Biology and therapeutic prospects

Abul K. Abbas; Eleonora Trotta; Dimitre R. Simeonov; Alexander Marson; Jeffrey A. Bluestone

Deeper insights into the biology of interleukin-2 and its receptors are leading to therapeutic strategies for selective Treg stimulation. Interleukin-2 (IL-2), the first cytokine that was molecularly cloned, was shown to be a T cell growth factor essential for the proliferation of T cells and the generation of effector and memory cells. On the basis of this activity, the earliest therapeutic application of IL-2 was to boost immune responses in cancer patients. Therefore, it was a surprise that genetic deletion of the cytokine or its receptor led not only to the expected immune deficiency but also to systemic autoimmunity and lymphoproliferation. Subsequent studies established that IL-2 is essential for the maintenance of Foxp3+ regulatory T cells (Treg cells), and in its absence, there is a profound deficiency of Treg cells and resulting autoimmunity. We now know that IL-2 promotes the generation, survival, and functional activity of Treg cells and thus has dual and opposing functions: maintaining Treg cells to control immune responses and stimulating conventional T cells to promote immune responses. It is well documented that certain IL-2 conformations result in selective targeting of Treg cells by increasing reliance on CD25 binding while compromising CD122 binding. Recent therapeutic strategies have emerged to use IL-2, monoclonal antibodies to IL-2, or IL-2 variants to boost Treg cell numbers and function to treat autoimmune diseases while dealing with the continuing challenges to minimize the generation of effector and memory cells, natural killer cells, and other innate lymphoid populations.


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.

Collaboration


Dive into the Dimitre R. Simeonov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William A. Gahl

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Adams

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eric Boyer

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rachel E. Gate

University of California

View shared research outputs
Top Co-Authors

Avatar

Thomas C. Markello

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge