Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
ACS Med Chem Lett ; 15(5): 714-721, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38746903

ABSTRACT

We herein report the discovery, synthesis, and evolution of a series of indazoles and azaindazoles as CNS-penetrant IRAK4 inhibitors. Described is the use of structure-based and property-based drug design strategically leveraged to guide the property profile of a key series into a favorable property space while maintaining potency and selectivity. Our rationale that led toward functionalities with potency improvements, CNS-penetration, solubility, and favorable drug-like properties is portrayed. In vivo evaluation of an advanced analogue showed significant, dose-dependent modulation of inflammatory cytokines in a mouse model. In pursuit of incorporating a highly engineered bridged ether that was crucial to metabolic stability in this series, significant synthetic challenges were overcome to enable the preparation of the analogues.

2.
J Med Chem ; 67(10): 8383-8395, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38695469

ABSTRACT

Interleukin receptor associated kinase 4 (IRAK4) plays an important role in innate immune signaling through Toll-like and interleukin-1 receptors and represents an attractive target for the treatment of inflammatory diseases and cancer. We previously reported the development of a potent, selective, and brain-penetrant imidazopyrimidine series of IRAK4 inhibitors. However, lead molecule BIO-7488 (1) suffered from low solubility which led to variable PK, compound accumulation, and poor in vivo tolerability. Herein, we describe the discovery of a series of pyridone analogs with improved solubility which are highly potent, selective and demonstrate desirable PK profiles including good oral bioavailability and excellent brain penetration. BIO-8169 (2) reduced the in vivo production of pro-inflammatory cytokines, was well tolerated in safety studies in rodents and dog at margins well above the predicted efficacious exposure and showed promising results in a mouse model for multiple sclerosis.


Subject(s)
Brain , Interleukin-1 Receptor-Associated Kinases , Protein Kinase Inhibitors , Animals , Dogs , Male , Mice , Rats , Brain/metabolism , Brain/drug effects , Drug Discovery , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/therapeutic use , Structure-Activity Relationship
3.
Brain ; 144(8): 2361-2374, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34145876

ABSTRACT

Autoantibodies are a hallmark of numerous neurological disorders, including multiple sclerosis, autoimmune encephalitides and neuromyelitis optica. Whilst well understood in peripheral myeloid cells, the pathophysiological significance of autoantibody-induced Fc receptor signalling in microglia remains unknown, in part due to the lack of a robust in vivo model. Moreover, the application of therapeutic antibodies for neurodegenerative disease also highlights the importance of understanding Fc receptor signalling in microglia. Here, we describe a novel in vivo experimental paradigm that allows for selective engagement of Fc receptors within the CNS by peripherally injecting anti-myelin oligodendrocyte glycoprotein (MOG) monoclonal antibodies into normal wild-type mice. MOG antigen-bound immunoglobulins were detected throughout the CNS and triggered a rapid and tightly regulated proliferative response in both brain and spinal cord microglia. This microglial response was abrogated when anti-MOG antibodies were deprived of Fc receptor effector function or injected into Fcγ receptor knockout mice and was associated with the downregulation of Fc receptors in microglia, but not peripheral myeloid cells, establishing that this response was dependent on central Fc receptor engagement. Downstream of the Fc receptors, BTK was a required signalling node for this response, as microglia proliferation was amplified in BtkE41K knock-in mice expressing a constitutively active form of the enzyme and blunted in mice treated with a CNS-penetrant small molecule inhibitor of BTK. Finally, this response was associated with transient and stringently regulated changes in gene expression predominantly related to cellular proliferation, which markedly differed from transcriptional programs typically associated with Fc receptor engagement in peripheral myeloid cells. Together, these results establish a physiologically-meaningful functional response to Fc receptor and BTK signalling in microglia, while providing a novel in vivo tool to further dissect the roles of microglia-specific Fc receptor and BTK-driven responses to both pathogenic and therapeutic antibodies in CNS homeostasis and disease.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Autoantibodies/immunology , Brain/pathology , Microglia/pathology , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Fc/metabolism , Spinal Cord/pathology , Animals , Brain/immunology , Brain/metabolism , Cell Proliferation/physiology , Mice , Microglia/immunology , Microglia/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
4.
Nature ; 568(7752): 336-343, 2019 04.
Article in English | MEDLINE | ID: mdl-30996318

ABSTRACT

The brains of humans and other mammals are highly vulnerable to interruptions in blood flow and decreases in oxygen levels. Here we describe the restoration and maintenance of microcirculation and molecular and cellular functions of the intact pig brain under ex vivo normothermic conditions up to four hours post-mortem. We have developed an extracorporeal pulsatile-perfusion system and a haemoglobin-based, acellular, non-coagulative, echogenic, and cytoprotective perfusate that promotes recovery from anoxia, reduces reperfusion injury, prevents oedema, and metabolically supports the energy requirements of the brain. With this system, we observed preservation of cytoarchitecture; attenuation of cell death; and restoration of vascular dilatory and glial inflammatory responses, spontaneous synaptic activity, and active cerebral metabolism in the absence of global electrocorticographic activity. These findings demonstrate that under appropriate conditions the isolated, intact large mammalian brain possesses an underappreciated capacity for restoration of microcirculation and molecular and cellular activity after a prolonged post-mortem interval.


Subject(s)
Autopsy , Brain/blood supply , Brain/cytology , Cerebrovascular Circulation , Microcirculation , Swine , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caspase 3/metabolism , Cell Survival , Cerebral Arteries/physiology , Disease Models, Animal , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Inflammation/metabolism , Inflammation/pathology , Neuroglia/cytology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Perfusion , Reperfusion Injury/prevention & control , Swine/blood , Synapses/metabolism , Synapses/pathology , Time Factors , Vasodilation
5.
Ann Neurol ; 81(4): 560-571, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28253550

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.


Subject(s)
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 , Animals , Cell Culture Techniques , Disease Models, Animal , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells
6.
Neuron ; 89(6): 1208-1222, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26924435

ABSTRACT

Trisomy 21, or Down syndrome (DS), is the most common genetic cause of developmental delay and intellectual disability. To gain insight into the underlying molecular and cellular pathogenesis, we conducted a multi-region transcriptome analysis of DS and euploid control brains spanning from mid-fetal development to adulthood. We found genome-wide alterations in the expression of a large number of genes, many of which exhibited temporal and spatial specificity and were associated with distinct biological processes. In particular, we uncovered co-dysregulation of genes associated with oligodendrocyte differentiation and myelination that were validated via cross-species comparison to Ts65Dn trisomy mice. Furthermore, we show that hypomyelination present in Ts65Dn mice is in part due to cell-autonomous effects of trisomy on oligodendrocyte differentiation and results in slower neocortical action potential transmission. Together, these results identify defects in white matter development and function in DS, and they provide a transcriptional framework for further investigating DS neuropathogenesis.


Subject(s)
Brain , Cell Differentiation/genetics , Down Syndrome/pathology , Gene Expression Regulation, Developmental/genetics , Myelin Sheath/metabolism , Oligodendroglia/pathology , Action Potentials/genetics , Adolescent , Adult , Animals , Brain/growth & development , Brain/metabolism , Brain/pathology , Cell Differentiation/physiology , Child , Child, Preschool , Chromosomes, Human, Pair 17/genetics , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/physiopathology , Female , Gene Expression Profiling , Humans , Infant , Infant, Newborn , Male , Mice , Mice, Transgenic , Mosaicism , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Neural Conduction/genetics , Postmortem Changes , Trisomy/genetics , White Matter/pathology , White Matter/ultrastructure , Young Adult
7.
Neuron ; 89(2): 248-68, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26796689

ABSTRACT

The human CNS follows a pattern of development typical of all mammals, but certain neurodevelopmental features are highly derived. Building the human CNS requires the precise orchestration and coordination of myriad molecular and cellular processes across a staggering array of cell types and over a long period of time. Dysregulation of these processes affects the structure and function of the CNS and can lead to neurological or psychiatric disorders. Recent technological advances and increased focus on human neurodevelopment have enabled a more comprehensive characterization of the human CNS and its development in both health and disease. The aim of this review is to highlight recent advancements in our understanding of the molecular and cellular landscapes of the developing human CNS, with focus on the cerebral neocortex, and the insights these findings provide into human neural evolution, function, and dysfunction.


Subject(s)
Central Nervous System/cytology , Central Nervous System/growth & development , Neurogenesis/physiology , Animals , Brain/cytology , Brain/embryology , Brain/growth & development , Central Nervous System/embryology , Humans , Neurodevelopmental Disorders/pathology , Organogenesis/physiology
8.
Cell ; 158(2): 383-396, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25018103

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , 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
9.
Nat Neurosci ; 17(4): 506-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24609463

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.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/physiopathology , Colonic Neoplasms/physiopathology , Hypoxia/metabolism , Leukoencephalopathies/metabolism , Oligodendroglia/physiology , Wnt Proteins/physiology , Wnt Signaling Pathway/physiology , 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
10.
Neuron ; 81(3): 574-87, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24507192

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Interneurons/physiology , Transcription Factors/metabolism , 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
11.
J Neurosci ; 32(26): 8930-9, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22745493

ABSTRACT

Premature children born with very low birth weight (VLBW) can suffer chronic hypoxic injury as a consequence of abnormal lung development and cardiovascular abnormalities, often leading to grave neurological and behavioral consequences. Emerging evidence suggests that environmental enrichment improves outcome in animal models of adult brain injury and disease; however, little is known about the impact of environmental enrichment following developmental brain injury. Intriguingly, data on socio-demographic factors from longitudinal studies that examined a number of VLBW cohorts suggest that early environment has a substantial impact on neurological and behavioral outcomes. In the current study, we demonstrate that environmental enrichment significantly enhances behavioral and neurobiological recovery from perinatal hypoxic injury. Using a genetic fate-mapping model that allows us to trace the progeny of GFAP+ astroglial cells, we show that hypoxic injury increases the proportion of astroglial cells that attain a neuronal fate. In contrast, environmental enrichment increases the stem cell pool, both through increased stem cell proliferation and stem cell survival. In mice subjected to hypoxia and subsequent enrichment there is an additive effect of both conditions on hippocampal neurogenesis from astroglia, resulting in a robust increase in the number of neurons arising from GFAP+ cells by the time these mice reach full adulthood.


Subject(s)
Cell Differentiation/physiology , Cognition Disorders/nursing , Cognition Disorders/pathology , Environment , Glial Fibrillary Acidic Protein/metabolism , Stem Cells/physiology , Analysis of Variance , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Count , Cell Differentiation/genetics , Cognition Disorders/etiology , Deoxyuridine/metabolism , Disease Models, Animal , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hypoxia/complications , Idoxuridine/metabolism , Ki-67 Antigen/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis , Neuroglia/metabolism , Receptors, Estrogen/genetics , Stem Cells/metabolism , Tamoxifen/pharmacology
12.
J Neurosci ; 31(25): 9205-21, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21697371

ABSTRACT

Glial fibrillary acidic protein-positive (GFAP(+)) cells give rise to new neurons in the neurogenic niches; whether they are able to generate neurons in the cortical parenchyma is not known. Here, we use genetic fate mapping to examine the progeny of GFAP(+) cells after postnatal hypoxia, a model for the brain injury observed in premature children. After hypoxia, immature cortical astroglia underwent a shift toward neuronal fate and generated cortical excitatory neurons that appeared synaptically integrated into the circuitry. Fate-mapped cortical GFAP(+) cells derived ex vivo from hypoxic, but not normoxic, mice were able to form pluripotent, long-term self-renewing neurospheres. Similarly, exposure to low oxygen conditions in vitro induced stem-cell-like potential in immature cortical GFAP(+) cells. Our data support the conclusion that hypoxia promotes pluripotency in GFAP(+) cells in the cortical parenchyma. Such plasticity possibly explains the cognitive recovery found in some preterm children.


Subject(s)
Glial Fibrillary Acidic Protein/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Cell Differentiation , Cells, Cultured , Mice , Mice, Transgenic
13.
Nat Neurosci ; 14(8): 1009-16, 2011 Jun 26.
Article in English | MEDLINE | ID: mdl-21706018

ABSTRACT

Permanent damage to white matter tracts, comprising axons and myelinating oligodendrocytes, is an important component of brain injuries of the newborn that cause cerebral palsy and cognitive disabilities, as well as multiple sclerosis in adults. However, regulatory factors relevant in human developmental myelin disorders and in myelin regeneration are unclear. We found that AXIN2 was expressed in immature oligodendrocyte progenitor cells (OLPs) in white matter lesions of human newborns with neonatal hypoxic-ischemic and gliotic brain damage, as well as in active multiple sclerosis lesions in adults. Axin2 is a target of Wnt transcriptional activation that negatively feeds back on the pathway, promoting ß-catenin degradation. We found that Axin2 function was essential for normal kinetics of remyelination. The small molecule inhibitor XAV939, which targets the enzymatic activity of tankyrase, acted to stabilize Axin2 levels in OLPs from brain and spinal cord and accelerated their differentiation and myelination after hypoxic and demyelinating injury. Together, these findings indicate that Axin2 is an essential regulator of remyelination and that it might serve as a pharmacological checkpoint in this process.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/therapy , Cytoskeletal Proteins/metabolism , Gene Expression Regulation/physiology , Myelin Proteins/metabolism , Adult , Animals , Animals, Newborn , Axin Protein , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Injuries/etiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/ultrastructure , Cerebral Cortex/cytology , Corpus Callosum/drug effects , Corpus Callosum/metabolism , Cytoskeletal Proteins/deficiency , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/therapy , Infant, Newborn , Ki-67 Antigen/metabolism , Lysophosphatidylcholines/toxicity , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Myelin Proteins/genetics , Myelin Proteins/therapeutic use , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Oligodendroglia/physiology , Organ Culture Techniques
14.
Dis Model Mech ; 3(11-12): 678-88, 2010.
Article in English | MEDLINE | ID: mdl-21030421

ABSTRACT

Newborn neurological injuries are the leading cause of intellectual and motor disabilities that are associated with cerebral palsy. Cerebral white matter injury is a common feature in hypoxic-ischemic encephalopathy (HIE), which affects full-term infants, and in periventricular leukomalacia (PVL), which affects preterm infants. This article discusses recent efforts to model neonatal white matter injury using mammalian systems. We emphasize that a comprehensive understanding of oligodendrocyte development and physiology is crucial for obtaining new insights into the pathobiology of HIE and PVL as well as for the generation of more sophisticated and faithful animal models.


Subject(s)
Cerebral Palsy/complications , Cerebral Palsy/pathology , Disease Models, Animal , Leukomalacia, Periventricular/complications , Leukomalacia, Periventricular/pathology , Animals , Biomedical Research , Humans , Infant, Newborn , Oligodendroglia/pathology
15.
Mol Cell Neurosci ; 44(4): 362-73, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20470892

ABSTRACT

It is well established that cerebellar granule cell precursors (GCPs) initially derive from progenitors in the rhombic lip of the embryonic cerebellar primordium. GCPs proliferate and migrate tangentially across the cerebellum to form the external granule cell layer (EGL) in late embryogenesis and early postnatal development. It is unclear whether GCPs are specified exclusively in the embryonic rhombic lip or whether their precursor persists in the neonate. Using transgenic mice expressing DsRed under the human glial fibrillary acidic protein (hGFAP) promoter, we found 2 populations of DsRed(+) cells in the EGL in the first postnatal week defined by bright and faint DsRed-fluorescent signal. Bright DsRed(+) cells have a protein expression profile and electrophysiological characteristics typical of astrocytes, but faint DsRed(+) cells in the EGL and internal granule cell layer (IGL) express markers and physiological properties of immature neurons. To determine if these astroglial cells gave rise to GCPs, we genetically tagged them with EGFP or betagal reporter genes at postnatal day (P)3-P5 using a hGFAP promoter driven inducible Cre recombinase. We found that GFAP promoter(+) cells in the EGL are proliferative and express glial and neural stem cell markers. In addition, immature granule cells (GCs) en route to the IGL at P12 as well as GCs in the mature cerebellum, 30days after recombination, express the reporter protein, suggesting that GFAP promoter(+) cells in the EGL generate a subset of granule cells. The identification of glial cells which function as neuronal progenitor cells profoundly impacts our understanding of cellular plasticity in the developing cerebellum.


Subject(s)
Astrocytes/metabolism , Cerebellum/cytology , Glial Fibrillary Acidic Protein/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Lineage/physiology , Genes, Reporter , Green Fluorescent Proteins , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/physiology , Neurons/cytology , Promoter Regions, Genetic/genetics , Stem Cells/cytology , Time Factors , beta-Galactosidase
16.
Stem Cells ; 27(5): 1152-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19418461

ABSTRACT

Neural stem or progenitor cells (NSC/NPCs) able to generate the different neuron and glial cell types of the cerebellum have been isolated in vitro, but their identity and location in the intact cerebellum are unclear. Here, we use inducible Cre recombination in GFAPCreER(T2) mice to irreversibly activate reporter gene expression at P2 (postnatal day 2), P5, and P12 in cells with GFAP (glial fibrillary acidic protein) promoter activity and analyze the fate of genetically tagged cells in vivo. We show that cells tagged at P2-P5 with beta-galactosidase or enhanced green fluorescent proteins reporter genes generate at least 30% of basket and stellate GABAergic interneurons in the molecular layer (ML) and that they lose their neurogenic potential by P12, after which they generate only glia. Tagged cells in the cerebellar white matter (WM) were initially GFAP/S100beta+ and expressed the NSC/NPCs proteins LeX, Musashi1, and Sox2 in vivo. One week after tagging, reporter+ cells in the WM upregulated the neuronal progenitor markers Mash1, Pax2, and Gad-67. These Pax2+ progenitors migrated throughout the cerebellar cortex, populating the ML and leaving the WM by P18. These data suggest that a pool of GFAP/S100beta+ glial cells located in the cerebellar WM generate a large fraction of cerebellar interneurons for the ML within the first postnatal 12 days of cerebellar development. This restricted critical period implies that powerful inhibitory factors may restrict their fate potential in vivo at later stages of development.


Subject(s)
Cerebellum/metabolism , Glial Fibrillary Acidic Protein/genetics , Interneurons/metabolism , Promoter Regions, Genetic/genetics , Stem Cells/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Biomarkers/metabolism , Cell Lineage/drug effects , Cerebellum/cytology , Gene Expression Regulation/drug effects , Genes, Reporter , Integrases/metabolism , Interneurons/cytology , Interneurons/drug effects , Mice , Mice, Inbred C57BL , Recombination, Genetic/genetics , Stem Cells/cytology , Stem Cells/drug effects , Tamoxifen/pharmacology , Time Factors
17.
Pediatrics ; 123(2): 503-11, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19171615

ABSTRACT

OBJECTIVE: Although preterm very low birth weight infants have a high prevalence of neuroanatomical abnormalities when evaluated at term-equivalent age, patterns of brain growth in prematurely born infants during school age and adolescence remain largely unknown. Our goal was to test the hypothesis that preterm birth results in long-term dynamic changes in the developing brain. METHODS: We performed serial volumetric MRI studies at ages 8 and 12 years in 55 preterm infants born weighing 600 to 1250 g and 20 term control children who participated in the follow-up component of a prospective, randomized, placebo-controlled intraventricular hemorrhage prevention study. RESULTS: Total brain volumes increased 2% to 3% between the ages of 8 and 12 years for both preterm and term children. These changes involved reductions in cerebral gray matter while white matter increased. Between 8 and 12 years of age, preterm subjects experienced a 2% decrease in left cerebral gray matter compared with a 10% reduction in left cerebral gray for term controls. For right cerebral gray matter, preterm children experienced a 3% decrease in volume between years 8 and 12, compared with 9% for term controls (group-by-time). In contrast, preterm subjects had a 10% increase in cerebral white matter volumes bilaterally between ages 8 and 12 years, compared with >26% increases for both hemispheres for term controls. Significant differences in regional volume changes between study groups were found in bilateral temporal gray and in parietal white matter. CONCLUSIONS: Preterm birth continues to perturb the trajectory of cerebral development during late childhood and early adolescence with preterm children, showing both lower gray matter reduction and less white matter gain over time compared with term control subjects.


Subject(s)
Brain/growth & development , Infant, Premature/growth & development , Magnetic Resonance Imaging , Adolescent , Age Factors , Child , Female , Humans , Infant, Newborn , Male , Organ Size , Term Birth
18.
J Neurosci ; 29(4): 1202-11, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19176828

ABSTRACT

Chronic postnatal hypoxia causes an apparent loss of cortical neurons that is reversed during recovery (Fagel et al., 2006). The cellular and molecular mechanisms underlying this plasticity are not understood. Here, we show that chronic hypoxia from postnatal days 3 (P3) to 10 causes a 30% decrease in cortical neurons and a 24% decrease in cortical volume. T-brain-1 (Tbr1)(+) and SMI-32(+) excitatory neuron numbers were completely recovered 1 month after the insult, but the mice showed a residual deficit in Parvalbumin(+) and Calretinin(+) GABAergic interneurons. In contrast, hypoxic mice carrying a disrupted fibroblast growth factor receptor-1 (Fgfr1) gene in GFAP+ cells [Fgfr1 conditional knock-out (cKO)], demonstrated a persistent loss of excitatory cortical neurons and a worsening of the interneuron defect. Labeling proliferating progenitors at P17 revealed increased generation of cortical NeuN(+) and Tbr1(+) excitatory neurons in wild-type mice subjected to hypoxic insult, whereas Fgfr1 cKO failed to mount a cortical neurogenetic response. Hypoxic wild-type mice also demonstrated a twofold increase in cell proliferation in the subventricular zone (SVZ) at P17 and a threefold increase in neurogenesis in the olfactory bulb (OB) at P48, compared with normoxic mice. In contrast, Fgfr1 cKO mice had decreased SVZ cell proliferation and curtailed reactive neurogenesis in the OB. Thus, the activation of FGFR-1 in GFAP+ cells is required for neuronal recovery after neonatal hypoxic injury, which is attributable in part to enhanced cortical and OB neurogenesis. In contrast, there is incomplete recovery of inhibitory neurons after injury, which may account for persistent behavioral deficits.


Subject(s)
Cerebral Cortex/pathology , Hypoxia/pathology , Nerve Regeneration/physiology , Neurons/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Proliferation , Cerebral Cortex/physiopathology , Creatinine/metabolism , DNA-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein/genetics , Hypoxia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/physiology , Olfactory Bulb , Parvalbumins/metabolism , Phosphopyruvate Hydratase/metabolism , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/metabolism , T-Box Domain Proteins
19.
J Pediatr ; 152(4): 513-20, 520.e1, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18346506

ABSTRACT

OBJECTIVES: To more precisely examine regional and subregional microstructural brain changes associated with preterm birth. STUDY DESIGN: We obtained brain volumes from 29 preterm children, age 12 years, with no ultrasound scanning evidence of intraventricular hemorrhage or cystic periventricular leukomalacia in the newborn period, and 22 age- and sex-matched term control subjects. RESULTS: Preterm male subjects demonstrated significantly lower white matter volumes in bilateral cingulum, corpus callosum, corticospinal tract, prefrontal cortex, superior and inferior longitudinal fasciculi compared with term male subjects. Gray matter volumes in prefrontal cortex, basal ganglia, and temporal lobe also were significantly reduced in preterm male subjects. Brain volumes of preterm female subjects were not significantly different from those of term female control subjects. Voxel-based morphometry results were not correlated with perinatal variables or cognitive outcome. Higher maternal education was associated with higher cognitive performance in preterm male subjects. CONCLUSIONS: Preterm male children continue to demonstrate abnormal neurodevelopment at 12 years of age. However, brain morphology in preterm female children may no longer differ from that of term female children. The neurodevelopmental abnormalities we detected in preterm male subjects appear to be relatively diffuse, involving multiple neural systems. The relationship between aberrant neurodevelopment and perinatal variables may be mediated by genetic factors, environmental factors, or both reflected in maternal education level.


Subject(s)
Brain/anatomy & histology , Infant, Premature , Intelligence , Birth Weight , Brain/growth & development , Case-Control Studies , Cerebral Hemorrhage/prevention & control , Child , Cognition Disorders , Female , Follow-Up Studies , Gestational Age , Humans , Indomethacin/therapeutic use , Infant, Newborn , Magnetic Resonance Imaging , Male , Organ Size , Regression Analysis , Sex Factors
20.
Proc Natl Acad Sci U S A ; 104(51): 20558-63, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18077357

ABSTRACT

The lifelong addition of neurons to the hippocampus is a remarkable form of structural plasticity, yet the molecular controls over proliferation, neuronal fate determination, survival, and maturation are poorly understood. Expression of Notch1 was found to change dynamically depending on the differentiation state of neural precursor cells. Through the use of inducible gain- and loss-of-function of Notch1 mice we show that this membrane receptor is essential to these distinct processes. We found in vivo that activated Notch1 overexpression induces proliferation, whereas gamma-secretase inhibition or genetic ablation of Notch1 promotes cell cycle exit, indicating that the level of activated Notch1 regulates the magnitude of neurogenesis from postnatal progenitor cells. Abrogation of Notch signaling in vivo or in vitro leads to a transition from neural stem or precursor cells to transit-amplifying cells or neurons. Further, genetic Notch1 manipulation modulates survival and dendritic morphology of newborn granule cells. These results provide evidence for the expansive prevalence of Notch signaling in hippocampal morphogenesis and plasticity, suggesting that Notch1 could be a target of diverse traumatic and environmental modulators of adult neurogenesis.


Subject(s)
Dendrites/ultrastructure , Dentate Gyrus/growth & development , Neurons/ultrastructure , Receptor, Notch1/physiology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Count , Cell Proliferation , Dentate Gyrus/cytology , Green Fluorescent Proteins , Hippocampus/growth & development , Mice , Mice, Knockout , Neuronal Plasticity , Neurons/physiology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL