Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 173(6): 1356-1369.e22, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856954

ABSTRACT

Genetic changes causing brain size expansion in human evolution have remained elusive. Notch signaling is essential for radial glia stem cell proliferation and is a determinant of neuronal number in the mammalian cortex. We find that three paralogs of human-specific NOTCH2NL are highly expressed in radial glia. Functional analysis reveals that different alleles of NOTCH2NL have varying potencies to enhance Notch signaling by interacting directly with NOTCH receptors. Consistent with a role in Notch signaling, NOTCH2NL ectopic expression delays differentiation of neuronal progenitors, while deletion accelerates differentiation into cortical neurons. Furthermore, NOTCH2NL genes provide the breakpoints in 1q21.1 distal deletion/duplication syndrome, where duplications are associated with macrocephaly and autism and deletions with microcephaly and schizophrenia. Thus, the emergence of human-specific NOTCH2NL genes may have contributed to the rapid evolution of the larger human neocortex, accompanied by loss of genomic stability at the 1q21.1 locus and resulting recurrent neurodevelopmental disorders.


Subject(s)
Brain/embryology , Cerebral Cortex/physiology , Neurogenesis/physiology , Receptor, Notch2/metabolism , Signal Transduction , Animals , Cell Differentiation , Embryonic Stem Cells/metabolism , Female , Gene Deletion , Genes, Reporter , Gorilla gorilla , HEK293 Cells , Humans , Neocortex/cytology , Neural Stem Cells/metabolism , Neuroglia/metabolism , Neurons/metabolism , Pan troglodytes , Receptor, Notch2/genetics , Sequence Analysis, RNA
2.
Genome Res ; 32(4): 656-670, 2022 04.
Article in English | MEDLINE | ID: mdl-35332097

ABSTRACT

Genome-wide association studies (GWAS) have been highly informative in discovering disease-associated loci but are not designed to capture all structural variations in the human genome. Using long-read sequencing data, we discovered widespread structural variation within SINE-VNTR-Alu (SVA) elements, a class of great ape-specific transposable elements with gene-regulatory roles, which represents a major source of structural variability in the human population. We highlight the presence of structurally variable SVAs (SV-SVAs) in neurological disease-associated loci, and we further associate SV-SVAs to disease-associated SNPs and differential gene expression using luciferase assays and expression quantitative trait loci data. Finally, we genetically deleted SV-SVAs in the BIN1 and CD2AP Alzheimer's disease-associated risk loci and in the BCKDK Parkinson's disease-associated risk locus and assessed multiple aspects of their gene-regulatory influence in a human neuronal context. Together, this study reveals a novel layer of genetic variation in transposable elements that may contribute to identification of the structural variants that are the actual drivers of disease associations of GWAS loci.


Subject(s)
DNA Transposable Elements , Genome-Wide Association Study , Alu Elements , DNA Transposable Elements/genetics , Genetic Predisposition to Disease , Genetic Variation , Genome, Human , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci
3.
Genome Res ; 31(4): 551-563, 2021 04.
Article in English | MEDLINE | ID: mdl-33722937

ABSTRACT

Transposable element (TE) invasions have shaped vertebrate genomes over the course of evolution. They have contributed an extra layer of species-specific gene regulation by providing novel transcription factor binding sites. In humans, SINE-VNTR-Alu (SVA) elements are one of three still active TE families; approximately 2800 SVA insertions exist in the human genome, half of which are human-specific. TEs are often silenced by KRAB zinc finger (KZNF) proteins recruiting corepressor proteins that establish a repressive chromatin state. A number of KZNFs have been reported to bind SVAs, but their individual contribution to repressing SVAs and their roles in suppressing SVA-mediated gene-regulatory effects remains elusive. We analyzed the genome-wide binding profile for ZNF91 in human cells and found that ZNF91 interacts with the VNTR region of SVAs. Through CRISPR-Cas9-mediated deletion of ZNF91 in human embryonic stem cells, we established that loss of ZNF91 results in increased transcriptional activity of SVAs. In contrast, SVA activation was not observed upon genetic deletion of the ZNF611 gene encoding another strong SVA interactor. Epigenetic profiling confirmed the loss of SVA repression in the absence of ZNF91 and revealed that mainly evolutionary young SVAs gain gene activation-associated epigenetic modifications. Genes close to activated SVAs showed a mild up-regulation, indicating SVAs adopt properties of cis-regulatory elements in the absence of repression. Notably, genome-wide derepression of SVAs elicited the communal up-regulation of KZNFs that reside in KZNF clusters. This phenomenon may provide new insights into the potential mechanisms used by the host genome to sense and counteract TE invasions.


Subject(s)
Human Embryonic Stem Cells , Kruppel-Like Transcription Factors/deficiency , Multigene Family/genetics , Repressor Proteins/genetics , Retroelements/genetics , Transcriptional Activation , Up-Regulation , Genome, Human , Humans , Zinc Fingers/genetics
4.
Mol Biol Evol ; 37(9): 2531-2548, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32330268

ABSTRACT

Ever since the availability of genomes from Neanderthals, Denisovans, and ancient humans, the field of evolutionary genomics has been searching for protein-coding variants that may hold clues to how our species evolved over the last ∼600,000 years. In this study, we identify such variants in the human-specific NOTCH2NL gene family, which were recently identified as possible contributors to the evolutionary expansion of the human brain. We find evidence for the existence of unique protein-coding NOTCH2NL variants in Neanderthals and Denisovans which could affect their ability to activate Notch signaling. Furthermore, in the Neanderthal and Denisovan genomes, we find unusual NOTCH2NL configurations, not found in any of the modern human genomes analyzed. Finally, genetic analysis of archaic and modern humans reveals ongoing adaptive evolution of modern human NOTCH2NL genes, identifying three structural variants acting complementary to drive our genome to produce a lower dosage of NOTCH2NL protein. Because copy-number variations of the 1q21.1 locus, encompassing NOTCH2NL genes, are associated with severe neurological disorders, this seemingly contradicting drive toward low levels of NOTCH2NL protein indicates that the optimal dosage of NOTCH2NL may have not yet been settled in the human population.


Subject(s)
Biological Evolution , Neanderthals/genetics , Receptor, Notch2/genetics , Animals , Genome, Human , Genomic Structural Variation , Humans , Multigene Family , Receptor, Notch2/metabolism
5.
Nature ; 516(7530): 242-5, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25274305

ABSTRACT

Throughout evolution primate genomes have been modified by waves of retrotransposon insertions. For each wave, the host eventually finds a way to repress retrotransposon transcription and prevent further insertions. In mouse embryonic stem cells, transcriptional silencing of retrotransposons requires KAP1 (also known as TRIM28) and its repressive complex, which can be recruited to target sites by KRAB zinc-finger (KZNF) proteins such as murine-specific ZFP809 which binds to integrated murine leukaemia virus DNA elements and recruits KAP1 to repress them. KZNF genes are one of the fastest growing gene families in primates and this expansion is hypothesized to enable primates to respond to newly emerged retrotransposons. However, the identity of KZNF genes battling retrotransposons currently active in the human genome, such as SINE-VNTR-Alu (SVA) and long interspersed nuclear element 1 (L1), is unknown. Here we show that two primate-specific KZNF genes rapidly evolved to repress these two distinct retrotransposon families shortly after they began to spread in our ancestral genome. ZNF91 underwent a series of structural changes 8-12 million years ago that enabled it to repress SVA elements. ZNF93 evolved earlier to repress the primate L1 lineage until ∼12.5 million years ago when the L1PA3-subfamily of retrotransposons escaped ZNF93's restriction through the removal of the ZNF93-binding site. Our data support a model where KZNF gene expansion limits the activity of newly emerged retrotransposon classes, and this is followed by mutations in these retrotransposons to evade repression, a cycle of events that could explain the rapid expansion of lineage-specific KZNF genes.


Subject(s)
Evolution, Molecular , Kruppel-Like Transcription Factors/metabolism , Primates/genetics , Retroelements/genetics , Animals , Base Sequence , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mutation/genetics , Zinc Fingers
6.
Development ; 138(23): 5213-22, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22069189

ABSTRACT

Development of meso-diencephalic dopamine (mdDA) neurons requires the combined actions of the orphan nuclear receptor Nurr1 and the paired-like homeobox transcription factor Pitx3. Whereas all mdDA neurons require Nurr1 for expression of Th and survival, dependence on Pitx3 is displayed only by the mdDA subpopulation that will form the substantia nigra (SNc). Previously, we have demonstrated that Pitx3(-/-) embryos lack the expression of the retinoic acid (RA)-generating enzyme Ahd2, which is normally selectively expressed in the Pitx3-dependent DA neurons of the SNc. Restoring RA signaling in Pitx3(-/-) embryos revealed a selective dependence of SNc neurons on the presence of RA for differentiation into Th-positive neurons and maintenance throughout embryonic development. Whereas these data are suggestive of an important developmental role for RA in neurons of the SNc, it remained unclear whether other Nurr1 and Pitx3 target genes depend on RA signaling in a manner similar to Th. In the search for genes that were affected in Pitx3-deficient mdDA neurons and restored upon embryonic RA treatment, we provide evidence that Delta-like 1, D2R (Drd2) and Th are regulated by Pitx3 and RA signaling, which influences the mdDA terminal differentiated phenotype. Furthermore, we show that regulation of Ahd2-mediated RA signaling represents only one aspect of the Pitx3 downstream cascade, as Vmat2, Dat, Ahd2 (Aldh1a1), En1, En2 and Cck were unaffected by RA treatment and are (subset) specifically modulated by Pitx3. In conclusion, our data reveal several RA-dependent and -independent aspects of the Pitx3-regulated gene cascade, suggesting that Pitx3 acts on multiple levels in the molecular subset-specification of mdDA neurons.


Subject(s)
Diencephalon/cytology , Diencephalon/embryology , Gene Regulatory Networks/drug effects , Homeodomain Proteins/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , Analysis of Variance , Animals , Calcium-Binding Proteins , Cell Differentiation/drug effects , Cell Differentiation/physiology , Female , Fluorescent Antibody Technique , Gene Regulatory Networks/physiology , Genotype , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Microarray Analysis , Neurons/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Polymerase Chain Reaction , Pregnancy , Receptors, Dopamine D2/metabolism , Transcription Factors/deficiency
7.
J Comp Neurol ; 532(7): e25648, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38958676

ABSTRACT

In this study, we investigated recurrent copy number variations (CNVs) in the 19p12 locus, which are associated with neurodevelopmental disorders. The two genes in this locus, ZNF675 and ZNF681, arose via gene duplication in primates, and their presence in several pathological CNVs in the human population suggests that either or both of these genes are required for normal human brain development. ZNF675 and ZNF681 are members of the Krüppel-associated box zinc finger (KZNF) protein family, a class of transcriptional repressors important for epigenetic silencing of specific genomic regions. About 170 primate-specific KZNFs are present in the human genome. Although KZNFs are primarily associated with repressing retrotransposon-derived DNA, evidence is emerging that they can be co-opted for other gene regulatory processes. We show that genetic deletion of ZNF675 causes developmental defects in cortical organoids, and our data suggest that part of the observed neurodevelopmental phenotype is mediated by a gene regulatory role of ZNF675 on the promoter of the neurodevelopmental gene Hes family BHLH transcription factor 1 (HES1). We also find evidence for the recently evolved regulation of genes involved in neurological disorders, microcephalin 1 and sestrin 3. We show that ZNF675 interferes with HES1 auto-inhibition, a process essential for the maintenance of neural progenitors. As a striking example of how some KZNFs have integrated into preexisting gene expression networks, these findings suggest the emergence of ZNF675 has caused a change in the balance of HES1 autoregulation. The association of ZNF675 CNV with human developmental disorders and ZNF675-mediated regulation of neurodevelopmental genes suggests that it evolved into an important factor for human brain development.


Subject(s)
Primates , Transcription Factor HES-1 , Humans , Animals , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Primates/genetics , Homeostasis/physiology , Homeostasis/genetics , DNA Copy Number Variations/genetics , Mice , Biological Evolution , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
8.
Nat Microbiol ; 9(5): 1189-1206, 2024 May.
Article in English | MEDLINE | ID: mdl-38548923

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with short- and long-term neurological complications. The variety of symptoms makes it difficult to unravel molecular mechanisms underlying neurological sequalae after coronavirus disease 2019 (COVID-19). Here we show that SARS-CoV-2 triggers the up-regulation of synaptic components and perturbs local electrical field potential. Using cerebral organoids, organotypic culture of human brain explants from individuals without COVID-19 and post-mortem brain samples from individuals with COVID-19, we find that neural cells are permissive to SARS-CoV-2 to a low extent. SARS-CoV-2 induces aberrant presynaptic morphology and increases expression of the synaptic components Bassoon, latrophilin-3 (LPHN3) and fibronectin leucine-rich transmembrane protein-3 (FLRT3). Furthermore, we find that LPHN3-agonist treatment with Stachel partially restored organoid electrical activity and reverted SARS-CoV-2-induced aberrant presynaptic morphology. Finally, we observe accumulation of relatively static virions at LPHN3-FLRT3 synapses, suggesting that local hindrance can contribute to synaptic perturbations. Together, our study provides molecular insights into SARS-CoV-2-brain interactions, which may contribute to COVID-19-related neurological disorders.


Subject(s)
Brain , COVID-19 , Homeostasis , Organoids , SARS-CoV-2 , Synapses , Humans , SARS-CoV-2/physiology , COVID-19/virology , COVID-19/metabolism , COVID-19/pathology , Brain/virology , Synapses/virology , Synapses/metabolism , Organoids/virology , Virion/metabolism , Neurons/virology , Neurons/metabolism , Receptors, Peptide/metabolism , Receptors, Peptide/genetics
9.
Genome Biol Evol ; 15(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37847041

ABSTRACT

Krüppel-associated box (KRAB) zinc finger proteins (KZNFs) recognize and repress transposable elements (TEs); TEs are DNA elements that are capable of replicating themselves throughout our genomes with potentially harmful consequences. However, genes from this family of transcription factors have a much wider potential for genomic regulation. KZNFs have become integrated into gene-regulatory networks through the control of TEs that function as enhancers and gene promoters; some KZNFs also bind directly to gene promoters, suggesting an additional, more direct layer of KZNF co-option into gene-regulatory networks. Binding site analysis of ZNF519, ZNF441, and ZNF468 suggests the structural evolution of KZNFs to recognize TEs can result in coincidental binding to gene promoters independent of TE sequences. We show a higher rate of sequence turnover in gene promoter KZNF binding sites than neighboring regions, implying a selective pressure is being applied by the binding of a KZNF. Through CRISPR/Cas9 mediated genetic deletion of ZNF519, ZNF441, and ZNF468, we provide further evidence for genome-wide co-option of the KZNF-mediated gene-regulatory functions; KZNF knockout leads to changes in expression of KZNF-bound genes in neuronal lineages. Finally, we show that the opposite can be established upon KZNF overexpression, further strengthening the support for the role of KZNFs as bona-fide gene regulators. With no eminent role for ZNF519 in controlling its TE target, our study may provide a snapshot into the early stages of the completed co-option of a KZNF, showing the lasting, multilayered impact that retrovirus invasions and host response mechanisms can have upon the evolution of our genomes.


Subject(s)
Primates , Zinc Fingers , Animals , Zinc Fingers/genetics , Primates/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , DNA Transposable Elements , Gene Regulatory Networks
10.
Philos Trans R Soc Lond B Biol Sci ; 375(1795): 20190333, 2020 03 30.
Article in English | MEDLINE | ID: mdl-32075554

ABSTRACT

The large family of KRAB zinc finger (KZNF) genes are transcription factors implicated in recognizing and repressing repetitive sequences such as transposable elements (TEs) in our genome. Through successive waves of retrotransposition-mediated insertions, various classes of TEs have invaded mammalian genomes at multiple timepoints throughout evolution. Even though most of the TE classes in our genome lost the capability to retrotranspose millions of years ago, it remains elusive why the KZNFs that evolved to repress them are still retained in our genome. One hypothesis is that KZNFs become repurposed for other regulatory roles. Here, we find evidence that evolutionary changes in KZNFs provide them not only with the ability to repress TEs, but also to bind to gene promoters independent of TEs. Using KZNF binding site data in conjunction with gene expression values from the Allen Brain Atlas, we show that KZNFs have the ability to regulate gene expression in the human brain in a region-specific manner. Our analysis shows that the expression of KZNFs shows correlation with the expression of their target genes, suggesting that KZNFs have a direct influence on gene expression in the developing human brain. The extent of this regulation and the impact it has on primate brain evolution are still to be determined, but our results imply that KZNFs have become widely integrated into neuronal gene regulatory networks. Our analysis predicts that gene expression networks have been repeatedly innovated throughout primate evolution, continuously gaining new layers of gene regulation mediated by both TEs and KZNFs in our genome. This article is part of a discussion meeting issue 'Crossroads between transposons and gene regulation'.


Subject(s)
Brain/growth & development , DNA Transposable Elements , Evolution, Molecular , Gene Expression Regulation, Developmental , Genes, Developmental , Primates/genetics , Zinc Fingers/genetics , Animals , Brain/metabolism , Humans , Protein Binding
11.
Stem Cell Reports ; 12(2): 245-257, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30639214

ABSTRACT

The cerebral cortex has expanded in size and complexity in primates, yet the molecular innovations that enabled primate-specific brain attributes remain obscure. We generated cerebral cortex organoids from human, chimpanzee, orangutan, and rhesus pluripotent stem cells and sequenced their transcriptomes at weekly time points for comparative analysis. We used transcript structure and expression conservation to discover gene regulatory long non-coding RNAs (lncRNAs). Of 2,975 human, multi-exonic lncRNAs, 2,472 were structurally conserved in at least one other species and 920 were conserved in all. Three hundred eighty-six human lncRNAs were transiently expressed (TrEx) and many were also TrEx in great apes (46%) and rhesus (31%). Many TrEx lncRNAs are expressed in specific cell types by single-cell RNA sequencing. Four TrEx lncRNAs selected based on cell-type specificity, gene structure, and expression pattern conservation were ectopically expressed in HEK293 cells by CRISPRa. All induced trans gene expression changes were consistent with neural gene regulatory activity.


Subject(s)
Cell Differentiation/genetics , Cerebral Cortex/physiology , Gene Expression Regulation/genetics , RNA, Long Noncoding/genetics , Animals , Cell Line , Gene Expression Profiling/methods , HEK293 Cells , Humans , Organoids/physiology , Pluripotent Stem Cells/physiology , Primates , Sequence Analysis, RNA , Transcriptome/genetics
12.
Nat Commun ; 10(1): 4430, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31562326

ABSTRACT

Zika virus (ZIKV) invades and persists in the central nervous system (CNS), causing severe neurological diseases. However the virus journey, from the bloodstream to tissues through a mature endothelium, remains unclear. Here, we show that ZIKV-infected monocytes represent suitable carriers for viral dissemination to the CNS using human primary monocytes, cerebral organoids derived from embryonic stem cells, organotypic mouse cerebellar slices, a xenotypic human-zebrafish model, and human fetus brain samples. We find that ZIKV-exposed monocytes exhibit higher expression of adhesion molecules, and higher abilities to attach onto the vessel wall and transmigrate across endothelia. This phenotype is associated to enhanced monocyte-mediated ZIKV dissemination to neural cells. Together, our data show that ZIKV manipulates the monocyte adhesive properties and enhances monocyte transmigration and viral dissemination to neural cells. Monocyte transmigration may represent an important mechanism required for viral tissue invasion and persistence that could be specifically targeted for therapeutic intervention.


Subject(s)
Cell Adhesion Molecules/metabolism , Monocytes/metabolism , Monocytes/virology , Neurons/metabolism , Transendothelial and Transepithelial Migration/physiology , Zika Virus Infection/metabolism , Zika Virus/physiology , Zika Virus/pathogenicity , Animals , Cell Adhesion/physiology , Cell Survival , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/virology , Cerebellum/pathology , Cerebellum/virology , Disease Models, Animal , Embryonic Stem Cells , Endothelium/virology , Female , Humans , Monocytes/pathology , Neurons/pathology , Neurons/virology , Organoids/metabolism , Organoids/pathology , Zebrafish , Zika Virus Infection/pathology , Zika Virus Infection/virology
13.
Noncoding RNA ; 4(2)2018 Apr 18.
Article in English | MEDLINE | ID: mdl-29670042

ABSTRACT

The expansion of long non-coding RNAs (lncRNAs) in organismal genomes has been associated with the emergence of sophisticated regulatory networks that may have contributed to more complex neuronal processes, such as higher-order cognition. In line with the important roles of lncRNAs in the normal functioning of the human brain, dysregulation of lncRNA expression has been implicated in aging and age-related neurodegenerative disorders. In this paper, we discuss the function and expression of known neuronal-associated lncRNAs, their impact on epigenetic changes, the contribution of transposable elements to lncRNA expression, and the implication of lncRNAs in maintaining the 3D nuclear architecture in neurons. Moreover, we discuss how the complex molecular processes that are orchestrated by lncRNAs in the aged brain may contribute to neuronal pathogenesis by promoting protein aggregation and neurodegeneration. Finally, this review explores the possibility that age-related disturbances of lncRNA expression change the genomic and epigenetic regulatory landscape of neurons, which may affect neuronal processes such as neurogenesis and synaptic plasticity.

14.
Cell Rep ; 24(4): 973-986.e8, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30044992

ABSTRACT

Endosomal sorting complex required for transport (ESCRT) complex proteins regulate biogenesis and release of extracellular vesicles (EVs), which enable cell-to-cell communication in the nervous system essential for development and adult function. We recently showed human loss-of-function (LOF) mutations in ESCRT-III member CHMP1A cause autosomal recessive microcephaly with pontocerebellar hypoplasia, but its mechanism was unclear. Here, we show Chmp1a is required for progenitor proliferation in mouse cortex and cerebellum and progenitor maintenance in human cerebral organoids. In Chmp1a null mice, this defect is associated with impaired sonic hedgehog (Shh) secretion and intraluminal vesicle (ILV) formation in multivesicular bodies (MVBs). Furthermore, we show CHMP1A is important for release of an EV subtype that contains AXL, RAB18, and TMED10 (ART) and SHH. Our findings show CHMP1A loss impairs secretion of SHH on ART-EVs, providing molecular mechanistic insights into the role of ESCRT proteins and EVs in the brain.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Extracellular Vesicles/metabolism , Hedgehog Proteins/metabolism , Adult , Animals , Brain/embryology , Brain/metabolism , Choroid Plexus/embryology , Choroid Plexus/growth & development , Choroid Plexus/metabolism , Humans , Infant, Newborn , Mice , NIH 3T3 Cells , Vesicular Transport Proteins
15.
Gene Expr Patterns ; 6(2): 134-40, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16326148

ABSTRACT

In order to obtain leads to molecular mechanisms of signal transduction pathways and controlled gene expression in neuronal development we have screened the adult mouse brain for expressed forkhead transcription factors using a degenerate RT-PCR approach. Here, we focus on three FoxO genes found to be expressed in the brain: FoxO1, FoxO3 and FoxO6. The FoxO subfamily of forkhead transcription family is emerging as a central keypoint in an array of cellular functions, such as metabolism, differentiation and transformation. In situ hybridization experiments on adult and embryonic mouse brain showed differential expression patterns for three FoxO members. FoxO1 was strongly expressed in the striatum and neuronal subsets of the hippocampus (dentate gyrus and the ventral/posterior part of the CA regions), whereas FoxO3 was more diffusely expressed throughout the brain including all hippocampal areas, cortex and cerebellum. FoxO6 expression was eminent in various parts of the adult mouse brain, including the entire hippocampus, the amygdalohippocampal area and the shell of the nucleus accumbens. Remarkably, all three FoxO transcription factors were expressed relatively late in the developing murine brain, starting between E12.5 and E14. In summary, the presented data show FoxO factors to be expressed in the adult and developing mouse brain, in a spatially end temporally restricted manner.


Subject(s)
Brain/embryology , Brain/metabolism , Forkhead Transcription Factors/genetics , Animals , Base Sequence , DNA, Complementary/genetics , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Mice, Inbred C57BL
16.
Biochem J ; 391(Pt 3): 623-9, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-15987244

ABSTRACT

Forkhead members of the 'O' class (FoxO) are transcription factors crucial for the regulation of metabolism, cell cycle, cell death and cell survival. FoxO factors are regulated by insulin-mediated activation of PI3K (phosphoinositide 3-kinase)-PKB (protein kinase B) signalling. Activation of PI3K-PKB signalling results in the phosphorylation of FoxO factors on three conserved phosphorylation motifs, which are essential for the translocation of FoxO factors from the nucleus to the cytosol. FoxO6, however, remains mostly nuclear due to the fact that its shuttling ability is dramatically impaired. FoxO1, FoxO3 and FoxO4 all contain an N- and C-terminal PKB motif and a motif located in the forkhead domain. FoxO6 lacks the conserved C-terminal PKB motif, which is the cause of the shuttling impairment. Since FoxO6 can be considered constitutively nuclear, we investigated whether it is also a constitutively active transcription factor. Our results show that FoxO6 transcriptional activity is inhibited by growth factors, independent of shuttling, indicating that it is not constitutively active. The PKB site in the forkhead domain (Ser184) regulated the DNA binding characteristics and the N-terminal PKB site acted as a growth factor sensor. In summary, FoxO6 is not a constitutively active transcription factor and can be regulated by growth factors in a Thr26- and Ser184-dependent manner, independent of shuttling to the cytosol.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Serine/metabolism , Threonine/metabolism , Transcription, Genetic , Amino Acid Motifs , Cell Line , Forkhead Transcription Factors/genetics , Humans , Mutation , Phosphorylation , Protein Structure, Tertiary , Protein Transport , Serine/genetics , Threonine/genetics
17.
Development ; 136(4): 531-40, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19144721

ABSTRACT

In recent years, the meso-diencephalic dopaminergic (mdDA) neurons have been extensively studied for their association with Parkinson's disease. Thus far, specification of the dopaminergic phenotype of mdDA neurons is largely attributed to the orphan nuclear receptor Nurr1. In this study, we provide evidence for extensive interplay between Nurr1 and the homeobox transcription factor Pitx3 in vivo. Both Nurr1 and Pitx3 interact with the co-repressor PSF and occupy the promoters of Nurr1 target genes in concert. Moreover, in vivo expression analysis reveals that Nurr1 alone is not sufficient to drive the dopaminergic phenotype in mdDA neurons but requires Pitx3 for full activation of target gene expression. In the absence of Pitx3, Nurr1 is kept in a repressed state through interaction with the co-repressor SMRT. Highly resembling the effect of ligand activation of nuclear receptors, recruitment of Pitx3 modulates the Nurr1 transcriptional complex by decreasing the interaction with SMRT, which acts through HDACs to keep promoters in a repressed deacetylated state. Indeed, interference with HDAC-mediated repression in Pitx3(-/-) embryos efficiently reactivates the expression of Nurr1 target genes, bypassing the necessity for Pitx3. These data position Pitx3 as an essential potentiator of Nurr1 in specifying the dopaminergic phenotype, providing novel insights into mechanisms underlying development of mdDA neurons in vivo, and the programming of stem cells as a future cell replacement therapy for Parkinson's disease.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Dopamine/metabolism , Homeodomain Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Embryo, Mammalian/enzymology , Gene Expression Regulation, Developmental , Genome/genetics , Histone Deacetylases/metabolism , Mice , Models, Biological , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Subfamily 4, Group A, Member 2 , PTB-Associated Splicing Factor , Promoter Regions, Genetic/genetics , Protein Binding , RNA-Binding Proteins/metabolism , Transcription Factors/deficiency
18.
Development ; 136(14): 2363-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19515692

ABSTRACT

The orphan nuclear receptor Nurr1 is essential for the development of meso-diencephalic dopamine (mdDA) neurons and is required, together with the homeobox transcription factor Pitx3, for the expression of genes involved in dopamine metabolism. In order to elucidate the molecular mechanisms that underlie the neuronal deficits in Nurr1(-/-) mice, we performed combined gene expression microarrays and ChIP-on-chip analysis and thereby identified Dlk1, Ptpru and Klhl1 as novel Nurr1 target genes in vivo. In line with the previously described cooperativity between Nurr1 and Pitx3, we show that the expression of Ptpru and Klhl1 in mdDA neurons is also dependent on Pitx3. Furthermore, we demonstrate that Nurr1 interacts with the Ptpru promoter directly and requires Pitx3 for full expression of Ptpru in mdDA neurons. By contrast, the expression of Dlk1 is maintained in Pitx3(-/-) embryos and is even expanded into the rostral part of the mdDA area, suggesting a unique position of Dlk1 in the Nurr1 and Pitx3 transcriptional cascades. Expression analysis in Dlk1(-/-) embryos reveals that Dlk1 is required to prevent premature expression of Dat in mdDA neuronal precursors as part of the multifaceted process of mdDA neuronal differentiation driven by Nurr1 and Pitx3. Taken together, the involvement of Nurr1 and Pitx3 in the expression of novel target genes involved in important neuronal processes such as neuronal patterning, axon outgrowth and terminal differentiation, opens up new avenues to study the properties of mdDA neurons during development and in neuronal pathology as observed in Parkinson's disease.


Subject(s)
DNA-Binding Proteins/metabolism , Diencephalon/cytology , Diencephalon/metabolism , Dopamine/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Microfilament Proteins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , Calcium-Binding Proteins , Cell Differentiation , Cells, Cultured , DNA Primers/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Diencephalon/embryology , Diencephalon/growth & development , Dopamine Plasma Membrane Transport Proteins/genetics , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Models, Neurological , Neurons/cytology , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2 , Oligonucleotide Array Sequence Analysis , Pregnancy , Promoter Regions, Genetic , Transcription Factors/deficiency , Transcription Factors/genetics
19.
Development ; 134(14): 2673-84, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17592014

ABSTRACT

Selective neuronal loss in the substantia nigra (SNc), as described for Parkinson's disease (PD) in humans and for Pitx3 deficiency in mice, highlights the existence of neuronal subpopulations. As yet unknown subset-specific gene cascades might underlie the observed differences in neuronal vulnerability. We identified a developmental cascade in mice in which Ahd2 (Aldh1a1) is under the transcriptional control of Pitx3. Interestingly, Ahd2 distribution is restricted to a subpopulation of the meso-diencephalic dopaminergic (mdDA) neurons that is affected by Pitx3 deficiency. Ahd2 is involved in the synthesis of retinoic acid (RA), which has a crucial role in neuronal patterning, differentiation and survival in the brain. Most intriguingly, restoring RA signaling in the embryonic mdDA area counteracts the developmental defects caused by Pitx3 deficiency. The number of tyrosine hydroxylase-positive (TH+) neurons was significantly increased after RA treatment in the rostral mdDA region of Pitx3-/- embryos. This effect was specific for the rostral part of the developing mdDA area, and was observed exclusively in Pitx3-/- embryos. The effect of RA treatment during the critical phase was preserved until later in development, and our data suggest that RA is required for the establishment of proper mdDA neuronal identity. This positions Pitx3 centrally in a mdDA developmental cascade linked to RA signaling. Here, we propose a novel mechanism in which RA is involved in mdDA neuronal development and maintenance, providing new insights into subset-specific vulnerability in PD.


Subject(s)
Homeodomain Proteins/metabolism , Substantia Nigra/embryology , Substantia Nigra/metabolism , Transcription Factors/metabolism , Tretinoin/physiology , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase 1 Family , Amino Acid Sequence , Animals , Cell Differentiation , Cell Lineage , Diencephalon/cytology , Diencephalon/embryology , Diencephalon/metabolism , Dopamine/metabolism , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Pregnancy , Retinal Dehydrogenase , Signal Transduction , Transcription Factors/genetics , Tretinoin/pharmacology , Tyrosine 3-Monooxygenase/metabolism
20.
J Biol Chem ; 278(38): 35959-67, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12857750

ABSTRACT

Forkhead transcription factors of the FoxO-group are associated with cellular processes like cell cycle progression and DNA-repair. FoxO function is regulated by protein kinase B (PKB) via the phosphatidylinositol 3-kinase/PKB survival pathway. Phosphorylation of serine and threonine residues in specific PKB phosphorylation motifs leads to exclusion of FoxO-proteins from the nucleus, which excludes them from exerting transactivating activity. Members of the FoxO-group have three highly conserved regions containing a PKB phosphorylation motif. This study describes the cloning and characterization of a novel forkhead domain gene from mouse that appeared to be highly related to the FoxO group of transcription factors and was therefore designated FoxO6. The FoxO6 gene was mapped in region D1 on mouse chromosome 4. In humans, FOXO6 is located on chromosomal region 1p34.1. Embryonic expression of FoxO6 is most apparent in the developing brain, and FoxO6 is expressed in a specific temporal and spatial pattern. Therefore it is probably involved in regulation of specific cellular differentiation. In the adult animal FoxO6 expression is maintained in areas of the nucleus accumbens, cingulate cortex, parts of the amygdala, and in the hippocampus. Structure function analysis of FoxO6 compared with its group members shows that the overall homology is high, but surprisingly a highly conserved region containing multiple phosphorylation sites is lacking. In transfection studies, FoxO6 coupled to GFP showed an unexpected high nuclear localization after stimulation with growth factors, in contrast to the predominant cytosolic localization of FoxO1 and FoxO3. We also show that nuclear export of FoxO6 is mediated through the phosphatidylinositol 3-kinase/PKB pathway. Furthermore, we show using a chimeric approach that we can fully restore the ability of FoxO6 to shuttle between nucleus and cytosol. In conclusion, the data presented here gives a new view on regulation of FoxO-function through multiple phosphorylation events and other mechanisms involved in the nuclear exclusion of FoxO-proteins.


Subject(s)
Protein Serine-Threonine Kinases , Transcription Factors/chemistry , Transcription Factors/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Brain/metabolism , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Chromosome Mapping , Cytosol/metabolism , DNA Repair , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors , Green Fluorescent Proteins , Humans , In Situ Hybridization , Insulin/pharmacology , Luciferases/metabolism , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Genetic , Molecular Sequence Data , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Biosynthesis , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA/metabolism , Sequence Homology, Amino Acid , Serine/metabolism , Structure-Activity Relationship , Threonine/metabolism , Time Factors , Tissue Distribution , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL