Network


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

Hotspot


Dive into the research topics where Stacey M. Glasgow is active.

Publication


Featured researches published by Stacey M. Glasgow.


Neuron | 2012

Sox9 and NFIA Coordinate a Transcriptional Regulatory Cascade during the Initiation of Gliogenesis

Peng Kang; Hyun Kyoung Lee; Stacey M. Glasgow; Meggie Finley; Tataka Donti; Zachary B. Gaber; Brett H. Graham; Aaron E. Foster; Bennett G. Novitch; Richard M. Gronostajski; Benjamin Deneen

Transcriptional cascades that operate over the course of lineage development are fundamental mechanisms that control cellular differentiation. In the developing central nervous system (CNS), these mechanisms are well characterized during neurogenesis, but remain poorly defined during neural stem cell commitment to the glial lineage. NFIA is a transcription factor that plays a crucial role in the onset of gliogenesis; we found that its induction is regulated by the transcription factor Sox9 and that this relationship mediates the initiation of gliogenesis. Subsequently, Sox9 and NFIA form a complex and coregulate a set of genes induced after glial initiation. Functional studies revealed that a subset of these genes, Apcdd1 and Mmd2, perform key migratory and metabolic roles during astro-gliogenesis, respectively. In sum, these studies delineate a transcriptional regulatory cascade that operates during the initiation of gliogenesis and identifies a unique set of genes that regulate key aspects of astro-glial precursor physiology during development.


Nature Neuroscience | 2014

Mutual antagonism between Sox10 and NFIA regulates diversification of glial lineages and glioma subtypes

Stacey M. Glasgow; Wenyi Zhu; C. Claus Stolt; Teng-Wei Huang; Fuyi Chen; Joseph J. LoTurco; Jeffrey L. Neul; Michael Wegner; Carrie A. Mohila; Benjamin Deneen

Lineage progression and diversification is regulated by the coordinated action of unique sets of transcription factors. Oligodendrocytes (OL) and astrocytes (AS) comprise the glial sub-lineages in the CNS, and the manner in which their associated regulatory factors orchestrate lineage diversification during development and disease remains an open question. Sox10 and NFIA are key transcriptional regulators of gliogenesis associated with OL and AS. We found that NFIA inhibited Sox10 induction of OL differentiation through direct association and antagonism of its function. Conversely, we found that Sox10 antagonized NFIA function and suppressed AS differentiation in mouse and chick systems. Using this developmental paradigm as a model for glioma, we found that this relationship similarly regulated the generation of glioma subtypes. Our results describe the antagonistic relationship between Sox10 and NFIA that regulates the balance of OL and AS fate during development and demonstrate for the first time, to the best of our knowledge, that the transcriptional processes governing glial sub-lineage diversification oversee the generation of glioma subtypes.


The Journal of Neuroscience | 2013

The miR-223/Nuclear Factor I-A Axis Regulates Glial Precursor Proliferation and Tumorigenesis in the CNS

Stacey M. Glasgow; Dylan Laug; Vita Brawley; Zhiyuan Zhang; Amanda Corder; Zheng Yin; Stephen T. C. Wong; Xiao Nan Li; Aaron E. Foster; Nabil Ahmed; Benjamin Deneen

Contemporary views of tumorigenesis regard its inception as a convergence of genetic mutation and developmental context. Glioma is the most common and deadly malignancy in the CNS; therefore, understanding how regulators of glial development contribute to its formation remains a key question. Previously we identified nuclear factor I-A (NFIA) as a key regulator of developmental gliogenesis, while miR-223 has been shown to repress NFIA expression in other systems. Using this relationship as a starting point, we found that miR-223 can suppress glial precursor proliferation via repression of NFIA during chick spinal cord development. This relationship is conserved in glioma, as miR-223 and NFIA expression is negatively correlated in human glioma tumors, and the miR-223/NFIA axis suppresses tumorigenesis in a human glioma cell line. Subsequent analysis of NFIA function revealed that it directly represses p21 and is required for tumorigenesis in a mouse neural stem cell model of glioma. These studies represent the first characterization of miR-223/NFIA axis function in glioma and demonstrate that it is a conserved proliferative mechanism across CNS development and tumorigenesis.


Nature Communications | 2015

miR-218 is essential to establish motor neuron fate as a downstream effector of Isl1-Lhx3

Karen P. Thiebes; Heejin Nam; Xiaolu A. Cambronne; Rongkun Shen; Stacey M. Glasgow; Hyong Ho Cho; Ji Sun Kwon; Richard H. Goodman; Jae W. Lee; Seunghee Lee; Soo Kyung Lee

While microRNAs have emerged as an important component of gene regulatory networks, it remains unclear how microRNAs collaborate with transcription factors in the gene networks that determines neuronal cell fate. Here, we show that in the developing spinal cord, the expression of miR-218 is directly upregulated by the Isl1-Lhx3 complex, which drives motor neuron fate. Inhibition of miR-218 suppresses the generation of motor neurons in both chick neural tube and mouse embryonic stem cells, suggesting that miR-218 plays a crucial role in motor neuron differentiation. Results from unbiased RISC-trap screens, in vivo reporter assays, and overexpression studies indicated that miR-218 directly represses transcripts that promote developmental programs for interneurons. Additionally, we found that miR-218 activity is required for Isl1-Lhx3 to effectively induce motor neurons and suppress interneuron fates. Together, our results reveal an essential role of miR-218 as a downstream effector of the Isl1-Lhx3 complex in establishing motor neuron identity.


Glia | 2013

Expression profiling of Aldh1l1-precursors in the developing spinal cord reveals glial lineage-specific genes and direct Sox9-Nfe2l1 interactions

Anna V. Molofsky; Stacey M. Glasgow; Lesley S. Chaboub; Hui-Hsin Tsai; Alice T. Murnen; Kevin W. Kelley; Stephen P.J. Fancy; Tracy J Yuen; Lohith Madireddy; Sergio E. Baranzini; Benjamin Deneen; David H. Rowitch; Michael C. Oldham

Developmental regulation of gliogenesis in the mammalian CNS is incompletely understood, in part due to a limited repertoire of lineage‐specific genes. We used Aldh1l1‐GFP as a marker for gliogenic radial glia and later‐stage precursors of developing astrocytes and performed gene expression profiling of these cells. We then used this dataset to identify candidate transcription factors that may serve as glial markers or regulators of glial fate. Our analysis generated a database of developmental stage‐related markers of Aldh1l1+ cells between murine embryonic day 13.5–18.5. Using these data we identify the bZIP transcription factor Nfe2l1 and demonstrate that it promotes glial fate under direct Sox9 regulatory control. Thus, this dataset represents a resource for identifying novel regulators of glial development.


Annals of Neurology | 2012

Evidence that nuclear factor IA inhibits repair after white matter injury

Stephen P.J. Fancy; Stacey M. Glasgow; Meggie Finley; David H. Rowitch; Benjamin Deneen

Chronic demyelination can result in axonopathy and is associated with human neurological conditions such as multiple sclerosis (MS) in adults and cerebral palsy in infants. In these disorders, myelin regeneration is inhibited by impaired differentiation of oligodendrocyte progenitors into myelin‐producing oligodendrocytes. However, regulatory factors relevant in human myelin disorders and in myelin regeneration remain poorly understood. Here we have investigated the role of the transcription factor nuclear factor IA (NFIA) in oligodendrocyte progenitor differentiation during developmental and regenerative myelination.


The Journal of Neuroscience | 2016

Temporal Profiling of Astrocyte Precursors Reveals Parallel Roles for Asef during Development and after Injury

Lesley S. Chaboub; Jeanne M. Manalo; Hyun Kyoung Lee; Stacey M. Glasgow; Fengju Chen; Yoshihiro Kawasaki; Tetsu Akiyama; Chay T. Kuo; Chad J. Creighton; Carrie A. Mohila; Benjamin Deneen

Lineage development is a stepwise process, governed by stage-specific regulatory factors and associated markers. Astrocytes are one of the principle cell types in the CNS and the stages associated with their development remain very poorly defined. To identify these stages, we performed gene-expression profiling on astrocyte precursor populations in the spinal cord, identifying distinct patterns of gene induction during their development that are strongly correlated with human astrocytes. Validation studies identified a new cohort of astrocyte-associated genes during development and demonstrated their expression in reactive astrocytes in human white matter injury (WMI). Functional studies on one of these genes revealed that mice lacking Asef exhibited impaired astrocyte differentiation during development and repair after WMI, coupled with compromised blood–brain barrier integrity in the adult CNS. These studies have identified distinct stages of astrocyte lineage development associated with human WMI and, together with our functional analysis of Asef, highlight the parallels between astrocyte development and their reactive counterparts associated with injury. SIGNIFICANCE STATEMENT Astrocytes play a central role in CNS function and associated diseases. Yet the mechanisms that control their development remain poorly defined. Using the developing mouse spinal cord as a model system, we identify molecular changes that occur in developing astrocytes. These molecular signatures are strongly correlated with human astrocyte expression profiles and validation in mouse spinal cord identifies a host of new genes associated with the astrocyte lineage. These genes are present in reactive astrocytes in human white matter injury, and functional studies reveal that one of these genes, Asef, contributes to reactive astrocyte responses after injury. These studies identify distinct stages of astrocyte lineage development and highlight the parallels between astrocyte development and their reactive counterparts associated with injury.


Nature Neuroscience | 2017

Glia-specific enhancers and chromatin structure regulate NFIA expression and glioma tumorigenesis

Stacey M. Glasgow; Jeffrey Carlson; Wenyi Zhu; Lesley S. Chaboub; Peng Kang; Hyun Kyoung Lee; Yoanne M. Clovis; Brittney E. Lozzi; Robert J. McEvilly; Michael G. Rosenfeld; Chad J. Creighton; Soo Kyung Lee; Carrie A. Mohila; Benjamin Deneen

Long-range enhancer interactions critically regulate gene expression, yet little is known about how their coordinated activities contribute to CNS development or how this may, in turn, relate to disease states. By examining the regulation of the transcription factor NFIA in the developing spinal cord, we identified long-range enhancers that recapitulate NFIA expression across glial and neuronal lineages in vivo. Complementary genetic studies found that Sox9–Brn2 and Isl1–Lhx3 regulate enhancer activity and NFIA expression in glial and neuronal populations. Chromatin conformation analysis revealed that these enhancers and transcription factors form distinct architectures within these lineages in the spinal cord. In glioma models, the glia-specific architecture is present in tumors, and these enhancers are required for NFIA expression and contribute to glioma formation. By delineating three-dimensional mechanisms of gene expression regulation, our studies identify lineage-specific chromatin architectures and associated enhancers that regulate cell fate and tumorigenesis in the CNS.


Nature Communications | 2015

Corrigendum: miR-218 is essential to establish motor neuron fate as a downstream effector of Isl1-Lhx3.

Karen P. Thiebes; Heejin Nam; Xiaolu A. Cambronne; Rongkun Shen; Stacey M. Glasgow; Hyong Ho Cho; Ji Sun Kwon; Richard H. Goodman; Jae W. Lee; Seunghee Lee; Soo Kyung Lee

Corrigendum: miR-218 is essential to establish motor neuron fate as a downstream effector of Isl1–Lhx3


Nature Communications | 2015

Erratum: Corrigendum: miR-218 is essential to establish motor neuron fate as a downstream effector of Isl1–Lhx3

Karen P. Thiebes; Heejin Nam; Xiaolu A. Cambronne; Rongkun Shen; Stacey M. Glasgow; Hyong-Ho Cho; Ji-sun Kwon; Richard H. Goodman; Jae W. Lee; Seunghee Lee; Soo-Kyung Lee

Corrigendum: miR-218 is essential to establish motor neuron fate as a downstream effector of Isl1–Lhx3

Collaboration


Dive into the Stacey M. Glasgow's collaboration.

Top Co-Authors

Avatar

Benjamin Deneen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Carrie A. Mohila

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lesley S. Chaboub

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heejin Nam

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jae W. Lee

Korea Aerospace University

View shared research outputs
Top Co-Authors

Avatar

Seunghee Lee

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge