Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Glia ; 71(11): 2664-2678, 2023 11.
Article in English | MEDLINE | ID: mdl-37519044

ABSTRACT

Mutations leading to colony-stimulating factor-1 receptor (CSF-1R) loss-of-function or haploinsufficiency cause CSF1R-related leukoencephalopathy (CRL), an adult-onset disease characterized by loss of myelin and neurodegeneration, for which there is no effective therapy. Symptom onset usually occurs in the fourth decade of life and the penetrance of disease in carriers is high. However, familial studies have identified a few carriers of pathogenic CSF1R mutations that remain asymptomatic even in their seventh decade of life, raising the possibility that the development and severity of disease might be influenced by environmental factors. Here we report new cases in which long-term glucocorticoid treatment is associated with asymptomatic status in elder carriers of pathogenic CSF-1R mutations. The main objective of the present study was to investigate the link between chronic immunosuppression initiated pre-symptomatically and resistance to the development of symptomatic CRL, in the Csf1r+/- mouse model. We show that chronic prednisone administration prevents the development of memory, motor coordination and social interaction deficits, as well as the demyelination, neurodegeneration and microgliosis associated with these deficits. These findings are in agreement with the preliminary clinical observations and support the concept that pre-symptomatic immunosuppression is protective in patients carrying pathogenic CSF1R variants associated with CRL. Proteomic analysis of microglia and oligodendrocytes indicates that prednisone suppresses processes involved in microglial activation and alleviates senescence and improves fitness of oligodendrocytes. This analysis also identifies new potential targets for therapeutic intervention.


Subject(s)
Leukoencephalopathies , Receptor, Macrophage Colony-Stimulating Factor , Mice , Animals , Prednisone/pharmacology , Proteomics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Leukoencephalopathies/genetics , Leukoencephalopathies/prevention & control , Microglia , Receptor Protein-Tyrosine Kinases , Immunosuppression Therapy
2.
Glia ; 69(3): 779-791, 2021 03.
Article in English | MEDLINE | ID: mdl-33079443

ABSTRACT

Adult onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is a dementia resulting from dominantly inherited CSF1R inactivating mutations. The Csf1r+/- mouse mimics ALSP symptoms and pathology. Csf1r is mainly expressed in microglia, but also in cortical layer V neurons that are gradually lost in Csf1r+/- mice with age. We therefore examined whether microglial or neuronal Csf1r loss caused neurodegeneration in Csf1r+/- mice. The behavioral deficits, pathologies and elevation of Csf2 expression contributing to disease, previously described in the Csf1r+/- ALSP mouse, were reproduced by microglial deletion (MCsf1rhet mice), but not by neural deletion. Furthermore, increased Csf2 expression by callosal astrocytes, oligodendrocytes, and microglia was observed in Csf1r+/- mice and, in MCsf1rhet mice, the densities of these three cell types were increased in supraventricular patches displaying activated microglia, an early site of disease pathology. These data confirm that ALSP is a primary microgliopathy and inform future therapeutic and experimental approaches.


Subject(s)
Demyelinating Diseases , Leukoencephalopathies , Neurodegenerative Diseases , Animals , Leukoencephalopathies/genetics , Mice , Microglia , Neuroglia , Receptor Protein-Tyrosine Kinases , Receptors, Colony-Stimulating Factor , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
3.
J Neurosci ; 39(10): 1892-1909, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30626701

ABSTRACT

Emerging studies are providing compelling evidence that the pathogenesis of Huntington's disease (HD), a neurodegenerative disorder with frequent midlife onset, encompasses developmental components. Moreover, our previous studies using a hypomorphic model targeting huntingtin during the neurodevelopmental period indicated that loss-of-function mechanisms account for this pathogenic developmental component (Arteaga-Bracho et al., 2016). In the present study, we specifically ascertained the roles of subpallial lineage species in eliciting the previously observed HD-like phenotypes. Accordingly, we used the Cre-loxP system to conditionally ablate the murine huntingtin gene (Httflx) in cells expressing the subpallial patterning markers Gsx2 (Gsx2-Cre) or Nkx2.1 (Nkx2.1-Cre) in Httflx mice of both sexes. These genetic manipulations elicited anxiety-like behaviors, hyperkinetic locomotion, age-dependent motor deficits, and weight loss in both Httflx;Gsx2-Cre and Httflx;Nkx2.1-Cre mice. In addition, these strains displayed unique but complementary spatial patterns of basal ganglia degeneration that are strikingly reminiscent of those seen in human cases of HD. Furthermore, we observed early deficits of somatostatin-positive and Reelin-positive interneurons in both Htt subpallial null strains, as well as early increases of cholinergic interneurons, Foxp2+ arkypallidal neurons, and incipient deficits with age-dependent loss of parvalbumin-positive neurons in Httflx;Nkx2.1-Cre mice. Overall, our findings indicate that selective loss-of-huntingtin function in subpallial lineages differentially disrupts the number, complement, and survival of forebrain interneurons and globus pallidus GABAergic neurons, thereby leading to the development of key neurological hallmarks of HD during adult life. Our findings have important implications for the establishment and deployment of neural circuitries and the integrity of network reserve in health and disease.SIGNIFICANCE STATEMENT Huntington's disease (HD) is a progressive degenerative disorder caused by aberrant trinucleotide expansion in the huntingtin gene. Mechanistically, this mutation involves both loss- and gain-of-function mechanisms affecting a broad array of cellular and molecular processes. Although huntingtin is widely expressed during adult life, the mutant protein only causes the demise of selective neuronal subtypes. The mechanisms accounting for this differential vulnerability remain elusive. In this study, we have demonstrated that loss-of-huntingtin function in subpallial lineages not only differentially disrupts distinct interneuron species early in life, but also leads to a pattern of neurological deficits that are reminiscent of HD. This work suggests that early disruption of selective neuronal subtypes may account for the profiles of enhanced regional cellular vulnerability to death in HD.


Subject(s)
Brain/growth & development , Huntingtin Protein/physiology , Huntington Disease/physiopathology , Interneurons/physiology , Neurons/physiology , Animals , Anxiety/physiopathology , Behavior, Animal , Brain/pathology , Corpus Striatum/growth & development , Corpus Striatum/pathology , Female , Globus Pallidus/growth & development , Globus Pallidus/pathology , Huntingtin Protein/genetics , Huntington Disease/pathology , Huntington Disease/psychology , Interneurons/ultrastructure , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Cortex/growth & development , Motor Cortex/pathology , Neurons/ultrastructure , Prosencephalon/growth & development , Prosencephalon/pathology , Reelin Protein
4.
Proc Natl Acad Sci U S A ; 113(20): 5736-41, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27140644

ABSTRACT

Recent studies have identified impairments in neural induction and in striatal and cortical neurogenesis in Huntington's disease (HD) knock-in mouse models and associated embryonic stem cell lines. However, the potential role of these developmental alterations for HD pathogenesis and progression is currently unknown. To address this issue, we used BACHD:CAG-Cre(ERT2) mice, which carry mutant huntingtin (mHtt) modified to harbor a floxed exon 1 containing the pathogenic polyglutamine expansion (Q97). Upon tamoxifen administration at postnatal day 21, the floxed mHtt-exon1 was removed and mHtt expression was terminated (Q97(CRE)). These conditional mice displayed similar profiles of impairments to those mice expressing mHtt throughout life: (i) striatal neurodegeneration, (ii) early vulnerability to NMDA-mediated excitotoxicity, (iii) impairments in motor coordination, (iv) temporally distinct abnormalities in striatal electrophysiological activity, and (v) altered corticostriatal functional connectivity and plasticity. These findings strongly suggest that developmental aberrations may play important roles in HD pathogenesis and progression.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Action Potentials , Animals , Apoptosis , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Female , GABAergic Neurons/physiology , Gene Expression , Gene Expression Regulation, Developmental , Humans , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle Strength , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Organ Specificity , Rotarod Performance Test
5.
Neurobiol Dis ; 96: 144-155, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27623015

ABSTRACT

The mutation in huntingtin (mHtt) leads to a spectrum of impairments in the developing forebrain of Huntington's disease (HD) mouse models. Whether these developmental alterations are due to loss- or gain-of-function mechanisms and contribute to HD pathogenesis is unknown. We examined the role of selective loss of huntingtin (Htt) function during development on postnatal vulnerability to cell death. We employed mice expressing very low levels of Htt throughout embryonic life to postnatal day 21 (Hdhd•hyp). We demonstrated that Hdhd•hyp mice exhibit: (1) late-life striatal and cortical neuronal degeneration; (2) neurological and skeletal muscle alterations; and (3) white matter tract impairments and axonal degeneration. Hdhd•hyp embryos also exhibited subpallial heterotopias, aberrant striatal maturation and deregulation of gliogenesis. These results indicate that developmental deficits associated with Htt functions render cells present at discrete neural foci increasingly susceptible to cell death, thus implying the potential existence of a loss-of-function developmental component to HD pathogenesis.


Subject(s)
Developmental Disabilities/genetics , Huntingtin Protein/deficiency , Huntington Disease/complications , Huntington Disease/genetics , Mutation/genetics , Neurodegenerative Diseases/etiology , Age Factors , Animals , Animals, Newborn , Cell Differentiation/genetics , Developmental Disabilities/complications , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Huntingtin Protein/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/complications , Psychomotor Disorders/etiology , Psychomotor Disorders/genetics , RNA, Messenger/metabolism , White Matter/pathology
6.
Neurobiol Dis ; 74: 219-28, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25497733

ABSTRACT

Mutations in the colony stimulating factor-1 receptor (CSF1R) that abrogate the expression of the affected allele or lead to the expression of mutant receptor chains devoid of kinase activity have been identified in both familial and sporadic cases of ALSP. To determine the validity of the Csf1r heterozygous mouse as a model of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) we performed behavioral, radiologic, histopathologic, ultrastructural and cytokine expression studies of young and old Csf1r+/- and control Csf1r+/+ mice. Six to 8-month old Csf1r+/- mice exhibit cognitive deficits, and by 9-11 months develop sensorimotor deficits and in male mice, depression and anxiety-like behavior. MRIs of one year-old Csf1r+/- mice reveal lateral ventricle enlargement and thinning of the corpus callosum. Ultrastructural analysis of the corpus callosum uncovers dysmyelinated axons as well as neurodegeneration, evidenced by the presence of axonal spheroids. Histopathological examination of 11-week-old mice reveals increased axonal and myelin staining in the cortex, increase of neuronal cell density in layer V and increase of microglial cell densities throughout the brain, suggesting that early developmental changes contribute to disease. By 10-months of age, the neuronal cell density normalizes, oligodendrocyte precursor cells increase in layers II-III and V and microglial densities remain elevated without an increase in astrocytes. Also, the age-dependent increase in CSF-1R+ neurons in cortical layer V is reduced. Moreover, the expression of Csf2, Csf3, Il27 and Il6 family cytokines is increased, consistent with microglia-mediated inflammation. These results demonstrate that the inactivation of one Csf1r allele is sufficient to cause an ALSP-like disease in mice. The Csf1r+/- mouse is a model of ALSP that will allow the critical events for disease development to be determined and permit rapid evaluation of therapeutic approaches. Furthermore, our results suggest that aberrant activation of microglia in Csf1r+/- mice may play a central role in ALSP pathology.


Subject(s)
Disease Models, Animal , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Animals , Anxiety/pathology , Anxiety/physiopathology , Brain/immunology , Brain/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Cytokines/metabolism , Depression/pathology , Depression/physiopathology , Disease Progression , Female , Leukoencephalopathies/pathology , Leukoencephalopathies/physiopathology , Leukoencephalopathies/psychology , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Neuroglia/immunology , Neuroglia/pathology , Neurons/immunology , Neurons/pathology , Olfactory Perception/physiology , Phenotype , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , White Matter/immunology , White Matter/pathology
7.
J Biol Chem ; 288(30): 21972-86, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23744080

ABSTRACT

Interleukin-34 (IL-34) is highly expressed in brain. IL-34 signaling via its cognate receptor, colony-stimulating factor-1 receptor (CSF-1R), is required for the development of microglia. However, the differential expression of IL-34 and the CSF-1R in brain suggests that IL-34 may signal via an alternate receptor. By IL-34 affinity chromatography of solubilized mouse brain membrane followed by mass spectrometric analysis, we identified receptor-type protein-tyrosine phosphatase ζ (PTP-ζ), a cell surface chondroitin sulfate (CS) proteoglycan, as a novel IL-34 receptor. PTP-ζ is primarily expressed on neural progenitors and glial cells and is highly expressed in human glioblastomas. IL-34 selectively bound PTP-ζ in CSF-1R-deficient U251 human glioblastoma cell lysates and inhibited the proliferation, clonogenicity, and motility of U251 cells in a PTP-ζ-dependent manner. These effects were correlated with an increase in tyrosine phosphorylation of the previously identified PTP-ζ downstream effectors focal adhesion kinase and paxillin. IL-34 binding to U251 cells was abrogated by chondroitinase ABC treatment, and CS competed with IL-34 for binding to the extracellular domain of PTP-ζ and to the cells, indicating a dependence of binding on PTP-ζ CS moieties. This study identifies an alternate receptor for IL-34 that may mediate its action on novel cellular targets.


Subject(s)
Interleukins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Receptors, Interleukin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Brain/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Interleukins/pharmacology , Mass Spectrometry , Mice , Microscopy, Fluorescence , Molecular Sequence Data , NIH 3T3 Cells , Paxillin/metabolism , Phosphorylation/drug effects , Protein Binding , RNA Interference , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Receptors, Interleukin/genetics , Tyrosine/metabolism
8.
Nat Commun ; 15(1): 6410, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080283

ABSTRACT

Adult neurogenesis is a unique form of neuronal plasticity in which newly generated neurons are integrated into the adult dentate gyrus in a process that is modulated by environmental stimuli. Adult-born neurons can contribute to spatial memory, but it is unknown whether they alter neural representations of space in the hippocampus. Using in vivo two-photon calcium imaging, we find that male and female mice previously housed in an enriched environment, which triggers an increase in neurogenesis, have increased spatial information encoding in the dentate gyrus. Ablating adult neurogenesis blocks the effect of enrichment and lowers spatial information, as does the chemogenetic silencing of adult-born neurons. Both ablating neurogenesis and silencing adult-born neurons decreases the calcium activity of dentate gyrus neurons, resulting in a decreased amplitude of place-specific responses. These findings are in contrast with previous studies that suggested a predominantly inhibitory action for adult-born neurons. We propose that adult neurogenesis improves representations of space by increasing the gain of dentate gyrus neurons and thereby improving their ability to tune to spatial features. This mechanism may mediate the beneficial effects of environmental enrichment on spatial learning and memory.


Subject(s)
Dentate Gyrus , Hippocampus , Neurogenesis , Neurons , Spatial Memory , Animals , Neurogenesis/physiology , Male , Female , Dentate Gyrus/physiology , Dentate Gyrus/cytology , Mice , Neurons/physiology , Neurons/metabolism , Hippocampus/physiology , Hippocampus/cytology , Hippocampus/metabolism , Spatial Memory/physiology , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Calcium/metabolism , Spatial Learning/physiology
9.
Dev Biol ; 367(2): 100-13, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22542597

ABSTRACT

The CSF-1 receptor (CSF-1R) regulates CNS microglial development. However, the localization and developmental roles of this receptor and its ligands, IL-34 and CSF-1, in the brain are poorly understood. Here we show that compared to wild type mice, CSF-1R-deficient (Csf1r-/-) mice have smaller brains of greater mass. They further exhibit an expansion of lateral ventricle size, an atrophy of the olfactory bulb and a failure of midline crossing of callosal axons. In brain, IL-34 exhibited a broader regional expression than CSF-1, mostly without overlap. Expression of IL-34, CSF-1 and the CSF-1R were maximal during early postnatal development. However, in contrast to the expression of its ligands, CSF-1R expression was very low in adult brain. Postnatal neocortical expression showed that CSF-1 was expressed in layer VI, whereas IL-34 was expressed in the meninges and layers II-V. The broader expression of IL-34 is consistent with its previously implicated role in microglial development. The differential expression of CSF-1R ligands, with respect to CSF-1R expression, could reflect their CSF-1R-independent signaling. Csf1r-/- mice displayed increased proliferation and apoptosis of neocortical progenitors and reduced differentiation of specific excitatory neuronal subtypes. Indeed, addition of CSF-1 or IL-34 to microglia-free, CSF-1R-expressing dorsal forebrain clonal cultures, suppressed progenitor self-renewal and enhanced neuronal differentiation. Consistent with a neural developmental role for the CSF-1R, ablation of the Csf1r gene in Nestin-positive neural progenitors led to a smaller brain size, an expanded neural progenitor pool and elevated cellular apoptosis in cortical forebrain. Thus our results also indicate novel roles for the CSF-1R in the regulation of corticogenesis.


Subject(s)
Brain/growth & development , Brain/metabolism , Interleukins/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Neural Stem Cells/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Apoptosis , Base Sequence , Brain/abnormalities , Brain/cytology , Cell Differentiation , Cell Proliferation , Cell Survival , DNA Primers/genetics , Gene Expression Regulation, Developmental , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/cytology , Receptor, Macrophage Colony-Stimulating Factor/deficiency , Receptor, Macrophage Colony-Stimulating Factor/genetics , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 107(38): 16685-90, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20823235

ABSTRACT

The repressor element-1 (RE1) silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) silences neuronal genes in neural stem cells (NSCs) and nonneuronal cells through its role as a dynamic modular platform for recruitment of transcriptional and epigenetic regulatory cofactors to RE1-containing promoters. In embryonic stem cells, the REST regulatory network is highly integrated with the transcriptional circuitry governing self-renewal and pluripotency, although its exact functional role is unclear. The C-terminal cofactor for REST, CoREST, also acts as a modular scaffold, but its cell type-specific roles have not been elucidated. We used chromatin immunoprecipitation-on-chip to examine CoREST and REST binding sites in NSCs and their proximate progenitor species. In NSCs, we identified a larger number of CoREST (1,820) compared with REST (322) target genes. The majority of these CoREST targets do not contain known RE1 motifs. Notably, these CoREST target genes do play important roles in pluripotency networks, in modulating NSC identity and fate decisions and in epigenetic processes previously associated with both REST and CoREST. Moreover, we found that NSC-mediated developmental transitions were associated primarily with liberation of CoREST from promoters with transcriptional repression favored in less lineage-restricted radial glia and transcriptional activation favored in more lineage-restricted neuronal-oligodendrocyte precursors. Clonal NSC REST and CoREST gene manipulation paradigms further revealed that CoREST has largely independent and previously uncharacterized roles in promoting NSC multilineage potential and modulating early neural fate decisions.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Binding Sites , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Co-Repressor Proteins , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors
11.
Front Cell Neurosci ; 17: 1275935, 2023.
Article in English | MEDLINE | ID: mdl-37964794

ABSTRACT

Recent studies have emphasized the role of microglia in the progression of many neurodegenerative diseases. The colony stimulating factors, CSF-1 (M-CSF), granulocyte-macrophage CSF (GM-CSF) and granulocyte CSF (G-CSF) regulate microglia through different cognate receptors. While the receptors for GM-CSF (GM-CSFR) and G-CSF (G-CSFR) are specific for their ligands, CSF-1 shares its receptor, the CSF-1 receptor-tyrosine kinase (CSF-1R), with interleukin-34 (IL-34). All four cytokines are expressed locally in the CNS. Activation of the CSF-1R in macrophages is anti-inflammatory. In contrast, the actions of GM-CSF and G-CSF elicit different activated states. We here review the roles of each of these cytokines in the CNS and how they contribute to the development of disease in a mouse model of CSF-1R-related leukodystrophy. Understanding their roles in this model may illuminate their contribution to the development or exacerbation of other neurodegenerative diseases.

12.
Biomedicines ; 11(8)2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37626591

ABSTRACT

Colony-stimulating factor-1 receptor (CSF-1R)-related leukoencephalopathy (CRL) is a neurodegenerative disease that triggers early demyelination, leading to an adult-onset dementia. Triggering receptor expressed on myeloid cells-2 (TREM2) is a microglial receptor that promotes the activation of microglia and phagocytic clearance of apoptotic neurons and myelin debris. We investigated the role of Trem2 in the demyelination observed in the Csf1r+/- mouse model of CRL. We show that elevation of Trem2 expression and callosal demyelination occur in 4-5-month-old Csf1r+/- mice, prior to the development of symptoms. Absence of Trem2 in the Csf1r+/- mouse attenuated myelin pathology and normalized microglial densities and morphology in the corpus callosum. Trem2 absence also prevented axonal degeneration and the loss of cortical layer V neurons observed in Csf1r+/- mice. Furthermore, the absence of Trem2 prevented the accumulation of myelin-derived lipids in Csf1r+/- macrophages and reduced the production of TNF-α after myelin engulfment. These data suggest that TREM2 contributes to microglial dyshomeostasis in CRL.

13.
Proc Natl Acad Sci U S A ; 106(51): 21900-5, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19955426

ABSTRACT

The pathogenesis of Huntington's disease (HD) remains elusive. The identification of increasingly early pathophysiological abnormalities in HD suggests the possibility that impairments of striatal medium spiny neuron (MSN) specification and maturation may underlie the etiology of HD. In fact, we demonstrate that HD knock-in (Hdh-Q111) mice exhibited delayed acquisition of early striatal cytoarchitecture with aberrant expression of progressive markers of MSN neurogenesis (Islet1, DARPP-32, mGluR1, and NeuN). Hdh-Q111 striatal progenitors also displayed delayed cell cycle exit between E13.5-15.5 (BrdU birth-dating) and an enhanced fraction of abnormal cycling cells in association with expansion of the pool of intermediate progenitors and over expression of the core pluripotency (PP) factor, Sox2. Clonal analysis further revealed that Hdh-Q111 neural stem cells (NSCs) displayed: impaired lineage restriction, reduced proliferative potential, enhanced late-stage self-renewal, and deregulated MSN subtype specification. Further, our analysis revealed that in addition to Sox2, the core PP factor, Nanog is expressed within the striatal generative and mantle regions, and in Hdh-Q111 embryos the fraction of Nanog-expressing MSN precursors was substantially increased. Moreover, compared to Hdh-Q18 embryos, the Hdh-Q111 striatal anlagen exhibited significantly higher levels of the essential PP cofactor, Stat3. These findings suggest that Sox2 and Nanog may play roles during a selective window of embryonic brain maturation, and alterations of these factors may, in part, be responsible for mediating the aberrant program of Hdh-Q111 striatal MSN specification and maturation. We propose that these HD-associated developmental abnormalities might compromise neuronal homeostasis and subsequently render MSNs more vulnerable to late life stressors.


Subject(s)
Corpus Striatum/pathology , Disease Models, Animal , Huntington Disease/pathology , Stem Cells/cytology , Animals , Mice , Mice, Transgenic , Microscopy, Fluorescence
14.
FEBS J ; 289(17): 5049-5073, 2022 09.
Article in English | MEDLINE | ID: mdl-34145972

ABSTRACT

The role of colony-stimulating factor-1 receptor (CSF-1R) in macrophage and organismal development has been extensively studied in mouse. Within the last decade, mutations in the CSF1R have been shown to cause rare diseases of both pediatric (Brain Abnormalities, Neurodegeneration, and Dysosteosclerosis, OMIM #618476) and adult (CSF1R-related leukoencephalopathy, OMIM #221820) onset. Here we review the genetics, penetrance, and histopathological features of these diseases and discuss to what extent the animal models of Csf1r deficiency currently available provide systems in which to study the underlying mechanisms involved.


Subject(s)
Leukoencephalopathies , Osteosclerosis , Animals , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Mice , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics
15.
BMC Neurosci ; 11: 14, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20137068

ABSTRACT

BACKGROUND: Long non-protein-coding RNAs (ncRNAs) are emerging as important regulators of cellular differentiation and are widely expressed in the brain. RESULTS: Here we show that many long ncRNAs exhibit dynamic expression patterns during neuronal and oligodendrocyte (OL) lineage specification, neuronal-glial fate transitions, and progressive stages of OL lineage elaboration including myelination. Consideration of the genomic context of these dynamically regulated ncRNAs showed they were part of complex transcriptional loci that encompass key neural developmental protein-coding genes, with which they exhibit concordant expression profiles as indicated by both microarray and in situ hybridization analyses. These included ncRNAs associated with differentiation-specific nuclear subdomains such as Gomafu and Neat1, and ncRNAs associated with developmental enhancers and genes encoding important transcription factors and homeotic proteins. We also observed changes in ncRNA expression profiles in response to treatment with trichostatin A, a histone deacetylase inhibitor that prevents the progression of OL progenitors into post-mitotic OLs by altering lineage-specific gene expression programs. CONCLUSION: This is the first report of long ncRNA expression in neuronal and glial cell differentiation and of the modulation of ncRNA expression by modification of chromatin architecture. These observations explicitly link ncRNA dynamics to neural stem cell fate decisions, specification and epigenetic reprogramming and may have important implications for understanding and treating neuropsychiatric diseases.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Neuroglia/physiology , Neurons/physiology , Oligodendroglia/physiology , RNA, Untranslated/metabolism , Animals , Brain/cytology , Brain/drug effects , Brain/physiology , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Chromatin/drug effects , Chromatin/physiology , Gene Expression/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Mice , Neurogenesis/genetics , Neurogenesis/physiology , Neuroglia/cytology , Neuroglia/drug effects , Neurons/cytology , Oligodendroglia/cytology , Oligodendroglia/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology , gamma-Aminobutyric Acid/metabolism
16.
Cell Rep ; 30(9): 3004-3019.e5, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130903

ABSTRACT

CSF-1R haploinsufficiency causes adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). Previous studies in the Csf1r+/- mouse model of ALSP hypothesized a central role of elevated cerebral Csf2 expression. Here, we show that monoallelic deletion of Csf2 rescues most behavioral deficits and histopathological changes in Csf1r+/- mice by preventing microgliosis and eliminating most microglial transcriptomic alterations, including those indicative of oxidative stress and demyelination. We also show elevation of Csf2 transcripts and of several CSF-2 downstream targets in the brains of ALSP patients, demonstrating that the mechanisms identified in the mouse model are functional in humans. Our data provide insights into the mechanisms underlying ALSP. Because increased CSF2 levels and decreased microglial Csf1r expression have also been reported in Alzheimer's disease and multiple sclerosis, we suggest that the unbalanced CSF-1R/CSF-2 signaling we describe in the present study may contribute to the pathogenesis of other neurodegenerative conditions.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Microglia/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Alleles , Animals , Anti-Inflammatory Agents/metabolism , Antioxidants/metabolism , Atrophy , Depression/prevention & control , Female , Gene Deletion , Gene Expression Regulation , Gliosis/pathology , Heterozygote , Homeostasis , Humans , Leukocytes/pathology , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Leukoencephalopathies/physiopathology , Mice, Inbred C57BL , Microglia/pathology , Motor Activity , Myelin Sheath/pathology , Olfactory Bulb/pathology , Olfactory Bulb/physiopathology , Oxidative Stress , Phenotype , Receptor, Macrophage Colony-Stimulating Factor/deficiency , Spatial Memory , Transcriptome/genetics , White Matter/pathology , White Matter/physiopathology
17.
J Neurosci ; 25(36): 8311-21, 2005 Sep 07.
Article in English | MEDLINE | ID: mdl-16148239

ABSTRACT

Recent studies suggest that specific neural basic helix-loop-helix (HLH; i.e., Olig1 and Olig2, Mash1), associated inhibitory HLH (i.e., Id2 and Id4), high-mobility group domain (i.e., Sox10), and homeodomain (i.e., Nkx2.2) transcription factors are involved in oligodendrocyte (OL) lineage specification and progressive stages of maturation including myelination. However, the developmental interplay among these lineage-selective determinants, in a cell- and maturational stage-specific context, has not yet been defined. We show here in vivo and in vitro developmental expression profiles for these distinct classes of transcriptional regulators of OLs. We show that progressive stages of OL lineage maturation are characterized by dynamic changes in the subcellular distribution of these transcription factors and by different permutations of combinatorial transcriptional codes. Transient transfections of these precise combinatorial codes with a luciferase reporter gene driven by the myelin basic protein promoter define how changes in the molecular composition of these transcriptional complexes modulate myelin gene expression. Our overall findings suggest that the dynamic interplay between developmental stage-specific classes of transcriptional activators and associated inhibitory factors orchestrate myelin gene expression during terminal maturation of the mammalian CNS.


Subject(s)
Gene Expression Regulation , Myelin Basic Protein/genetics , Oligodendroglia/physiology , Prosencephalon/physiology , Trans-Activators/metabolism , Transcription, Genetic , 3T3 Cells , Animals , Animals, Newborn , Culture Media , Gene Expression Regulation/drug effects , Genes, Reporter , Homeobox Protein Nkx-2.2 , Luciferases/genetics , Mice , Neuroglia/physiology , Neurons/physiology , Oligodendroglia/drug effects , Platelet-Derived Growth Factor/pharmacology
18.
Trends Neurosci ; 39(6): 378-393, 2016 06.
Article in English | MEDLINE | ID: mdl-27083478

ABSTRACT

The colony-stimulating factor-1 receptor (CSF-1R) kinase regulates tissue macrophage homeostasis, osteoclastogenesis, and Paneth cell development. However, recent studies in mice have revealed that CSF-1R signaling directly controls the development and maintenance of microglia, and cell autonomously regulates neuronal differentiation and survival. While the CSF-1R-cognate ligands, CSF-1 and interleukin-34 (IL-34) compete for binding to the CSF-1R, they are expressed in a largely non-overlapping manner by mature neurons. The recent identification of a dominantly inherited, adult-onset, progressive dementia associated with inactivating mutations in the CSF-1R highlights the importance of CSF-1R signaling in the brain. We review the roles of the CSF-1R and its ligands in microglial and neural development and function, and their relevance to our understanding of neurodegenerative disease.


Subject(s)
Brain/metabolism , Ligands , Macrophage Colony-Stimulating Factor/metabolism , Microglia/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Humans , Phosphorylation
19.
Br J Radiol ; 89(1057): 20150762, 2016.
Article in English | MEDLINE | ID: mdl-26514377

ABSTRACT

We reviewed the literature for studies pertaining to impaired adult neurogenesis leading to neurocognitive impairment following cranial irradiation in rodent models. This compendium was compared with respect to radiation dose, converted to equivalent dose in 2 Gy fractions (EQD2) to allow for direct comparison between studies. The effects of differences between animal species and the dependence on animal age as well as for time after irradiation were also considered. One of the major sites of de novo adult neurogenesis is the hippocampus, and as such, this review also focuses on assessing evidence related to the expression and potential effects of inflammatory cytokines on neural stem cells in the subgranular zone of the dentate gyrus and whether this correlates with neurocognitive impairment. This review also discusses potential strategies to mitigate the detrimental effects on neurogenesis and neurocognition resulting from cranial irradiation, and how the rationale for these strategies compares with the current outcome of pre-clinical studies.


Subject(s)
Behavior, Animal/radiation effects , Cognition Disorders/etiology , Cranial Irradiation/adverse effects , Hippocampus/radiation effects , Radiation Injuries, Experimental/complications , Age Factors , Animals , Neurogenesis/radiation effects , Radiation Dosage , Rats
20.
Sci Rep ; 5: 14384, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26399509

ABSTRACT

Cancer patients undergoing cranial irradiation are at risk of developing neurocognitive impairments. Recent evidence suggests that radiation-induced injury to the hippocampi could play an important role in this cognitive decline. As a tool for studying the mechanisms of hippocampal-dependent cognitive decline, we developed a mouse model replicating the results of the recent clinical RTOG 0933 study of hippocampal sparing whole-brain irradiation. We irradiated 16-week-old female C57BL/6J mice to a single dose of 10 Gy using either whole-brain irradiation (WBRT) or hippocampal sparing irradiation (HSI). These animals, as well as sham-irradiated controls, were subjected to behavioral/cognitive assessments distinguishing between hippocampal-dependent and hippocampal-independent functions. Irradiation was well tolerated by all animals and only limited cell death of proliferating cells was found within the generative zones. Animals exposed to WBRT showed significant deficits compared to sham-irradiated controls in the hippocampal-dependent behavioral task. In contrast, HSI mice did not perform significantly different from sham-irradiated mice (control group) and performed significantly better when compared to WBRT mice. This is consistent with the results from the RTOG 0933 clinical trial, and as such this animal model could prove a helpful tool for exploring new strategies for mitigating cognitive decline in cancer patients receiving cranial irradiation.


Subject(s)
Cranial Irradiation , Hippocampus , Organ Sparing Treatments , Animals , Behavior, Animal/radiation effects , Cell Proliferation/radiation effects , Cognition Disorders/diagnosis , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Cranial Irradiation/adverse effects , Cranial Irradiation/methods , Female , Humans , Mice , Models, Animal , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Neurogenesis/radiation effects , Radiation Dosage , Spatial Memory/radiation effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL