Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
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.
Int J Mol Sci ; 23(17)2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36077416

ABSTRACT

Neurofibromatosis type 2 is an autosomal dominant tumor-prone disorder mainly caused by NF2 point mutations or intragenic deletions. Few individuals with a complex phenotype and 22q12 microdeletions have been described. The 22q12 microdeletions' pathogenic effects at the genetic and epigenetic levels are currently unknown. We here report on 22q12 microdeletions' characterization in three NF2 patients with different phenotype complexities. A possible effect of the position was investigated by in silico analysis of 22q12 topologically associated domains (TADs) and regulatory elements, and by expression analysis of 12 genes flanking patients' deletions. A 147 Kb microdeletion was identified in the patient with the mildest phenotype, while two large deletions of 561 Kb and 1.8 Mb were found in the other two patients, showing a more severe symptomatology. The last two patients displayed intellectual disability, possibly related to AP1B1 gene deletion. The microdeletions change from one to five TADs, and the 22q12 chromatin regulatory landscape, according to the altered expression levels of four deletion-flanking genes, including PIK3IP1, are likely associated with an early ischemic event occurring in the patient with the largest deletion. Our results suggest that the identification of the deletion extent can provide prognostic markers, predictive of NF2 phenotypes, and potential therapeutic targets, thus overall improving patient management.


Subject(s)
Intellectual Disability , Neurofibromatosis 2 , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex beta Subunits , Humans , Intellectual Disability/genetics , Neurofibromatosis 2/genetics , Phenotype
3.
Opt Lett ; 46(10): 2453-2456, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33988608

ABSTRACT

We demonstrate optically tunable control of second-harmonic generation in all-dielectric nanoantennas: by using a control beam that is absorbed by the nanoresonator, we thermo-optically change the refractive index of the radiating element to modulate the amplitude of the second-harmonic signal. For a moderate temperature increase of roughly 40 K, modulation of the efficiency up to 60% is demonstrated; this large tunability of the single meta-atom response paves the way to exciting avenues for reconfigurable homogeneous and heterogeneous metasurfaces.

4.
Nucleic Acids Res ; 47(10): 5325-5340, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30937446

ABSTRACT

Stem cell identity and plasticity are controlled by master regulatory genes and complex circuits also involving non-coding RNAs. Circular RNAs (circRNAs) are a class of RNAs generated from protein-coding genes by backsplicing, resulting in stable RNA structures devoid of free 5' and 3' ends. Little is known of the mechanisms of action of circRNAs, let alone in stem cell biology. In this study, for the first time, we determined that a circRNA controls mesenchymal stem cell (MSC) identity and differentiation. High-throughput MSC expression profiling from different tissues revealed a large number of expressed circRNAs. Among those, circFOXP1 was enriched in MSCs compared to differentiated mesodermal derivatives. Silencing of circFOXP1 dramatically impaired MSC differentiation in culture and in vivo. Furthermore, we demonstrated a direct interaction between circFOXP1 and miR-17-3p/miR-127-5p, which results in the modulation of non-canonical Wnt and EGFR pathways. Finally, we addressed the interplay between canonical and non-canonical Wnt pathways. Reprogramming to pluripotency of MSCs reduced circFOXP1 and non-canonical Wnt, whereas canonical Wnt was boosted. The opposing effect was observed during generation of MSCs from human pluripotent stem cells. Our results provide unprecedented evidence for a regulatory role for circFOXP1 as a gatekeeper of pivotal stem cell molecular networks.


Subject(s)
Forkhead Transcription Factors/metabolism , MicroRNAs/metabolism , RNA , Repressor Proteins/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , ErbB Receptors/metabolism , Exoribonucleases/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Gene Silencing , HEK293 Cells , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , RNA, Circular , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Stem Cells/cytology , Wnt Proteins/metabolism
5.
J Neurochem ; 155(1): 98-110, 2020 10.
Article in English | MEDLINE | ID: mdl-32141088

ABSTRACT

Acute environmental stress rarely implies long-lasting neurophysiological and behavioral alterations. On the contrary, chronic stress exerts a potent toxic effect at the glutamatergic synapse whose altered physiology has been recognized as a core trait of neuropsychiatric disorders. The endocannabinoid system (ECS) plays an important role in the homeostatic response to acute stress. In particular, stress induces synthesis of endocannabinoid (eCB) 2-arachidonyl glycerol (2-AG). 2-AG stimulates presynaptic cannabinoid 1 (CB1) receptor contributing to stress response termination through inhibition of glutamate release, restraining thereafter anxiety arousal. We employ mouse models of stress response coupled to gene expression analyses, unravelling that in response to acute psychosocial stress in the mouse hippocampus, ECS-mediated synaptic modulation is enhanced via transcriptional repression of two enzymes involved in 2-AG degradation: α/ß-hydrolase domain containing 6 (ABHD6) and monoacylglycerol lipase (MAGL). Such a process is orchestrated by the epigenetic corepressor LSD1 who directly interacts with promoter regulatory regions of Abhd6 and Magl. Remarkably, negative transcriptional control of Abhd6 and Magl is lost in the hippocampus upon chronic psychosocial stress, possibly contributing to trauma-induced drift of synapse physiology toward uncontrolled glutamate transmission. We previously showed that in mice lysine-specific demethylase 1 (LSD1) increases its hippocampal expression in response to psychosocial stress preventing excessive consolidation of anxiety-related plasticity. In this work, we unravel a nodal epigenetic modulation of eCB turn over, shedding new light on the molecular substrate of converging stress-terminating effects displayed by ECS and LSD1.


Subject(s)
Endocannabinoids/physiology , Histone Demethylases/metabolism , Stress Disorders, Traumatic, Acute/physiopathology , Animals , Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , Epigenetic Repression , Gene Expression Regulation , Glycerides/pharmacology , Hippocampus/metabolism , Histone Demethylases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoacylglycerol Lipases/biosynthesis , Monoacylglycerol Lipases/genetics , Receptor, Cannabinoid, CB1/agonists , Social Environment , Stress, Psychological
6.
Opt Express ; 28(15): 22186-22199, 2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32752485

ABSTRACT

A novel spectroscopy technique to enable the rapid characterization of discrete mid-infrared integrated photonic waveguides is demonstrated. The technique utilizes lithography patterned polymer blocks that absorb light strongly within the molecular fingerprint region. These act as integrated waveguide detectors when combined with an atomic force microscope that measures the photothermal expansion when infrared light is guided to the block. As a proof of concept, the technique is used to experimentally characterize propagation loss and grating coupler response of Ge-on-Si waveguides at wavelengths from 6 to 10 µm. In addition, when the microscope is operated in scanning mode at fixed wavelength, the guided mode exiting the output facet is imaged with a lateral resolution better than 500 nm i.e. below the diffraction limit. The characterization technique can be applied to any mid-infrared waveguide platform and can provide non-destructive in-situ testing of discrete waveguide components.

7.
Mar Drugs ; 18(8)2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32781644

ABSTRACT

Collagen-based skin-like scaffolds (CBSS) are promising alternatives to skin grafts to repair wounds and injuries. In this work, we propose that the common marine invertebrate sea urchin represents a promising and eco-friendly source of native collagen to develop innovative CBSS for skin injury treatment. Sea urchin food waste after gonad removal was here used to extract fibrillar glycosaminoglycan (GAG)-rich collagen to produce bilayer (2D + 3D) CBSS. Microstructure, mechanical stability, permeability to water and proteins, ability to exclude bacteria and act as scaffolding for fibroblasts were evaluated. Our data show that the thin and dense 2D collagen membrane strongly reduces water evaporation (less than 5% of water passes through the membrane after 7 days) and protein diffusion (less than 2% of BSA passes after 7 days), and acts as a barrier against bacterial infiltration (more than 99% of the different tested bacterial species is retained by the 2D collagen membrane up to 48 h), thus functionally mimicking the epidermal layer. The thick sponge-like 3D collagen scaffold, structurally and functionally resembling the dermal layer, is mechanically stable in wet conditions, biocompatible in vitro (seeded fibroblasts are viable and proliferate), and efficiently acts as a scaffold for fibroblast infiltration. Thus, thanks to their chemical and biological properties, CBSS derived from sea urchins might represent a promising, eco-friendly, and economically sustainable biomaterial for tissue regenerative medicine.


Subject(s)
Fibrillar Collagens/pharmacology , Fibroblasts/physiology , Regenerative Medicine , Sea Urchins/chemistry , Seafood , Skin, Artificial , Tissue Scaffolds , Waste Products , Animals , Cell Culture Techniques , Cell Line , Cell Proliferation , Cell Survival , Cricetinae , Fibrillar Collagens/chemistry , Fibrillar Collagens/isolation & purification , Fibroblasts/metabolism , Food Handling
8.
Nano Lett ; 19(5): 3104-3114, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30950626

ABSTRACT

Photosensitive proteins embedded in the cell membrane (about 5 nm thickness) act as photoactivated proton pumps, ion gates, enzymes, or more generally, as initiators of stimuli for the cell activity. They are composed of a protein backbone and a covalently bound cofactor (e.g. the retinal chromophore in bacteriorhodopsin (BR), channelrhodopsin, and other opsins). The light-induced conformational changes of both the cofactor and the protein are at the basis of the physiological functions of photosensitive proteins. Despite the dramatic development of microscopy techniques, investigating conformational changes of proteins at the membrane monolayer level is still a big challenge. Techniques based on atomic force microscopy (AFM) can detect electric currents through protein monolayers and even molecular binding forces in single-protein molecules but not the conformational changes. For the latter, Fourier-transform infrared spectroscopy (FTIR) using difference-spectroscopy mode is typically employed, but it is performed on macroscopic liquid suspensions or thick films containing large amounts of purified photosensitive proteins. In this work, we develop AFM-assisted, tip-enhanced infrared difference-nanospectroscopy to investigate light-induced conformational changes of the bacteriorhodopsin mutant D96N in single submicrometric native purple membrane patches. We obtain a significant improvement compared with the signal-to-noise ratio of standard IR nanospectroscopy techniques by exploiting the field enhancement in the plasmonic nanogap that forms between a gold-coated AFM probe tip and an ultraflat gold surface, as further supported by electromagnetic and thermal simulations. IR difference-spectra in the 1450-1800 cm-1 range are recorded from individual patches as thin as 10 nm, with a diameter of less than 500 nm, well beyond the diffraction limit for FTIR microspectroscopy. We find clear spectroscopic evidence of a branching of the photocycle for BR molecules in direct contact with the gold surfaces, with equal amounts of proteins either following the standard proton-pump photocycle or being trapped in an intermediate state not directly contributing to light-induced proton transport. Our results are particularly relevant for BR-based optoelectronic and energy-harvesting devices, where BR molecular monolayers are put in contact with metal surfaces, and, more generally, for AFM-based IR spectroscopy studies of conformational changes of proteins embedded in intrinsically heterogeneous native cell membranes.


Subject(s)
Bacteriorhodopsins/ultrastructure , Membrane Proteins/ultrastructure , Mutant Proteins/ultrastructure , Proton Pumps/ultrastructure , Bacteriorhodopsins/chemistry , Bacteriorhodopsins/genetics , Electromagnetic Fields , Ion Transport/genetics , Membrane Proteins/chemistry , Microscopy, Atomic Force , Mutant Proteins/chemistry , Mutant Proteins/genetics , Nanotechnology/methods , Protein Conformation , Proton Pumps/chemistry , Purple Membrane/chemistry , Purple Membrane/ultrastructure , Spectroscopy, Fourier Transform Infrared
9.
Int J Mol Sci ; 21(17)2020 Aug 29.
Article in English | MEDLINE | ID: mdl-32872402

ABSTRACT

There is no argument with regard to the physical and psychological stress-related nature of neuropsychiatric disorders. Yet, the mechanisms that facilitate disease onset starting from molecular stress responses are elusive. Environmental stress challenges individuals' equilibrium, enhancing homeostatic request in the attempt to steer down arousal-instrumental molecular pathways that underlie hypervigilance and anxiety. A relevant homeostatic pathway is the endocannabinoid system (ECS). In this review, we summarize recent discoveries unambiguously listing ECS as a stress coping mechanism. As stress evokes huge excitatory responses in emotional-relevant limbic areas, the ECS limits glutamate release via 2-arachydonilglycerol (2-AG) stress-induced synthesis and retrograde cannabinoid 1 (CB1)-receptor activation at the synapse. However, ECS shows intrinsic vulnerability as 2-AG overstimulation by chronic stress rapidly leads to CB1-receptor desensitization. In this review, we emphasize the protective role of 2-AG in stress-response termination and stress resiliency. Interestingly, we discuss ECS regulation with a further nuclear homeostatic system whose nature is exquisitely epigenetic, orchestrated by Lysine Specific Demethylase 1. We here emphasize a remarkable example of stress-coping network where transcriptional homeostasis subserves synaptic and behavioral adaptation, aiming at reducing psychiatric effects of traumatic experiences.


Subject(s)
Adaptation, Psychological , Endocannabinoids/metabolism , Histone Demethylases/metabolism , Animals , Arachidonic Acids/metabolism , Epigenesis, Genetic , Glutamic Acid/metabolism , Glycerides/metabolism , Homeostasis , Humans , Receptor, Cannabinoid, CB1/metabolism
10.
Int J Mol Sci ; 21(9)2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344798

ABSTRACT

Psychiatric disorders represent a heterogeneous class of multifactorial mental diseases whose origin entails a pathogenic integration of genetic and environmental influences. Incidence of these pathologies is dangerously high, as more than 20% of the Western population is affected. Despite the diverse origins of specific molecular dysfunctions, these pathologies entail disruption of fine synaptic regulation, which is fundamental to behavioral adaptation to the environment. The synapses, as functional units of cognition, represent major evolutionary targets. Consistently, fine synaptic tuning occurs at several levels, involving a novel class of molecular regulators known as long non-coding RNAs (lncRNAs). Non-coding RNAs operate mainly in mammals as epigenetic modifiers and enhancers of proteome diversity. The prominent evolutionary expansion of the gene number of lncRNAs in mammals, particularly in primates and humans, and their preferential neuronal expression does represent a driving force that enhanced the layering of synaptic control mechanisms. In the last few years, remarkable alterations of the expression of lncRNAs have been reported in psychiatric conditions such as schizophrenia, autism, and depression, suggesting unprecedented mechanistic insights into disruption of fine synaptic tuning underlying severe behavioral manifestations of psychosis. In this review, we integrate literature data from rodent pathological models and human evidence that proposes the biology of lncRNAs as a promising field of neuropsychiatric investigation.


Subject(s)
Epigenesis, Genetic , Mental Disorders/genetics , RNA, Long Noncoding/genetics , Synaptic Transmission/genetics , Animals , Central Nervous System/metabolism , Disease Models, Animal , Endocannabinoids/physiology , Evolution, Molecular , Gene Expression Regulation/genetics , Humans , Hypothalamo-Hypophyseal System/physiopathology , Mammals/genetics , Mental Disorders/epidemiology , Mental Disorders/metabolism , Mental Disorders/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neuronal Plasticity/genetics , Pituitary-Adrenal System/physiopathology , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/classification , Synaptic Transmission/physiology
11.
Int J Mol Sci ; 21(20)2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33050350

ABSTRACT

Emotional and cognitive information processing represent higher-order brain functions. They require coordinated interaction of specialized brain areas via a complex spatial and temporal equilibrium among neuronal cell-autonomous, circuitry, and network mechanisms. The delicate balance can be corrupted by stressful experiences, increasing the risk of developing psychopathologies in vulnerable individuals. Neuropsychiatric disorders affect twenty percent of the western world population, but therapies are still not effective for some patients. Elusive knowledge of molecular pathomechanisms and scarcity of objective biomarkers in humans present complex challenges, while the adoption of rodent models helps to improve our understanding of disease correlate and aids the search for novel pharmacological targets. Stress administration represents a strategy to induce, trace, and modify molecular and behavioral endophenotypes of mood disorders in animals. However, a mouse or rat model will only display one or a few endophenotypes of a specific human psychopathology, which cannot be in any case recapitulated as a whole. To override this issue, shared criteria have been adopted to deconstruct neuropsychiatric disorders, i.e., depression, into specific behavioral aspects, and inherent neurobiological substrates, also recognizable in lower mammals. In this work, we provide a rationale for rodent models of stress administration. In particular, comparing each rodent model with a real-life human traumatic experience, we intend to suggest an introductive guide to better comprehend and interpret these paradigms.


Subject(s)
Mental Disorders/etiology , Stress, Physiological , Stress, Psychological/complications , Animals , Disease Management , Disease Models, Animal , Disease Susceptibility , Environment , Humans , Mental Disorders/diagnosis , Mental Disorders/therapy , Prognosis , Research
12.
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
13.
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
14.
Cereb Cortex ; 27(3): 2226-2248, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27005990

ABSTRACT

Alterations in the balance of inhibitory and excitatory synaptic transmission have been implicated in the pathogenesis of neurological disorders such as epilepsy. Eukaryotic elongation factor 2 kinase (eEF2K) is a highly regulated, ubiquitous kinase involved in the control of protein translation. Here, we show that eEF2K activity negatively regulates GABAergic synaptic transmission. Indeed, loss of eEF2K increases GABAergic synaptic transmission by upregulating the presynaptic protein Synapsin 2b and α5-containing GABAA receptors and thus interferes with the excitation/inhibition balance. This cellular phenotype is accompanied by an increased resistance to epilepsy and an impairment of only a specific hippocampal-dependent fear conditioning. From a clinical perspective, our results identify eEF2K as a potential novel target for antiepileptic drugs, since pharmacological and genetic inhibition of eEF2K can revert the epileptic phenotype in a mouse model of human epilepsy.


Subject(s)
Elongation Factor 2 Kinase/metabolism , Epilepsy/enzymology , Neurons/enzymology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Conditioning, Psychological/physiology , Disease Models, Animal , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/genetics , Epilepsy/pathology , Fear/physiology , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Neurons/pathology , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Synapsins/genetics , Synapsins/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
15.
Int J Mol Sci ; 19(7)2018 Jul 11.
Article in English | MEDLINE | ID: mdl-29997370

ABSTRACT

Cyclin-dependent kinase 5 regulatory subunit 1 (CDK5R1) gene encodes for p35, the main activator of Cyclin-dependent kinase 5 (CDK5). The active p35/CDK5 complex is involved in numerous aspects of brain development and function, and its deregulation is closely associated to Alzheimer's disease (AD) onset and progression. We recently showed that miR-15/107 family can negatively regulate CDK5R1 expression modifying mRNA stability. Interestingly, miRNAs belonging to miR-15/107 family are downregulated in AD brain while CDK5R1 is upregulated. Long non-coding RNAs (lncRNAs) are emerging as master regulators of gene expression, including miRNAs, and their dysregulation has been implicated in the pathogenesis of AD. Here, we evaluated the existence of an additional layer of CDK5R1 expression regulation provided by lncRNAs. In particular, we focused on three lncRNAs potentially regulating CDK5R1 expression levels, based on existing data: NEAT1, HOTAIR, and MALAT1. We demonstrated that NEAT1 and HOTAIR negatively regulate CDK5R1 mRNA levels, while MALAT1 has a positive effect. We also showed that all three lncRNAs positively control miR-15/107 family of miRNAs. Moreover, we evaluated the expression of NEAT1, HOTAIR, and MALAT1 in AD and control brain tissues. Interestingly, NEAT1 displayed increased expression levels in temporal cortex and hippocampus of AD patients. Interestingly, we observed a strong positive correlation between CDK5R1 and NEAT1 expression levels in brain tissues, suggesting a possible neuroprotective role of NEAT1 in AD to compensate for increased CDK5R1 levels. Overall, our work provides evidence of another level of CDK5R1 expression regulation mediated by lncRNAs and points to NEAT1 as a biomarker, as well as a potential pharmacological target for AD therapy.


Subject(s)
Alzheimer Disease/metabolism , Nerve Tissue Proteins/genetics , RNA, Long Noncoding/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Disease Progression , Gene Expression Regulation , Genetic Markers , HeLa Cells , Hippocampus/metabolism , Humans , Temporal Lobe/metabolism
16.
Hum Genet ; 136(10): 1329-1339, 2017 10.
Article in English | MEDLINE | ID: mdl-28776093

ABSTRACT

Neurofibromatosis type I (NF1) microdeletion syndrome, which is present in 4-11% of NF1 patients, is associated with a severe phenotype as it is caused by the deletion of NF1 and other genes in the 17q11.2 region. The variable expressivity of the disease makes it challenging to establish genotype-phenotype correlations, which also affects prognosis and counselling. We here describe a 3-year-old NF1 patient with an atypical deletion and a complex phenotype. The patient showed overgrowth, café au lait spots, inguinal freckling, and neurological abnormalities. The extent of the deletion was determined by means of array comparative genomic hybridisation, and its breakpoints were isolated by means of long-range polymerase chain reaction. Sequence analysis of the deletion junction fragment revealed the occurrence of an Alu-mediated recombination that led to the generation of a chimeric gene consisting of three exons of RNF135 and eleven exons of SUZ12. Interestingly, the deletion shares a common RNF135-centred region with another deletion described in a non-NF1 patient with overgrowth. In comparison with the normal RNF135 allele, the chimeric transcript was 350-fold over-expressed in peripheral blood, and the ADAP2 gene located upstream of RNF135 was also up-regulated. In line with this, the deletion causes the loss of a chromatin TD boundary, which entails the aberrant adoption of distal cis-acting regulatory elements. These findings suggest that RNF135 haploinsufficiency is related to overgrowth in patients with NF1 microdeletion syndrome and, for the first time, strongly indicate a position effect that warrants further genotype-phenotype correlation studies to investigate the possible existence of previously unknown pathogenic mechanisms.


Subject(s)
Chromosomal Position Effects , Chromosome Deletion , GTPase-Activating Proteins , Gene Expression Regulation, Neoplastic , Neurofibromatosis 1 , Polycomb Repressive Complex 2 , Recombination, Genetic , Ubiquitin-Protein Ligases , Alleles , Child, Preschool , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/genetics , Humans , Male , Neoplasm Proteins , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Oncogene Proteins, Fusion , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
17.
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
18.
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
19.
Cell Rep ; 39(8): 110857, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35613587

ABSTRACT

Protocadherin-19 (PCDH19) is a synaptic cell-adhesion molecule encoded by X-linked PCDH19, a gene linked with epilepsy. Here, we report a synapse-to-nucleus signaling pathway through which PCDH19 bridges neuronal activity with gene expression. In particular, we describe the NMDA receptor (NMDAR)-dependent proteolytic cleavage of PCDH19, which leads to the generation of a PCDH19 C-terminal fragment (CTF) able to enter the nucleus. We demonstrate that PCDH19 CTF associates with chromatin and with the chromatin remodeler lysine-specific demethylase 1 (LSD1) and regulates expression of immediate-early genes (IEGs). Our results are consistent with a model whereby PCDH19 favors maintenance of neuronal homeostasis via negative feedback regulation of IEG expression and provide a key to interpreting PCDH19-related hyperexcitability.


Subject(s)
Cadherins , Epilepsy , Genes, Immediate-Early , Protocadherins , Cadherins/genetics , Cadherins/metabolism , Chromatin/genetics , Chromatin/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Gene Expression Regulation , Humans , Protocadherins/genetics , Protocadherins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
20.
Environ Int ; 163: 107200, 2022 05.
Article in English | MEDLINE | ID: mdl-35349910

ABSTRACT

Three-dimensional (3D) structured organoids are the most advanced in vitro models for studying human health effects, but their application to evaluate the biological effects associated with microplastic exposure was neglected until now. Fibers from synthetic clothes and fabrics are a major source of airborne microplastics, and their release from dryer machines is poorly understood. We quantified and characterized the microplastic fibers (MPFs) released in the exhaust filter of a household dryer and tested their effects on airway organoids (1, 10, and 50 µg mL-1) by optical microscopy, scanning electron microscopy (SEM), confocal microscopy and quantitative reverse transcription-polymerase chain reaction (qRT-PCR). While the presence of MPFs did not inhibit organoid growth, we observed a significant reduction of SCGB1A1 gene expression related to club cell functionality and a polarized cell growth along the fibers. The MPFs did not cause relevant inflammation or oxidative stress but were coated with a cellular layer, resulting in the inclusion of fibers in the organoid. This effect could have long-term implications regarding lung epithelial cells undergoing repair. This exposure study using human airway organoids proved suitability of the model for studying the effects of airborne microplastic contamination on humans and could form the basis for further research regarding the toxicological assessment of emerging contaminants such as micro- or nanoplastics.


Subject(s)
Microplastics , Plastics , Humans , Organoids , Textiles
SELECTION OF CITATIONS
SEARCH DETAIL