Network


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

Hotspot


Dive into the research topics where Casey A. Gifford is active.

Publication


Featured researches published by Casey A. Gifford.


Science | 2006

A virulence locus of Pseudomonas aeruginosa encodes a protein secretion apparatus.

Joseph D. Mougous; Marianne E. Cuff; Stefan Raunser; Aimee Shen; Min Zhou; Casey A. Gifford; Andrew L. Goodman; Grazyna Joachimiak; Claudia L. Ordoñez; Stephen Lory; Thomas Walz; Andrzej Joachimiak; John J. Mekalanos

Bacterial pathogens frequently use protein secretion to mediate interactions with their hosts. Here we found that a virulence locus (HSI-I) of Pseudomonas aeruginosa encodes a protein secretion apparatus. The apparatus assembled in discrete subcellular locations and exported Hcp1, a hexameric protein that forms rings with a 40 angstrom internal diameter. Regulatory patterns of HSI-I suggested that the apparatus functions during chronic infections. We detected Hcp1 in pulmonary secretions of cystic fibrosis (CF) patients and Hcp1-specific antibodies in their sera. Thus, HSI-I likely contributes to the pathogenesis of P. aeruginosa in CF patients. HSI-I–related loci are widely distributed among bacterial pathogens and may play a general role in mediating host interactions.


Cell | 2013

Transcriptional and epigenetic dynamics during specification of human embryonic stem cells.

Casey A. Gifford; Michael J. Ziller; Hongcang Gu; Cole Trapnell; Julie Donaghey; Alexander M. Tsankov; Alex K. Shalek; David R. Kelley; Alexander A. Shishkin; Robbyn Issner; Xiaolan Zhang; Michael J. Coyne; Jennifer L. Fostel; Laurie Holmes; Jim Meldrim; Mitchell Guttman; Charles B. Epstein; Hongkun Park; Oliver Kohlbacher; John L. Rinn; Andreas Gnirke; Eric S. Lander; Bradley E. Bernstein; Alexander Meissner

Differentiation of human embryonic stem cells (hESCs) provides a unique opportunity to study the regulatory mechanisms that facilitate cellular transitions in a human context. To that end, we performed comprehensive transcriptional and epigenetic profiling of populations derived through directed differentiation of hESCs representing each of the three embryonic germ layers. Integration of whole-genome bisulfite sequencing, chromatin immunoprecipitation sequencing, and RNA sequencing reveals unique events associated with specification toward each lineage. Lineage-specific dynamic alterations in DNA methylation and H3K4me1 are evident at putative distal regulatory elements that are frequently bound by pluripotency factors in the undifferentiated hESCs. In addition, we identified germ-layer-specific H3K27me3 enrichment at sites exhibiting high DNA methylation in the undifferentiated state. A better understanding of these initial specification events will facilitate identification of deficiencies in current approaches, leading to more faithful differentiation strategies as well as providing insights into the rewiring of human regulatory programs during cellular transitions.


Nature Cell Biology | 2007

Threonine phosphorylation post-translationally regulates protein secretion in Pseudomonas aeruginosa.

Joseph D. Mougous; Casey A. Gifford; Talia L. Ramsdell; John J. Mekalanos

Secreted proteins are crucial to the arsenal of bacterial pathogens. Although optimal activity of these proteins is likely to require precise regulation of release, the signalling events that trigger secretion are poorly understood. Here, we identify a threonine phosphorylation event that post-translationally regulates the Hcp secretion island-I-encoded type VI secretion system of Pseudomonas aeruginosa (H-T6SS). We show that a serine–threonine kinase, PpkA, is required for assembly of the H-T6SS and for secretion of Hcp1. PpkA activity is antagonized by PppA, a Ser–Thr phosphatase. These proteins exhibit reciprocal effects on the H-T6SS by acting on an FHA domain-containing protein, termed Fha1. Colocalization experiments with the T6S AAA+ family protein, ClpV1, indicate that Fha1 is a core scaffolding protein of the H-T6SS. Mutations affecting this H-T6S regulatory pathway provide a molecular explanation for the variation in Hcp1 secretion among clinical P. aeruginosa isolates. This mechanism of triggering secretion may be general, as many T6SSs contain orthologues of these proteins. Post-translational regulation of protein secretion by Thr phosphorylation is unprecedented in bacteria, and is likely to reflect the requirement for T6S to respond rapidly and reversibly to its environment.


Nature Genetics | 2015

Targeted disruption of DNMT1, DNMT3A and DNMT3B in human embryonic stem cells

Jing Liao; Rahul Karnik; Hongcang Gu; Michael J. Ziller; Kendell Clement; Alexander M. Tsankov; Veronika Akopian; Casey A. Gifford; Julie Donaghey; Christina Galonska; Ramona Pop; Deepak Reyon; Shengdar Q. Tsai; William Mallard; J. Keith Joung; John L. Rinn; Andreas Gnirke; Alexander Meissner

DNA methylation is a key epigenetic modification involved in regulating gene expression and maintaining genomic integrity. Here we inactivated all three catalytically active DNA methyltransferases (DNMTs) in human embryonic stem cells (ESCs) using CRISPR/Cas9 genome editing to further investigate the roles and genomic targets of these enzymes. Disruption of DNMT3A or DNMT3B individually as well as of both enzymes in tandem results in viable, pluripotent cell lines with distinct effects on the DNA methylation landscape, as assessed by whole-genome bisulfite sequencing. Surprisingly, in contrast to findings in mouse, deletion of DNMT1 resulted in rapid cell death in human ESCs. To overcome this immediate lethality, we generated a doxycycline-responsive tTA-DNMT1* rescue line and readily obtained homozygous DNMT1-mutant lines. However, doxycycline-mediated repression of exogenous DNMT1* initiates rapid, global loss of DNA methylation, followed by extensive cell death. Our data provide a comprehensive characterization of DNMT-mutant ESCs, including single-base genome-wide maps of the targets of these enzymes.


Nature | 2015

Dissecting neural differentiation regulatory networks through epigenetic footprinting

Michael J. Ziller; Reuven Edri; Yakey Yaffe; Julie Donaghey; Ramona Pop; William Mallard; Robbyn Issner; Casey A. Gifford; Alon Goren; Jeffrey Xing; Hongcang Gu; Davide Cacchiarelli; Alexander M. Tsankov; John L. Rinn; Tarjei S. Mikkelsen; Oliver Kohlbacher; Andreas Gnirke; Bradley E. Bernstein; Yechiel Elkabetz; Alexander Meissner

Models derived from human pluripotent stem cells that accurately recapitulate neural development in vitro and allow for the generation of specific neuronal subtypes are of major interest to the stem cell and biomedical community. Notch signalling, particularly through the Notch effector HES5, is a major pathway critical for the onset and maintenance of neural progenitor cells in the embryonic and adult nervous system. Here we report the transcriptional and epigenomic analysis of six consecutive neural progenitor cell stages derived from a HES5::eGFP reporter human embryonic stem cell line. Using this system, we aimed to model cell-fate decisions including specification, expansion and patterning during the ontogeny of cortical neural stem and progenitor cells. In order to dissect regulatory mechanisms that orchestrate the stage-specific differentiation process, we developed a computational framework to infer key regulators of each cell-state transition based on the progressive remodelling of the epigenetic landscape and then validated these through a pooled short hairpin RNA screen. We were also able to refine our previous observations on epigenetic priming at transcription factor binding sites and suggest here that they are mediated by combinations of core and stage-specific factors. Taken together, we demonstrate the utility of our system and outline a general framework, not limited to the context of the neural lineage, to dissect regulatory circuits of differentiation.


Cell | 2015

Integrative Analyses of Human Reprogramming Reveal Dynamic Nature of Induced Pluripotency

Davide Cacchiarelli; Cole Trapnell; Michael J. Ziller; Magali Soumillon; Marcella Cesana; Rahul Karnik; Julie Donaghey; Zachary D. Smith; Sutheera Ratanasirintrawoot; Xiaolan Zhang; Shannan J. Ho Sui; Zhaoting Wu; Veronika Akopian; Casey A. Gifford; John G. Doench; John L. Rinn; George Q. Daley; Alexander Meissner; Eric S. Lander; Tarjei S. Mikkelsen

Induced pluripotency is a promising avenue for disease modeling and therapy, but the molecular principles underlying this process, particularly in human cells, remain poorly understood due to donor-to-donor variability and intercellular heterogeneity. Here, we constructed and characterized a clonal, inducible human reprogramming system that provides a reliable source of cells at any stage of the process. This system enabled integrative transcriptional and epigenomic analysis across the human reprogramming timeline at high resolution. We observed distinct waves of gene network activation, including the ordered re-activation of broad developmental regulators followed by early embryonic patterning genes and culminating in the emergence of a signature reminiscent of pre-implantation stages. Moreover, complementary functional analyses allowed us to identify and validate novel regulators of the reprogramming process. Altogether, this study sheds light on the molecular underpinnings of induced pluripotency in human cells and provides a robust cell platform for further studies. PAPERCLIP.


Nature Biotechnology | 2015

A qPCR ScoreCard quantifies the differentiation potential of human pluripotent stem cells.

Alexander M. Tsankov; Veronika Akopian; Ramona Pop; Sundari Chetty; Casey A. Gifford; Laurence Daheron; Nadejda M. Tsankova; Alexander Meissner

Research on human pluripotent stem cells has been hampered by the lack of a standardized, quantitative, scalable assay of pluripotency. We previously described an assay called ScoreCard that used gene expression signatures to quantify differentiation efficiency. Here we report an improved version of the assay based on qPCR that enables faster, more quantitative assessment of functional pluripotency. We provide an in-depth characterization of the revised signature panel (commercially available as the TaqMan hPSC Scorecard Assay) through embryoid body and directed differentiation experiments as well as a detailed comparison to the teratoma assay. We further show that the improved ScoreCard enables a wider range of applications, such as screening of small molecules, genetic perturbations and assessment of culture conditions. Our approach can be extended beyond stem cell applications to characterize and assess the utility of other cell types and lineages.


Current Opinion in Genetics & Development | 2012

Epigenetic obstacles encountered by transcription factors: reprogramming against all odds.

Casey A. Gifford; Alexander Meissner

Reprogramming of a somatic nucleus to an induced pluripotent state can be achieved in vitro through ectopic expression of Oct4 (Pou5f1), Sox2, Klf4 and c-Myc. While the ability of these factors to regulate transcription in a pluripotent context has been studied extensively, their ability to interact with and remodel a somatic genome remains underexplored. Several recent studies have begun to provide mechanistic insights that will eventually lead to a more rational design and improved understanding of nuclear reprogramming.


Nature Genetics | 2018

Genetic determinants and epigenetic effects of pioneer-factor occupancy

Julie Donaghey; Sudhir Thakurela; Jocelyn Charlton; Jennifer S. Chen; Zachary D. Smith; Hongcang Gu; Ramona Pop; Kendell Clement; Elena K. Stamenova; Rahul Karnik; David R. Kelley; Casey A. Gifford; Davide Cacchiarelli; John L. Rinn; Andreas Gnirke; Michael J. Ziller; Alexander Meissner

Transcription factors (TFs) direct developmental transitions by binding to target DNA sequences, influencing gene expression and establishing complex gene-regultory networks. To systematically determine the molecular components that enable or constrain TF activity, we investigated the genomic occupancy of FOXA2, GATA4 and OCT4 in several cell types. Despite their classification as pioneer factors, all three TFs exhibit cell-type-specific binding, even when supraphysiologically and ectopically expressed. However, FOXA2 and GATA4 can be distinguished by low enrichment at loci that are highly occupied by these factors in alternative cell types. We find that expression of additional cofactors increases enrichment at a subset of these sites. Finally, FOXA2 occupancy and changes to DNA accessibility can occur in G1-arrested cells, but subsequent loss of DNA methylation requires DNA replication.Investigation of FOXA2, GATA4 and OCT4 binding across several cell types provides insights into the genetic determinants and epigenetic effects of pioneer-factor occupancy. The data suggest that FOXA2 samples most of its potential binding sites but is stabilized at only a subset of targets.


Scientific Reports | 2015

In vivo Monitoring of Transcriptional Dynamics After Lower-Limb Muscle Injury Enables Quantitative Classification of Healing

Carlos A. Aguilar; Anna Shcherbina; Darrell O. Ricke; Ramona Pop; Christopher T. Carrigan; Casey A. Gifford; Maria L. Urso; Melissa A. Kottke; Alexander Meissner

Traumatic lower-limb musculoskeletal injuries are pervasive amongst athletes and the military and typically an individual returns to activity prior to fully healing, increasing a predisposition for additional injuries and chronic pain. Monitoring healing progression after a musculoskeletal injury typically involves different types of imaging but these approaches suffer from several disadvantages. Isolating and profiling transcripts from the injured site would abrogate these shortcomings and provide enumerative insights into the regenerative potential of an individual’s muscle after injury. In this study, a traumatic injury was administered to a mouse model and healing progression was examined from 3 hours to 1 month using high-throughput RNA-Sequencing (RNA-Seq). Comprehensive dissection of the genome-wide datasets revealed the injured site to be a dynamic, heterogeneous environment composed of multiple cell types and thousands of genes undergoing significant expression changes in highly regulated networks. Four independent approaches were used to determine the set of genes, isoforms, and genetic pathways most characteristic of different time points post-injury and two novel approaches were developed to classify injured tissues at different time points. These results highlight the possibility to quantitatively track healing progression in situ via transcript profiling using high- throughput sequencing.

Collaboration


Dive into the Casey A. Gifford's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ramona Pop

University of Massachusetts Medical School

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge