Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(6): 114329, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38850535

ABSTRACT

Many autism spectrum disorder (ASD)-associated genes act as transcriptional regulators (TRs). Chromatin immunoprecipitation sequencing (ChIP-seq) was used to identify the regulatory targets of ARID1B, BCL11A, FOXP1, TBR1, and TCF7L2, ASD-associated TRs in the developing human and mouse cortex. These TRs shared substantial overlap in the binding sites, especially within open chromatin. The overlap within a promoter region, 1-2,000 bp upstream of the transcription start site, was highly predictive of brain-expressed genes. This signature was observed in 96 out of 102 ASD-associated genes. In vitro CRISPRi against ARID1B and TBR1 delineated downstream convergent biology in mouse cortical cultures. After 8 days, NeuN+ and CALB+ cells were decreased, GFAP+ cells were increased, and transcriptomic signatures correlated with the postmortem brain samples from individuals with ASD. We suggest that functional convergence across five ASD-associated TRs leads to shared neurodevelopmental outcomes of haploinsufficient disruption.

2.
Proc Natl Acad Sci U S A ; 119(15): e2108760119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377797

ABSTRACT

Enhancers integrate transcription factor signaling pathways that drive cell fate specification in the developing brain. We paired enhancer labeling and single-cell RNA-sequencing (scRNA-seq) to delineate and distinguish specification of neuronal lineages in mouse medial, lateral, and caudal ganglionic eminences (MGE, LGE, and CGE) at embryonic day (E)11.5. We show that scRNA-seq clustering using transcription factors improves resolution of regional and developmental populations, and that enhancer activities identify specific and overlapping GE-derived neuronal populations. First, we mapped the activities of seven evolutionarily conserved brain enhancers at single-cell resolution in vivo, finding that the selected enhancers had diverse activities in specific progenitor and neuronal populations across the GEs. We then applied enhancer-based labeling, scRNA-seq, and analysis of in situ hybridization data to distinguish transcriptionally distinct and spatially defined subtypes of MGE-derived GABAergic and cholinergic projection neurons and interneurons. Our results map developmental origins and specification paths underlying neurogenesis in the embryonic basal ganglia and showcase the power of scRNA-seq combined with enhancer-based labeling to resolve the complex paths of neuronal specification underlying mouse brain development.


Subject(s)
Basal Ganglia , Cholinergic Neurons , Enhancer Elements, Genetic , GABAergic Neurons , Neurogenesis , Animals , Basal Ganglia/cytology , Basal Ganglia/embryology , Cell Lineage/genetics , Cholinergic Neurons/metabolism , GABAergic Neurons/metabolism , Mice , Neurogenesis/genetics , RNA-Seq , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34921112

ABSTRACT

We uncovered a transcription factor (TF) network that regulates cortical regional patterning in radial glial stem cells. Screening the expression of hundreds of TFs in the developing mouse cortex identified 38 TFs that are expressed in gradients in the ventricular zone (VZ). We tested whether their cortical expression was altered in mutant mice with known patterning defects (Emx2, Nr2f1, and Pax6), which enabled us to define a cortical regionalization TF network (CRTFN). To identify genomic programming underlying this network, we performed TF ChIP-seq and chromatin-looping conformation to identify enhancer-gene interactions. To map enhancers involved in regional patterning of cortical progenitors, we performed assays for epigenomic marks and DNA accessibility in VZ cells purified from wild-type and patterning mutant mice. This integrated approach has identified a CRTFN and VZ enhancers involved in cortical regional patterning in the mouse.


Subject(s)
Cerebral Cortex/embryology , Gene Regulatory Networks , Regulatory Elements, Transcriptional , Transcription Factors/metabolism , Animals , COUP Transcription Factor I/metabolism , Cerebral Cortex/metabolism , Epigenome , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Mice , PAX6 Transcription Factor/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Transcription Factors/genetics
4.
Cell Rep ; 37(10): 110089, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34879283

ABSTRACT

Deleterious genetic variants in POGZ, which encodes the chromatin regulator Pogo Transposable Element with ZNF Domain protein, are strongly associated with autism spectrum disorder (ASD). Although it is a high-confidence ASD risk gene, the neurodevelopmental functions of POGZ remain unclear. Here we reveal the genomic binding of POGZ in the developing forebrain at euchromatic loci and gene regulatory elements (REs). We profile chromatin accessibility and gene expression in Pogz-/- mice and show that POGZ promotes the active chromatin state and transcription of clustered synaptic genes. We further demonstrate that POGZ forms a nuclear complex and co-occupies loci with ADNP, another high-confidence ASD risk gene, and provide evidence that POGZ regulates other neurodevelopmental disorder risk genes as well. Our results reveal a neurodevelopmental function of an ASD risk gene and identify molecular targets that may elucidate its function in ASD.


Subject(s)
Autistic Disorder/enzymology , Brain/enzymology , Cell Cycle Proteins/physiology , Chromatin Assembly and Disassembly , DNA-Binding Proteins/physiology , Euchromatin/metabolism , Synapses/enzymology , Transposases/metabolism , Animals , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Binding Sites , Brain/growth & development , Cell Cycle Proteins/genetics , DNA Transposable Elements , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Euchromatin/genetics , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Promoter Regions, Genetic , Synapses/genetics , Transposases/genetics
5.
Nat Genet ; 53(4): 521-528, 2021 04.
Article in English | MEDLINE | ID: mdl-33782603

ABSTRACT

Ultraconserved enhancer sequences show perfect conservation between human and rodent genomes, suggesting that their functions are highly sensitive to mutation. However, current models of enhancer function do not sufficiently explain this extreme evolutionary constraint. We subjected 23 ultraconserved enhancers to different levels of mutagenesis, collectively introducing 1,547 mutations, and examined their activities in transgenic mouse reporter assays. Overall, we find that the regulatory properties of ultraconserved enhancers are robust to mutation. Upon mutagenesis, nearly all (19/23, 83%) still functioned as enhancers at one developmental stage, as did most of those tested again later in development (5/9, 56%). Replacement of endogenous enhancers with mutated alleles in mice corroborated results of transgenic assays, including the functional resilience of ultraconserved enhancers to mutation. Our findings show that the currently known activities of ultraconserved enhancers do not necessarily require the perfect conservation observed in evolution and suggest that additional regulatory or other functions contribute to their sequence constraint.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Mutation , Transcription Factors/genetics , Alleles , Animals , Base Sequence , Conserved Sequence , Embryo, Mammalian , Humans , Mice , Mutagenesis, Site-Directed , Rats , Transcription Factors/metabolism
6.
Cell ; 172(3): 491-499.e15, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29358049

ABSTRACT

Non-coding "ultraconserved" regions containing hundreds of consecutive bases of perfect sequence conservation across mammalian genomes can function as distant-acting enhancers. However, initial deletion studies in mice revealed that loss of such extraordinarily constrained sequences had no immediate impact on viability. Here, we show that ultraconserved enhancers are required for normal development. Focusing on some of the longest ultraconserved sites genome wide, located near the essential neuronal transcription factor Arx, we used genome editing to create an expanded series of knockout mice lacking individual or combinations of ultraconserved enhancers. Mice with single or pairwise deletions of ultraconserved enhancers were viable and fertile but in nearly all cases showed neurological or growth abnormalities, including substantial alterations of neuron populations and structural brain defects. Our results demonstrate the functional importance of ultraconserved enhancers and indicate that remarkably strong sequence conservation likely results from fitness deficits that appear subtle in a laboratory setting.


Subject(s)
Conserved Sequence , Embryonic Development/genetics , Enhancer Elements, Genetic , Animals , Brain/abnormalities , Brain/embryology , Brain/metabolism , Female , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
7.
J Comp Neurol ; 524(3): 609-29, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26304102

ABSTRACT

The development of the cortex is an elaborate process that integrates a plethora of finely tuned molecular processes ranging from carefully regulated gradients of transcription factors, dynamic changes in the chromatin landscape, or formation of protein complexes to elicit and regulate transcription. Combined with cellular processes such as cell type specification, proliferation, differentiation, and migration, all of these developmental processes result in the establishment of an adult mammalian cortex with its typical lamination and regional patterning. By examining in-depth the role of one transcription factor, Pax6, on the regulation of cortical development, its integration in the regulation of chromatin state, and its regulation by cis-regulatory elements, we aim to demonstrate the importance of integrating each level of regulation in our understanding of cortical development.


Subject(s)
Cerebral Cortex/growth & development , Epigenesis, Genetic , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Biological Evolution , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Humans , PAX6 Transcription Factor
8.
Neuron ; 88(6): 1192-1207, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671461

ABSTRACT

We demonstrate using conditional mutagenesis that Pbx1, with and without Pbx2(+/-) sensitization, regulates regional identity and laminar patterning of the developing mouse neocortex in cortical progenitors (Emx1-Cre) and in newly generated neurons (Nex1-Cre). Pbx1/2 mutants have three salient molecular phenotypes of cortical regional and laminar organization: hypoplasia of the frontal cortex, ventral expansion of the dorsomedial cortex, and ventral expansion of Reelin expression in the cortical plate of the frontal cortex, concomitant with an inversion of cortical layering in the rostral cortex. Molecular analyses, including PBX ChIP-seq, provide evidence that PBX promotes frontal cortex identity by repressing genes that promote dorsocaudal fate.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Homeodomain Proteins/physiology , Neural Stem Cells/physiology , Neurons/physiology , Transcription Factors/physiology , Animals , Animals, Newborn , Mice , Mice, Transgenic , Mitosis/physiology , Pre-B-Cell Leukemia Transcription Factor 1 , Reelin Protein , Stem Cells/physiology
9.
Nat Commun ; 6: 6887, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25904499

ABSTRACT

Guidepost cells present at and surrounding the midline provide guidance cues that orient the growing axons through commissures. Here we show that the transcription factor Nkx2.1 known to control the specification of GABAergic interneurons also regulates the differentiation of astroglia and polydendrocytes within the mouse anterior commissure (AC). Nkx2.1-positive glia were found to originate from three germinal regions of the ventral telencephalon. Nkx2.1-derived glia were observed in and around the AC region by E14.5. Thereafter, a selective cell ablation strategy showed a synergistic role of Nkx2.1-derived cells, both GABAergic interneurons and astroglia, towards the proper formation of the AC. Finally, our results reveal that the Nkx2.1-regulated cells mediate AC axon guidance through the expression of the repellent cue, Slit2. These results bring forth interesting insights about the spatial and temporal origin of midline telencephalic glia, and highlight the importance of neurons and astroglia towards the formation of midline commissures.


Subject(s)
Anterior Commissure, Brain/embryology , Astrocytes/metabolism , GABAergic Neurons/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Interneurons/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Anterior Commissure, Brain/cytology , Anterior Commissure, Brain/metabolism , Astrocytes/cytology , Axons , Cell Movement , Electroporation , Embryo, Mammalian , GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Immunohistochemistry , In Vitro Techniques , Interneurons/cytology , Mice , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Telencephalon/cytology , Telencephalon/embryology , Telencephalon/metabolism , Thyroid Nuclear Factor 1
10.
Adv Neurobiol ; 8: 133-64, 2014.
Article in English | MEDLINE | ID: mdl-25300136

ABSTRACT

Roundabout receptors (Robo) and their Slit ligands were discovered in the 1990s and found to be key players in axon guidance. Slit was initially described s an extracellular matrix protein that was expressed by midline glia in Drosophila. A few years later, it was shown that, in vertebrates and invertebrates, Slits acted as chemorepellents for axons crossing the midline. Robo proteins were originally discovered in Drosophila in a mutant screen for genes involved in the regulation of midline crossing. This ligand-receptor pair has since been implicated in a variety of other neuronal and non-neuronal processes ranging from cell migration to angiogenesis, tumourigenesis and even organogenesis of tissues such as kidneys, lungs and breasts.


Subject(s)
Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Axons/physiology , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Models, Biological , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Signal Transduction/physiology , Roundabout Proteins
11.
J Neurosci ; 34(16): 5717-31, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741061

ABSTRACT

The elaborate cytoarchitecture of the mammalian neocortex requires the timely production of its constituent pyramidal neurons and interneurons and their disposition in appropriate layers. Numerous chemotropic factors present in the forebrain throughout cortical development play important roles in the orchestration of these events. The Roundabout (Robo) family of receptors and their ligands, the Slit proteins, are expressed in the developing forebrain, and are known to play important roles in the generation and migration of cortical interneurons. However, few studies have investigated their function(s) in the development of pyramidal cells. Here, we observed expression of Robo1 and Slit genes (Slit1, Slit2) in cells lining the telencephalic ventricles, and found significant increases in progenitor cells (basal and apical) at embryonic day (E)12.5 and E14.5 in the developing cortex of Robo1(-/-), Slit1(-/-), and Slit1(-/-)/Slit2(-/-), but not in mice lacking the other Robo or Slit genes. Using layer-specific markers, we found that both early- and late-born pyramidal neuron populations were significantly increased in the cortices of Robo1(-/-) mice at the end of corticogenesis (E18.5). The excess number of cortical pyramidal neurons generated prenatally appears to die in early postnatal life. The observed increase in pyramidal neurons was due to prolonged proliferative activity of their progenitors and not due to changes in cell cycle events. This finding, confirmed by in utero electroporation with Robo1 short hairpin RNA (shRNA) or control constructs into progenitors along the ventricular zone as well as in dissociated cortical cell cultures, points to a novel role for Robo1 in regulating the proliferation and generation of pyramidal neurons.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Developmental/genetics , Neocortex , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/physiology , Receptors, Immunologic/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Cerebral Ventricles/growth & development , Embryo, Mammalian , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/cytology , Neocortex/embryology , Neocortex/growth & development , Nerve Tissue Proteins/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , Roundabout Proteins
12.
Circ Res ; 112(3): 465-75, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23255421

ABSTRACT

RATIONALE: The Slit-Roundabout (Robo) signaling pathway has pleiotropic functions during Drosophila heart development. However, its role in mammalian heart development is largely unknown. OBJECTIVE: To analyze the role of Slit-Robo signaling in the formation of the pericardium and the systemic venous return in the murine heart. METHODS AND RESULTS: Expression of genes encoding Robo1 and Robo2 receptors and their ligands Slit2 and Slit3 was found in or around the systemic venous return and pericardium during development. Analysis of embryos lacking Robo1 revealed partial absence of the pericardium, whereas Robo1/2 double mutants additionally showed severely reduced sinus horn myocardium, hypoplastic caval veins, and a persistent left inferior caval vein. Mice lacking Slit3 recapitulated the defects in the myocardialization, alignment, and morphology of the caval veins. Ligand binding assays confirmed Slit3 as the preferred ligand for the Robo1 receptor, whereas Slit2 showed preference for Robo2. Sinus node development was mostly unaffected in all mutants. In addition, we show absence of cross-regulation with previously identified regulators Tbx18 and Wt1. We provide evidence that pericardial defects are created by abnormal localization of the caval veins combined with ectopic pericardial cavity formation. Local increase in neural crest cell death and impaired neural crest adhesive and migratory properties underlie the ectopic pericardium formation. CONCLUSIONS: A novel Slit-Robo signaling pathway is involved in the development of the pericardium, the sinus horn myocardium, and the alignment of the caval veins. Reduced Slit3 binding in the absence of Robo1, causing impaired cardiac neural crest survival, adhesion, and migration, underlies the pericardial defects.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pericardium/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Venae Cavae/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Movement , Gene Expression Regulation, Developmental , Gestational Age , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Crest/abnormalities , Neural Crest/metabolism , Pericardium/abnormalities , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Sinoatrial Node/abnormalities , Sinoatrial Node/metabolism , T-Box Domain Proteins/metabolism , Tissue Culture Techniques , Venae Cavae/abnormalities , WT1 Proteins/metabolism , Roundabout Proteins
13.
J Neurosci ; 32(47): 16892-905, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175841

ABSTRACT

In the postnatal forebrain, the subventricular zone (SVZ) contains a pool of undifferentiated cells, which proliferate and migrate along the rostral migratory stream (RMS) to the olfactory bulb and differentiate into granule cells and periglomerular cells. Plexin-B2 is a semaphorin receptor previously known to act on neuronal proliferation in the embryonic brain and neuronal migration in the cerebellum. We show here that, in the postnatal and adult CNS, Plexin-B2 is expressed in the subventricular zone lining the telencephalic ventricles and in the rostral migratory stream. We analyzed Plxnb2(-/-) mice and found that there is a marked reduction in the proliferation of SVZ cells in the mutant. Plexin-B2 expression is downregulated in the olfactory bulb as interneurons initiate radial migration. BrdU labeling and GFP electroporation into postnatal SVZ, in addition to time-lapse videomicroscopy, revealed that neuroblasts deficient for Plexin-B2 migrate faster than control ones and leave the RMS more rapidly. Overall, these results show that Plexin-B2 plays a role in postnatal neurogenesis and in the migration of SVZ-derived neuroblasts.


Subject(s)
Cell Movement/physiology , Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Neurons/physiology , Prosencephalon/physiology , Animals , Antimetabolites , Brain Tissue Transplantation , Bromodeoxyuridine , Cell Movement/genetics , Electroporation , Female , Glial Cell Line-Derived Neurotrophic Factor/physiology , Hepatocyte Growth Factor/physiology , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Knockout , Mutation/physiology , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Olfactory Bulb/physiology , Prosencephalon/cytology
14.
Development ; 139(18): 3326-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22912413

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons are born in the nasal placode and migrate along olfactory and vomeronasal axons to reach the forebrain and settle in the hypothalamus, where they control reproduction. The molecular cues that guide their migration have not been fully identified, but are thought to control either cell movement directly or the patterning of their axonal substrates. Using genetically altered mouse models we show that the migration of GnRH neurons is directly modulated by Slit2 and Robo3, members of the axon guidance Slit ligand and Robo receptor families. Mice lacking Slit2 or Robo3 have a reduced number of GnRH neurons in the forebrain, but a normal complement of their supporting axons, pointing to a direct role for these molecules in GnRH neuron migration.


Subject(s)
Cell Movement/physiology , Gonadotropin-Releasing Hormone/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , COS Cells , Cell Movement/genetics , Chlorocebus aethiops , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Receptors, Cell Surface
15.
Development ; 137(12): 1939-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501589

ABSTRACT

In most tissues, the precise control of cell migration and cell-cell interaction is of paramount importance to the development of a functional structure. Several families of secreted molecules have been implicated in regulating these aspects of development, including the Slits and their Robo receptors. These proteins have well described roles in axon guidance but by influencing cell polarity and adhesion, they participate in many developmental processes in diverse cell types. We review recent progress in understanding both the molecular mechanisms that modulate Slit/Robo expression and their functions in neural and non-neural tissue.


Subject(s)
Cell Movement/genetics , Cell Polarity/genetics , Cytoskeleton/metabolism , Nervous System/metabolism , Neurogenesis/physiology , Animals , Cytoskeleton/genetics , Drosophila/genetics , Drosophila/metabolism , Mice , Mice, Knockout
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 34(4): 669-73, 2010 May 30.
Article in English | MEDLINE | ID: mdl-20227455

ABSTRACT

Abnormal lipid profiles have been reported in the central nervous system (CNS) in individuals with schizophrenia, although the etiology of these changes remains to be elucidated. While treatment with second-generation antipsychotics has been associated with alterations in peripheral lipid levels and changes in erythrocyte membrane composition, the relationship between peripheral and CNS lipid levels is complex and the effect of antipsychotics on CNS lipid regulation is not yet understood. In this study we investigated whether sub-chronic administration of the second-generation antipsychotic clozapine and the first-generation antipsychotic haloperidol alters brain membrane lipid composition in male Sprague-Dawley rats. The relationship between brain membrane lipid composition and plasma cholesterol concentrations was also assessed. Our results indicate that brain lipid composition and plasma cholesterol concentrations are not altered following administration of antipsychotics. No correlation was observed between plasma and brain membrane cholesterol levels. Our findings suggest that observed alterations in brain lipid profiles in individuals with schizophrenia are not a consequence of treatment with antipsychotic medications.


Subject(s)
Brain Chemistry/drug effects , Cholesterol/analysis , Clozapine/pharmacology , Haloperidol/pharmacology , Phospholipids/analysis , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , Body Weight/drug effects , Cholesterol/blood , Male , Rats , Rats, Sprague-Dawley
17.
Neuropsychopharmacology ; 35(5): 1226-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20072114

ABSTRACT

Abnormalities of amount and function of presynaptic terminals may have an important role in the mechanism of illness in schizophrenia. The SNARE proteins (SNAP-25, syntaxin, and VAMP) are enriched in presynaptic terminals, where they interact to form a functional complex to facilitate vesicle fusion. SNARE protein amounts are altered in the cortical regions in schizophrenia, but studies of protein-protein interactions are limited. We extended these investigations to the striatal regions (such as the nucleus accumbens, ventromedial caudate (VMC), and dorsal caudate) relevant to disease symptoms. In addition to measuring SNARE protein levels, we studied SNARE protein-protein interactions using a novel ELISA method. The possible effect of antipsychotic treatment was investigated in parallel in the striatum of rodents that were administered haloperidol and clozapine. In schizophrenia samples, compared with controls, SNAP-25 was 32% lower (P=0.015) and syntaxin was 26% lower (P=0.006) in the VMC. In contrast, in the same region, SNARE protein-protein interactions were higher in schizophrenia (P=0.008). Confocal microscopy of schizophrenia and control VMC showed qualitatively similar SNARE protein immunostaining. Haloperidol treatment of rats increased levels of SNAP-25 (mean 24%, P=0.003), syntaxin (mean 18%, P=0.010), and VAMP (mean 16%, P=0.001), whereas clozapine increased only the VAMP level (mean 13%, P=0.004). Neither drug altered SNARE protein-protein interactions. These results indicate abnormalities of amount and interactions of proteins directly related to presynaptic function in the VMC in schizophrenia. SNARE proteins and their interactions may be a novel target for the development of therapeutics.


Subject(s)
Corpus Striatum/drug effects , Corpus Striatum/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Female , Haloperidol/pharmacology , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley , SNARE Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...