Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(15): e2321502121, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38564636

RESUMO

The release of paused RNA polymerase II (RNAPII) from promoter-proximal regions is tightly controlled to ensure proper regulation of gene expression. The elongation factor PTEF-b is known to release paused RNAPII via phosphorylation of the RNAPII C-terminal domain by its cyclin-dependent kinase component, CDK9. However, the signal and stress-specific roles of the various RNAPII-associated macromolecular complexes containing PTEF-b/CDK9 are not yet clear. Here, we identify and characterize the CDK9 complex required for transcriptional response to hypoxia. Contrary to previous reports, our data indicate that a CDK9 complex containing BRD4 but not AFF1/4 is essential for this hypoxic stress response. We demonstrate that BRD4 bromodomains (BET) are dispensable for the release of paused RNAPII at hypoxia-activated genes and that BET inhibition by JQ1 is insufficient to impair hypoxic gene response. Mechanistically, we demonstrate that the C-terminal region of BRD4 is required for Polymerase-Associated Factor-1 Complex (PAF1C) recruitment to establish an elongation-competent RNAPII complex at hypoxia-responsive genes. PAF1C disruption using a small-molecule inhibitor (iPAF1C) impairs hypoxia-induced, BRD4-mediated RNAPII release. Together, our results provide insight into potentially targetable mechanisms that control the hypoxia-responsive transcriptional elongation.


Assuntos
Proteínas Nucleares , Fatores de Transcrição , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regulação da Expressão Gênica , Quinases Ciclina-Dependentes/metabolismo , Quinase 9 Dependente de Ciclina/genética , Quinase 9 Dependente de Ciclina/metabolismo , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fosforilação , Hipóxia , Transcrição Gênica , Fator B de Elongação Transcricional Positiva/genética , Fator B de Elongação Transcricional Positiva/metabolismo , Proteínas que Contêm Bromodomínio , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
2.
J Biol Rhythms ; 39(2): 208-214, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38305093

RESUMO

Atopic dermatitis (AD) is symptomatically worse in the evening, but the mechanism driving nocturnal eczema remains elusive. Our objective was to determine the circadian rhythm of skin barrier function measured by transepidermal water loss (TEWL) in AD patients and explore the molecular underpinnings. A pilot study was performed on a diverse group of AD (n = 4) and control (n = 2) young patients. We used an inpatient tightly controlled, modified, constant routine protocol. TEWL was measured at least every 90 min in the antecubital fossa (lesional) and forearm, while whole blood samples were collected every 4 h. Results show a significant difference in the antecubital fossa TEWL in the AD group versus controls. TEWL in control skin decreases starting a few hours prior to bedtime, both in the antecubital fossa and in the forearm, while in the AD forearm skin, pre-bedtime TEWL increases. We identified 1576 differentially expressed genes using a time-dependent model. The top 20 upregulated gene ontology pathways included neuronal pathways, while the downregulated functional terms included innate immune signaling and viral response. Similar pathways positively correlated with forearm TEWL in controls and inversely with the AD group. Upregulation in sensory perception pathways correlated with increases in lesional (antecubital fossa) TEWL in the evening. Results show skin barrier function worsens in the evening in the AD group, at a time when barrier is normally rejuvenating in healthy skin. This timing and the detection of transcriptomic signatures of sensory perception and diminished viral response might correspond to the nocturnal itch. Larger studies are needed to evaluate these associations in the skin.


Assuntos
Dermatite Atópica , Humanos , Dermatite Atópica/diagnóstico , Projetos Piloto , Perda Insensível de Água/fisiologia , Ritmo Circadiano , Pele
3.
Sci Adv ; 9(47): eadj1261, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-37992162

RESUMO

The biological role of the repetitive DNA sequences in the human genome remains an outstanding question. Recent long-read human genome assemblies have allowed us to identify a function for one of these repetitive regions. We have uncovered a tandem array of conserved primate-specific retrogenes encoding the protein Elongin A3 (ELOA3), a homolog of the RNA polymerase II (RNAPII) elongation factor Elongin A (ELOA). Our genomic analysis shows that the ELOA3 gene cluster is conserved among primates and the number of ELOA3 gene repeats is variable in the human population and across primate species. Moreover, the gene cluster has undergone concerted evolution and homogenization within primates. Our biochemical studies show that ELOA3 functions as a promoter-associated RNAPII pause-release elongation factor with distinct biochemical and functional features from its ancestral homolog, ELOA. We propose that the ELOA3 gene cluster has evolved to fulfil a transcriptional regulatory function unique to the primate lineage that can be targeted to regulate cellular hyperproliferation.


Assuntos
Fatores de Alongamento de Peptídeos , RNA Polimerase II , Animais , Humanos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fatores de Alongamento de Peptídeos/genética , Primatas/genética , Elonguina/genética , Família Multigênica , Sequências de Repetição em Tandem/genética
4.
Mol Cell ; 83(16): 2896-2910.e4, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37442129

RESUMO

The BET family protein BRD4, which forms the CDK9-containing BRD4-PTEFb complex, is considered to be a master regulator of RNA polymerase II (Pol II) pause release. Because its tandem bromodomains interact with acetylated histone lysine residues, it has long been thought that BRD4 requires these bromodomains for its recruitment to chromatin and transcriptional regulatory function. Here, using rapid depletion and genetic complementation with domain deletion mutants, we demonstrate that BRD4 bromodomains are dispensable for Pol II pause release. A minimal, bromodomain-less C-terminal BRD4 fragment containing the PTEFb-interacting C-terminal motif (CTM) is instead both necessary and sufficient to mediate Pol II pause release in the absence of full-length BRD4. Although BRD4-PTEFb can associate with chromatin through acetyl recognition, our results indicate that a distinct, active BRD4-PTEFb population functions to regulate transcription independently of bromodomain-mediated chromatin association. These findings may enable more effective pharmaceutical modulation of BRD4-PTEFb activity.


Assuntos
Proteínas Nucleares , Fatores de Transcrição , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Histonas/metabolismo , Regulação da Expressão Gênica , Cromatina/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
5.
Sci Adv ; 9(10): eadf2468, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36888719

RESUMO

The polymerase-associated factor 1 complex (PAF1C) is a key, post-initiation transcriptional regulator of both promoter-proximal pausing and productive elongation catalyzed by RNA Pol II and is also involved in transcriptional repression of viral gene expression during human immunodeficiency virus-1 (HIV-1) latency. Using a molecular docking-based compound screen in silico and global sequencing-based candidate evaluation in vivo, we identified a first-in-class, small-molecule inhibitor of PAF1C (iPAF1C) that disrupts PAF1 chromatin occupancy and induces global release of promoter-proximal paused RNA Pol II into gene bodies. Transcriptomic analysis revealed that iPAF1C treatment mimics acute PAF1 subunit depletion and impairs RNA Pol II pausing at heat shock-down-regulated genes. Furthermore, iPAF1C enhances the activity of diverse HIV-1 latency reversal agents both in cell line latency models and in primary cells from persons living with HIV-1. In sum, this study demonstrates that efficient disruption of PAF1C by a first-in-class, small-molecule inhibitor may have therapeutic potential for improving current HIV-1 latency reversal strategies.


Assuntos
HIV-1 , RNA Polimerase II , Humanos , RNA Polimerase II/metabolismo , HIV-1/genética , HIV-1/metabolismo , Simulação de Acoplamento Molecular , Linhagem Celular , Transcrição Gênica , Fatores de Transcrição/genética
6.
bioRxiv ; 2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36711760

RESUMO

Circadian clocks may mediate lifespan extension by caloric or dietary restriction (DR). We find that the core clock transcription factor Clock is crucial for a robust longevity and fecundity response to DR in Drosophila. To identify clock-controlled mediators, we performed RNA-sequencing from abdominal fat bodies across the 24 h day after just 5 days under control or DR diets. In contrast to more chronic DR regimens, we did not detect significant changes in the rhythmic expression of core clock genes. Yet we discovered that DR induced de novo rhythmicity or increased expression of rhythmic clock output genes. Network analysis revealed that DR increased network connectivity in one module comprised of genes encoding proteasome subunits. Adult, fat body specific RNAi knockdown demonstrated that proteasome subunits contribute to DR-mediated lifespan extension. Thus, clock control of output links DR-mediated changes in rhythmic transcription to lifespan extension.

7.
PLoS Biol ; 20(1): e3001456, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35081110

RESUMO

In traumatic brain injury (TBI), the initial injury phase is followed by a secondary phase that contributes to neurodegeneration, yet the mechanisms leading to neuropathology in vivo remain to be elucidated. To address this question, we developed a Drosophila head-specific model for TBI termed Drosophila Closed Head Injury (dCHI), where well-controlled, nonpenetrating strikes are delivered to the head of unanesthetized flies. This assay recapitulates many TBI phenotypes, including increased mortality, impaired motor control, fragmented sleep, and increased neuronal cell death. TBI results in significant changes in the transcriptome, including up-regulation of genes encoding antimicrobial peptides (AMPs). To test the in vivo functional role of these changes, we examined TBI-dependent behavior and lethality in mutants of the master immune regulator NF-κB, important for AMP induction, and found that while sleep and motor function effects were reduced, lethality effects were enhanced. Similarly, loss of most AMP classes also renders flies susceptible to lethal TBI effects. These studies validate a new Drosophila TBI model and identify immune pathways as in vivo mediators of TBI effects.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Drosophila melanogaster , Neuroglia/imunologia , Animais , Peptídeos Antimicrobianos/genética , Peptídeos Antimicrobianos/metabolismo , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/mortalidade , Modelos Animais de Doenças , Imunidade Inata , Locomoção , Masculino , Mutação , NF-kappa B/genética , NF-kappa B/metabolismo , Transtornos do Sono-Vigília , Transcriptoma
8.
Mol Cell ; 81(21): 4413-4424.e5, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34480849

RESUMO

Based on in vitro studies, it has been demonstrated that the DSIF complex, composed of SPT4 and SPT5, regulates the elongation stage of transcription catalyzed by RNA polymerase II (RNA Pol II). The precise cellular function of SPT5 is not clear, because conventional gene depletion strategies for SPT5 result in loss of cellular viability. Using an acute inducible protein depletion strategy to circumvent this issue, we report that SPT5 loss triggers the ubiquitination and proteasomal degradation of the core RNA Pol II subunit RPB1, a process that we show to be evolutionarily conserved from yeast to human cells. RPB1 degradation requires the E3 ligase Cullin 3, the unfoldase VCP/p97, and a novel form of CDK9 kinase complex. Our study demonstrates that SPT5 stabilizes RNA Pol II specifically at promoter-proximal regions, permitting RNA Pol II release from promoters into gene bodies and providing mechanistic insight into the cellular function of SPT5 in safeguarding accurate gene expression.


Assuntos
Proteínas Culina/metabolismo , Proteínas Nucleares/metabolismo , RNA Polimerase II/química , RNA Polimerase II/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Animais , Sobrevivência Celular , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Culina/química , Fibroblastos/metabolismo , Humanos , Ácidos Indolacéticos/química , Camundongos , Ubiquitina-Proteína Ligases Nedd4/química , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/química , Proteoma , Proteômica/métodos , Ubiquitina-Proteína Ligases/química , Proteína com Valosina/química , Proteína com Valosina/metabolismo
9.
Nat Methods ; 18(3): 303-308, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33589837

RESUMO

Current proteomic approaches disassemble and digest nucleosome particles, blurring readouts of the 'histone code'. To preserve nucleosome-level information, we developed Nuc-MS, which displays the landscape of histone variants and their post-translational modifications (PTMs) in a single mass spectrum. Combined with immunoprecipitation, Nuc-MS quantified nucleosome co-occupancy of histone H3.3 with variant H2A.Z (sixfold over bulk) and the co-occurrence of oncogenic H3.3K27M with euchromatic marks (for example, a >15-fold enrichment of dimethylated H3K79me2). Nuc-MS is highly concordant with chromatin immunoprecipitation-sequencing (ChIP-seq) and offers a new readout of nucleosome-level biology.


Assuntos
Histonas/metabolismo , Nucleossomos/metabolismo , Proteômica/métodos , Espectrometria de Massas por Ionização por Electrospray/métodos , Linhagem Celular , Imunoprecipitação da Cromatina/métodos , Células HEK293 , Código das Histonas , Humanos , Metilação
10.
Genes Dev ; 35(3-4): 273-285, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33446572

RESUMO

The regulation of gene expression catalyzed by RNA polymerase II (Pol II) requires a host of accessory factors to ensure cell growth, differentiation, and survival under environmental stress. Here, using the auxin-inducible degradation (AID) system to study transcriptional activities of the bromodomain and extraterminal domain (BET) and super elongation complex (SEC) families, we found that the CDK9-containing BRD4 complex is required for the release of Pol II from promoter-proximal pausing for most genes, while the CDK9-containing SEC is required for activated transcription in the heat shock response. By using both the proteolysis targeting chimera (PROTAC) dBET6 and the AID system, we found that dBET6 treatment results in two major effects: increased pausing due to BRD4 loss, and reduced enhancer activity attributable to BRD2 loss. In the heat shock response, while auxin-mediated depletion of the AFF4 subunit of the SEC has a more severe defect than AFF1 depletion, simultaneous depletion of AFF1 and AFF4 leads to a stronger attenuation of the heat shock response, similar to treatment with the SEC inhibitor KL-1, suggesting a possible redundancy among SEC family members. This study highlights the usefulness of orthogonal acute depletion/inhibition strategies to identify distinct and redundant biological functions among Pol II elongation factor paralogs.


Assuntos
Expressão Gênica/genética , Fatores de Alongamento de Peptídeos/metabolismo , RNA Polimerase II/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Células HCT116 , Resposta ao Choque Térmico , Humanos , Fatores de Alongamento de Peptídeos/genética , Proteínas/genética , Proteínas/metabolismo , RNA Polimerase II/genética , Fatores de Transcrição/genética
11.
Crit Care Med ; 48(12): e1294-e1299, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33031153

RESUMO

OBJECTIVES: Core clock genes regulate tissue-specific transcriptome oscillations that synchronize physiologic processes throughout the body, held in phase by the central circadian rhythm. The central circadian rhythm rapidly dampens with onset of critical illness, but the effect of critical illness on gene expression oscillations is unknown. The objective of this study was to characterize the rhythmicity and phase coherence of core clock genes and the broader transcriptome after onset of critical illness. DESIGN: Cross-sectional study. SETTING: ICUs and hospital clinical research unit. PATIENTS: Critically ill patients within the first day of presenting from the community and healthy volunteers. INTERVENTIONS: Usual care (critically ill patients) and modified constant routine (healthy volunteers). MEASUREMENTS AND MAIN RESULTS: We studied 15 critically ill patients, including 10 with sepsis and five with intracerebral hemorrhage, and 11 healthy controls. The central circadian rhythm and rest-activity rhythms were profiled by continuous wrist actigraphy, and serum melatonin sampled every 2 hours along with whole blood for RNA isolation over 24 hours. The gene expression transcriptome was obtained by RNA sequencing. Core clock genes were analyzed for rhythmicity by cosinor fit. Significant circadian rhythmicity was identified in five of six core clock genes in healthy controls, but none in critically ill patients. TimeSignature, a validated algorithm based on 41 genes, was applied to assess overall transcriptome phase coherence. Median absolute error of TimeSignature was higher in individual critically ill patients than healthy patients (4.90 vs 1.48 hr) and was correlated with encephalopathy severity by Glasgow Coma Scale in critically ill patients (rho, -0.54; p = 0.036). CONCLUSIONS: Gene expression rhythms rapidly become abnormal during critical illness. The association between disrupted transcriptome rhythms and encephalopathy suggests a path for future work to elucidate the underlying pathophysiology.


Assuntos
Ritmo Circadiano , Estado Terminal , Expressão Gênica , Adulto , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Unidades de Terapia Intensiva , Masculino , Pessoa de Meia-Idade , Transcriptoma
12.
Genes Dev ; 34(21-22): 1493-1502, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33033055

RESUMO

Catalytic-inactivating mutations within the Drosophila enhancer H3K4 mono-methyltransferase Trr and its mammalian homologs, MLL3/4, cause only minor changes in gene expression compared with whole-gene deletions for these COMPASS members. To identify essential histone methyltransferase-independent functions of Trr, we screened to identify a minimal Trr domain sufficient to rescue Trr-null lethality and demonstrate that this domain binds and stabilizes Utx in vivo. Using the homologous MLL3/MLL4 human sequences, we mapped a short ∼80-amino-acid UTX stabilization domain (USD) that promotes UTX stability in the absence of the rest of MLL3/4. Nuclear UTX stability is enhanced when the USD is fused with the MLL4 HMG-box. Thus, COMPASS-dependent UTX stabilization is an essential noncatalytic function of Trr/MLL3/MLL4, suggesting that stabilizing UTX could be a therapeutic strategy for cancers with MLL3/4 loss-of-function mutations.


Assuntos
Sequência Conservada/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Genes Letais/genética , Histona-Lisina N-Metiltransferase/genética , Oxirredutases N-Desmetilantes/genética , Animais , Deleção de Genes , Regulação da Expressão Gênica/genética , Células HCT116 , Humanos , Domínios Proteicos , Estabilidade Proteica
13.
J Biol Rhythms ; 35(5): 439-451, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32613882

RESUMO

The circadian rhythm drives the oscillatory expression of thousands of genes across all tissues, coordinating physiological processes. The effect of this rhythm on health has generated increasing interest in discovering genes under circadian control by searching for periodic patterns in transcriptomic time-series experiments. While algorithms for detecting cycling transcripts have advanced, there remains little guidance quantifying the effect of experimental design and analysis choices on cycling detection accuracy. We present TimeTrial, a user-friendly benchmarking framework using both real and synthetic data to investigate cycle detection algorithms' performance and improve circadian experimental design. Results show that the optimal choice of analysis method depends on the sampling scheme, noise level, and shape of the waveform of interest and provides guidance on the impact of sampling frequency and duration on cycling detection accuracy. The TimeTrial software is freely available for download and may also be accessed through a web interface. By supplying a tool to vary and optimize experimental design considerations, TimeTrial will enhance circadian transcriptomics studies.


Assuntos
Cronobiologia/métodos , Ritmo Circadiano , Perfilação da Expressão Gênica/métodos , Software , Transcriptoma , Algoritmos , Animais , Humanos , Camundongos , Fatores de Tempo
14.
PLoS Genet ; 15(10): e1008356, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31593562

RESUMO

Disrupted circadian rhythms is a prominent and early feature of neurodegenerative diseases including Huntington's disease (HD). In HD patients and animal models, striatal and hypothalamic neurons expressing molecular circadian clocks are targets of mutant Huntingtin (mHtt) pathogenicity. Yet how mHtt disrupts circadian rhythms remains unclear. In a genetic screen for modifiers of mHtt effects on circadian behavior in Drosophila, we discovered a role for the neurodegenerative disease gene Ataxin2 (Atx2). Genetic manipulations of Atx2 modify the impact of mHtt on circadian behavior as well as mHtt aggregation and demonstrate a role for Atx2 in promoting mHtt aggregation as well as mHtt-mediated neuronal dysfunction. RNAi knockdown of the Fragile X mental retardation gene, dfmr1, an Atx2 partner, also partially suppresses mHtt effects and Atx2 effects depend on dfmr1. Atx2 knockdown reduces the cAMP response binding protein A (CrebA) transcript at dawn. CrebA transcript level shows a prominent diurnal regulation in clock neurons. Loss of CrebA also partially suppresses mHtt effects on behavior and cell loss and restoration of CrebA can suppress Atx2 effects. Our results indicate a prominent role of Atx2 in mediating mHtt pathology, specifically via its regulation of CrebA, defining a novel molecular pathway in HD pathogenesis.


Assuntos
Ataxina-2/genética , Relógios Circadianos/genética , Proteína A de Ligação a Elemento de Resposta do AMP Cíclico/genética , Proteínas de Drosophila/genética , Proteína Huntingtina/genética , Doença de Huntington/genética , Animais , Ritmo Circadiano/genética , Modelos Animais de Doenças , Drosophila melanogaster/genética , Proteína do X Frágil de Retardo Mental/genética , Humanos , Doença de Huntington/patologia , Proteínas Mutantes/genética , Neurônios/metabolismo , Transdução de Sinais/genética
15.
Cell Rep ; 27(1): 59-70.e4, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30943415

RESUMO

Neurodegenerative diseases commonly involve the disruption of circadian rhythms. Studies indicate that mutant Huntingtin (mHtt), the cause of Huntington's disease (HD), disrupts circadian rhythms often before motor symptoms are evident. Yet little is known about the molecular mechanisms by which mHtt impairs circadian rhythmicity and whether circadian clocks can modulate HD pathogenesis. To address this question, we used a Drosophila HD model. We found that both environmental and genetic perturbations of the circadian clock alter mHtt-mediated neurodegeneration. To identify potential genetic pathways that mediate these effects, we applied a behavioral platform to screen for clock-regulated HD suppressors, identifying a role for Heat Shock Protein 70/90 Organizing Protein (Hop). Hop knockdown paradoxically reduces mHtt aggregation and toxicity. These studies demonstrate a role for the circadian clock in a neurodegenerative disease model and reveal a clock-regulated molecular and cellular pathway that links clock function to neurodegenerative disease.


Assuntos
Relógios Circadianos/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Choque Térmico/metabolismo , Resposta ao Choque Térmico/fisiologia , Proteína Huntingtina/metabolismo , Proteína Huntingtina/toxicidade , Agregação Patológica de Proteínas , Animais , Animais Geneticamente Modificados , Relógios Circadianos/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Embrião não Mamífero , Feminino , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiologia , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/metabolismo , Masculino , Proteínas Mutantes/fisiologia , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia
17.
Sleep ; 42(5)2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30753650

RESUMO

Slow-wave sleep (SWS) is important for overall health since it affects many physiological processes including cardio-metabolic function. Sleep and autonomic nervous system (ANS) activity are closely coupled at anatomical and physiological levels. Sleep-related changes in autonomic function are likely the main pathway through which SWS affects many systems within the body. There are characteristic changes in ANS activity across sleep stages. Notably, in non-rapid eye-movement sleep, the progression into SWS is characterized by increased parasympathetic activity, an important measure of cardiovascular health. Experimental manipulations that enhance slow-wave activity (SWA, 0.5-4 Hz) can improve sleep-mediated memory and immune function. However, effects of SWA enhancement on autonomic regulation have not been investigated. Here, we employed an adaptive algorithm to deliver 50 ms sounds phase-locked to slow-waves, with regular pauses in stimulation (~5 s ON/~5 s OFF), in healthy young adults. We sought to determine whether acoustic enhancement of SWA altered parasympathetic activity during SWS assessed with heart rate variability (HRV), and evening-to-morning changes in HRV, plasma cortisol, and blood pressure. Stimulation, compared with a sham condition, increased SWA during ON versus OFF intervals. This ON/OFF SWA enhancement was associated with a reduction in evening-to-morning change of cortisol levels and indices of sympathetic activity. Furthermore, the enhancement of SWA in ON intervals during sleep cycles 2-3 was accompanied by an increase in parasympathetic activity (high-frequency, HRV). Together these findings suggest that acoustic enhancement of SWA has a positive effect on autonomic function in sleep. Approaches to strengthen brain-heart interaction during sleep could have important implications for cardiovascular health.


Assuntos
Estimulação Acústica/métodos , Ondas Encefálicas/fisiologia , Encéfalo/fisiologia , Frequência Cardíaca/fisiologia , Sono de Ondas Lentas/fisiologia , Adolescente , Adulto , Pressão Sanguínea/fisiologia , Estudos Cross-Over , Eletroencefalografia/métodos , Feminino , Humanos , Masculino , Fases do Sono/fisiologia , Adulto Jovem
18.
PLoS Comput Biol ; 15(1): e1006664, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30615612

RESUMO

Cancer development is driven by series of events involving mutations, which may become fixed in a tumor via genetic drift and selection. This process usually includes a limited number of driver (advantageous) mutations and a greater number of passenger (neutral or mildly deleterious) mutations. We focus on a real-world leukemia model evolving on the background of a germline mutation. Severe congenital neutropenia (SCN) evolves to secondary myelodysplastic syndrome (sMDS) and/or secondary acute myeloid leukemia (sAML) in 30-40%. The majority of SCN cases are due to a germline ELANE mutation. Acquired mutations in CSF3R occur in >70% sMDS/sAML associated with SCN. Hypotheses underlying our model are: an ELANE mutation causes SCN; CSF3R mutations occur spontaneously at a low rate; in fetal life, hematopoietic stem and progenitor cells expands quickly, resulting in a high probability of several tens to several hundreds of cells with CSF3R truncation mutations; therapeutic granulocyte colony-stimulating factor (G-CSF) administration early in life exerts a strong selective pressure, providing mutants with a growth advantage. Applying population genetics theory, we propose a novel two-phase model of disease development from SCN to sMDS. In Phase 1, hematopoietic tissues expand and produce tens to hundreds of stem cells with the CSF3R truncation mutation. Phase 2 occurs postnatally through adult stages with bone marrow production of granulocyte precursors and positive selection of mutants due to chronic G-CSF therapy to reverse the severe neutropenia. We predict the existence of the pool of cells with the mutated truncated receptor before G-CSF treatment begins. The model does not require increase in mutation rate under G-CSF treatment and agrees with age distribution of sMDS onset and clinical sequencing data.


Assuntos
Modelos Genéticos , Mutação/genética , Síndromes Mielodisplásicas , Neutropenia/congênito , Ciclo Celular/genética , Biologia Computacional , Síndrome Congênita de Insuficiência da Medula Óssea , Neoplasias Hematológicas/genética , Humanos , Elastase de Leucócito/genética , Taxa de Mutação , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/genética , Neutropenia/complicações , Neutropenia/genética , Neutropenia/fisiopatologia , Receptores de Fator Estimulador de Colônias/genética , Seleção Genética/genética
19.
Proc Natl Acad Sci U S A ; 115(39): E9247-E9256, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30201705

RESUMO

Circadian clocks play a key role in regulating a vast array of biological processes, with significant implications for human health. Accurate assessment of physiological time using transcriptional biomarkers found in human blood can significantly improve diagnosis of circadian disorders and optimize the delivery time of therapeutic treatments. To be useful, such a test must be accurate, minimally burdensome to the patient, and readily generalizable to new data. A major obstacle in development of gene expression biomarker tests is the diversity of measurement platforms and the inherent variability of the data, often resulting in predictors that perform well in the original datasets but cannot be universally applied to new samples collected in other settings. Here, we introduce TimeSignature, an algorithm that robustly infers circadian time from gene expression. We demonstrate its application in data from three independent studies using distinct microarrays and further validate it against a new set of samples profiled by RNA-sequencing. Our results show that TimeSignature is more accurate and efficient than competing methods, estimating circadian time to within 2 h for the majority of samples. Importantly, we demonstrate that once trained on data from a single study, the resulting predictor can be universally applied to yield highly accurate results in new data from other studies independent of differences in study population, patient protocol, or assay platform without renormalizing the data or retraining. This feature is unique among expression-based predictors and addresses a major challenge in the development of generalizable, clinically useful tests.


Assuntos
Relógios Circadianos/genética , Perfilação da Expressão Gênica/métodos , Aprendizado de Máquina , Biomarcadores/sangue , Ritmo Circadiano/genética , Expressão Gênica , Genes/genética , Humanos , Modelos Estatísticos , Reprodutibilidade dos Testes , Sono , Transcriptoma
20.
Proc Natl Acad Sci U S A ; 115(26): 6816-6821, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29891655

RESUMO

The successful treatment of chronic dermal wounds, such as diabetic foot ulcers (DFU), depends on the development of safe, effective, and affordable regenerative tools that the surgeon can rely on to promote wound closure. Although promising, strategies that involve cell-based therapies and the local release of exogenous growth factors are costly, require very long development times, and result in modest improvements in patient outcome. We describe the development of an antioxidant shape-conforming regenerative wound dressing that uses the laminin-derived dodecapeptide A5G81 as a potent tethered cell adhesion-, proliferation-, and haptokinesis-inducing ligand to locally promote wound closure. A5G81 immobilized within a thermoresponsive citrate-based hydrogel facilitates integrin-mediated spreading, migration, and proliferation of dermal and epidermal cells, resulting in faster tissue regeneration in diabetic wounds. This peptide-hydrogel system represents a paradigm shift in dermoconductive and dermoinductive strategies for treating DFU without the need for soluble biological or pharmacological factors.


Assuntos
Antioxidantes , Bandagens , Diabetes Mellitus Experimental/terapia , Pé Diabético/terapia , Hidrogéis , Laminina , Oligopeptídeos , Cicatrização , Animais , Antioxidantes/química , Antioxidantes/farmacologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Pé Diabético/metabolismo , Pé Diabético/patologia , Hidrogéis/química , Hidrogéis/farmacologia , Laminina/química , Laminina/farmacologia , Camundongos , Oligopeptídeos/química , Oligopeptídeos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...