211 results on '"Rowitch DH"'
Search Results
52. Challenges to curing primary brain tumours.
- Author
-
Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, Gottardo N, Gutmann DH, Hargrave D, Holland EC, Jones DTW, Joyce JA, Kearns P, Kieran MW, Mellinghoff IK, Merchant M, Pfister SM, Pollard SM, Ramaswamy V, Rich JN, Robinson GW, Rowitch DH, Sampson JH, Taylor MD, Workman P, and Gilbertson RJ
- Subjects
- Humans, Brain Neoplasms therapy
- Abstract
Despite decades of research, brain tumours remain among the deadliest of all forms of cancer. The ability of these tumours to resist almost all conventional and novel treatments relates, in part, to the unique cell-intrinsic and microenvironmental properties of neural tissues. In an attempt to encourage progress in our understanding and ability to successfully treat patients with brain tumours, Cancer Research UK convened an international panel of clinicians and laboratory-based scientists to identify challenges that must be overcome if we are to cure all patients with a brain tumour. The seven key challenges summarized in this Position Paper are intended to serve as foci for future research and investment.
- Published
- 2019
- Full Text
- View/download PDF
53. Single-cell genomics identifies cell type-specific molecular changes in autism.
- Author
-
Velmeshev D, Schirmer L, Jung D, Haeussler M, Perez Y, Mayer S, Bhaduri A, Goyal N, Rowitch DH, and Kriegstein AR
- Subjects
- Adolescent, Cell Nucleus metabolism, Child, Child, Preschool, Female, Gene Expression Profiling, Genomics methods, Humans, Male, Microglia metabolism, Neurons metabolism, Sequence Analysis, RNA, Single-Cell Analysis methods, Young Adult, Autistic Disorder genetics, Autistic Disorder psychology, Gene Expression Regulation, Neocortex metabolism
- Abstract
Despite the clinical and genetic heterogeneity of autism, bulk gene expression studies show that changes in the neocortex of autism patients converge on common genes and pathways. However, direct assessment of specific cell types in the brain affected by autism has not been feasible until recently. We used single-nucleus RNA sequencing of cortical tissue from patients with autism to identify autism-associated transcriptomic changes in specific cell types. We found that synaptic signaling of upper-layer excitatory neurons and the molecular state of microglia are preferentially affected in autism. Moreover, our results show that dysregulation of specific groups of genes in cortico-cortical projection neurons correlates with clinical severity of autism. These findings suggest that molecular changes in upper-layer cortical circuits are linked to behavioral manifestations of autism., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
54. The neurointensive nursery: concept, development, and insights gained.
- Author
-
Glass HC, Ferriero DM, Rowitch DH, and Shimotake TK
- Subjects
- Brain, Humans, Infant, Infant, Newborn, Magnetic Resonance Imaging, Neurology trends, Hypothermia, Induced, Hypoxia-Ischemia, Brain prevention & control
- Abstract
Purpose of Review: With the advent of therapeutic hypothermia for treatment of hypoxic ischemic encephalopathy, and improvements in neuroimaging and bedside neuromonitoring, a new era of neonatal brain-focused care has emerged in recent years. We describe the development of the first neurointensive care nursery (NICN) as a model for comanagement of neonates with identified neurologic risk factors by a multidisciplinary team constituted of neurologists, neonatologists, specialized nurses, and others with the goal of optimizing management, preventing secondary injury and maximizing long-term outcomes., Recent Findings: Optimizing brain metabolic environment and perfusion and preventing secondary brain injury are key to neurocritical care. This includes close management of temperature, blood pressure, oxygenation, carbon dioxide, and glucose levels. Early developmental interventions and involvement of physical and occupational therapy provide additional assessment information. Finally, long-term follow-up is essential for any neurocritical care program., Summary: The NICN model aims to optimize evidence-based care of infants at risk for neurologic injury. Results from ongoing hypothermia and neuroprotective trials are likely to yield additional treatments. New technologies, such as functional MRI, continuous neurophysiological assessment, and whole genomic approaches to rapid diagnosis may further enhance clinical protocols and neonatal precision medicine. Importantly, advances in neurocritical care improve our ability to provide comprehensive information when counseling families. Long-term follow-up data will determine if the NICN/Neuro-NICU provides enduring benefit to infants at risk for neurologic injury.
- Published
- 2019
- Full Text
- View/download PDF
55. Single-cell reconstruction of the early maternal-fetal interface in humans.
- Author
-
Vento-Tormo R, Efremova M, Botting RA, Turco MY, Vento-Tormo M, Meyer KB, Park JE, Stephenson E, Polański K, Goncalves A, Gardner L, Holmqvist S, Henriksson J, Zou A, Sharkey AM, Millar B, Innes B, Wood L, Wilbrey-Clark A, Payne RP, Ivarsson MA, Lisgo S, Filby A, Rowitch DH, Bulmer JN, Wright GJ, Stubbington MJT, Haniffa M, Moffett A, and Teichmann SA
- Subjects
- Cell Differentiation genetics, Decidua cytology, Decidua immunology, Decidua metabolism, Female, Fetus immunology, Fetus metabolism, Humans, Killer Cells, Natural cytology, Killer Cells, Natural immunology, Ligands, Placenta immunology, RNA, Small Cytoplasmic genetics, Sequence Analysis, RNA, Stromal Cells cytology, Stromal Cells metabolism, Transcriptome, Trophoblasts cytology, Trophoblasts immunology, Trophoblasts metabolism, Cell Communication immunology, Fetus cytology, Histocompatibility, Maternal-Fetal immunology, Placenta cytology, Placenta metabolism, Pregnancy immunology, Single-Cell Analysis
- Abstract
During early human pregnancy the uterine mucosa transforms into the decidua, into which the fetal placenta implants and where placental trophoblast cells intermingle and communicate with maternal cells. Trophoblast-decidual interactions underlie common diseases of pregnancy, including pre-eclampsia and stillbirth. Here we profile the transcriptomes of about 70,000 single cells from first-trimester placentas with matched maternal blood and decidual cells. The cellular composition of human decidua reveals subsets of perivascular and stromal cells that are located in distinct decidual layers. There are three major subsets of decidual natural killer cells that have distinctive immunomodulatory and chemokine profiles. We develop a repository of ligand-receptor complexes and a statistical tool to predict the cell-type specificity of cell-cell communication via these molecular interactions. Our data identify many regulatory interactions that prevent harmful innate or adaptive immune responses in this environment. Our single-cell atlas of the maternal-fetal interface reveals the cellular organization of the decidua and placenta, and the interactions that are critical for placentation and reproductive success.
- Published
- 2018
- Full Text
- View/download PDF
56. Dlx1 and Dlx2 Promote Interneuron GABA Synthesis, Synaptogenesis, and Dendritogenesis.
- Author
-
Pla R, Stanco A, Howard MA, Rubin AN, Vogt D, Mortimer N, Cobos I, Potter GB, Lindtner S, Price JD, Nord AS, Visel A, Schreiner CE, Baraban SC, Rowitch DH, and Rubenstein JLR
- Subjects
- Animals, Cerebral Cortex cytology, Female, GABAergic Neurons cytology, Gene Expression Regulation, Developmental, Glutamate Decarboxylase metabolism, Homeodomain Proteins genetics, Interneurons cytology, Male, Mice, Knockout, Miniature Postsynaptic Potentials, Transcription Factors genetics, Vesicular Inhibitory Amino Acid Transport Proteins metabolism, Cerebral Cortex growth & development, GABAergic Neurons physiology, Homeodomain Proteins physiology, Interneurons physiology, Synapses physiology, Transcription Factors physiology, gamma-Aminobutyric Acid biosynthesis
- Abstract
The postnatal functions of the Dlx1&2 transcription factors in cortical interneurons (CINs) are unknown. Here, using conditional Dlx1, Dlx2, and Dlx1&2 knockouts (CKOs), we defined their roles in specific CINs. The CKOs had dendritic, synaptic, and survival defects, affecting even PV+ CINs. We provide evidence that DLX2 directly drives Gad1, Gad2, and Vgat expression, and show that mutants had reduced mIPSC amplitude. In addition, the mutants formed fewer GABAergic synapses on excitatory neurons and had reduced mIPSC frequency. Furthermore, Dlx1/2 CKO had hypoplastic dendrites, fewer excitatory synapses, and reduced excitatory input. We provide evidence that some of these phenotypes were due to reduced expression of GRIN2B (a subunit of the NMDA receptor), a high confidence Autism gene. Thus, Dlx1&2 coordinate key components of CIN postnatal development by promoting their excitability, inhibitory output, and survival.
- Published
- 2018
- Full Text
- View/download PDF
57. New Recipes for Myelinating Oligodendrocytes.
- Author
-
Nobuta H, Stockley JH, and Rowitch DH
- Subjects
- Cell Differentiation, Humans, Myelin Sheath, Oligodendroglia
- Abstract
While myelinating oligodendrocytes are attractive candidates for cell-based regenerative therapies, producing them in adequate quantities and regulation of progenitor differentiation pathways has proven limiting. Recently, Hubler et al. (2018) and Madhavan et al. (2018) generated cerebral organoids with myelinating oligodendrocytes and manipulated sterol pathway small molecules to promote myelin synthesis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
58. Oligodendrocyte-encoded Kir4.1 function is required for axonal integrity.
- Author
-
Schirmer L, Möbius W, Zhao C, Cruz-Herranz A, Ben Haim L, Cordano C, Shiow LR, Kelley KW, Sadowski B, Timmons G, Pröbstel AK, Wright JN, Sin JH, Devereux M, Morrison DE, Chang SM, Sabeur K, Green AJ, Nave KA, Franklin RJ, and Rowitch DH
- Subjects
- Animals, Axons pathology, Humans, Leukoencephalopathies physiopathology, Mice, Mice, Knockout, Neuroglia metabolism, Neuroglia pathology, Neurons metabolism, Neurons pathology, Oligodendroglia metabolism, Oligodendroglia pathology, Seizures physiopathology, Spinal Cord metabolism, Spinal Cord physiopathology, Axons metabolism, Leukoencephalopathies genetics, Potassium Channels, Inwardly Rectifying genetics, Seizures genetics
- Abstract
Glial support is critical for normal axon function and can become dysregulated in white matter (WM) disease. In humans, loss-of-function mutations of KCNJ10, which encodes the inward-rectifying potassium channel KIR4.1, causes seizures and progressive neurological decline. We investigated Kir4.1 functions in oligodendrocytes (OLs) during development, adulthood and after WM injury. We observed that Kir4.1 channels localized to perinodal areas and the inner myelin tongue, suggesting roles in juxta-axonal K
+ removal. Conditional knockout (cKO) of OL- Kcnj10 resulted in late onset mitochondrial damage and axonal degeneration. This was accompanied by neuronal loss and neuro-axonal dysfunction in adult OL- Kcnj10 cKO mice as shown by delayed visual evoked potentials, inner retinal thinning and progressive motor deficits. Axon pathologies in OL- Kcnj10 cKO were exacerbated after WM injury in the spinal cord. Our findings point towards a critical role of OL-Kir4.1 for long-term maintenance of axonal function and integrity during adulthood and after WM injury., Competing Interests: LS filed a patent for the detection of antibodies against KIR4.1 in a subpopulation of patients with multiple sclerosis (WO2015166057A1), WM, CZ, AC, LB, CC, LS, KK, BS, GT, AP, JW, JS, MD, DM, SC, KS, AG, RF, DR No competing interests declared, KN Reviewing editor, eLife, (© 2018, Schirmer et al.)- Published
- 2018
- Full Text
- View/download PDF
59. A Glial Signature and Wnt7 Signaling Regulate Glioma-Vascular Interactions and Tumor Microenvironment.
- Author
-
Griveau A, Seano G, Shelton SJ, Kupp R, Jahangiri A, Obernier K, Krishnan S, Lindberg OR, Yuen TJ, Tien AC, Sabo JK, Wang N, Chen I, Kloepper J, Larrouquere L, Ghosh M, Tirosh I, Huillard E, Alvarez-Buylla A, Oldham MC, Persson AI, Weiss WA, Batchelor TT, Stemmer-Rachamimov A, Suvà ML, Phillips JJ, Aghi MK, Mehta S, Jain RK, and Rowitch DH
- Subjects
- Animals, Bevacizumab pharmacology, Blood-Brain Barrier metabolism, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Glioma drug therapy, Glioma metabolism, Humans, Mice, Neoplasm Transplantation, Oligodendrocyte Transcription Factor 2 genetics, Temozolomide pharmacology, Tumor Cells, Cultured, Tumor Microenvironment, Wnt Proteins genetics, Wnt Signaling Pathway drug effects, Brain Neoplasms blood supply, Glioma blood supply, Oligodendrocyte Transcription Factor 2 metabolism, Oligodendroglia microbiology, Wnt Proteins metabolism
- Abstract
Gliomas comprise heterogeneous malignant glial and stromal cells. While blood vessel co-option is a potential mechanism to escape anti-angiogenic therapy, the relevance of glial phenotype in this process is unclear. We show that Olig2
+ oligodendrocyte precursor-like glioma cells invade by single-cell vessel co-option and preserve the blood-brain barrier (BBB). Conversely, Olig2-negative glioma cells form dense perivascular collections and promote angiogenesis and BBB breakdown, leading to innate immune cell activation. Experimentally, Olig2 promotes Wnt7b expression, a finding that correlates in human glioma profiling. Targeted Wnt7a/7b deletion or pharmacologic Wnt inhibition blocks Olig2+ glioma single-cell vessel co-option and enhances responses to temozolomide. Finally, Olig2 and Wnt7 become upregulated after anti-VEGF treatment in preclinical models and patients. Thus, glial-encoded pathways regulate distinct glioma-vascular microenvironmental interactions., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
60. Sonic Hedgehog Agonist Protects Against Complex Neonatal Cerebellar Injury.
- Author
-
Nguyen V, Sabeur K, Maltepe E, Ameri K, Bayraktar O, and Rowitch DH
- Subjects
- Amino Acids, Dicarboxylic pharmacology, Animals, Animals, Newborn, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Calcium-Binding Proteins metabolism, Cell Proliferation drug effects, Cell Proliferation genetics, Cells, Cultured, Cerebellum drug effects, Developmental Disabilities etiology, Disease Models, Animal, Gene Expression Regulation, Developmental drug effects, Gene Expression Regulation, Developmental genetics, Glucocorticoids pharmacology, Hypoxia, Brain complications, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microfilament Proteins metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Nervous System Malformations etiology, Prednisolone therapeutic use, Purkinje Cells drug effects, Purkinje Cells metabolism, Von Hippel-Lindau Tumor Suppressor Protein genetics, Von Hippel-Lindau Tumor Suppressor Protein metabolism, Zinc Finger Protein GLI1 genetics, Zinc Finger Protein GLI1 metabolism, Anti-Inflammatory Agents therapeutic use, Cerebellum abnormalities, Cyclohexylamines therapeutic use, Hedgehog Proteins agonists, Hedgehog Proteins metabolism, Neuroprotective Agents therapeutic use, Thiophenes therapeutic use
- Abstract
The cerebellum undergoes rapid growth during the third trimester and is vulnerable to injury and deficient growth in infants born prematurely. Factors associated with preterm cerebellar hypoplasia include chronic lung disease and postnatal glucocorticoid administration. We modeled chronic hypoxemia and glucocorticoid administration in neonatal mice to study whole cerebellar and cell type-specific effects of dual exposure. Chronic neonatal hypoxia resulted in permanent cerebellar hypoplasia. This was compounded by administration of prednisolone as shown by greater volume loss and Purkinje cell death. In the setting of hypoxia and prednisolone, administration of a small molecule Smoothened-Hedgehog agonist (SAG) preserved cerebellar volume and protected against Purkinje cell death. Such protective effects were observed even when SAG was given as a one-time dose after dual insult. To model complex injury and determine cell type-specific roles for the hypoxia inducible factor (HIF) pathway, we performed conditional knockout of von Hippel Lindau (VHL) to hyperactivate HIF1α in cerebellar granule neuron precursors (CGNP) or Purkinje cells. Surprisingly, HIF activation in either cell type resulted in no cerebellar deficit. However, in mice administered prednisolone, HIF overactivation in CGNPs resulted in significant cerebellar hypoplasia, whereas HIF overactivation in Purkinje cells caused cell death. Together, these findings indicate that HIF primes both cell types for injury via glucocorticoids, and that hypoxia/HIF + postnatal glucocorticoid administration act on distinct cellular pathways to cause cerebellar injury. They further suggest that SAG is neuroprotective in the setting of complex neonatal cerebellar injury.
- Published
- 2018
- Full Text
- View/download PDF
61. Origin and dynamics of oligodendrocytes in the developing brain: Implications for perinatal white matter injury.
- Author
-
van Tilborg E, de Theije CGM, van Hal M, Wagenaar N, de Vries LS, Benders MJ, Rowitch DH, and Nijboer CH
- Subjects
- Animals, Brain growth & development, Cell Differentiation physiology, Cell Movement physiology, Female, Humans, Infant, Newborn, Myelin Sheath pathology, Pregnancy, White Matter growth & development, Brain pathology, Brain Injuries pathology, Oligodendroglia pathology, White Matter injuries, White Matter pathology
- Abstract
Infants born prematurely are at high risk to develop white matter injury (WMI), due to exposure to hypoxic and/or inflammatory insults. Such perinatal insults negatively impact the maturation of oligodendrocytes (OLs), thereby causing deficits in myelination. To elucidate the precise pathophysiology underlying perinatal WMI, it is essential to fully understand the cellular mechanisms contributing to healthy/normal white matter development. OLs are responsible for myelination of axons. During brain development, OLs are generally derived from neuroepithelial zones, where neural stem cells committed to the OL lineage differentiate into OL precursor cells (OPCs). OPCs, in turn, develop into premyelinating OLs and finally mature into myelinating OLs. Recent studies revealed that OPCs develop in multiple waves and form potentially heterogeneous populations. Furthermore, it has been shown that myelination is a dynamic and plastic process with an excess of OPCs being generated and then abolished if not integrated into neural circuits. Myelination patterns between rodents and humans show high spatial and temporal similarity. Therefore, experimental studies on OL biology may provide novel insights into the pathophysiology of WMI in the preterm infant and offers new perspectives on potential treatments for these patients., (© 2017 The Authors GLIA Published by Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
62. Systematic Three-Dimensional Coculture Rapidly Recapitulates Interactions between Human Neurons and Astrocytes.
- Author
-
Krencik R, Seo K, van Asperen JV, Basu N, Cvetkovic C, Barlas S, Chen R, Ludwig C, Wang C, Ward ME, Gan L, Horner PJ, Rowitch DH, and Ullian EM
- Subjects
- Astrocytes metabolism, Cell Lineage genetics, Cell Lineage physiology, Cells, Cultured, Humans, Neural Stem Cells cytology, Neural Stem Cells metabolism, Neurons metabolism, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism, Synapses metabolism, Synapses physiology, Astrocytes cytology, Cell Differentiation genetics, Coculture Techniques, Neurons cytology
- Abstract
Human astrocytes network with neurons in dynamic ways that are still poorly defined. Our ability to model this relationship is hampered by the lack of relevant and convenient tools to recapitulate this complex interaction. To address this barrier, we have devised efficient coculture systems utilizing 3D organoid-like spheres, termed asteroids, containing pre-differentiated human pluripotent stem cell (hPSC)-derived astrocytes (hAstros) combined with neurons generated from hPSC-derived neural stem cells (hNeurons) or directly induced via Neurogenin 2 overexpression (iNeurons). Our systematic methods rapidly produce structurally complex hAstros and synapses in high-density coculture with iNeurons in precise numbers, allowing for improved studies of neural circuit function, disease modeling, and drug screening. We conclude that these bioengineered neural circuit model systems are reliable and scalable tools to accurately study aspects of human astrocyte-neuron functional properties while being easily accessible for cell-type-specific manipulations and observations., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
63. Fibrinogen Activates BMP Signaling in Oligodendrocyte Progenitor Cells and Inhibits Remyelination after Vascular Damage.
- Author
-
Petersen MA, Ryu JK, Chang KJ, Etxeberria A, Bardehle S, Mendiola AS, Kamau-Devers W, Fancy SPJ, Thor A, Bushong EA, Baeza-Raja B, Syme CA, Wu MD, Rios Coronado PE, Meyer-Franke A, Yahn S, Pous L, Lee JK, Schachtrup C, Lassmann H, Huang EJ, Han MH, Absinta M, Reich DS, Ellisman MH, Rowitch DH, Chan JR, and Akassoglou K
- Subjects
- Activin Receptors, Type I drug effects, Activin Receptors, Type I genetics, Activin Receptors, Type I metabolism, Animals, Blood Vessels drug effects, Blood Vessels pathology, Fibrinogen antagonists & inhibitors, Lysophosphatidylcholines pharmacology, Mice, Mice, Inbred C57BL, Mice, Knockout, Microarray Analysis, Myelin Sheath metabolism, Oligodendrocyte Precursor Cells drug effects, Plasmids genetics, Signal Transduction drug effects, Bone Morphogenetic Proteins metabolism, Fibrinogen pharmacology, Oligodendrocyte Precursor Cells metabolism, Remyelination drug effects
- Abstract
Blood-brain barrier (BBB) disruption alters the composition of the brain microenvironment by allowing blood proteins into the CNS. However, whether blood-derived molecules serve as extrinsic inhibitors of remyelination is unknown. Here we show that the coagulation factor fibrinogen activates the bone morphogenetic protein (BMP) signaling pathway in oligodendrocyte progenitor cells (OPCs) and suppresses remyelination. Fibrinogen induces phosphorylation of Smad 1/5/8 and inhibits OPC differentiation into myelinating oligodendrocytes (OLs) while promoting an astrocytic fate in vitro. Fibrinogen effects are rescued by BMP type I receptor inhibition using dorsomorphin homolog 1 (DMH1) or CRISPR/Cas9 activin A receptor type I (ACVR1) knockout in OPCs. Fibrinogen and the BMP target Id2 are increased in demyelinated multiple sclerosis (MS) lesions. Therapeutic depletion of fibrinogen decreases BMP signaling and enhances remyelination in vivo. Targeting fibrinogen may be an upstream therapeutic strategy to promote the regenerative potential of CNS progenitors in diseases with remyelination failure., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
64. Reactive astrocyte COX2-PGE2 production inhibits oligodendrocyte maturation in neonatal white matter injury.
- Author
-
Shiow LR, Favrais G, Schirmer L, Schang AL, Cipriani S, Andres C, Wright JN, Nobuta H, Fleiss B, Gressens P, and Rowitch DH
- Subjects
- Animals, Animals, Newborn, Astrocytes drug effects, Cells, Cultured, Female, Fetus cytology, Gene Expression Regulation, Developmental drug effects, Gene Expression Regulation, Developmental physiology, Glial Fibrillary Acidic Protein metabolism, Humans, Interleukin-1beta pharmacology, L-Lactate Dehydrogenase metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Oligodendroglia drug effects, Rats, Signal Transduction drug effects, Signal Transduction physiology, Spatial Memory drug effects, Spatial Memory physiology, White Matter growth & development, White Matter metabolism, Cyclooxygenase 2 metabolism, Dinoprostone metabolism, Oligodendroglia metabolism, White Matter cytology, White Matter drug effects
- Abstract
Inflammation is a major risk factor for neonatal white matter injury (NWMI), which is associated with later development of cerebral palsy. Although recent studies have demonstrated maturation arrest of oligodendrocyte progenitor cells (OPCs) in NWMI, the identity of inflammatory mediators with direct effects on OPCs has been unclear. Here, we investigated downstream effects of pro-inflammatory IL-1β to induce cyclooxygenase-2 (COX2) and prostaglandin E2 (PGE2) production in white matter. First, we assessed COX2 expression in human fetal brain and term neonatal brain affected by hypoxic-ischemic encephalopathy (HIE). In the developing human brain, COX2 was expressed in radial glia, microglia, and endothelial cells. In human term neonatal HIE cases with subcortical WMI, COX2 was strongly induced in reactive astrocytes with "A2" reactivity. Next, we show that OPCs express the EP1 receptor for PGE2, and PGE2 acts directly on OPCs to block maturation in vitro. Pharmacologic blockade with EP1-specific inhibitors (ONO-8711, SC-51089), or genetic deficiency of EP1 attenuated effects of PGE2. In an IL-1β-induced model of NWMI, astrocytes also exhibit "A2" reactivity and induce COX2. Furthermore, in vivo inhibition of COX2 with Nimesulide rescues hypomyelination and behavioral impairment. These findings suggest that neonatal white matter astrocytes can develop "A2" reactivity that contributes to OPC maturation arrest in NWMI through induction of COX2-PGE2 signaling, a pathway that can be targeted for neonatal neuroprotection., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
65. Olig1 is required for noggin-induced neonatal myelin repair.
- Author
-
Sabo JK, Heine V, Silbereis JC, Schirmer L, Levison SW, and Rowitch DH
- Subjects
- Animals, Cell Culture Techniques, Disease Models, Animal, Humans, Infant, Infant, Newborn, Infant, Newborn, Diseases, Mice, Mice, Inbred C57BL, Mice, Knockout, Neural Stem Cells, Basic Helix-Loop-Helix Transcription Factors metabolism, Bone Morphogenetic Proteins metabolism, Carrier Proteins metabolism, Hypoxia-Ischemia, Brain metabolism, Nerve Tissue Proteins metabolism, Oligodendroglia metabolism, Stroke metabolism
- Abstract
Objective: Neonatal white matter injury (NWMI) is a lesion found in preterm infants that can lead to cerebral palsy. Although antagonists of bone morphogenetic protein (BMP) signaling, such as Noggin, promote oligodendrocyte precursor cell (OPC) production after hypoxic-ischemic (HI) injury, the downstream functional targets are poorly understood. The basic helix-loop-helix protein, oligodendrocyte transcription factor 1 (Olig1), promotes oligodendrocyte (OL) development and is essential during remyelination in adult mice. Here, we investigated whether Olig1 function is required downstream of BMP antagonism for response to injury in the neonatal brain., Methods: We used wild-type and Olig1-null mice subjected to neonatal stroke and postnatal neural progenitor cultures, and we analyzed Olig1 expression in human postmortem samples from neonates that suffered HI encephalopathy (HIE)., Results: Olig1-null neonatal mice showed significant hypomyelination after moderate neonatal stroke. Surprisingly, damaged white matter tracts in Olig1-null mice lacked Olig2
+ OPCs, and instead proliferating neuronal precursors and GABAergic interneurons were present. We demonstrate that Noggin-induced OPC production requires Olig1 function. In postnatal neural progenitors, Noggin governs production of OLs versus interneurons through Olig1-mediated repression of Dlx1/2 transcription factors. Additionally, we observed that Olig1 and the BMP signaling effector, phosphorylated SMADs (Sma- and Mad-related proteins) 1, 5, and 8, were elevated in the subventricular zone of human infants with HIE compared to controls., Interpretation: These findings indicate that Olig1 has a critical function in regulation of postnatal neural progenitor cell production in response to Noggin. Ann Neurol 2017;81:560-571., (© 2017 American Neurological Association.)- Published
- 2017
- Full Text
- View/download PDF
66. A Sequentially Priming Phosphorylation Cascade Activates the Gliomagenic Transcription Factor Olig2.
- Author
-
Zhou J, Tien AC, Alberta JA, Ficarro SB, Griveau A, Sun Y, Deshpande JS, Card JD, Morgan-Smith M, Michowski W, Hashizume R, James CD, Ligon KL, Snider WD, Sicinski P, Marto JA, Rowitch DH, and Stiles CD
- Subjects
- Animals, Casein Kinase II metabolism, Cell Line, Tumor, Cyclin-Dependent Kinases metabolism, Disease Models, Animal, Glioma pathology, Glycogen Synthase Kinase 3 metabolism, Humans, Mice, Phosphorylation drug effects, Phosphoserine metabolism, Small Molecule Libraries pharmacology, Tumor Suppressor Protein p53 metabolism, Carcinogenesis metabolism, Carcinogenesis pathology, Glioma metabolism, Oligodendrocyte Transcription Factor 2 metabolism
- Abstract
During development of the vertebrate CNS, the basic helix-loop-helix (bHLH) transcription factor Olig2 sustains replication competence of progenitor cells that give rise to neurons and oligodendrocytes. A pathological counterpart of this developmental function is seen in human glioma, wherein Olig2 is required for maintenance of stem-like cells that drive tumor growth. The mitogenic/gliomagenic functions of Olig2 are regulated by phosphorylation of a triple serine motif (S10, S13, and S14) in the amino terminus. Here, we identify a set of three serine/threonine protein kinases (glycogen synthase kinase 3α/β [GSK3α/β], casein kinase 2 [CK2], and cyclin-dependent kinases 1/2 [CDK1/2]) that are, collectively, both necessary and sufficient to phosphorylate the triple serine motif. We show that phosphorylation of the motif itself serves as a template to prime phosphorylation of additional serines and creates a highly charged "acid blob" in the amino terminus of Olig2. Finally, we show that small molecule inhibitors of this forward-feeding phosphorylation cascade have potential as glioma therapeutics., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
67. Concise Review: Stem Cell-Based Treatment of Pelizaeus-Merzbacher Disease.
- Author
-
Osorio MJ, Rowitch DH, Tesar P, Wernig M, Windrem MS, and Goldman SA
- Subjects
- Animals, Disease Models, Animal, Humans, Mutation genetics, Myelin Sheath metabolism, Pelizaeus-Merzbacher Disease pathology, Pelizaeus-Merzbacher Disease therapy, Stem Cell Transplantation, Stem Cells cytology
- Abstract
Pelizaeus-Merzbacher disease (PMD) is an X-linked disorder caused by mutation in the proteolipid protein-1 (PLP1) gene, which encodes the proteolipid protein of myelinating oligodendroglia. PMD exhibits phenotypic variability that reflects its considerable genotypic heterogeneity, but all forms of the disease result in central hypomyelination, associated in most cases with early neurological dysfunction, progressive deterioration, and ultimately death. PMD may present as a connatal, classic and transitional forms, or as the less severe spastic paraplegia type 2 and PLP-null phenotypes. These disorders are most often associated with duplications of the PLP1 gene, but can also be caused by coding and noncoding point mutations as well as full or partial deletion of the gene. A number of genetically-distinct but phenotypically-similar disorders of hypomyelination exist which, like PMD, lack any effective therapy. Yet as relatively pure CNS hypomyelinating disorders, with limited involvement of the PNS and relatively little attendant neuronal pathology, PMD and similar hypomyelinating disorders are attractive therapeutic targets for neural stem cell and glial progenitor cell transplantation, efforts at which are now underway in a number of research centers. Stem Cells 2017;35:311-315., (© 2016 AlphaMed Press.)
- Published
- 2017
- Full Text
- View/download PDF
68. Neurotoxic reactive astrocytes are induced by activated microglia.
- Author
-
Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Münch AE, Chung WS, Peterson TC, Wilton DK, Frouin A, Napier BA, Panicker N, Kumar M, Buckwalter MS, Rowitch DH, Dawson VL, Dawson TM, Stevens B, and Barres BA
- Subjects
- Animals, Astrocytes metabolism, Axotomy, Cell Culture Techniques, Cell Survival, Complement C1q metabolism, Disease Progression, Humans, Inflammation pathology, Interleukin-1alpha metabolism, Mice, Mice, Inbred C57BL, Microglia metabolism, Neurodegenerative Diseases pathology, Oligodendroglia pathology, Phagocytosis, Phenotype, Rats, Rats, Sprague-Dawley, Synapses pathology, Toxins, Biological metabolism, Tumor Necrosis Factor-alpha metabolism, Astrocytes classification, Astrocytes pathology, Cell Death, Central Nervous System pathology, Microglia pathology, Neurons pathology
- Abstract
Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.
- Published
- 2017
- Full Text
- View/download PDF
69. Functional diversity of astrocytes in neural circuit regulation.
- Author
-
Ben Haim L and Rowitch DH
- Subjects
- Animals, Humans, Mammals, Astrocytes physiology, Brain physiology, Cell Communication physiology, Nerve Net physiology, Neurons physiology
- Abstract
Although it is well established that all brain regions contain various neuronal subtypes with different functions, astrocytes have traditionally been thought to be homogenous. However, recent evidence has shown that astrocytes in the mammalian CNS display distinct inter- and intra-regional features, as well as functional diversity. In the CNS, astrocyte processes fill the local environment in non-overlapping domains. Therefore, a potential advantage of region-specified astrocytes might be their capacity to regulate local development or optimize local neural circuit function. An overview of the regional heterogeneity of neuron-astrocyte interactions indicates novel ways in which they could regulate normal neurological function and shows how they might become dysregulated in disease.
- Published
- 2017
- Full Text
- View/download PDF
70. Sustaining careers of physician-scientists in neonatology and pediatric critical care medicine: formulating supportive departmental policies.
- Author
-
Christou H, Dizon ML, Farrow KN, Jadcherla SR, Leeman KT, Maheshwari A, Rubin LP, Stansfield BK, and Rowitch DH
- Subjects
- Academic Medical Centers organization & administration, Career Choice, Focus Groups, Guidelines as Topic, Hospitals, Pediatric organization & administration, Humans, Job Satisfaction, Medical Staff, Hospital, Mentors, Program Development, Surveys and Questionnaires, Workforce, Critical Care organization & administration, Neonatology organization & administration, Pediatrics organization & administration, Physicians, Translational Research, Biomedical organization & administration
- Abstract
Understanding mechanisms of childhood disease and development of rational therapeutics are fundamental to progress in pediatric intensive care specialties. However, Division Chiefs and Department Chairs face unique challenges when building effective laboratory-based research programs in Neonatal and Pediatric Intensive Care, owing to high clinical demands necessary to maintain competence as well as financial pressures arising from fund flow models and the current extramural funding climate. Given these factors, the role of institutional support that could facilitate successful transition of promising junior faculty to independent research careers is ever more important. Would standardized guidelines of such support provide greater consistency among institutions? We addressed preliminary questions during a national focus group, a workshop and a survey of junior and senior academicians to solicit recommendations for optimal levels of protected time and resources when starting an independent laboratory. The consensus was that junior faculty should be assigned no more than 8 wk clinical service and should obtain start-up funds of $500K-1M exclusive of a 5-y committed salary support. Senior respondents placed a higher premium on protected time than junior faculty.
- Published
- 2016
- Full Text
- View/download PDF
71. Extensive migration of young neurons into the infant human frontal lobe.
- Author
-
Paredes MF, James D, Gil-Perotin S, Kim H, Cotter JA, Ng C, Sandoval K, Rowitch DH, Xu D, McQuillen PS, Garcia-Verdugo JM, Huang EJ, and Alvarez-Buylla A
- Subjects
- Doublecortin Domain Proteins, Frontal Lobe cytology, Gyrus Cinguli cytology, Humans, Infant, Interneurons cytology, Interneurons physiology, Lateral Ventricles cytology, Lateral Ventricles growth & development, Microtubule-Associated Proteins metabolism, Neuropeptides metabolism, Cell Movement, Frontal Lobe growth & development, Neurogenesis, Neuronal Plasticity, Neurons cytology, Neurons physiology
- Abstract
The first few months after birth, when a child begins to interact with the environment, are critical to human brain development. The human frontal lobe is important for social behavior and executive function; it has increased in size and complexity relative to other species, but the processes that have contributed to this expansion are unknown. Our studies of postmortem infant human brains revealed a collection of neurons that migrate and integrate widely into the frontal lobe during infancy. Chains of young neurons move tangentially close to the walls of the lateral ventricles and along blood vessels. These cells then individually disperse long distances to reach cortical tissue, where they differentiate and contribute to inhibitory circuits. Late-arriving interneurons could contribute to developmental plasticity, and the disruption of their postnatal migration or differentiation may underlie neurodevelopmental disorders., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
72. Lineage-Restricted OLIG2-RTK Signaling Governs the Molecular Subtype of Glioma Stem-like Cells.
- Author
-
Kupp R, Shtayer L, Tien AC, Szeto E, Sanai N, Rowitch DH, and Mehta S
- Subjects
- Animals, Brain Neoplasms metabolism, Cell Cycle, Cell Nucleus metabolism, ErbB Receptors metabolism, Glioma metabolism, Humans, Intercellular Signaling Peptides and Proteins metabolism, Mesoderm metabolism, Mice, Neoplastic Stem Cells metabolism, Neural Stem Cells metabolism, Neural Stem Cells pathology, Oligodendrocyte Transcription Factor 2, Phosphorylation, Receptors, Platelet-Derived Growth Factor metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Brain Neoplasms pathology, Cell Lineage, Glioma pathology, Neoplastic Stem Cells pathology, Nerve Tissue Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction
- Abstract
The basic helix-loop-helix (bHLH) transcription factor OLIG2 is a master regulator of oligodendroglial fate decisions and tumorigenic competence of glioma stem-like cells (GSCs). However, the molecular mechanisms underlying dysregulation of OLIG2 function during gliomagenesis remains poorly understood. Here, we show that OLIG2 modulates growth factor signaling in two distinct populations of GSCs, characterized by expression of either the epidermal growth factor receptor (EGFR) or platelet-derived growth factor receptor alpha (PDGFRα). Biochemical analyses of OLIG2 function in normal and malignant neural progenitors reveal a positive feedforward loop between OLIG2 and EGFR to sustain co-expression. Furthermore, loss of OLIG2 function results in mesenchymal transformation in PDGFRα(HIGH) GSCs, a phenomenon that appears to be circumscribed in EGFR(HIGH) GSCs. Exploitation of OLIG2's dual and antithetical, pro-mitotic (EGFR-driven), and lineage-specifying (PDGFRα-driven) functions by glioma cells appears to be critical for sustaining growth factor signaling and GSC molecular subtype., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
73. The Role of the Neurointensive Care Nursery for Neonatal Encephalopathy.
- Author
-
Glass HC and Rowitch DH
- Subjects
- Cooperative Behavior, Hospital Units, Humans, Infant, Newborn, Neurology, Hypoxia-Ischemia, Brain therapy, Intensive Care Units, Neonatal, Neonatologists, Neurologists
- Abstract
Neonatal encephalopathy due to intrapartum events is estimated at 1 to 2 per 1000 live births in high-income countries. Outcomes have improved over the past decade due to implementation of therapeutic hypothermia, the only clinically available neuroprotective strategy for hypoxic-ischemic encephalopathy. Neonatal encephalopathy is the most common condition treated within a neonatal neurocritical care unit. Neonates with encephalopathy benefit from a neurocritical care approach due to prevention of secondary brain injury through attention to basic physiology, earlier recognition and treatment of neurologic complications, consistent management using guidelines and protocols, and use of optimized teams at dedicated referral centers., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
74. Identification of proliferative progenitors associated with prominent postnatal growth of the pons.
- Author
-
Lindquist RA, Guinto CD, Rodas-Rodriguez JL, Fuentealba LC, Tate MC, Rowitch DH, and Alvarez-Buylla A
- Subjects
- Animals, Animals, Newborn growth & development, Cell Proliferation, Fourth Ventricle cytology, Mice, Neurogenesis, Oligodendrocyte Transcription Factor 2 metabolism, Oligodendroglia physiology, Pons cytology, SOXB1 Transcription Factors metabolism, Oligodendrocyte Precursor Cells physiology, Pons growth & development
- Abstract
The pons controls crucial sensorimotor and autonomic functions. In humans, it grows sixfold postnatally and is a site of paediatric gliomas; however, the mechanisms of pontine growth remain poorly understood. We show that the murine pons quadruples in volume postnatally; growth is fastest during postnatal days 0-4 (P0-P4), preceding most myelination. We identify three postnatal proliferative compartments: ventricular, midline and parenchymal. We find no evidence of postnatal neurogenesis in the pons, but each progenitor compartment produces new astroglia and oligodendroglia; the latter expand 10- to 18-fold postnatally, and are derived mostly from the parenchyma. Nearly all parenchymal progenitors at P4 are Sox2(+)Olig2(+), but by P8 a Sox2(-) subpopulation emerges, suggesting a lineage progression from Sox2(+) 'early' to Sox2(-) 'late' oligodendrocyte progenitor. Fate mapping reveals that >90% of adult oligodendrocytes derive from P2-P3 Sox2(+) progenitors. These results demonstrate the importance of postnatal Sox2(+)Olig2(+) progenitors in pontine growth and oligodendrogenesis.
- Published
- 2016
- Full Text
- View/download PDF
75. Astrocytes: The Final Frontier….
- Author
-
Kelley KW and Rowitch DH
- Subjects
- Animals, Humans, Astrocytes cytology, Brain cytology, Microglia cytology, Neurons cytology, Oligodendroglia cytology
- Abstract
In this issue of Neuron, Zhang et al. (2016) develop a novel approach to generate populations of human astrocytes to uncover their uniquely human traits., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
76. Dysregulation of astrocyte extracellular signaling in Costello syndrome.
- Author
-
Krencik R, Hokanson KC, Narayan AR, Dvornik J, Rooney GE, Rauen KA, Weiss LA, Rowitch DH, and Ullian EM
- Subjects
- Animals, Astrocytes metabolism, Cell Differentiation, Cell Line, Gene Expression Regulation, Genes, ras, Genotype, Hippocampus metabolism, Humans, Mass Spectrometry, Mice, Mice, Transgenic, Mutation, Neuronal Plasticity, Neurons cytology, Neurons metabolism, Oligonucleotide Array Sequence Analysis, Phenotype, Proteoglycans metabolism, Snail Family Transcription Factors, Transcription Factors genetics, Transcription Factors metabolism, ras Proteins metabolism, Astrocytes cytology, Costello Syndrome metabolism, Extracellular Matrix metabolism, Induced Pluripotent Stem Cells cytology, Signal Transduction
- Abstract
Astrocytes produce an assortment of signals that promote neuronal maturation according to a precise developmental timeline. Is this orchestrated timing and signaling altered in human neurodevelopmental disorders? To address this question, the astroglial lineage was investigated in two model systems of a developmental disorder with intellectual disability caused by mutant Harvey rat sarcoma viral oncogene homolog (HRAS) termed Costello syndrome: mutant HRAS human induced pluripotent stem cells (iPSCs) and transgenic mice. Human iPSCs derived from patients with Costello syndrome differentiated to astroglia more rapidly in vitro than those derived from wild-type cell lines with normal HRAS, exhibited hyperplasia, and also generated an abundance of extracellular matrix remodeling factors and proteoglycans. Acute treatment with a farnesyl transferase inhibitor and knockdown of the transcription factor SNAI2 reduced expression of several proteoglycans in Costello syndrome iPSC-derived astrocytes. Similarly, mice in which mutant HRAS was expressed selectively in astrocytes exhibited experience-independent increased accumulation of perineuronal net proteoglycans in cortex, as well as increased parvalbumin expression in interneurons, when compared to wild-type mice. Our data indicate that astrocytes expressing mutant HRAS dysregulate cortical maturation during development as shown by abnormal extracellular matrix remodeling and implicate excessive astrocyte-to-neuron signaling as a possible drug target for treating mental impairment and enhancing neuroplasticity., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
77. Postnatal growth of the human pons: a morphometric and immunohistochemical analysis.
- Author
-
Tate MC, Lindquist RA, Nguyen T, Sanai N, Barkovich AJ, Huang EJ, Rowitch DH, and Alvarez-Buylla A
- Subjects
- Adolescent, Analysis of Variance, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Proliferation physiology, Child, Child, Preschool, Female, Humans, Infant, Infant, Newborn, Ki-67 Antigen metabolism, Magnetic Resonance Imaging, Male, Myelin Sheath metabolism, Oligodendrocyte Transcription Factor 2, Nerve Tissue Proteins metabolism, Pons anatomy & histology, Pons growth & development, Pons metabolism
- Abstract
Despite its critical importance to global brain function, the postnatal development of the human pons remains poorly understood. In the present study, we first performed magnetic resonance imaging (MRI)-based morphometric analyses of the postnatal human pons (0-18 years; n = 6-14/timepoint). Pons volume increased 6-fold from birth to 5 years, followed by continued slower growth throughout childhood. The observed growth was primarily due to expansion of the basis pontis. T2-based MRI analysis suggests that this growth is linked to increased myelination, and histological analysis of myelin basic protein in human postmortem specimens confirmed a dramatic increase in myelination during infancy. Analysis of cellular proliferation revealed many Ki67(+) cells during the first 7 months of life, particularly during the first month, where proliferation was increased in the basis relative to tegmentum. The majority of proliferative cells in the postnatal pons expressed the transcription factor Olig2, suggesting an oligodendrocyte lineage. The proportion of proliferating cells that were Olig2(+) was similar through the first 7 months of life and between basis and tegmentum. The number of Ki67(+) cells declined dramatically from birth to 7 months and further decreased by 3 years, with a small number of Ki67(+) cells observed throughout childhood. In addition, two populations of vimentin/nestin-expressing cells were identified: a dorsal group near the ventricular surface, which persists throughout childhood, and a parenchymal population that diminishes by 7 months and was not evident later in childhood. Together, our data reveal remarkable postnatal growth in the ventral pons, particularly during infancy when cells are most proliferative and myelination increases., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
78. Astrocyte development and heterogeneity.
- Author
-
Bayraktar OA, Fuentealba LC, Alvarez-Buylla A, and Rowitch DH
- Subjects
- Adult, Animals, Cerebral Ventricles cytology, Humans, Neuroglia cytology, Astrocytes physiology, Cerebral Ventricles embryology, Mammals physiology, Models, Neurological, Neural Stem Cells physiology, Neuroglia physiology, Spinal Cord cytology
- Abstract
Astrocytes have many roles within the brain parenchyma, and a subpopulation restricted to germinal niches functions as neural stem cells (NSCs) that produce various types of neuronal progeny in relation to spatiotemporal factors. A growing body of evidence supports the concept of morphological and molecular differences between astrocytes in different brain regions, which might relate to their derivation from regionally patterned radial glia. Indeed, the notion that astrocytes are molecularly and functionally heterogeneous could help explain how the central nervous system (CNS) retains embryonic positional information into adulthood. Here, we discuss recent evidence for regionally encoded functions of astrocytes in the developing and adult CNS to provide an integrated concept of the origin and possible function of astrocyte heterogeneity. We focus on the regionalization of NSCs in the ventricular-subventricular zone (V-SVZ) of the adult mammalian brain and emerging evidence for a segmental organization of astrocytes in the developing spinal cord and forebrain. We propose that astrocytes' diversity will provide fundamental clues to understand regional brain organization and function., (Copyright © 2015 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
79. Oligodendrocyte-encoded HIF function couples postnatal myelination and white matter angiogenesis.
- Author
-
Yuen TJ, Silbereis JC, Griveau A, Chang SM, Daneman R, Fancy SPJ, Zahed H, Maltepe E, and Rowitch DH
- Subjects
- Animals, Cell Differentiation, Corpus Callosum metabolism, Endothelial Cells cytology, In Vitro Techniques, Mice, Neovascularization, Physiologic, Neural Stem Cells, Oxygen metabolism, Paracrine Communication, Proto-Oncogene Proteins metabolism, Von Hippel-Lindau Tumor Suppressor Protein metabolism, Wnt Proteins metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Myelin Sheath metabolism, Oligodendroglia metabolism
- Abstract
Myelin sheaths provide critical functional and trophic support for axons in white matter tracts of the brain. Oligodendrocyte precursor cells (OPCs) have extraordinary metabolic requirements during development as they differentiate to produce multiple myelin segments, implying that they must first secure adequate access to blood supply. However, mechanisms that coordinate myelination and angiogenesis are unclear. Here, we show that oxygen tension, mediated by OPC-encoded hypoxia-inducible factor (HIF) function, is an essential regulator of postnatal myelination. Constitutive HIF1/2α stabilization resulted in OPC maturation arrest through autocrine activation of canonical Wnt7a/7b. Surprisingly, such OPCs also show paracrine activity that induces excessive postnatal white matter angiogenesis in vivo and directly stimulates endothelial cell proliferation in vitro. Conversely, OPC-specific HIF1/2α loss of function leads to insufficient angiogenesis in corpus callosum and catastrophic axon loss. These findings indicate that OPC-intrinsic HIF signaling couples postnatal white matter angiogenesis, axon integrity, and the onset of myelination in mammalian forebrain., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
80. An amino terminal phosphorylation motif regulates intranuclear compartmentalization of Olig2 in neural progenitor cells.
- Author
-
Meijer DH, Sun Y, Liu T, Kane MF, Alberta JA, Adelmant G, Kupp R, Marto JA, Rowitch DH, Nakatani Y, Stiles CD, and Mehta S
- Subjects
- Amino Acid Motifs physiology, Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Nucleus genetics, Cells, Cultured, Female, Male, Mice, Mice, Knockout, Nerve Tissue Proteins genetics, Oligodendrocyte Transcription Factor 2, Phosphorylation genetics, Pregnancy, Basic Helix-Loop-Helix Transcription Factors metabolism, Basic Helix-Loop-Helix Transcription Factors physiology, Cell Nucleus chemistry, Cell Nucleus metabolism, Nerve Tissue Proteins metabolism, Nerve Tissue Proteins physiology, Neural Stem Cells chemistry, Neural Stem Cells metabolism
- Abstract
The bHLH transcription factor Olig2 is expressed in cycling neural progenitor cells but also in terminally differentiated, myelinating oligodendrocytes. Sustained expression of Olig2 is counterintuitive because all known functions of the protein in expansion of neural progenitors and specification of oligodendrocyte progenitors are completed with the formation of mature white matter. How are the biological functions of Olig2 suppressed in terminally differentiated oligodendrocytes? In previous studies, we have shown that a triple serine motif in the amino terminus of Olig2 is phosphorylated in cycling neural progenitors but not in their differentiated progeny. We now show that phosphorylation of the triple serine motif regulates intranuclear compartmentalization of murine Olig2. Phosphorylated Olig2 is preferentially localized to a transcriptionally active "open" chromatin compartment together with coregulator proteins essential for regulation of gene expression. Unphosphorylated Olig2, as seen in mature white matter, is localized mainly within a transcriptionally inactive, chromatin fraction characterized by condensed and inaccessible DNA. Of special note is the observation that the p53 tumor suppressor protein is confined to the open chromatin fraction. Proximity ligation assays show that phosphorylation brings Olig2 within 30 nm of p53 within the open chromatin compartment. The data thus shed light on previously noted promitogenic functions of phosphorylated Olig2, which reflect, at least in part, an oppositional relationship with p53 functions., (Copyright © 2014 the authors 0270-6474/14/348507-12$15.00/0.)
- Published
- 2014
- Full Text
- View/download PDF
81. Astrocyte-encoded positional cues maintain sensorimotor circuit integrity.
- Author
-
Molofsky AV, Kelley KW, Tsai HH, Redmond SA, Chang SM, Madireddy L, Chan JR, Baranzini SE, Ullian EM, and Rowitch DH
- Subjects
- Animals, Astrocytes cytology, Axons physiology, Cell Polarity, Cell Survival drug effects, Humans, Mice, Motor Neurons cytology, Motor Neurons drug effects, Semaphorin-3A deficiency, Semaphorin-3A genetics, Semaphorin-3A metabolism, Semaphorin-3A pharmacology, Sensory Receptor Cells cytology, Spinal Cord cytology, Synapses metabolism, Astrocytes physiology, Motor Neurons physiology, Neural Pathways physiology, Sensory Receptor Cells physiology
- Abstract
Astrocytes, the most abundant cells in the central nervous system, promote synapse formation and help to refine neural connectivity. Although they are allocated to spatially distinct regional domains during development, it is unknown whether region-restricted astrocytes are functionally heterogeneous. Here we show that postnatal spinal cord astrocytes express several region-specific genes, and that ventral astrocyte-encoded semaphorin 3a (Sema3a) is required for proper motor neuron and sensory neuron circuit organization. Loss of astrocyte-encoded Sema3a leads to dysregulated α-motor neuron axon initial segment orientation, markedly abnormal synaptic inputs, and selective death of α- but not of adjacent γ-motor neurons. In addition, a subset of TrkA(+) sensory afferents projects to ectopic ventral positions. These findings demonstrate that stable maintenance of a positional cue by developing astrocytes influences multiple aspects of sensorimotor circuit formation. More generally, they suggest that regional astrocyte heterogeneity may help to coordinate postnatal neural circuit refinement.
- Published
- 2014
- Full Text
- View/download PDF
82. Parallel states of pathological Wnt signaling in neonatal brain injury and colon cancer.
- Author
-
Fancy SP, Harrington EP, Baranzini SE, Silbereis JC, Shiow LR, Yuen TJ, Huang EJ, Lomvardas S, and Rowitch DH
- Subjects
- Adenomatous Polyposis Coli Protein genetics, Adenomatous Polyposis Coli Protein metabolism, Animals, Biomarkers metabolism, Brain Injuries pathology, Cell Differentiation, Colonic Neoplasms pathology, Female, Gene Expression Regulation physiology, Genetic Association Studies, Humans, Infant, Newborn, Infant, Newborn, Diseases, Mice, Mice, Transgenic, Oligodendroglia metabolism, Random Allocation, Up-Regulation, Wnt Proteins genetics, Wnt Proteins metabolism, Wnt Signaling Pathway genetics, Brain Injuries metabolism, Brain Injuries physiopathology, Colonic Neoplasms physiopathology, Hypoxia metabolism, Leukoencephalopathies metabolism, Oligodendroglia physiology, Wnt Proteins physiology, Wnt Signaling Pathway physiology
- Abstract
In colon cancer, mutation of the Wnt repressor APC (encoding adenomatous polyposis coli) leads to a state of aberrant and unrestricted high-activity signaling. However, the relevance of high Wnt tone in non-genetic human disease is unknown. Here we demonstrate that distinct functional states of Wnt activity determine oligodendrocyte precursor cell (OPC) differentiation and myelination. Mouse OPCs with genetic Wnt dysregulation (high tone) express multiple genes in common with colon cancer, including Lef1, Sp5, Ets2, Rnf43 and Dusp4. Surprisingly, we found that OPCs in lesions of hypoxic human neonatal white matter injury upregulated markers of high Wnt activity and lacked expression of APC. We also found that lack of Wnt repressor tone promoted permanent white matter injury after mild hypoxic insult. These findings suggest a state of pathological high-activity Wnt signaling in human disease tissues that lack predisposing genetic mutation.
- Published
- 2014
- Full Text
- View/download PDF
83. Olig1 function is required to repress dlx1/2 and interneuron production in Mammalian brain.
- Author
-
Silbereis JC, Nobuta H, Tsai HH, Heine VM, McKinsey GL, Meijer DH, Howard MA, Petryniak MA, Potter GB, Alberta JA, Baraban SC, Stiles CD, Rubenstein JL, and Rowitch DH
- Subjects
- Action Potentials genetics, Action Potentials physiology, Age Factors, Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Brain embryology, Brain growth & development, Cell Count, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Embryo, Mammalian, Gene Expression Regulation, Developmental genetics, Glutamate Decarboxylase metabolism, Homeodomain Proteins genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mutation genetics, Nerve Tissue Proteins metabolism, Neuropeptides metabolism, Organ Culture Techniques, Patch-Clamp Techniques, Synapses physiology, Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Brain cytology, Gene Expression Regulation, Developmental physiology, Homeodomain Proteins metabolism, Interneurons physiology, Transcription Factors metabolism
- Abstract
Abnormal GABAergic interneuron density, and imbalance of excitatory versus inhibitory tone, is thought to result in epilepsy, neurodevelopmental disorders, and psychiatric disease. Recent studies indicate that interneuron cortical density is determined primarily by the size of the precursor pool in the embryonic telencephalon. However, factors essential for regulating interneuron allocation from telencephalic multipotent precursors are poorly understood. Here we report that Olig1 represses production of GABAergic interneurons throughout the mouse brain. Olig1 deletion in mutant mice results in ectopic expression and upregulation of Dlx1/2 genes in the ventral medial ganglionic eminences and adjacent regions of the septum, resulting in an ∼30% increase in adult cortical interneuron numbers. We show that Olig1 directly represses the Dlx1/2 I12b intergenic enhancer and that Dlx1/2 functions genetically downstream of Olig1. These findings establish Olig1 as an essential repressor of Dlx1/2 and interneuron production in developing mammalian brain., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
84. Evolving concepts of gliogenesis: a look way back and ahead to the next 25 years.
- Author
-
Freeman MR and Rowitch DH
- Subjects
- Animals, History, 20th Century, History, 21st Century, Humans, Neuroglia classification, Biological Evolution, Cell Differentiation physiology, Neuroglia physiology, Neurosciences history, Neurosciences trends
- Abstract
Glial cells are present in all organisms with a CNS and, with increasing brain complexity, glial cells have undergone substantive increases in cell number, diversity, and functions. Invertebrates, such as Drosophila, possess glial subtypes with similarity to mammalian astrocytes in their basic morphology and function, representing fertile ground for unraveling fundamental aspects of glial biology. Although glial subtypes in simple organisms may be relatively homogenous, emerging evidence suggests the possibility that mammalian astrocytes might be highly diversified to match the needs of local neuronal subtypes. In this Perspective, we review classic and new roles identified for astrocytes and oligodendrocytes by recent studies. We propose that delineating genetic and developmental programs across species will be essential to understand the core functions of glia that allow enhanced neuronal function and to achieve new insights into glial roles in higher-order brain function and neurological disease., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
85. Missense mutation in mouse GALC mimics human gene defect and offers new insights into Krabbe disease.
- Author
-
Potter GB, Santos M, Davisson MT, Rowitch DH, Marks DL, Bongarzone ER, and Petryniak MA
- Subjects
- Animals, Brain metabolism, COS Cells, Child, Child, Preschool, Chlorocebus aethiops, Disease Models, Animal, Genetic Variation, Gliosis genetics, Gliosis metabolism, Humans, Leukodystrophy, Globoid Cell pathology, Mice, Myelin Sheath genetics, Myelin Sheath metabolism, Myelin Sheath pathology, Psychosine metabolism, Spinal Cord metabolism, Galactosylceramidase genetics, Galactosylceramidase metabolism, Leukodystrophy, Globoid Cell genetics, Leukodystrophy, Globoid Cell metabolism, Mutation, Missense
- Abstract
Krabbe disease is a devastating pediatric leukodystrophy caused by mutations in the galactocerebrosidase (GALC) gene. A significant subset of the infantile form of the disease is due to missense mutations that result in aberrant protein production. The currently used mouse model, twitcher, has a nonsense mutation not found in Krabbe patients, although it is similar to the human 30 kb deletion in abrogating GALC expression. Here, we identify a spontaneous mutation in GALC, GALCtwi-5J, that precisely matches the E130K missense mutation in patients with infantile Krabbe disease. GALCtwi-5J homozygotes show loss of enzymatic activity despite normal levels of precursor protein, and manifest a more severe phenotype than twitcher, with half the life span. Although neuropathological hallmarks such as gliosis, globoid cells and psychosine accumulation are present throughout the nervous system, the CNS does not manifest significant demyelination. In contrast, the PNS is severely hypomyelinated and lacks large diameter axons, suggesting primary dysmyelination, rather than a demyelinating process. Our data indicate that early demise is due to mechanisms other than myelin loss and support an important role for neuroinflammation in Krabbe disease progression. Furthermore, our results argue against a causative relationship between psychosine accumulation, white matter loss and gliosis.
- Published
- 2013
- Full Text
- View/download PDF
86. Expression profiling of Aldh1l1-precursors in the developing spinal cord reveals glial lineage-specific genes and direct Sox9-Nfe2l1 interactions.
- Author
-
Molofsky AV, Glasgow SM, Chaboub LS, Tsai HH, Murnen AT, Kelley KW, Fancy SP, Yuen TJ, Madireddy L, Baranzini S, Deneen B, Rowitch DH, and Oldham MC
- Subjects
- Age Factors, Aldehyde Dehydrogenase 1 Family, Animals, Cell Differentiation, Cells, Cultured, Chickens, Computational Biology, Electroporation, Embryo, Mammalian, Flow Cytometry, Gene Expression Profiling, Glial Fibrillary Acidic Protein, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Isoenzymes genetics, Mice, Mice, Transgenic, NF-E2-Related Factor 1 genetics, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Neurons classification, Neurons metabolism, Oligonucleotide Array Sequence Analysis, Retinal Dehydrogenase genetics, SOX9 Transcription Factor genetics, Spinal Cord embryology, Transcription Factors genetics, Transcription Factors metabolism, Gene Expression Regulation, Developmental physiology, Isoenzymes metabolism, NF-E2-Related Factor 1 metabolism, Neuroglia metabolism, Retinal Dehydrogenase metabolism, SOX9 Transcription Factor metabolism, Spinal Cord cytology
- Abstract
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., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
87. A dramatic increase of C1q protein in the CNS during normal aging.
- Author
-
Stephan AH, Madison DV, Mateos JM, Fraser DA, Lovelett EA, Coutellier L, Kim L, Tsai HH, Huang EJ, Rowitch DH, Berns DS, Tenner AJ, Shamloo M, and Barres BA
- Subjects
- Animals, Behavior, Animal, Blotting, Western, Brain physiology, Electrophysiology, Excitatory Postsynaptic Potentials, Humans, Immunohistochemistry, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Immunoelectron, Aging metabolism, Brain metabolism, Complement C1q biosynthesis
- Abstract
The decline of cognitive function has emerged as one of the greatest health threats of old age. Age-related cognitive decline is caused by an impacted neuronal circuitry, yet the molecular mechanisms responsible are unknown. C1q, the initiating protein of the classical complement cascade and powerful effector of the peripheral immune response, mediates synapse elimination in the developing CNS. Here we show that C1q protein levels dramatically increase in the normal aging mouse and human brain, by as much as 300-fold. This increase was predominantly localized in close proximity to synapses and occurred earliest and most dramatically in certain regions of the brain, including some but not all regions known to be selectively vulnerable in neurodegenerative diseases, i.e., the hippocampus, substantia nigra, and piriform cortex. C1q-deficient mice exhibited enhanced synaptic plasticity in the adult and reorganization of the circuitry in the aging hippocampal dentate gyrus. Moreover, aged C1q-deficient mice exhibited significantly less cognitive and memory decline in certain hippocampus-dependent behavior tests compared with their wild-type littermates. Unlike in the developing CNS, the complement cascade effector C3 was only present at very low levels in the adult and aging brain. In addition, the aging-dependent effect of C1q on the hippocampal circuitry was independent of C3 and unaccompanied by detectable synapse loss, providing evidence for a novel, complement- and synapse elimination-independent role for C1q in CNS aging.
- Published
- 2013
- Full Text
- View/download PDF
88. Nuclear localization of the mitochondrial factor HIGD1A during metabolic stress.
- Author
-
Ameri K, Rajah AM, Nguyen V, Sanders TA, Jahangiri A, Delay M, Donne M, Choi HJ, Tormos KV, Yeghiazarians Y, Jeffrey SS, Rinaudo PF, Rowitch DH, Aghi M, and Maltepe E
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Disease Models, Animal, Female, Glioblastoma metabolism, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Hypoxia-Ischemia, Brain metabolism, Infant, Newborn, Intracellular Signaling Peptides and Proteins, Mice, Myocardial Infarction metabolism, Protein Binding, Protein Transport, Transplantation, Heterologous, bcl-2 Homologous Antagonist-Killer Protein metabolism, bcl-2-Associated X Protein metabolism, Cell Nucleus metabolism, Mitochondrial Proteins metabolism, Neoplasm Proteins metabolism, Stress, Physiological
- Abstract
Cellular stress responses are frequently governed by the subcellular localization of critical effector proteins. Apoptosis-inducing Factor (AIF) or Glyceraldehyde 3-Phosphate Dehydrogenase (GAPDH), for example, can translocate from mitochondria to the nucleus, where they modulate apoptotic death pathways. Hypoxia-inducible gene domain 1A (HIGD1A) is a mitochondrial protein regulated by Hypoxia-inducible Factor-1α (HIF1α). Here we show that while HIGD1A resides in mitochondria during physiological hypoxia, severe metabolic stress, such as glucose starvation coupled with hypoxia, in addition to DNA damage induced by etoposide, triggers its nuclear accumulation. We show that nuclear localization of HIGD1A overlaps with that of AIF, and is dependent on the presence of BAX and BAK. Furthermore, we show that AIF and HIGD1A physically interact. Additionally, we demonstrate that nuclear HIGD1A is a potential marker of metabolic stress in vivo, frequently observed in diverse pathological states such as myocardial infarction, hypoxic-ischemic encephalopathy (HIE), and different types of cancer. In summary, we demonstrate a novel nuclear localization of HIGD1A that is commonly observed in human disease processes in vivo.
- Published
- 2013
- Full Text
- View/download PDF
89. STAT3-mediated astrogliosis protects myelin development in neonatal brain injury.
- Author
-
Nobuta H, Ghiani CA, Paez PM, Spreuer V, Dong H, Korsak RA, Manukyan A, Li J, Vinters HV, Huang EJ, Rowitch DH, Sofroniew MV, Campagnoni AT, de Vellis J, and Waschek JA
- Subjects
- Age Factors, Animals, Animals, Newborn, Astrocytes chemistry, Astrocytes drug effects, Benzamides pharmacology, Benzamides therapeutic use, Cell Differentiation drug effects, Cell Differentiation genetics, Cell Proliferation drug effects, Cells, Cultured, Culture Media, Conditioned pharmacology, Dioxoles pharmacology, Dioxoles therapeutic use, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Female, Gene Expression Regulation, Developmental drug effects, Glial Fibrillary Acidic Protein metabolism, Gliosis drug therapy, Humans, Infant, Infant, Newborn, Lipopolysaccharides toxicity, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Postmortem Changes, Receptors, Transforming Growth Factor beta antagonists & inhibitors, STAT3 Transcription Factor deficiency, Signal Transduction drug effects, Signal Transduction genetics, Smad2 Protein metabolism, Stem Cells drug effects, Transforming Growth Factor beta1 metabolism, Astrocytes metabolism, Brain Injuries complications, Brain Injuries pathology, Gliosis etiology, Myelin Sheath metabolism, STAT3 Transcription Factor metabolism
- Abstract
Objective: Pathological findings in neonatal brain injury associated with preterm birth include focal and/or diffuse white matter injury (WMI). Despite the heterogeneous nature of this condition, reactive astrogliosis and microgliosis are frequently observed. Thus, molecular mechanisms by which glia activation contribute to WMI were investigated., Methods: Postmortem brains of neonatal brain injury were investigated to identify molecular features of reactive astrocytes. The contribution of astrogliosis to WMI was further tested in a mouse model in genetically engineered mice., Results: Activated STAT3 signaling in reactive astrocytes was found to be a common feature in postmortem brains of neonatal brain injury. In a mouse model of neonatal WMI, conditional deletion of STAT3 in astrocytes resulted in exacerbated WMI, which was associated with delayed maturation of oligodendrocytes. Mechanistically, the delay occurred in association with overexpression of transforming growth factor (TGF)β-1 in microglia, which in healthy controls decreased with myelin maturation in an age-dependent manner. TGFβ-1 directly and dose-dependently inhibited the maturation of purified oligodendrocyte progenitors, and pharmacological inhibition of TGFβ-1 signaling in vivo reversed the delay in myelin development. Factors secreted from STAT3-deficient astrocytes promoted elevated TGFβ-1 production in cultured microglia compared to wild-type astrocytes., Interpretation: These results suggest that myelin development is regulated by a mechanism involving crosstalk between microglia and oligodendrocyte progenitors. Reactive astrocytes may modify this signaling in a STAT3-dependent manner, preventing the pathological expression of TGFβ-1 in microglia and the impairment of oligodendrocyte maturation., (Copyright © 2012 American Neurological Association.)
- Published
- 2012
- Full Text
- View/download PDF
90. Oligodendrocyte regeneration after neonatal hypoxia requires FoxO1-mediated p27Kip1 expression.
- Author
-
Jablonska B, Scafidi J, Aguirre A, Vaccarino F, Nguyen V, Borok E, Horvath TL, Rowitch DH, and Gallo V
- Subjects
- Animals, Animals, Newborn, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p27 genetics, Forkhead Box Protein O1, Humans, Hypoxia, Brain pathology, Infant, Infant, Newborn, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Oligodendroglia cytology, Cell Differentiation physiology, Cyclin-Dependent Kinase Inhibitor p27 biosynthesis, Forkhead Transcription Factors physiology, Gene Expression Regulation, Developmental, Hypoxia, Brain metabolism, Nerve Regeneration physiology, Oligodendroglia physiology
- Abstract
Diffuse white matter injury (DWMI) caused by hypoxia is associated with permanent neurodevelopmental disabilities in preterm infants. The cellular and molecular mechanisms producing DWMI are poorly defined. Using a mouse model of neonatal hypoxia, we demonstrate a biphasic effect on oligodendrocyte development, resulting in hypomyelination. Oligodendrocyte death and oligodendrocyte progenitor cell (OPC) proliferation during the week after hypoxia were followed by delayed oligodendrocyte differentiation and abnormal myelination, as demonstrated by electron microscopy. Cdk2 activation was essential for the regenerative OPC response after hypoxia and was accompanied by reduced FoxO1-dependent p27(Kip1) expression. p27(Kip1) was also reduced in OPCs in human infant white matter lesions after hypoxia. The negative effects of hypoxia on oligodendrogenesis and myelination were more pronounced in p27(Kip1)-null mice; conversely, overexpression of FoxO1 or p27(Kip1) in OPCs after hypoxia promoted oligodendrogenesis. Our studies demonstrate for the first time that neonatal hypoxia affects the Foxo1/p27(Kip1) pathway during white matter development. We also show that molecular manipulation of this pathway enhances oligodendrocyte regeneration during a critical developmental time window after DWMI. Thus, FoxO1 and p27(Kip1) may serve as promising target molecules for promoting timely oligodendrogenesis in neonatal DWMI.
- Published
- 2012
- Full Text
- View/download PDF
91. Voltage-gated potassium channel EAG2 controls mitotic entry and tumor growth in medulloblastoma via regulating cell volume dynamics.
- Author
-
Huang X, Dubuc AM, Hashizume R, Berg J, He Y, Wang J, Chiang C, Cooper MK, Northcott PA, Taylor MD, Barnes MJ, Tihan T, Chen J, Hackett CS, Weiss WA, James CD, Rowitch DH, Shuman MA, Jan YN, and Jan LY
- Subjects
- Animals, COS Cells, Cell Cycle Checkpoints genetics, Cell Proliferation, Cells, Cultured, Chlorocebus aethiops, Enzyme Activation genetics, Ether-A-Go-Go Potassium Channels genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, HEK293 Cells, Humans, MAP Kinase Signaling System, Medulloblastoma mortality, Mice, Survival Analysis, Cell Size, Ether-A-Go-Go Potassium Channels metabolism, Medulloblastoma physiopathology, Mitosis
- Abstract
Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.
- Published
- 2012
- Full Text
- View/download PDF
92. Evidence that nuclear factor IA inhibits repair after white matter injury.
- Author
-
Fancy SP, Glasgow SM, Finley M, Rowitch DH, and Deneen B
- Subjects
- Adenomatous Polyposis Coli Protein metabolism, Animals, Arabidopsis Proteins metabolism, Cell Differentiation drug effects, Cells, Cultured, Cerebral Cortex cytology, Chromatin Immunoprecipitation, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Disease Models, Animal, Electroporation, Embryo, Mammalian, Gene Expression Regulation, Developmental genetics, Homeodomain Proteins metabolism, Humans, Hypoxia-Ischemia, Brain metabolism, Infant, Infant, Newborn, Intramolecular Transferases metabolism, Leukoencephalopathies chemically induced, Lysophosphatidylcholines toxicity, Mice, Mice, Transgenic, Multiple Sclerosis metabolism, Myelin Basic Protein metabolism, NFI Transcription Factors genetics, Oligodendroglia drug effects, Spinal Cord pathology, Stem Cells drug effects, Stem Cells metabolism, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Gene Expression Regulation, Developmental physiology, Leukoencephalopathies metabolism, Leukoencephalopathies pathology, NFI Transcription Factors metabolism, Oligodendroglia metabolism
- Abstract
Objective: 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., Methods: NFIA expression patterns in human neonatal hypoxic-ischemic encephalopathy (HIE) and MS as well as developmental expression in mice were evaluated. Functional studies during remyelination were performed using a lysolecithin model, coupled with lentiviral misexpression of NFIA. The role of NFIA during oligodendrocyte lineage development was characterized using chick and mouse models and in vitro culture of oligodendrocyte progenitors. Biochemical mechanism of NFIA function was evaluated using chromatin immunoprecipitation and reporter assays., Results: NFIA is expressed in oligodendrocyte progenitors, but not differentiated oligodendrocytes during mouse embryonic development. Examination of NFIA expression in white matter lesions of human newborns with neonatal HIE, as well active MS lesions in adults, revealed that it is similarly expressed in oligodendrocyte progenitors and not oligodendrocytes. Functional studies indicate that NFIA is sufficient to suppress oligodendrocyte progenitor differentiation during adult remyelination and embryonic development through direct repression of myelin gene expression., Interpretation: These studies suggest that NFIA participates in the control of oligodendrocyte progenitor differentiation and may contribute to the inhibition of remyelination in human myelin disorders., (Copyright © 2012 American Neurological Association.)
- Published
- 2012
- Full Text
- View/download PDF
93. Regional astrocyte allocation regulates CNS synaptogenesis and repair.
- Author
-
Tsai HH, Li H, Fuentealba LC, Molofsky AV, Taveira-Marques R, Zhuang H, Tenney A, Murnen AT, Fancy SP, Merkle F, Kessaris N, Alvarez-Buylla A, Richardson WD, and Rowitch DH
- Subjects
- Animals, Bacterial Proteins metabolism, Basic Helix-Loop-Helix Transcription Factors genetics, Brain abnormalities, Brain physiology, Brain Injuries physiopathology, Green Fluorescent Proteins, Homeobox Protein Nkx-2.2, Homeodomain Proteins metabolism, Integrases genetics, Luminescent Proteins metabolism, Mice, Mice, Transgenic, Nerve Tissue Proteins genetics, Proteins metabolism, RNA, Untranslated, Spinal Cord abnormalities, Spinal Cord physiology, Spinal Cord Injuries physiopathology, Transcription Factors metabolism, Transcription, Genetic, Zebrafish Proteins, Astrocytes physiology, Brain cytology, Cell Movement, Motor Neurons physiology, Spinal Cord cytology, Synapses physiology
- Abstract
Astrocytes, the most abundant cell population in the central nervous system (CNS), are essential for normal neurological function. We show that astrocytes are allocated to spatial domains in mouse spinal cord and brain in accordance with their embryonic sites of origin in the ventricular zone. These domains remain stable throughout life without evidence of secondary tangential migration, even after acute CNS injury. Domain-specific depletion of astrocytes in ventral spinal cord resulted in abnormal motor neuron synaptogenesis, which was not rescued by immigration of astrocytes from adjoining regions. Our findings demonstrate that region-restricted astrocyte allocation is a general CNS phenomenon and reveal intrinsic limitations of the astroglial response to injury.
- Published
- 2012
- Full Text
- View/download PDF
94. Regulated temporal-spatial astrocyte precursor cell proliferation involves BRAF signalling in mammalian spinal cord.
- Author
-
Tien AC, Tsai HH, Molofsky AV, McMahon M, Foo LC, Kaul A, Dougherty JD, Heintz N, Gutmann DH, Barres BA, and Rowitch DH
- Subjects
- Aldehyde Dehydrogenase metabolism, Animals, Astrocytes metabolism, Cell Proliferation, Cells, Cultured, Flow Cytometry, Immunohistochemistry, Mice, Spinal Cord embryology, Astrocytes cytology, Spinal Cord cytology
- Abstract
Expansion of astrocyte populations in the central nervous system is characteristic of evolutionarily more complex organisms. However, regulation of mammalian astrocyte precursor proliferation during development remains poorly understood. Here, we used Aldh1L1-GFP to identify two morphologically distinct types of proliferative astrocyte precursors: radial glia (RG) in the ventricular zone and a second cell type we call an 'intermediate astrocyte precursor' (IAP) located in the mantle region of the spinal cord. Astrogenic RG and IAP cells proliferated in a progressive ventral-to-dorsal fashion in a tight window from embryonic day 13.5 until postnatal day 3, which correlated precisely with the pattern of active ERK signalling. Conditional loss of BRAF function using BLBP-cre resulted in a 20% decrease in astrocyte production, whereas expression of activated BRAFV600E resulted in astrocyte hyperproliferation. Interestingly, BRAFV600E mitogenic effects in astrocytes were restricted, in part, by the function of p16INK4A-p19(ARF), which limited the temporal epoch for proliferation. Together, these findings suggest that astrocyte precursor proliferation involves distinct RG and IAP cells; is subjected to temporal and spatial control; and depends in part on BRAF signalling at early stages of mammalian spinal cord development.
- Published
- 2012
- Full Text
- View/download PDF
95. Cooperative interactions of BRAFV600E kinase and CDKN2A locus deficiency in pediatric malignant astrocytoma as a basis for rational therapy.
- Author
-
Huillard E, Hashizume R, Phillips JJ, Griveau A, Ihrie RA, Aoki Y, Nicolaides T, Perry A, Waldman T, McMahon M, Weiss WA, Petritsch C, James CD, and Rowitch DH
- Subjects
- Animals, Apoptosis drug effects, Astrocytoma genetics, Astrocytoma pathology, Blotting, Western, Cell Differentiation drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cells, Cultured, Child, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Immunohistochemistry, Indoles pharmacology, Mice, Mice, Inbred BALB C, Mice, Knockout, Mice, Nude, Mice, SCID, Neural Stem Cells cytology, Neural Stem Cells drug effects, Neural Stem Cells metabolism, Phosphorylation drug effects, Piperazines pharmacology, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf metabolism, Pyridines pharmacology, Sulfonamides pharmacology, Astrocytoma drug therapy, Cyclin-Dependent Kinase Inhibitor p16 antagonists & inhibitors, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Xenograft Model Antitumor Assays
- Abstract
Although malignant astrocytomas are a leading cause of cancer-related death in children, rational therapeutic strategies are lacking. We previously identified activating mutations of v-raf murine sarcoma viral oncogene homolog B1 (BRAF) (BRAF(T1799A) encoding BRAF(V600E)) in association with homozygous cyclin-dependent kinase inhibitor 2A (CDKN2A, encoding p14ARF and p16Ink4a) deletions in pediatric infiltrative astrocytomas. Here we report that BRAF(V600E) expression in neural progenitors (NPs) is insufficient for tumorigenesis and increases NP cellular differentiation as well as apoptosis. In contrast, astrocytomas are readily generated from NPs with additional Ink4a-Arf deletion. The BRAF(V600E) inhibitor PLX4720 significantly increased survival of mice after intracranial transplant of genetically relevant murine or human astrocytoma cells. Moreover, combination therapy using PLX4720 plus the Cyclin-dependent kinase (CDK) 4/6-specific inhibitor PD0332991 further extended survival relative to either monotherapy. Our findings indicate a rational therapeutic strategy for treating a subset of pediatric astrocytomas with BRAF(V600E) mutation and CDKN2A deficiency.
- Published
- 2012
- Full Text
- View/download PDF
96. Species-dependent posttranscriptional regulation of NOS1 by FMRP in the developing cerebral cortex.
- Author
-
Kwan KY, Lam MM, Johnson MB, Dube U, Shim S, Rašin MR, Sousa AM, Fertuzinhos S, Chen JG, Arellano JI, Chan DW, Pletikos M, Vasung L, Rowitch DH, Huang EJ, Schwartz ML, Willemsen R, Oostra BA, Rakic P, Heffer M, Kostović I, Judaš M, and Sestan N
- Subjects
- Animals, Cerebral Cortex metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome metabolism, Fragile X Syndrome physiopathology, Gene Expression Regulation, Humans, Mice, Mice, Knockout, Neurogenesis, Pyramidal Cells metabolism, RNA Processing, Post-Transcriptional, Species Specificity, Cerebral Cortex embryology, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome embryology, Nitric Oxide Synthase Type I metabolism
- Abstract
Fragile X syndrome (FXS), the leading monogenic cause of intellectual disability and autism, results from loss of function of the RNA-binding protein FMRP. Here, we show that FMRP regulates translation of neuronal nitric oxide synthase 1 (NOS1) in the developing human neocortex. Whereas NOS1 mRNA is widely expressed, NOS1 protein is transiently coexpressed with FMRP during early synaptogenesis in layer- and region-specific pyramidal neurons. These include midfetal layer 5 subcortically projecting neurons arranged into alternating columns in the prospective Broca's area and orofacial motor cortex. Human NOS1 translation is activated by FMRP via interactions with coding region binding motifs absent from mouse Nos1 mRNA, which is expressed in mouse pyramidal neurons, but not efficiently translated. Correspondingly, neocortical NOS1 protein levels are severely reduced in developing human FXS cases, but not FMRP-deficient mice. Thus, alterations in FMRP posttranscriptional regulation of NOS1 in developing neocortical circuits may contribute to cognitive dysfunction in FXS., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
97. Astrocytes and disease: a neurodevelopmental perspective.
- Author
-
Molofsky AV, Krencik R, Ullian EM, Tsai HH, Deneen B, Richardson WD, Barres BA, and Rowitch DH
- Subjects
- Astrocytes pathology, Heredodegenerative Disorders, Nervous System pathology, Humans, Mental Disorders pathology, Neural Stem Cells pathology, Neural Stem Cells physiology, Astrocytes physiology, Heredodegenerative Disorders, Nervous System etiology, Mental Disorders etiology, Neurogenesis
- Abstract
Astrocytes are no longer seen as a homogenous population of cells. In fact, recent studies indicate that astrocytes are morphologically and functionally diverse and play critical roles in neurodevelopmental diseases such as Rett syndrome and fragile X mental retardation. This review summarizes recent advances in astrocyte development, including the role of neural tube patterning in specification and developmental functions of astrocytes during synaptogenesis. We propose here that a precise understanding of astrocyte development is critical to defining heterogeneity and could lead advances in understanding and treating a variety of neuropsychiatric diseases.
- Published
- 2012
- Full Text
- View/download PDF
98. Identification of molecular compartments and genetic circuitry in the developing mammalian kidney.
- Author
-
Yu J, Valerius MT, Duah M, Staser K, Hansard JK, Guo JJ, McMahon J, Vaughan J, Faria D, Georgas K, Rumballe B, Ren Q, Krautzberger AM, Junker JP, Thiagarajan RD, Machanick P, Gray PA, van Oudenaarden A, Rowitch DH, Stiles CD, Ma Q, Grimmond SM, Bailey TL, Little MH, and McMahon AP
- Subjects
- Animals, Gene Expression Profiling, Gene Expression Regulation, Developmental genetics, Gene Expression Regulation, Developmental physiology, In Situ Hybridization, Kidney metabolism, Mice, Urogenital System metabolism
- Abstract
Lengthy developmental programs generate cell diversity within an organotypic framework, enabling the later physiological actions of each organ system. Cell identity, cell diversity and cell function are determined by cell type-specific transcriptional programs; consequently, transcriptional regulatory factors are useful markers of emerging cellular complexity, and their expression patterns provide insights into the regulatory mechanisms at play. We performed a comprehensive genome-scale in situ expression screen of 921 transcriptional regulators in the developing mammalian urogenital system. Focusing on the kidney, analysis of regional-specific expression patterns identified novel markers and cell types associated with development and patterning of the urinary system. Furthermore, promoter analysis of synexpressed genes predicts transcriptional control mechanisms that regulate cell differentiation. The annotated informational resource (www.gudmap.org) will facilitate functional analysis of the mammalian kidney and provides useful information for the generation of novel genetic tools to manipulate emerging cell populations.
- Published
- 2012
- Full Text
- View/download PDF
99. Olig1 function is required for remyelination potential of transplanted neural progenitor cells in a model of viral-induced demyelination.
- Author
-
Whitman LM, Blanc CA, Schaumburg CS, Rowitch DH, and Lane TE
- Subjects
- Animals, Astrocytes metabolism, Astrocytes pathology, Axons metabolism, Axons pathology, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Differentiation physiology, Cell Movement, Cells, Cultured, Coronavirus Infections metabolism, Coronavirus Infections pathology, Demyelinating Diseases pathology, Demyelinating Diseases virology, Disease Models, Animal, Male, Mice, Mice, Knockout, Murine hepatitis virus, Myelin Sheath pathology, Nerve Fibers, Myelinated pathology, Oligodendroglia pathology, Basic Helix-Loop-Helix Transcription Factors metabolism, Demyelinating Diseases metabolism, Myelin Sheath metabolism, Nerve Fibers, Myelinated metabolism, Neural Stem Cells transplantation, Oligodendroglia metabolism
- Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in cumulative neurologic deficits associated with progressive myelin loss. We have previously shown that transplantation of neural progenitor cells (NPCs) into mice persistently infected with the JHM strain of mouse hepatitis virus (JHMV) results in enhanced differentiation into oligodendrocyte progenitor cells (OPCs) that is associated with remyelination and axonal sparing. The current study examines the contributions of the transcription factor Olig1 on NPC differentiation and remyelination. Under defined conditions, NPCs preferentially differentiate into oligodendroglia whereas NPCs isolated from Olig1-deficient (Olig1-/-) mice exhibit enhanced differentiation into astrocytes. Transplantation of Olig1-/- and Olig1+/+ NPCs into JHMV-infected mice resulted in similar cell survival, proliferation, and selective migration to areas of demyelination. However, only recipients of wild type NPCs exhibited extensive remyelination compared to mice receiving Olig1-/- NPCs. In vivo characterization of NPCs revealed that Olig1+/+ NPCs preferentially differentiated into NG2-positive OPCs and formed processes expressing myelin basic protein that encircled axons. In contrast, the majority of transplanted Olig1-/- NPCs differentiated into GFAP-positive cells consistent with the astrocyte lineage. These results indicate that exogenous NPCs contribute to improved clinical and histological outcome and this is associated with remyelination by this donor population. Further, these findings reveal that Olig1function is required for the remyelination potential of NPCs after transplant, through specification and/or maintenance of oligodendroglial identity., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
100. Pro-neural miR-128 is a glioma tumor suppressor that targets mitogenic kinases.
- Author
-
Papagiannakopoulos T, Friedmann-Morvinski D, Neveu P, Dugas JC, Gill RM, Huillard E, Liu C, Zong H, Rowitch DH, Barres BA, Verma IM, and Kosik KS
- Subjects
- Animals, Cell Differentiation genetics, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic, Down-Regulation, Gene Expression Regulation, Neoplastic, Humans, Mice, Mice, Nude, Mice, SCID, Neural Stem Cells physiology, Brain Neoplasms genetics, ErbB Receptors genetics, Genes, Tumor Suppressor, Glioma genetics, MicroRNAs physiology, Receptor, Platelet-Derived Growth Factor alpha genetics
- Abstract
MicroRNAs (miRNAs) carry out post-transcriptional control of a multitude of cellular processes. Aberrant expression of miRNA can lead to diseases, including cancer. Gliomas are aggressive brain tumors that are thought to arise from transformed glioma-initiating neural stem cells (giNSCs). With the use of giNSCs and human glioblastoma cells, we investigated the function of miRNAs in gliomas. We identified pro-neuronal miR-128 as a candidate glioma tumor suppressor miRNA. Decreased expression of miR-128 correlates with aggressive human glioma subtypes. With a combination of molecular, cellular and in vivo approaches, we characterize miR-128's tumor suppressive role. miR-128 represses giNSC growth by enhancing neuronal differentiation. miR-128 represses growth and mediates differentiation by targeting oncogenic receptor tyrosine kinases (RTKs) epithelial growth factor receptor and platelet-derived growth factor receptor-α. Using an autochthonous glioma mouse model, we demonstrated that miR-128 repressed gliomagenesis. We identified miR-128 as a glioma tumor suppressor that targets RTK signaling to repress giNSC self-renewal and enhance differentiation.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.