Network


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

Hotspot


Dive into the research topics where Daniel C. Factor is active.

Publication


Featured researches published by Daniel C. Factor.


Nature Biotechnology | 2013

Transcription factor–mediated reprogramming of fibroblasts to expandable, myelinogenic oligodendrocyte progenitor cells

Fadi J. Najm; Angela M. Lager; Anita Zaremba; Krysta Wyatt; Andrew V. Caprariello; Daniel C. Factor; Robert T. Karl; Tadao Maeda; Robert H. Miller; Paul J. Tesar

Cell-based therapies for myelin disorders, such as multiple sclerosis and leukodystrophies, require technologies to generate functional oligodendrocyte progenitor cells. Here we describe direct conversion of mouse embryonic and lung fibroblasts to induced oligodendrocyte progenitor cells (iOPCs) using sets of either eight or three defined transcription factors. iOPCs exhibit a bipolar morphology and global gene expression profile consistent with bona fide OPCs. They can be expanded in vitro for at least five passages while retaining the ability to differentiate into multiprocessed oligodendrocytes. When transplanted to hypomyelinated mice, iOPCs are capable of ensheathing host axons and generating compact myelin. Lineage conversion of somatic cells to expandable iOPCs provides a strategy to study the molecular control of oligodendrocyte lineage identity and may facilitate neurological disease modeling and autologous remyelinating therapies.


Nature | 2015

Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo

Fadi J. Najm; Mayur Madhavan; Anita Zaremba; Elizabeth Shick; Robert T. Karl; Daniel C. Factor; Tyler E. Miller; Zachary S. Nevin; Christopher Kantor; Alex Sargent; Kevin L. Quick; Daniela Schlatzer; Hong Tang; Ruben Papoian; Kyle R. Brimacombe; Min Shen; Matthew B. Boxer; Ajit Jadhav; Andrew P. Robinson; Joseph R. Podojil; Stephen D. Miller; Robert H. Miller; Paul J. Tesar

Multiple sclerosis involves an aberrant autoimmune response and progressive failure of remyelination in the central nervous system. Prevention of neural degeneration and subsequent disability requires remyelination through the generation of new oligodendrocytes, but current treatments exclusively target the immune system. Oligodendrocyte progenitor cells are stem cells in the central nervous system and the principal source of myelinating oligodendrocytes. These cells are abundant in demyelinated regions of patients with multiple sclerosis, yet fail to differentiate, thereby representing a cellular target for pharmacological intervention. To discover therapeutic compounds for enhancing myelination from endogenous oligodendrocyte progenitor cells, we screened a library of bioactive small molecules on mouse pluripotent epiblast stem-cell-derived oligodendrocyte progenitor cells. Here we show seven drugs function at nanomolar doses selectively to enhance the generation of mature oligodendrocytes from progenitor cells in vitro. Two drugs, miconazole and clobetasol, are effective in promoting precocious myelination in organotypic cerebellar slice cultures, and in vivo in early postnatal mouse pups. Systemic delivery of each of the two drugs significantly increases the number of new oligodendrocytes and enhances remyelination in a lysolecithin-induced mouse model of focal demyelination. Administering each of the two drugs at the peak of disease in an experimental autoimmune encephalomyelitis mouse model of chronic progressive multiple sclerosis results in striking reversal of disease severity. Immune response assays show that miconazole functions directly as a remyelinating drug with no effect on the immune system, whereas clobetasol is a potent immunosuppressant as well as a remyelinating agent. Mechanistic studies show that miconazole and clobetasol function in oligodendrocyte progenitor cells through mitogen-activated protein kinase and glucocorticoid receptor signalling, respectively. Furthermore, both drugs enhance the generation of human oligodendrocytes from human oligodendrocyte progenitor cells in vitro. Collectively, our results provide a rationale for testing miconazole and clobetasol, or structurally modified derivatives, to enhance remyelination in patients.


Cell Stem Cell | 2014

Epigenomic Comparison Reveals Activation of “Seed” Enhancers during Transition from Naive to Primed Pluripotency

Daniel C. Factor; Olivia Corradin; Gabriel E. Zentner; Alina Saiakhova; Lingyun Song; Josh G. Chenoweth; Ronald D. G. McKay; Gregory E. Crawford; Peter C. Scacheri; Paul J. Tesar

Naive mouse embryonic stem cells (mESCs) and primed epiblast stem cells (mEpiSCs) represent successive snapshots of pluripotency during embryogenesis. Using transcriptomic and epigenomic mapping we show that a small fraction of transcripts are differentially expressed between mESCs and mEpiSCs and that these genes show expected changes in chromatin at their promoters and enhancers. Unexpectedly, the cis-regulatory circuitry of genes that are expressed at identical levels between these cell states also differs dramatically. In mESCs, these genes are associated with dominant proximal enhancers and dormant distal enhancers, which we term seed enhancers. In mEpiSCs, the naive-dominant enhancers are lost, and the seed enhancers take up primary transcriptional control. Seed enhancers have increased sequence conservation and show preferential usage in downstream somatic tissues, often expanding into super enhancers. We propose that seed enhancers ensure proper enhancer utilization and transcriptional fidelity as mammalian cells transition from naive pluripotency to a somatic regulatory program.


Nature | 2017

Transcription elongation factors represent in vivo cancer dependencies in glioblastoma

Tyler E. Miller; Brian B. Liau; Lisa C Wallace; Andrew R. Morton; Qi Xie; Deobrat Dixit; Daniel C. Factor; Leo Kim; James J. Morrow; Qiulian Wu; Stephen C. Mack; Christopher G. Hubert; Shawn M. Gillespie; William A. Flavahan; Thomas Hoffmann; Rohit Thummalapalli; Michael T. Hemann; Patrick J. Paddison; Craig Horbinski; Johannes Zuber; Peter C. Scacheri; Bradley E. Bernstein; Paul J. Tesar; Jeremy N. Rich

Glioblastoma is a universally lethal cancer with a median survival time of approximately 15 months. Despite substantial efforts to define druggable targets, there are no therapeutic options that notably extend the lifespan of patients with glioblastoma. While previous work has largely focused on in vitro cellular models, here we demonstrate a more physiologically relevant approach to target discovery in glioblastoma. We adapted pooled RNA interference (RNAi) screening technology for use in orthotopic patient-derived xenograft models, creating a high-throughput negative-selection screening platform in a functional in vivo tumour microenvironment. Using this approach, we performed parallel in vivo and in vitro screens and discovered that the chromatin and transcriptional regulators needed for cell survival in vivo are non-overlapping with those required in vitro. We identified transcription pause–release and elongation factors as one set of in vivo-specific cancer dependencies, and determined that these factors are necessary for enhancer-mediated transcriptional adaptations that enable cells to survive the tumour microenvironment. Our lead hit, JMJD6, mediates the upregulation of in vivo stress and stimulus response pathways through enhancer-mediated transcriptional pause–release, promoting cell survival specifically in vivo. Targeting JMJD6 or other identified elongation factors extends survival in orthotopic xenograft mouse models, suggesting that targeting transcription elongation machinery may be an effective therapeutic strategy for glioblastoma. More broadly, this study demonstrates the power of in vivo phenotypic screening to identify new classes of ‘cancer dependencies’ not identified by previous in vitro approaches, and could supply new opportunities for therapeutic intervention.


Journal of Clinical Investigation | 2016

RBPJ maintains brain tumor–initiating cells through CDK9-mediated transcriptional elongation

Qi Xie; Qiulian Wu; Leo Kim; Tyler E. Miller; Brian B. Liau; Stephen C. Mack; Kailin Yang; Daniel C. Factor; Xiaoguang Fang; Zhi Huang; Wenchao Zhou; Kareem Alazem; Xiuxing Wang; Bradley E. Bernstein; Shideng Bao; Jeremy N. Rich

Glioblastomas co-opt stem cell regulatory pathways to maintain brain tumor-initiating cells (BTICs), also known as cancer stem cells. NOTCH signaling has been a molecular target in BTICs, but NOTCH antagonists have demonstrated limited efficacy in clinical trials. Recombining binding protein suppressor of hairless (RBPJ) is considered a central transcriptional mediator of NOTCH activity. Here, we report that pharmacologic NOTCH inhibitors were less effective than targeting RBPJ in suppressing tumor growth. While NOTCH inhibitors decreased canonical NOTCH gene expression, RBPJ regulated a distinct profile of genes critical to BTIC stemness and cell cycle progression. RBPJ was preferentially expressed by BTICs and required for BTIC self-renewal and tumor growth. MYC, a key BTIC regulator, bound the RBPJ promoter and treatment with a bromodomain and extraterminal domain (BET) family bromodomain inhibitor decreased MYC and RBPJ expression. Proteomic studies demonstrated that RBPJ binds CDK9, a component of positive transcription elongation factor b (P-TEFb), to target gene promoters, enhancing transcriptional elongation. Collectively, RBPJ links MYC and transcriptional control through CDK9, providing potential nodes of fragility for therapeutic intervention, potentially distinct from NOTCH.


Nature | 2017

Therapeutic targeting of ependymoma as informed by oncogenic enhancer profiling

Stephen C. Mack; Kristian W. Pajtler; Lukas Chavez; Konstantin Okonechnikov; Kelsey C. Bertrand; Xiuxing Wang; Serap Erkek; Alexander J. Federation; Anne Song; Christine Lee; Xin Wang; Laura McDonald; James J. Morrow; Alina Saiakhova; Patrick Sin-Chan; Qiulian Wu; Kulandaimanuvel Antony Michaelraj; Tyler E. Miller; Christopher G. Hubert; Marina Ryzhova; Livia Garzia; Laura K. Donovan; Stephen M. Dombrowski; Daniel C. Factor; Betty Luu; Claudia L.L. Valentim; Ryan C. Gimple; Andrew R. Morton; Leo Kim; Briana Prager

Genomic sequencing has driven precision-based oncology therapy; however, the genetic drivers of many malignancies remain unknown or non-targetable, so alternative approaches to the identification of therapeutic leads are necessary. Ependymomas are chemotherapy-resistant brain tumours, which, despite genomic sequencing, lack effective molecular targets. Intracranial ependymomas are segregated on the basis of anatomical location (supratentorial region or posterior fossa) and further divided into distinct molecular subgroups that reflect differences in the age of onset, gender predominance and response to therapy. The most common and aggressive subgroup, posterior fossa ependymoma group A (PF-EPN-A), occurs in young children and appears to lack recurrent somatic mutations. Conversely, posterior fossa ependymoma group B (PF-EPN-B) tumours display frequent large-scale copy number gains and losses but have favourable clinical outcomes. More than 70% of supratentorial ependymomas are defined by highly recurrent gene fusions in the NF-κB subunit gene RELA (ST-EPN-RELA), and a smaller number involve fusion of the gene encoding the transcriptional activator YAP1 (ST-EPN-YAP1). Subependymomas, a distinct histologic variant, can also be found within the supratetorial and posterior fossa compartments, and account for the majority of tumours in the molecular subgroups ST-EPN-SE and PF-EPN-SE. Here we describe mapping of active chromatin landscapes in 42 primary ependymomas in two non-overlapping primary ependymoma cohorts, with the goal of identifying essential super-enhancer-associated genes on which tumour cells depend. Enhancer regions revealed putative oncogenes, molecular targets and pathways; inhibition of these targets with small molecule inhibitors or short hairpin RNA diminished the proliferation of patient-derived neurospheres and increased survival in mouse models of ependymomas. Through profiling of transcriptional enhancers, our study provides a framework for target and drug discovery in other cancers that lack known genetic drivers and are therefore difficult to treat.


Methods of Molecular Biology | 2013

Generation and Characterization of Epiblast Stem Cells from Blastocyst-Stage Mouse Embryos

Daniel C. Factor; Fadi J. Najm; Paul J. Tesar

Mouse epiblast stem cells (EpiSCs) are pluripotent embryonic cells that can be used to interrogate developmental transitions that occur during gastrulation. EpiSCs can also be robustly differentiated into functional somatic and germ cell derivatives making them a useful tool for studying development and regenerative medicine. Typically, mouse EpiSCs are isolated from the early postimplantation epiblast around 5.5 days post coitum (dpc). This chapter describes the methods for isolation of mouse EpiSCs from preimplantation blastocyst-stage mouse embryos (3.5 dpc). This technique enables the routine ability to derive EpiSC lines as it is much less labor intensive than isolation of EpiSCs from the postimplantation epiblast. We also detail relevant assays used to characterize new EpiSC lines and distinguish them from mouse embryonic stem cells.


Nature Methods | 2018

Induction of myelinating oligodendrocytes in human cortical spheroids

Mayur Madhavan; Zachary S. Nevin; H. Elizabeth Shick; Eric Garrison; Cheryl Clarkson-Paredes; Molly Karl; Benjamin L. L. Clayton; Daniel C. Factor; Kevin C. Allan; Lilianne Barbar; Tanya Jain; Panagiotis Douvaras; Valentina Fossati; Robert H. Miller; Paul J. Tesar

Cerebral organoids provide an accessible system for investigations of cellular composition, interactions, and organization but have lacked oligodendrocytes, the myelinating glia of the central nervous system. Here we reproducibly generated oligodendrocytes and myelin in ‘oligocortical spheroids’ derived from human pluripotent stem cells. Molecular features consistent with those of maturing oligodendrocytes and early myelin appeared by week 20 in culture, with further maturation and myelin compaction evident by week 30. Promyelinating drugs enhanced the rate and extent of oligodendrocyte generation and myelination, and spheroids generated from human subjects with a genetic myelin disorder recapitulated human disease phenotypes. Oligocortical spheroids provide a versatile platform for studies of myelination of the developing central nervous system and offer new opportunities for disease modeling and therapeutic development.A method for generating cortical spheroids from human pluripotent stem cells produces maturing oligodendrocytes that can myelinate axons and model myelin disease and drug effects.


Nature | 2018

Accumulation of 8,9-unsaturated sterols drives oligodendrocyte formation and remyelination

Zita Hubler; Dharmaraja Allimuthu; Ilya R. Bederman; Matthew S. Elitt; Mayur Madhavan; Kevin C. Allan; H. Elizabeth Shick; Eric Garrison; Molly Karl; Daniel C. Factor; Zachary S. Nevin; Joel L. Sax; Matthew Thompson; Yuriy Fedorov; Jing Jin; William K. Wilson; Martin Giera; Franz Bracher; Robert H. Miller; Paul J. Tesar; Drew J. Adams

Regeneration of myelin is mediated by oligodendrocyte progenitor cells—an abundant stem cell population in the central nervous system (CNS) and the principal source of new myelinating oligodendrocytes. Loss of myelin-producing oligodendrocytes in the CNS underlies a number of neurological diseases, including multiple sclerosis and diverse genetic diseases1–3. High-throughput chemical screening approaches have been used to identify small molecules that stimulate the formation of oligodendrocytes from oligodendrocyte progenitor cells and functionally enhance remyelination in vivo4–10. Here we show that a wide range of these pro-myelinating small molecules function not through their canonical targets but by directly inhibiting CYP51, TM7SF2, or EBP, a narrow range of enzymes within the cholesterol biosynthesis pathway. Subsequent accumulation of the 8,9-unsaturated sterol substrates of these enzymes is a key mechanistic node that promotes oligodendrocyte formation, as 8,9-unsaturated sterols are effective when supplied to oligodendrocyte progenitor cells in purified form whereas analogous sterols that lack this structural feature have no effect. Collectively, our results define a unifying sterol-based mechanism of action for most known small-molecule enhancers of oligodendrocyte formation and highlight specific targets to propel the development of optimal remyelinating therapeutics.Many small molecules that stimulate oligodendrocyte formation act not through their canonical pathways but by inhibiting enzymes within the cholesterol biosynthesis pathway and thereby inducing the accumulation of 8,9-unsaturated sterols.


Stem cell reports | 2018

Chemical Screening Identifies Enhancers of Mutant Oligodendrocyte Survival and Unmasks a Distinct Pathological Phase in Pelizaeus-Merzbacher Disease

Matthew S. Elitt; H. Elizabeth Shick; Mayur Madhavan; Kevin C. Allan; Benjamin L. L. Clayton; Chen Weng; Tyler E. Miller; Daniel C. Factor; Lilianne Barbar; Baraa S. Nawash; Zachary S. Nevin; Angela M. Lager; Yan Li; Fulai Jin; Drew J. Adams; Paul J. Tesar

Summary Pelizaeus-Merzbacher disease (PMD) is a fatal X-linked disorder caused by loss of myelinating oligodendrocytes and consequent hypomyelination. The underlying cellular and molecular dysfunctions are not fully defined, but therapeutic enhancement of oligodendrocyte survival could restore functional myelination in patients. Here we generated pure, scalable quantities of induced pluripotent stem cell-derived oligodendrocyte progenitor cells (OPCs) from a severe mouse model of PMD, Plp1jimpy. Temporal phenotypic and transcriptomic studies defined an early pathological window characterized by endoplasmic reticulum (ER) stress and cell death as OPCs exit their progenitor state. High-throughput phenotypic screening identified a compound, Ro 25–6981, which modulates the ER stress response and rescues mutant oligodendrocyte survival in jimpy, in vitro and in vivo, and in human PMD oligocortical spheroids. Surprisingly, increasing oligodendrocyte survival did not restore subsequent myelination, revealing a second pathological phase. Collectively, our work shows that PMD oligodendrocyte loss can be rescued pharmacologically and defines a need for multifactorial intervention to restore myelination.

Collaboration


Dive into the Daniel C. Factor's collaboration.

Top Co-Authors

Avatar

Paul J. Tesar

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Tyler E. Miller

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Zachary S. Nevin

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Leo Kim

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Mayur Madhavan

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert H. Miller

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Stephen C. Mack

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Andrew R. Morton

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge