Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neurosci ; 43(13): 2398-2423, 2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-36849418

RESUMO

The severity of Alzheimer's disease (AD) progression involves a complex interplay of genetics, age, and environmental factors orchestrated by histone acetyltransferase (HAT)-mediated neuroepigenetic mechanisms. While disruption of Tip60 HAT action in neural gene control is implicated in AD, alternative mechanisms underlying Tip60 function remain unexplored. Here, we report a novel RNA binding function for Tip60 in addition to its HAT function. We show that Tip60 preferentially interacts with pre-mRNAs emanating from its chromatin neural gene targets in the Drosophila brain and this RNA binding function is conserved in human hippocampus and disrupted in Drosophila brains that model AD pathology and in AD patient hippocampus of either sex. Since RNA splicing occurs co-transcriptionally and alternative splicing (AS) defects are implicated in AD, we investigated whether Tip60-RNA targeting modulates splicing decisions and whether this function is altered in AD. Replicate multivariate analysis of transcript splicing (rMATS) analysis of RNA-Seq datasets from wild-type and AD fly brains revealed a multitude of mammalian-like AS defects. Strikingly, over half of these altered RNAs are identified as bona-fide Tip60-RNA targets that are enriched for in the AD-gene curated database, with some of these AS alterations prevented against by increasing Tip60 in the fly brain. Further, human orthologs of several Tip60-modulated splicing genes in Drosophila are well characterized aberrantly spliced genes in human AD brains, implicating disruption of Tip60's splicing function in AD pathogenesis. Our results support a novel RNA interaction and splicing regulatory function for Tip60 that may underly AS impairments that hallmark AD etiology.SIGNIFICANCE STATEMENT Alzheimer's disease (AD) has recently emerged as a hotbed for RNA alternative splicing (AS) defects that alter protein function in the brain yet causes remain unclear. Although recent findings suggest convergence of epigenetics with co-transcriptional AS, whether epigenetic dysregulation in AD pathology underlies AS defects remains unknown. Here, we identify a novel RNA interaction and splicing regulatory function for Tip60 histone acetyltransferase (HAT) that is disrupted in Drosophila brains modeling AD pathology and in human AD hippocampus. Importantly, mammalian orthologs of several Tip60-modulated splicing genes in Drosophila are well characterized aberrantly spliced genes in human AD brain. We propose that Tip60-mediated AS modulation is a conserved critical posttranscriptional step that may underlie AS defects now characterized as hallmarks of AD.


Assuntos
Doença de Alzheimer , Proteínas de Drosophila , Animais , Humanos , Doença de Alzheimer/metabolismo , Proteínas de Drosophila/metabolismo , Precursores de RNA/genética , Precursores de RNA/metabolismo , Processamento Alternativo/genética , DNA Recombinante/metabolismo , Drosophila/fisiologia , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Mamíferos
2.
Mol Cell Neurosci ; 127: 103888, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37598897

RESUMO

Nucleocytoplasmic transport (NCT) in neurons is critical for enabling proteins to enter the nucleus and regulate plasticity genes in response to environmental cues. Such experience-dependent (ED) neural plasticity is central for establishing memory formation and cognitive function and can influence the severity of neurodegenerative disorders like Alzheimer's disease (AD). ED neural plasticity is driven by histone acetylation (HA) mediated epigenetic mechanisms that regulate dynamic activity-dependent gene transcription profiles in response to neuronal stimulation. Yet, how histone acetyltransferases (HATs) respond to extracellular cues in the in vivo brain to drive HA-mediated activity-dependent gene control remains unclear. We previously demonstrated that extracellular stimulation of rat hippocampal neurons in vitro triggers Tip60 HAT nuclear import with concomitant synaptic gene induction. Here, we focus on investigating Tip60 HAT subcellular localization and NCT specifically in neuronal activity-dependent gene control by using the learning and memory mushroom body (MB) region of the Drosophila brain as a powerful in vivo cognitive model system. We used immunohistochemistry (IHC) to compare the subcellular localization of Tip60 HAT in the Drosophila brain under normal conditions and in response to stimulation of fly brain neurons in vivo either by genetically inducing potassium channels activation or by exposure to natural positive ED conditions. Furthermore, we found that both inducible and ED condition-mediated neural induction triggered Tip60 nuclear import with concomitant induction of previously identified Tip60 target genes and that Tip60 levels in both the nucleus and cytoplasm were significantly decreased in our well-characterized Drosophila AD model. Mutagenesis of a putative nuclear localization signal (NLS) sequence and nuclear export signal (NES) sequence that we identified in the Drosophila Tip60 protein revealed that both are functionally required for appropriate Tip60 subcellular localization. Our results support a model by which neuronal stimulation triggers Tip60 NCT via its NLS and NES sequences to promote induction of activity-dependent neuroplasticity gene transcription and that this process may be disrupted in AD.


Assuntos
Doença de Alzheimer , Proteínas de Drosophila , Animais , Ratos , Transporte Ativo do Núcleo Celular , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Regulação da Expressão Gênica , Drosophila/metabolismo , Doença de Alzheimer/metabolismo , Plasticidade Neuronal/genética , Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Histona Acetiltransferases
3.
Mol Cell Neurosci ; 109: 103570, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33160016

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disorder hallmarked by amyloid-ß (Aß) plaque accumulation, neuronal cell death, and cognitive deficits that worsen during disease progression. Histone acetylation dysregulation, caused by an imbalance between reduced histone acetyltransferases (HAT) Tip60 and increased histone deacetylase 2 (HDAC2) levels, can directly contribute to AD pathology. However, whether such AD-associated neuroepigenetic alterations occur in response to Aß peptide production and can be protected against by increasing Tip60 levels over the course of neurodegenerative progression remains unknown. Here we profile Tip60 HAT/HDAC2 dynamics and transcriptome-wide changes across early and late stage AD pathology in the Drosophila brain produced solely by human amyloid-ß42. We show that early Aß42 induction leads to disruption of Tip60 HAT/HDAC2 balance during early neurodegenerative stages preceding Aß plaque accumulation that persists into late AD stages. Correlative transcriptome-wide studies reveal alterations in biological processes we classified as transient (early-stage only), late-onset (late-stage only), and constant (both). Increasing Tip60 HAT levels in the Aß42 fly brain protects against AD functional pathologies that include Aß plaque accumulation, neural cell death, cognitive deficits, and shorter life-span. Strikingly, Tip60 protects against Aß42-induced transcriptomic alterations via distinct mechanisms during early and late stages of neurodegeneration. Our findings reveal distinct modes of neuroepigenetic gene changes and Tip60 neuroprotection in early versus late stages in AD that can serve as early biomarkers for AD, and support the therapeutic potential of Tip60 over the course of AD progression.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Histona Acetiltransferases/fisiologia , Degeneração Neural/genética , Fragmentos de Peptídeos/toxicidade , Transcriptoma , Acetilação , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Apoptose , Aprendizagem por Associação/fisiologia , Modelos Animais de Doenças , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Epigênese Genética , Regulação da Expressão Gênica , Código das Histonas , Histona Desacetilase 2/fisiologia , Larva , Locomoção , Longevidade , Aprendizagem em Labirinto , Odorantes , Processamento de Proteína Pós-Traducional , Olfato/fisiologia
4.
Mol Cell Neurosci ; 101: 103412, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31682915

RESUMO

Genomic reorganizations mediating the engagement of target genes to transcription factories (TFs), characterized as specialized nuclear subcompartments enriched in hyperphosphorylated RNA polymerase II (RNAPII) and transcriptional regulators, act as an important layer of control in coordinating efficient gene transcription. However, their presence in hippocampal neurons and potential role in activity-dependent coregulation of genes within the brain remains unclear. Here, we investigate whether the well-characterized role for the histone acetyltransferase (HAT) Tip60 in mediating epigenetic control of inducible neuroplasticity genes involves TF associated chromatin reorganization in the hippocampus. We show that Tip60 shuttles into the nucleus following extracellular stimulation of rat hippocampal neurons with concomitant enhancement of Tip60 binding and activation of specific synaptic plasticity genes. Multicolor three-dimensional (3D) DNA fluorescent in situ hybridization (DNA-FISH) reveals that hippocampal stimulation mobilizes these same synaptic plasticity genes and Tip60 to RNAPII-rich TFs. Our data support a model by which external hippocampal stimulation promotes intracellular Tip60 HAT dynamics with concomitant TF associated genome reorganization to initiate Tip60mediated synaptic gene activation.


Assuntos
Hipocampo/metabolismo , Lisina Acetiltransferase 5/metabolismo , Plasticidade Neuronal/genética , Potenciais de Ação , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Montagem e Desmontagem da Cromatina , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Lisina Acetiltransferase 5/genética , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Ratos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
J Neurosci ; 38(19): 4569-4583, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29654189

RESUMO

Cognitive decline is a debilitating hallmark during preclinical stages of Alzheimer's disease (AD), yet the causes remain unclear. Because histone acetylation homeostasis is critical for mediating epigenetic gene control throughout neuronal development, we postulated that its misregulation contributes to cognitive impairment preceding AD pathology. Here, we show that disruption of Tip60 histone acetlytransferase (HAT)/histone deacetylase 2 (HDAC2) homeostasis occurs early in the brain of an AD-associated amyloid precursor protein (APP) Drosophila model and triggers epigenetic repression of neuroplasticity genes well before Aß plaques form in male and female larvae. Repressed genes display enhanced HDAC2 binding and reduced Tip60 and histone acetylation enrichment. Increasing Tip60 in the AD-associated APP brain restores Tip60 HAT/HDAC2 balance by decreasing HDAC2 levels, reverses neuroepigenetic alterations to activate synaptic plasticity genes, and reinstates brain morphology and cognition. Such Drosophila neuroplasticity gene epigenetic signatures are conserved in male and female mouse hippocampus and their expression and Tip60 function is compromised in hippocampus from AD patients. We suggest that Tip60 HAT/HDAC2-mediated epigenetic gene disruption is a critical initial step in AD that is reversed by restoring Tip60 in the brain.SIGNIFICANCE STATEMENT Mild cognitive impairment is a debilitating hallmark during preclinical stages of Alzheimer's disease (AD), yet its causes remain unclear. Although recent findings support elevated histone deacetylase 2 (HDAC2) as a cause for epigenetic repression of synaptic genes that contribute to cognitive deficits, whether alterations in histone acetlytransferase (HAT) levels that counterbalance HDAC2 repressor action occur and the identity of these HATs remain unknown. We demonstrate that disruption of Tip60 HAT/HDAC2 homeostasis occurs early in the AD Drosophila brain and triggers epigenetic repression of neuroplasticity genes before Aß plaques form. Increasing Tip60 in the AD brain restores Tip60 HAT/HDAC2 balance, reverses neuroepigenetic alterations to activate synaptic genes, and reinstates brain morphology and cognition. Our data suggest that disruption of the Tip60 HAT/HDAC2 balance is a critical initial step in AD.


Assuntos
Transtornos Cognitivos/genética , Transtornos Cognitivos/terapia , Repressão Epigenética/genética , Histona Desacetilase 2/genética , Lisina Acetiltransferase 5/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Idoso de 80 Anos ou mais , Animais , Drosophila melanogaster , Feminino , Homeostase/genética , Homeostase/fisiologia , Humanos , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/genética , Placa Amiloide/genética , Placa Amiloide/prevenção & controle , Olfato
6.
J Neurochem ; 143(3): 320-333, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28628228

RESUMO

Schizophrenia (SCZ) is characterized not only by psychosis, but also by working memory and executive functioning deficiencies, processes that rely on the prefrontal cortex (PFC). Because these cognitive impairments emerge prior to psychosis onset, we investigated synaptic function during development in the neurodevelopmental methylazoxymethanol (MAM) model for SCZ. Specifically, we hypothesize that N-methyl-D-aspartate receptor (NMDAR) hypofunction is attributable to reductions in the NR2B subunit through aberrant epigenetic regulation of gene expression, resulting in deficient synaptic physiology and PFC-dependent cognitive dysfunction, a hallmark of SCZ. Using western blot and whole-cell patch-clamp electrophysiology, we found that the levels of synaptic NR2B protein are significantly decreased in juvenile MAM animals, and the function of NMDARs is substantially compromised. Both NMDA-mEPSCs and synaptic NMDA-eEPSCs are significantly reduced in prelimbic PFC (plPFC). This protein loss during the juvenile period is correlated with an aberrant increase in enrichment of the epigenetic transcriptional repressor RE1-silencing transcription factor (REST) and the repressive histone marker H3K27me3 at the Grin2b promoter, as assayed by ChIP-quantitative polymerase chain reaction. Glutamate hypofunction has been a prominent hypothesis in the understanding of SCZ pathology; however, little attention has been given to the NMDAR system in the developing PFC in models for SCZ. Our work is the first to confirm that NMDAR hypofunction is a feature of early postnatal development, with epigenetic hyper-repression of the Grin2b promoter being a contributing factor. The selective loss of NR2B protein and subsequent synaptic dysfunction weakens plPFC function during development and may underlie early cognitive impairments in SCZ models and patients. Read the Editorial Highlight for this article on page 264.


Assuntos
Epigênese Genética/fisiologia , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/patologia , Animais , Animais Recém-Nascidos , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Epigênese Genética/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Técnicas In Vitro , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/toxicidade , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/efeitos dos fármacos , Gravidez , Ratos , Receptores de N-Metil-D-Aspartato/genética , Esquizofrenia/induzido quimicamente , Esquizofrenia/complicações , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
7.
J Neurosci ; 33(17): 7535-47, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23616558

RESUMO

Axonal transport defects and axonopathy are prominent in early preclinical stages of Alzheimer's disease (AD), often preceding known disease-related pathology by over a year. As epigenetic transcriptional regulatory mechanisms, such as histone acetylation, are critical for neurogenesis, it is postulated that their misregulation might be linked to early pathophysiological mechanisms that contribute to AD. The histone acetyltransferase (HAT) Tip60 epigenetically regulates genes enriched for neuronal functions and is implicated in AD via its formation of a transcriptional regulatory complex with the amyloid precursor protein (APP) intracellular domain. Disruption of APP function is associated with axonal transport defects, raising the possibility that an epigenetic role for Tip60 might also be involved. Here, we examine whether Tip60 HAT activity functions in axonal transport using Drosophila CNS motor neurons as a well-characterized transport model. We show that reduction of Tip60 HAT activity in the nervous system causes axonopathy and transport defects associated with epigenetic misregulation of certain axonal transport-linked Tip60 target genes. Functional consequences of these defects are evidenced by reduced locomotion activity of the mutant Tip60 larvae, and these phenotypes can be partially rescued with certain histone deacetylase inhibitors. Finally, we demonstrate that Tip60 function in axonal transport is mediated by APP and that, remarkably, excess Tip60 exerts a neuroprotective role in APP-induced axonal transport and functional locomotion defects. Our observations highlight a novel functional interactive role between Tip60 HAT activity and APP in axonal transport and provide insight into the importance of specific HAT modulators for cognitive disorder treatment.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Transporte Axonal/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Histona Acetiltransferases/genética , Fenótipo , Doença de Alzheimer/prevenção & controle , Animais , Animais Geneticamente Modificados , Drosophila , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/fisiologia , Histona Acetiltransferases/biossíntese , Histona Acetiltransferases/fisiologia , Atividade Motora/genética , Regulação para Cima/genética
8.
Front Behav Neurosci ; 17: 1176777, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37351153

RESUMO

Introduction: Glutamate excitotoxicity is causal in striatal neurodegeneration underlying motor dysfunction and cognitive deficits in Huntington's disease (HD). Excitatory amino acid transporter 2 (EAAT2), the predominant glutamate transporter accounting for >90% of glutamate transport, plays a key role in preventing excitotoxicity by clearing excess glutamate from the intrasynaptic cleft. Accordingly, EAAT2 has emerged as a promising therapeutic target for prevention of neuronal excitotoxicity underlying HD and other neurodegenerative diseases. Methods: We have previously designed novel EAAT2 positive allosteric modulator GT951, GTS467, and GTS551, with low nanomolar efficacy in glutamate uptake and favorable pharmacokinetic properties. In this study, we test the neuroprotective abilities of these novel EAAT2 activators in vivo using the robust Drosophila HD transgenic model expressing human huntingtin gene with expanded repeats (Htt128Q). Results: All three compounds significantly restored motor function impaired under HD pathology over a wide dose range. Additionally, treatment with all three compounds significantly improved HD-associated olfactory associative learning and short-term memory defects, while GT951 and GTS551 also improved middle-term memory in low-performing group. Similarly, treatment with GT951 and GTS551 partially protected against early mortality observed in our HD model. Further, treatment with all three EAAT2 activators induced epigenetic expression of EAAT2 Drosophila homolog and several cognition-associated genes. Conclusion: Together, these results highlight the efficacy of GT951, GTS467 and GTS551 in treating motor and cognitive impairments under HD pathology and support their development for treatment of HD.

9.
Animal Model Exp Med ; 6(1): 3-9, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36872303

RESUMO

ß-Amyloid (Aß) is a specific pathological hallmark of Alzheimer's disease (AD). Because of its neurotoxicity, AD patients exhibit multiple brain dysfunctions. Disease-modifying therapy (DMT) is the central concept in the development of AD therapeutics today, and most DMT drugs that are currently in clinical trials are anti-Aß drugs, such as aducanumab and lecanemab. Therefore, understanding Aß's neurotoxic mechanism is crucial for Aß-targeted drug development. Despite its total length of only a few dozen amino acids, Aß is incredibly diverse. In addition to the well-known Aß1-42 , N-terminally truncated, glutaminyl cyclase (QC) catalyzed, and pyroglutamate-modified Aß (pEAß) is also highly amyloidogenic and far more cytotoxic. The extracellular monomeric Aßx-42 (x = 1-11) initiates the aggregation to form fibrils and plaques and causes many abnormal cellular responses through cell membrane receptors and receptor-coupled signal pathways. These signal cascades further influence many cellular metabolism-related processes, such as gene expression, cell cycle, and cell fate, and ultimately cause severe neural cell damage. However, endogenous cellular anti-Aß defense processes always accompany the Aß-induced microenvironment alterations. Aß-cleaving endopeptidases, Aß-degrading ubiquitin-proteasome system (UPS), and Aß-engulfing glial cell immune responses are all essential self-defense mechanisms that we can leverage to develop new drugs. This review discusses some of the most recent advances in understanding Aß-centric AD mechanisms and suggests prospects for promising anti-Aß strategies.


Assuntos
Doença de Alzheimer , Síndromes Neurotóxicas , Humanos , Peptídeos beta-Amiloides , Membrana Celular , Citoplasma
10.
Epigenetics ; 17(7): 786-807, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34369292

RESUMO

Disruption of histone acetylation-mediated gene control is a critical step in Alzheimer's Disease (AD), yet chromatin analysis of antagonistic histone acetyltransferases (HATs) and histone deacetylases (HDACs) causing these alterations remains uncharacterized. We report the first Tip60 HAT versus HDAC2 chromatin (ChIP-seq) and transcriptional (RNA-seq) profiling study in Drosophila melanogaster brains that model early human AD. We find Tip60 and HDAC2 predominantly recruited to identical neuronal genes. Moreover, AD brains exhibit robust genome-wide early alterations that include enhanced HDAC2 and reduced Tip60 binding and transcriptional dysregulation. Orthologous human genes to co-Tip60/HDAC2 D. melanogaster neural targets exhibit conserved disruption patterns in AD patient hippocampi. Notably, we discovered distinct transcription factor binding sites close or within Tip60/HDAC2 co-peaks in neuronal genes, implicating them in coenzyme recruitment. Increased Tip60 protects against transcriptional dysregulation and enhanced HDAC2 enrichment genome-wide. We advocate Tip60 HAT/HDAC2 mediated epigenetic neuronal gene disruption as a genome-wide initial causal event in AD.


Assuntos
Doença de Alzheimer , Proteínas de Drosophila , Histona Acetiltransferases , Histona Desacetilase 2 , Acetilação , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Cromatina/metabolismo , Metilação de DNA , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Epigenômica , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Humanos , Transcriptoma
11.
J Neurochem ; 115(2): 493-504, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20626565

RESUMO

The histone acetyltransferase Elp3 (Elongator Protein 3) is the catalytic subunit of the highly conserved Elongator complex. Elp3 is essential for the complex functions of Elongator in both the nucleus and cytoplasm of neurons, including the epigenetic control of neuronal motility genes and the acetylation of α-tubulin that affects axonal branching and cortical neuron migration. Accordingly, misregulation of Elp3 has been implicated in human disorders that specifically affect neuronal function, including familial dysautonomia, a disease characterized by degeneration of the sensory and autonomic nervous system, and the motor neuron degenerative disorder amyotrophic lateral sclerosis. These studies underscore the importance of Elp3 in neurodevelopment and disease, and the need to further characterize the multiple nuclear and cytoplasmic based roles of ELP3 required for neurogenesis in animal models, in vivo. In this report, we investigate the behavioral and morphological consequences that result from targeted reduction of ELP3 specifically in the developing Drosophila nervous system. We demonstrate that loss of Elp3 during neurodevelopment leads to a hyperactive phenotype and sleep loss in the adult flies, a significant expansion in synaptic bouton number and axonal length and branching in the larval neuromuscular junction as well as the misregulation of certain genes known to be involved in these processes. Our results uncover a novel role for Elp3 in the regulation of synaptic bouton expansion during neurogenesis that may be linked with a requirement for sleep.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histona Acetiltransferases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Junção Neuromuscular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Distúrbios do Início e da Manutenção do Sono/genética , Animais , Animais Geneticamente Modificados , Comportamento Animal , Drosophila , Proteínas de Drosophila/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Histona Acetiltransferases/genética , Humanos , Larva , Masculino , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Sistema Nervoso/citologia , Sistema Nervoso/enzimologia , Sistema Nervoso/crescimento & desenvolvimento , Junção Neuromuscular/genética , Interferência de RNA/fisiologia , Privação do Sono/genética
12.
Front Mol Neurosci ; 13: 577622, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33304239

RESUMO

Amyloid-ß (Aß) peptides can form protease-resistant aggregates within and outside of neurons. Accumulation of these aggregates is a hallmark of Alzheimer's disease (AD) neuropathology and contributes to devastating cognitive deficits associated with this disorder. The primary etiological factor for Aß aggregation is either an increase in Aß production or a decrease in its clearance. Aß is produced by the sequential activity of ß- and γ-secretase on the amyloid precursor protein (APP) and the clearance is mediated by chaperone-mediated mechanisms. The Aß aggregates vary from soluble monomers and oligomers to insoluble senile plaques. While excess intraneuronal oligomers can transduce neurotoxic signals into neurons causing cellular defects like oxidative stress and neuroepigenetic mediated transcriptional dysregulation, extracellular senile plaques cause neurodegeneration by impairing neural membrane permeabilization and cell signaling pathways. Paradoxically, senile plaque formation is hypothesized to be an adaptive mechanism to sequester excess toxic soluble oligomers while leaving native functional Aß levels intact. This hypothesis is strengthened by the absence of positive outcomes and side effects from immunotherapy clinical trials aimed at complete Aß clearance, and support beneficial physiological roles for native Aß in cellular function. Aß has been shown to modulate synaptic transmission, consolidate memory, and protect against excitotoxicity. We discuss the current understanding of beneficial and detrimental roles for Aß in synaptic function and epigenetic gene control and the future promising prospects of early therapeutic interventions aimed at mediating Aß induced neuroepigenetic and synaptic dysfunctions to delay AD onset.

13.
Sci Rep ; 10(1): 18265, 2020 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-33106538

RESUMO

Epigenetic dysregulation is a common mechanism shared by molecularly and clinically heterogenous neurodegenerative diseases (NDs). Histone acetylation homeostasis, maintained by the antagonistic activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs), is necessary for appropriate gene expression and neuronal function. Disruption of neural acetylation homeostasis has been implicated in multiple types of NDs including Alzheimer's disease (AD), yet mechanisms underlying alterations remain unclear. We show that like AD, disruption of Tip60 HAT/HDAC2 balance with concomitant epigenetic repression of common Tip60 target neuroplasticity genes occurs early in multiple types of Drosophila ND models such as Parkinson's Disease (PD), Huntington's Disease (HD) and Amyotrophic Lateral Sclerosis (ALS). Repressed neuroplasticity genes show reduced enrichment of Tip60 and epigentic acetylation signatures at all gene loci examined with certain genes showing inappropriate HDAC2 repressor enrichment. Functional neuronal consequences for these disease conditions are reminiscent of human pathology and include locomotion, synapse morphology, and short-term memory deficits. Increasing Tip60 HAT levels specifically in the mushroom body learning and memory center in the Drosophila brain protects against locomotion and short-term memory function deficits in multiple NDs. Together, our results support a model by which Tip60 protects against neurological impairments in different NDs via similar modes of action.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas de Drosophila/metabolismo , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/metabolismo , Acetilação , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Drosophila , Histonas/química , Homeostase , Aprendizagem/fisiologia , Memória/fisiologia , Doenças Neurodegenerativas/patologia , Doença de Parkinson/patologia
14.
Biol Psychiatry ; 87(8): 745-755, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-31892408

RESUMO

BACKGROUND: Cortical dopaminergic systems are critically involved in prefrontal cortex (PFC) functions, especially in working memory and neurodevelopmental disorders such as schizophrenia. GSK-3ß (glycogen synthase kinase-3ß) is highly associated with cAMP (cyclic adenosine monophosphate)-independent dopamine D2 receptor (D2R)-mediated signaling to affect dopamine-dependent behaviors. However, the mechanisms underlying the GSK-3ß modulation of cognitive function via D2Rs remains unclear. METHODS: This study explored how conditional cell-type-specific ablation of GSK-3ß in D2R+ neurons (D2R-GSK-3ß-/-) in the brain affects synaptic function in the medial PFC (mPFC). Both male and female (postnatal days 60-90) mice, including 140 D2R, 24 D1R, and 38 DISC1 mice, were used. RESULTS: This study found that NMDA receptor (NMDAR) function was significantly increased in layer V pyramidal neurons in mPFC of D2R-GSK-3ß-/- mice, along with increased dopamine modulation of NMDAR-mediated current. Consistently, NR2A and NR2B protein levels were elevated in mPFC of D2R-GSK-3ß-/- mice. This change was accompanied by a significant increase in enrichment of activator histone mark H3K27ac at the promoters of both Grin2a and Grin2b genes. In addition, altered short- and long-term synaptic plasticity, along with an increased spine density in layer V pyramidal neurons, were detected in D2R-GSK-3ß-/- mice. Indeed, D2R-GSK-3ß-/- mice also exhibited a resistance of working memory impairment induced by injection of NMDAR antagonist MK-801. Notably, either inhibiting GSK-3ß or disrupting the D2R-DISC1 complex was able to reverse the mutant DISC1-induced decrease of NMDAR-mediated currents in the mPFC. CONCLUSIONS: This study demonstrates that GSK-3ß modulates cognition via D2R-DISC1 interaction and epigenetic regulation of NMDAR expression and function.


Assuntos
Disfunção Cognitiva , Receptores de N-Metil-D-Aspartato , Animais , Epigênese Genética , Feminino , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Camundongos , Proteínas do Tecido Nervoso , Plasticidade Neuronal , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
15.
Sci Rep ; 9(1): 1471, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30728362

RESUMO

Adult hippocampal dentate gyrus (DG) neural stem cells (NSCs) continuously undergo proliferation and differentiation, producing new functional neurons that remodel existing synaptic circuits. Although proliferation of these adult DG NSCs has been implicated in opiate dependence, whether NSC neuronal differentiation and subsequent dendritogenesis are also involved in such addictive behavior remains unknown. Here, we ask whether opiate exposure alters differentiation and dendritogenesis of DG NSCs and investigate the possibility that these alterations contribute to opiate addiction. We show that rat morphine self-administration (MSA), a paradigm that effectively mimics human opiate addiction, increases NSC neuronal differentiation and promotes neuronal dendrite growth in the adult DG. Further, we demonstrate that the µ-opioid receptor (MOR) is expressed on DG NSCs and that MSA leads to a two-fold elevation of endogenous MOR levels in doublecortin expressing (DCX+) NSC progenies in the rat DG. MOR expression is also detected in the cultured rat NSCs and morphine treatment in vitro increases NSC neuronal differentiation and dendritogenesis, suggesting that MOR mediates the effect of morphine on NSC neuronal differentiation and maturation. Finally, we show that conditional overexpression of MOR in DG NSCs under a doxycycline inducible system leads to facilitation of the acquisition of MSA in rats, without affecting the extinction process. We advocate that targeting MOR selectively in the DG NSC population might offer a novel therapeutic intervention for morphine addiction.


Assuntos
Morfina/efeitos adversos , Células-Tronco Neurais/citologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores Opioides mu/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Giro Denteado/metabolismo , Modelos Animais de Doenças , Proteína Duplacortina , Humanos , Masculino , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese , Ratos , Automedicação/efeitos adversos
16.
Genetics ; 175(3): 1229-40, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17179074

RESUMO

Chromatin packaging directly influences gene programming as it permits only certain portions of the genome to be activated in any given developmental stage, cell, and tissue type. Histone acetyltransferases (HATs) are a key class of chromatin regulatory proteins that mediate such developmental chromatin control; however, their specific roles during multicellular development remain unclear. Here, we report the first isolation and developmental characterization of a Drosophila HAT gene (Dmel\TIP60) that is the homolog of the human HAT gene TIP60. We show that Dmel\TIP60 is differentially expressed during Drosophila development, with transcript levels significantly peaking during embryogenesis. We further demonstrate that reducing endogenous Dmel\TIP60 expression in Drosophila embryonic cells by RNAi results in cellular defects and lethality. Finally, using a GAL4-targeted RNAi system in Drosophila, we show that ubiquitous or mesoderm/muscle-specific reduction of Dmel\TIP60 expression results in lethality during fly development. Our results suggest a mechanism for HAT regulation involving developmental control of HAT expression profiles and show that Dmel\TIP60 is essential for multicellular development. Significantly, our inducible and targeted HAT knockdown system in Drosophila now provides a powerful tool for effectively studying the roles of TIP60 in specific tissues and cell types during development.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Histona Acetiltransferases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Cruzamentos Genéticos , Primers do DNA , DNA Complementar/genética , Drosophila melanogaster/embriologia , Genes Essenciais/genética , Dados de Sequência Molecular , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Análise de Sequência de DNA
18.
PLoS One ; 11(7): e0159623, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27454757

RESUMO

Environmental enrichment (EE) conditions have beneficial effects for reinstating cognitive ability in neuropathological disorders like Alzheimer's disease (AD). While EE benefits involve epigenetic gene control mechanisms that comprise histone acetylation, the histone acetyltransferases (HATs) involved remain largely unknown. Here, we examine a role for Tip60 HAT action in mediating activity- dependent beneficial neuroadaptations to EE using the Drosophila CNS mushroom body (MB) as a well-characterized cognition model. We show that flies raised under EE conditions display enhanced MB axonal outgrowth, synaptic marker protein production, histone acetylation induction and transcriptional activation of cognition linked genes when compared to their genotypically identical siblings raised under isolated conditions. Further, these beneficial changes are impaired in both Tip60 HAT mutant flies and APP neurodegenerative flies. While EE conditions provide some beneficial neuroadaptive changes in the APP neurodegenerative fly MB, such positive changes are significantly enhanced by increasing MB Tip60 HAT levels. Our results implicate Tip60 as a critical mediator of EE-induced benefits, and provide broad insights into synergistic behavioral and epigenetic based therapeutic approaches for treatment of cognitive disorder.


Assuntos
Cognição , Proteínas de Drosophila/genética , Meio Ambiente , Histona Acetiltransferases/genética , Corpos Pedunculados/fisiologia , Acetilação , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Biomarcadores , Núcleo Celular/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Transporte Proteico , Células Piramidais/metabolismo , Ratos , Sinapses/metabolismo , Transcrição Gênica
19.
Fly (Austin) ; 9(1): 22-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26327426

RESUMO

Disruption of epigenetic gene control mechanisms involving histone acetylation in the brain causes cognitive impairment, a debilitating hallmark of most neurodegenerative disorders. Histone acetylation regulates cognitive gene expression via chromatin packaging control in neurons. Unfortunately, the histone acetyltransferases (HATs) that generate such neural epigenetic signatures and their mechanisms of action remain unclear. Our recent findings provide insight into this question by demonstrating that Tip60 HAT action is critical for morphology and function of the mushroom body (MB), the learning and memory center in the Drosophila brain. We show that Tip60 is robustly produced in MB Kenyon cells and extending axonal lobes and that targeted MB Tip60 HAT loss results in axonal outgrowth disruption. Functional consequences of loss and gain of Tip60 HAT levels in the MB are evidenced by defects in memory. Tip60 ChIP-Seq analysis reveals enrichment for genes that function in cognitive processes and accordingly, key genes representing these pathways are misregulated in the Tip60 HAT mutant fly brain. Remarkably, increasing levels of Tip60 in the MB rescues learning and memory deficits resulting from Alzheimer's disease associated amyloid precursor protein (APP) induced neurodegeneration. Our studies highlight the potential of HAT activators as a therapeutic option for cognitive disorders.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Epigênese Genética , Histona Acetiltransferases/metabolismo , Memória/fisiologia , Corpos Pedunculados/fisiologia , Animais
20.
Genetics ; 198(4): 1571-86, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25326235

RESUMO

Disruption of epigenetic gene control mechanisms in the brain causes significant cognitive impairment that is a debilitating hallmark of most neurodegenerative disorders, including Alzheimer's disease (AD). Histone acetylation is one of the best characterized of these epigenetic mechanisms that is critical for regulating learning- and memory- associated gene expression profiles, yet the specific histone acetyltransferases (HATs) that mediate these effects have yet to be fully characterized. Here, we investigate an epigenetic role for the HAT Tip60 in learning and memory formation using the Drosophila CNS mushroom body (MB) as a well-characterized cognition model. We show that Tip60 is endogenously expressed in the Kenyon cells, the intrinsic neurons of the MB, and in the MB axonal lobes. Targeted loss of Tip60 HAT activity in the MB causes thinner and shorter axonal lobes while increasing Tip60 HAT levels cause no morphological defects. Functional consequences of both loss and gain of Tip60 HAT levels in the MB are evidenced by defects in immediate-recall memory. Our ChIP-Seq analysis reveals that Tip60 target genes are enriched for functions in cognitive processes, and, accordingly, key genes representing these pathways are misregulated in the Tip60 HAT mutant fly brain. Remarkably, we find that both learning and immediate-recall memory deficits that occur under AD-associated, amyloid precursor protein (APP)-induced neurodegenerative conditions can be effectively rescued by increasing Tip60 HAT levels specifically in the MB. Together, our findings uncover an epigenetic transcriptional regulatory role for Tip60 in cognitive function and highlight the potential of HAT activators as a therapeutic option for neurodegenerative disorders.


Assuntos
Proteínas de Drosophila/genética , Drosophila/fisiologia , Epigênese Genética , Histona Acetiltransferases/genética , Aprendizagem , Memória , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Ativação Enzimática , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Histona Acetiltransferases/metabolismo , Corpos Pedunculados/embriologia , Corpos Pedunculados/metabolismo , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa