Network


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

Hotspot


Dive into the research topics where Lyra Griffiths is active.

Publication


Featured researches published by Lyra Griffiths.


Cell Reports | 2014

Targeting the DNA Repair Pathway in Ewing Sarcoma

Elizabeth Stewart; Ross Goshorn; Cori Bradley; Lyra Griffiths; Claudia A. Benavente; Nathaniel R. Twarog; Gregory Miller; William Caufield; Burgess B. Freeman; Armita Bahrami; Alberto S. Pappo; Jianrong Wu; Amos Loh; Åsa Karlström; Chris Calabrese; Brittney Gordon; Lyudmila Tsurkan; M. Jason Hatfield; Philip M. Potter; Scott E. Snyder; Suresh Thiagarajan; Abbas Shirinifard; András Sablauer; Anang A. Shelat; Michael A. Dyer

Ewing sarcoma (EWS) is a tumor of the bone and soft tissue that primarily affects adolescents and young adults. With current therapies, 70% of patients with localized disease survive, but patients with metastatic or recurrent disease have a poor outcome. We found that EWS cell lines are defective in DNA break repair and are sensitive to PARP inhibitors (PARPis). PARPi-induced cytotoxicity in EWS cells was 10- to 1,000-fold higher after administration of the DNA-damaging agents irinotecan or temozolomide. We developed an orthotopic EWS mouse model and performed pharmacokinetic and pharmacodynamic studies using three different PARPis that are in clinical development for pediatric cancer. Irinotecan administered on a low-dose, protracted schedule previously optimized for pediatric patients was an effective DNA-damaging agent when combined with PARPis; it was also better tolerated than combinations with temozolomide. Combining PARPis with irinotecan and temozolomide gave complete and durable responses in more than 80% of the mice.


Cell Stem Cell | 2015

Quantification of Retinogenesis in 3D Cultures Reveals Epigenetic Memory and Higher Efficiency in iPSCs Derived from Rod Photoreceptors

Daniel Hiler; Xiang Chen; Jennifer L. Hazen; Sergey Kupriyanov; Patrick A. Carroll; Chunxu Qu; Beisi Xu; Dianna A. Johnson; Lyra Griffiths; Sharon Frase; Alberto R. Rodriguez; Greg Martin; Jiakun Zhang; Jongrye Jeon; Yiping Fan; David Finkelstein; Robert N. Eisenman; Kristin K. Baldwin; Michael A. Dyer

Cell-based therapies to treat retinal degeneration are now being tested in clinical trials. However, it is not known whether the source of stem cells is important for the production of differentiated cells suitable for transplantation. To test this, we generated induced pluripotent stem cells (iPSCs) from murine rod photoreceptors (r-iPSCs) and scored their ability to make retinae by using a standardized quantitative protocol called STEM-RET. We discovered that r-iPSCs more efficiently produced differentiated retinae than did embryonic stem cells (ESCs) or fibroblast-derived iPSCs (f-iPSCs). Retinae derived from f-iPSCs had fewer amacrine cells and other inner nuclear layer cells. Integrated epigenetic analysis showed that DNA methylation contributes to the defects in f-iPSC retinogenesis and that rod-specific CTCF insulator protein-binding sites may promote r-iPSC retinogenesis. Together, our data suggest that the source of stem cells is important for producing retinal neurons in three-dimensional (3D) organ cultures.


Neuron | 2017

The Dynamic Epigenetic Landscape of the Retina During Development, Reprogramming, and Tumorigenesis

Issam Aldiri; Beisi Xu; Lu Wang; Xiang Chen; Daniel Hiler; Lyra Griffiths; Marc Valentine; Abbas Shirinifard; Suresh Thiagarajan; András Sablauer; Marie-Elizabeth Barabas; Jiakun Zhang; Dianna A. Johnson; Sharon Frase; Xin Zhou; John Easton; Jinghui Zhang; Elaine R. Mardis; Richard Wilson; James R. Downing; Michael A. Dyer

SUMMARY In the developing retina, multipotent neural progenitors undergo unidirectional differentiation in a precise spatiotemporal order. Here we profile the epigenetic and transcriptional changes that occur during retinogenesis in mice and humans. Although some progenitor genes and cell cycle genes were epigenetically silenced during retinogenesis, the most dramatic change was derepression of cell type–specific differentiation programs. We identified developmental stage–specific super-enhancers and showed that most epigenetic changes are conserved in humans and mice. To determine how the epigenome changes during tumorigenesis and reprogramming, we performed integrated epigenetic analysis of murine and human retinoblastomas and induced pluripotent stem cells (iPSCs) derived from murine rod photoreceptors. The retinoblastoma epigenome mapped to the developmental stage when retinal progenitors switch from neurogenic to a terminal patterns of cell division. The epigenome of retinoblastomas was more similar to that of normal retina than was that of retina-derived iPSCs, and we identified retina-specific epigenetic memory.


The EMBO Journal | 2014

Post‐transcriptional gene expression control by NANOS is up‐regulated and functionally important in pRb‐deficient cells

Wayne O. Miles; Michael Korenjak; Lyra Griffiths; Michael A. Dyer; Paolo Provero; Nicholas J. Dyson

Inactivation of the retinoblastoma tumor suppressor (pRb) is a common oncogenic event that alters the expression of genes important for cell cycle progression, senescence, and apoptosis. However, in many contexts, the properties of pRb‐deficient cells are similar to wild‐type cells suggesting there may be processes that counterbalance the transcriptional changes associated with pRb inactivation. Therefore, we have looked for sets of evolutionary conserved, functionally related genes that are direct targets of pRb/E2F proteins. We show that the expression of NANOS, a key facilitator of the Pumilio (PUM) post‐transcriptional repressor complex, is directly repressed by pRb/E2F in flies and humans. In both species, NANOS expression increases following inactivation of pRb/RBF1 and becomes important for tissue homeostasis. By analyzing datasets from normal retinal tissue and pRb‐null retinoblastomas, we find a strong enrichment for putative PUM substrates among genes de‐regulated in tumors. These include pro‐apoptotic genes that are transcriptionally down‐regulated upon pRb loss, and we characterize two such candidates, MAP2K3 and MAP3K1, as direct PUM substrates. Our data suggest that NANOS increases in importance in pRb‐deficient cells and helps to maintain homeostasis by repressing the translation of transcripts containing PUM Regulatory Elements (PRE).


Journal of Molecular Biology | 2016

Monitoring Ligand-Induced Protein Ordering in Drug Discovery.

Christy Rani R. Grace; David Ban; Jaeki Min; Anand Mayasundari; Lie Min; Kristin Finch; Lyra Griffiths; Nagakumar Bharatham; Donald Bashford; R. Kiplin Guy; Michael A. Dyer; Richard W. Kriwacki

While the gene for p53 is mutated in many human cancers causing loss of function, many others maintain a wild-type gene but exhibit reduced p53 tumor suppressor activity through overexpression of the negative regulators, Mdm2 and/or MdmX. For the latter mechanism of loss of function, the activity of endogenous p53 can be restored through inhibition of Mdm2 or MdmX with small molecules. We previously reported a series of compounds based upon the Nutlin-3 chemical scaffold that bind to both MdmX and Mdm2 [Vara, B. A. et al. (2014) Organocatalytic, diastereo- and enantioselective synthesis of nonsymmetric cis-stilbene diamines: A platform for the preparation of single-enantiomer cis-imidazolines for protein-protein inhibition. J. Org. Chem. 79, 6913-6938]. Here we present the first solution structures based on data from NMR spectroscopy for MdmX in complex with four of these compounds and compare them with the MdmX:p53 complex. A p53-derived peptide binds with high affinity (Kd value of 150nM) and causes the formation of an extensive network of hydrogen bonds within MdmX; this constitutes the induction of order within MdmX through ligand binding. In contrast, the compounds bind more weakly (Kd values from 600nM to 12μM) and induce an incomplete hydrogen bond network within MdmX. Despite relatively weak binding, the four compounds activated p53 and induced p21(Cip1) expression in retinoblastoma cell lines that overexpress MdmX, suggesting that they specifically target MdmX and/or Mdm2. Our results document structure-activity relationships for lead-like small molecules targeting MdmX and suggest a strategy for their further optimization in the future by using NMR spectroscopy to monitor small-molecule-induced protein order as manifested through hydrogen bond formation.


Pharmaceutical Research | 2014

Pharmacokinetics and efficacy of the spleen tyrosine kinase inhibitor r406 after ocular delivery for retinoblastoma.

Eleanor M. Pritchard; Elizabeth Stewart; Fangyi Zhu; Cori Bradley; Lyra Griffiths; Lei Yang; Praveen Kumar Suryadevara; Jiakun Zhang; Burgess B. Freeman; R. Kiplin Guy; Michael A. Dyer

PurposeRetinoblastoma is a childhood cancer of the retina. Clinical trials have shown that local delivery of broad spectrum chemotherapeutic agents is efficacious. Recent studies characterizing the genomic and epigenomic landscape of retinoblastoma identified spleen tyrosine kinase (SYK) as a promising candidate for targeted therapy. The purpose of this study was to conduct preclinical testing of the SYK antagonist R406 to evaluate it as a candidate for retinoblastoma treatment.MethodsThe efficacy of the SYK antagonist R406 delivered locally in a human orthotopic xenograft mouse model of retinoblastoma was tested. Intraocular exposure of R406 was determined for various routes and formulations.ResultsThere was no evidence of efficacy for subconjunctival. R406. Maximal vitreal concentration was 10-fold lower than the minimal concentration required to kill retinoblastoma cells in vitro. Dosage of R406 subconjunctivally from emulsion or suspension formulations, direct intravitreal injection of the soluble prodrug of R406 (R788), and repeated topical administration of R406 all increased vitreal exposure, but failed to reach the exposure required for retinoblastoma cell death in culture.ConclusionTaken together, these data suggest that R406 is not a viable clinical candidate for the treatment of retinoblastoma. This study highlights the importance of pharmacokinetic testing of molecular targeted retinoblastoma therapeutics.


Cell Reports | 2018

Retinal Cell Type DNA Methylation and Histone Modifications Predict Reprogramming Efficiency and Retinogenesis in 3D Organoid Cultures

Lu Wang; Daniel Hiler; Beisi Xu; Issam Aldiri; Xiang Chen; Xin Zhou; Lyra Griffiths; Marc Valentine; Abbas Shirinifard; András Sablauer; Suresh Thiagarajan; Marie-Elizabeth Barabas; Jiakun Zhang; Dianna A. Johnson; Sharon Frase; Michael A. Dyer

SUMMARY Diverse cell types can be reprogrammed into pluripotent stem cells by ectopic expression of Oct4 (Pou5f1), Klf4, Sox3, and Myc. Many of these induced pluripotent stem cells (iPSCs) retain memory, in terms of DNA methylation and histone modifications (epigenetic memory), of their cellular origins, and this may bias subsequent differentiation. Neurons are difficult to reprogram, and there has not been a systematic side-by-side characterization of reprogramming efficiency or epigenetic memory across different neuronal subtypes. Here, we compare reprogramming efficiency of five different retinal cell types at two different stages of development. Retinal differentiation from each iPSC line was measured using a quantitative standardized scoring system called STEM-RET and compared to the epigenetic memory. Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins. In addition, we identified biomarkers of iPSCs that are predictive of retinal differentiation.


Nature Protocols | 2016

Reprogramming of mouse retinal neurons and standardized quantification of their differentiation in 3D retinal cultures

Daniel Hiler; Marie E Barabas; Lyra Griffiths; Michael A. Dyer

Postmitotic differentiated neurons are among the most difficult cells to reprogram into induced pluripotent stem cells (iPSCs) because they have poor viability when cultured as dissociated cells. To overcome this, other protocols have required the inactivation of the p53 tumor suppressor to reprogram postmitotic neurons, which can result in tumorigenesis of the cells. We describe a method that does not require p53 inactivation but induces reprogramming in retinal cells from reprogrammable mice grown in aggregates with wild-type mouse retinal cells. After the first 10 d of reprogramming, the aggregates are then dispersed and plated on irradiated feeder cells to propagate and isolate individual iPSC clones. The reprogramming efficiency of different neuronal populations at any stage of development can be quantified using this protocol. Reprogramming retinal neurons using this protocol will take 56 d, and these retina-derived iPSCs can undergo retinal differentiation to produce retinae in 34 d. In addition, we describe a quantitative assessment of retinal differentiation from these neuron-derived iPSCs called STEM-RET. The procedure quantifies eye field specification, optic cup formation and retinal differentiation in 3D cultures using molecular, cellular and morphological criteria. An advanced level of cell culture experience is required to carry out this protocol.


SpringerPlus | 2014

The ATRX cDNA is prone to bacterial IS10 element insertions that alter its structure

David Valle-Garcia; Lyra Griffiths; Michael A. Dyer; Emily Bernstein; Félix Recillas-Targa

The SWI/SNF-like chromatin-remodeling protein ATRX has emerged as a key factor in the regulation of α-globin gene expression, incorporation of histone variants into the chromatin template and, more recently, as a frequently mutated gene across a wide spectrum of cancers. Therefore, the availability of a functional ATRX cDNA for expression studies is a valuable tool for the scientific community. We have identified two independent transposon insertions of a bacterial IS10 element into exon 8 of ATRX isoform 2 coding sequence in two different plasmids derived from a single source. We demonstrate that these insertion events are common and there is an insertion hotspot within the ATRX cDNA. Such IS10 insertions produce a truncated form of ATRX, which significantly compromises its nuclear localization. In turn, we describe ways to prevent IS10 insertion during propagation and cloning of ATRX-containing vectors, including optimal growth conditions, bacterial strains, and suggested sequencing strategies. Finally, we have generated an insertion-free plasmid that is available to the community for expression studies of ATRX.


bioRxiv | 2018

The Nucleome of Developing Murine Rod Photoreceptors

Issam Al Diri; Marc Valentine; Beisi Xu; Daniel Putnam; Lyra Griffiths; Marybeth Lupo; Jackie Norrie; Jiakun Zhang; Dianna Johnson; John Easton; Abbas Shirinifard; Ying Shao; Victoria Honnell; Sharon Frase; Shondra Miller; Valerie Stewart; Xiang Chen; Michael A. Dyer

The nuclei of rod photoreceptors in mice and other nocturnal species have an unusual inverted chromatin structure: the heterochromatin is centrally located to help focus light and improve photosensitivity. To better understand this unique nuclear organization, we performed ultra-deep Hi-C analysis on murine retina at 3 stages of development and on purified rod photoreceptors. Predicted looping interactions from the Hi-C data were validated with fluorescence in situ hybridization (FISH). We discovered that a subset of retinal genes that are important for retinal development, cancer, and stress response are localized to the facultative heterochromatin domain. We also used machine learning to develop an algorithm based on our chromatin Hidden Markov Modeling (chromHMM) of retinal development to predict heterochromatin domains and study their dynamics during retinogenesis. FISH data for 264 genomic loci were used to train and validate the algorithm. The integrated data were then used to identify a developmental stage– and cell type-specific core regulatory circuit super-enhancer (CRC-SE) upstream of the Vsx2 gene, which is required for bipolar neuron expression. Deletion of the Vsx2 CRC-SE in mice led to the loss of bipolar neurons in the retina.

Collaboration


Dive into the Lyra Griffiths's collaboration.

Top Co-Authors

Avatar

Michael A. Dyer

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Beisi Xu

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Xiang Chen

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Stewart

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jiakun Zhang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Abbas Shirinifard

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Burgess B. Freeman

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Daniel Hiler

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

John Easton

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Marc Valentine

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge