Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 42(18): 3689-3703, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35351830

ABSTRACT

Recent branching (100 MYA) of the mammalian evolutionary tree has enhanced brain complexity and functions at the putative cost of increased emotional circuitry vulnerability. Thus, to better understand psychopathology, a burden for the modern society, novel approaches should exploit evolutionary aspects of psychiatric-relevant molecular pathways. A handful of genes is nowadays tightly associated to psychiatric disorders. Among them, neuronal-enriched RbFOX1 modifies the activity of synaptic regulators in response to neuronal activity, keeping excitability within healthy domains. We here dissect a higher primates-restricted interaction between RbFOX1 and the transcriptional corepressor Lysine Specific Demethylase 1 (LSD1/KDM1A). A single nucleotide variation (AA to AG) in LSD1 gene appeared in higher primates and humans, endowing RbFOX1 with the ability to promote the alternative usage of a novel 3' AG splice site, which extends LSD1 exon E9 in the upstream intron (E9-long). Exon E9-long regulates LSD1 levels by Nonsense-Mediated mRNA Decay. As reintroduction of the archaic LSD1 variant (AA) abolishes E9-long splicing, the novel 3' AG splice site is necessary for RbFOX1 to control LSD1 levels. LSD1 is a homeostatic immediate early genes (IEGs) regulator playing a relevant part in environmental stress-response. In primates and humans, inclusion of LSD1 as RbFOX1 target provides RbFOX1 with the additional ability to regulate the IEGs. These data, together with extensive RbFOX1 involvement in psychiatric disorders and its stress-dependent regulation in male mice, suggest the RbFOX1-LSD1-IEGs axis as an evolutionary recent psychiatric-relevant pathway. Notably, outside the nervous system, RbFOX2-dependent LSD1 modulation could be a candidate deregulated mechanism in cancer.SIGNIFICANCE STATEMENT To be better understood, anxiety and depression need large human genetics studies aimed at further resolving the often ambiguous, aberrant neuronal pathomechanisms that impact corticolimbic circuitry physiology. Several genetic associations of the alternative splicing regulator RbFOX1 with psychiatric conditions suggest homeostatic unbalance as a neuronal signature of psychopathology. Here we move a step forward, characterizing a disease-relevant higher primates-specific pathway by which RbFOX1 acquires the ability to regulate neuronal levels of Lysine Specific Demethylase 1, an epigenetic modulator of environmental stress response. Thus, two brain-enriched enzymes, independently shown to homeostatically protect neurons with a clear readout in terms of emotional behavior in lower mammals, establish in higher primates and humans a new functional cooperation enhancing the complexity of environmental adaptation and stress vulnerability.


Subject(s)
Alternative Splicing , Lysine , Alternative Splicing/genetics , Animals , Brain/metabolism , Histone Demethylases/genetics , Humans , Lysine/metabolism , Male , Mammals , Mice , Primates , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Repressor Proteins/genetics
2.
Mol Neurobiol ; 57(1): 393-407, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31364026

ABSTRACT

Ten to 20% of western countries population suffers from major depression disorder (MDD). Stressful life events represent the main environmental risk factor contributing to the onset of MDD and other stress-related neuropsychiatric disorders. In this regard, investigating brain physiology of stress response underlying the remarkable individual variability in terms of behavioral outcome may uncover stress-vulnerability pathways as a source of candidate targets for conceptually new antidepressant treatments. Serum response factor (SRF) has been addressed as a stress transducer via promoting inherent experience-induced Immediate Early Genes (IEGs) expression in neurons. However, in resting conditions, SRF also represents a transcriptional repressor able to assemble the core LSD1/CoREST/HDAC2 corepressor complex, including demethylase and deacetylase activities. We here show that dominant negative SRF splicing isoform lacking most part of the transactivation domain, namely SRFΔ5, owes its transcriptional repressive behavior to the ability of assembling LSD1/CoREST/HDAC2 corepressor complex meanwhile losing its affinity for transcription-permissive cofactor ELK1. SRFΔ5 is highly expressed in the brain and developmentally regulated. In the light of its activity as negative modulator of dendritic spine density, SRFΔ5 increase along with brain maturation suggests a role in synaptic pruning. Upon acute psychosocial stress, SRFΔ5 isoform transiently increases its levels. Remarkably, when stress is chronically repeated, a different picture occurs where SRF protein becomes stably upregulated in vulnerable mice but not in resilient animals. These data suggest a role for SRFΔ5 that is restricted to acute stress response, while positive modulation of SRF during chronic stress matches the criteria for stress-vulnerability hallmark.


Subject(s)
Alternative Splicing/genetics , Co-Repressor Proteins/metabolism , Histone Demethylases/metabolism , Neuronal Plasticity , Serum Response Factor/genetics , Stress, Physiological , Animals , Cell Shape , Dendritic Spines/metabolism , HeLa Cells , Hippocampus/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice, Inbred C57BL , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Serum Response Factor/metabolism , Stress, Psychological/pathology
3.
Trends Neurosci ; 40(1): 28-38, 2017 01.
Article in English | MEDLINE | ID: mdl-27986293

ABSTRACT

The acquisition and maintenance of the specific neuronal functions underlying learning, memory, and emotion require transduction of environmental stimuli into remodeling of neuronal circuitry. This process occurs via induction of plasticity-related transcriptional programs. The epigenetic enzyme lysine-specific demethylase-1 (LSD1), also known as lysine demethylase 1A (KDM1A), and its neurospecific splicing variant neuroLSD1 have been implicated in this process through an antagonistic mechanism. Specifically, LSD1/neuroLSD1 are involved in the negative and positive regulation of activity-evoked transcription of immediate early genes (IEGs) impacting memory formation and emotional behavior. Remarkably, the splicing process generating neuroLSD1 is homeostatically modulated by environmental contingencies, further implicating the LSD1/neuroLSD1 dual system as a modifier of information processing in the brain.


Subject(s)
Alternative Splicing , Epigenesis, Genetic , Histone Demethylases/metabolism , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Animals , Humans
4.
Hum Mol Genet ; 25(12): 2578-2587, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27094131

ABSTRACT

Genetic diseases often lead to rare and severe syndromes and the identification of the genetic and protein alterations responsible for the pathogenesis is essential to understand both the physiological and pathological role of the gene product. Recently, de novo variants have been mapped on the gene encoding for the lysine-specific histone demethylase 1 (LSD1)/lysine(K)-specific histone demethylase 1A in three patients characterized by a new genetic disorder. We have analyzed the effects of these pathological mutations on the structure, stability and activity of LSD1 using both in vitro and cellular approaches. The three mutations (Glu403Lys, Asp580Gly and Tyr785His) affect active-site residues and lead to a partial impairment of catalytic activity. They also differentially perturb the ability of LSD1 to engage transcription factors that orchestrate key developmental programs. Moreover, cellular data indicate a decrease in the protein cellular half-life. Taken together, these results demonstrate the relevance of LSD1 in gene regulation and how even moderate alterations in its stability, catalytic activity and binding properties can strongly affect organism development. This depicts a perturbed interplay of catalytic and non-catalytic processes at the origin of the pathology.


Subject(s)
Histone Demethylases/chemistry , Histone Demethylases/genetics , Intellectual Disability/genetics , Transcription, Genetic , Catalysis , Catalytic Domain/genetics , Gene Expression Regulation , Histone Demethylases/metabolism , Humans , Intellectual Disability/pathology , Mutation , Protein Binding , Protein Conformation , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/genetics
5.
Proc Natl Acad Sci U S A ; 113(13): 3651-6, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976584

ABSTRACT

Behavioral changes in response to stressful stimuli can be controlled via adaptive epigenetic changes in neuronal gene expression. Here we indicate a role for the transcriptional corepressor Lysine-Specific Demethylase 1 (LSD1) and its dominant-negative splicing isoform neuroLSD1, in the modulation of emotional behavior. In mouse hippocampus, we show that LSD1 and neuroLSD1 can interact with transcription factor serum response factor (SRF) and set the chromatin state of SRF-targeted genes early growth response 1 (egr1) and c-fos Deletion or reduction of neuro LSD1 in mutant mice translates into decreased levels of activating histone marks at egr1 and c-fos promoters, dampening their psychosocial stress-induced transcription and resulting in low anxiety-like behavior. Administration of suberoylanilide hydroxamine to neuroLSD1(KO)mice reactivates egr1 and c-fos transcription and restores the behavioral phenotype. These findings indicate that LSD1 is a molecular transducer of stressful stimuli as well as a stress-response modifier. Indeed, LSD1 expression itself is increased acutely at both the transcriptional and splicing levels by psychosocial stress, suggesting that LSD1 is involved in the adaptive response to stress.


Subject(s)
Emotions/physiology , Genes, Immediate-Early , Histone Demethylases/physiology , Alternative Splicing , Animals , Early Growth Response Protein 1/genetics , Epigenesis, Genetic , Genes, fos , Histone Demethylases/deficiency , Histone Demethylases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neuronal Plasticity , Phenotype , Serum Response Factor/physiology , Stress, Psychological , Transcription, Genetic
6.
Cereb Cortex ; 25(9): 2729-40, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24735673

ABSTRACT

Alternative splicing in the brain is dynamic and instrumental to adaptive changes in response to stimuli. Lysine-specific demethylase 1 (LSD1/KDM1A) is a ubiquitously expressed histone H3Lys4 demethylase that acts as a transcriptional co-repressor in complex with its molecular partners CoREST and HDAC1/2. In mammalian brain, alternative splicing of LSD1 mini-exon E8a gives rise to neuroLSD1, a neurospecific isoform that, upon phosphorylation, acts as a dominant-negative causing disassembly of the co-repressor complex and de-repression of target genes. Here we show that the LSD1/neuroLSD1 ratio changes in response to neuronal activation and such effect is mediated by neurospecific splicing factors NOVA1 and nSR100/SRRM4 together with a novel cis-silencer. Indeed, we found that, in response to epileptogenic stimuli, downregulation of NOVA1 reduces exon E8a splicing and expression of neuroLSD1. Using behavioral and EEG analyses we observed that neuroLSD1-specific null mice are hypoexcitable and display decreased seizure susceptibility. Conversely, in a mouse model of Rett syndrome characterized by hyperexcitability, we measured higher levels of NOVA1 protein and upregulation of neuroLSD1. In conclusion, we propose that, in the brain, correct ratio between LSD1 and neuroLSD1 contributes to excitability and, when altered, could represent a pathogenic event associated with neurological disorders involving altered E/I.


Subject(s)
Alternative Splicing/genetics , Brain/pathology , Down-Regulation/genetics , Epilepsy/genetics , Histone Demethylases/metabolism , Neurons/physiology , Analysis of Variance , Animals , Antigens, Neoplasm/metabolism , Brain/physiopathology , Cell Line, Tumor , Chromatin Immunoprecipitation , Disease Models, Animal , Electroencephalography , Histone Demethylases/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuro-Oncological Ventral Antigen , Neuroblastoma/pathology , RNA-Binding Proteins/metabolism , Transfection
7.
Mol Cell Biol ; 34(14): 2760-70, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24820421

ABSTRACT

Mammalian genomes harbor three CoREST genes. rcor1 encodes CoREST (CoREST1), and the paralogues rcor2 and rcor3 encode CoREST2 and CoREST3, respectively. Here, we describe specific properties of transcriptional complexes formed by CoREST proteins with the histone demethylase LSD1/KDM1A and histone deacetylases 1 and 2 (HDAC1/2) and the finding that all three CoRESTs are expressed in the adult rat brain. CoRESTs interact equally strongly with LSD1/KDM1A. Structural analysis shows that the overall conformation of CoREST3 is similar to that of CoREST1 complexed with LSD1/KDM1A. Nonetheless, transcriptional repressive capacity of CoREST3 is lower than that of CoREST1, which correlates with the observation that CoREST3 leads to a reduced LSD1/KDM1A catalytic efficiency. Also, CoREST2 shows a lower transcriptional repression than CoREST1, which is resistant to HDAC inhibitors. CoREST2 displays lower interaction with HDAC1/2, which is barely present in LSD1/KDM1A-CoREST2 complexes. A nonconserved leucine in the first SANT domain of CoREST2 severely weakens its association with HDAC1/2. Furthermore, CoREST2 mutants with increased HDAC1/2 interaction and those without HDAC1/2 interaction exhibit equivalent transcriptional repression capacities, indicating that CoREST2 represses in an HDAC-independent manner. In conclusion, differences among CoREST proteins are instrumental in the modulation of protein-protein interactions and catalytic activities of LSD1/KDM1A-CoREST-HDAC complexes, fine-tuning gene expression regulation.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Animals , Catalytic Domain , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , Histone Deacetylases/metabolism , Histone Demethylases/metabolism , Humans , Male , Models, Molecular , Nerve Tissue Proteins/chemistry , Protein Conformation , Rats , Rats, Sprague-Dawley
8.
J Neurochem ; 128(5): 603-16, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24111946

ABSTRACT

Epigenetic mechanisms play important roles in brain development, orchestrating proliferation, differentiation, and morphogenesis. Lysine-Specific Demethylase 1 (LSD1 also known as KDM1A and AOF2) is a histone modifier involved in transcriptional repression, forming a stable core complex with the corepressors corepressor of REST (CoREST) and histone deacetylases (HDAC1/2). Importantly, in the mammalian CNS, neuronal LSD1-8a, an alternative splicing isoform of LSD1 including the mini-exon E8a, sets alongside LSD1 and is capable of enhancing neurite growth and morphogenesis. Here, we describe that the morphogenic properties of neuronal LSD1-8a require switching off repressive activity and this negative modulation is mediated in vivo by phosphorylation of the Thr369b residue coded by exon E8a. Three-dimensional crystal structure analysis using a phospho-mimetic mutant (Thr369bAsp), indicate that phosphorylation affects the residues surrounding the exon E8a-coded amino acids, causing a local conformational change. We suggest that phosphorylation, without affecting demethylase activity, causes in neurons CoREST and HDAC1/2 corepressors detachment from LSD1-8a and impairs neuronal LSD1-8a repressive activity. In neurons, Thr369b phosphorylation is required for morphogenic activity, converting neuronal LSD1-8a in a dominant-negative isoform, challenging LSD1-mediated transcriptional repression on target genes.


Subject(s)
Co-Repressor Proteins/biosynthesis , Co-Repressor Proteins/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histone Demethylases/biosynthesis , Histone Demethylases/genetics , Transcription, Genetic/genetics , Animals , Brain Chemistry/physiology , Cells, Cultured , Chromatin/metabolism , Enzyme Repression , Exons/genetics , Gene Expression Regulation, Enzymologic/genetics , Genes, Reporter , Immunoprecipitation , Isoenzymes/metabolism , Mass Spectrometry , Mutagenesis, Site-Directed , Neurites/metabolism , Phosphorylation , Protein Conformation , Rats
9.
J Neurosci ; 30(7): 2521-32, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20164337

ABSTRACT

A variety of chromatin remodeling complexes are thought to orchestrate transcriptional programs that lead neuronal precursors from earliest commitment to terminal differentiation. Here we show that mammalian neurons have a specialized chromatin remodeling enzyme arising from a neurospecific splice variant of LSD1/KDM1, histone lysine specific demethylase 1, whose demethylase activity on Lys4 of histone H3 has been related to gene repression. We found that alternative splicing of LSD1 transcript generates four full-length isoforms from combinatorial retention of two identified exons: the 4 aa exon E8a is internal to the amine oxidase domain, and its inclusion is restricted to the nervous system. Remarkably, the expression of LSD1 splice variants is dynamically regulated throughout cortical development, particularly during perinatal stages, with a progressive increase of LSD1 neurospecific isoforms over the ubiquitous ones. Notably, the same LSD1 splice dynamics can be fairly recapitulated in cultured cortical neurons. Functionally, LSD1 isoforms display in vitro a comparable demethylase activity, yet the inclusion of the sole exon E8a reduces LSD1 repressor activity on a reporter gene. Additional distinction among isoforms is supported by the knockdown of neurospecific variants in cortical neurons resulting in the inhibition of neurite maturation, whereas overexpression of the same variants enhances it. Instead, perturbation of LSD1 isoforms that are devoid of the neurospecific exon elicits no morphogenic effect. Collectively, results demonstrate that the arousal of neuronal LSD1 isoforms pacemakes early neurite morphogenesis, conferring a neurospecific function to LSD1 epigenetic activity.


Subject(s)
Alternative Splicing/genetics , Histone Demethylases/genetics , Morphogenesis/genetics , Neurites/physiology , Neurons/cytology , Animals , Brain/cytology , Cells, Cultured , Embryo, Mammalian , Exons/genetics , Gene Expression Regulation, Developmental/physiology , Genome , Green Fluorescent Proteins/genetics , Guanylate Kinases/metabolism , HeLa Cells , Humans , Immunoprecipitation/methods , Models, Molecular , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptors, Glutamate/metabolism , SAP90-PSD95 Associated Proteins , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...