Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 531
Filtrar
1.
Adv Sci (Weinh) ; 10(28): e2301367, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37565374

RESUMO

Lysine demethylase KDM7A removes histone modifications H3K9me1/2 and H3K27me1/2. KDM7A plays critical roles in gene expression and contribute to biological processes including tumorigenesis, metabolism, and embryonic development. However, the functions of KDM7A in mammalian nervous system are still poorly explored. In this study, functional roles of KDM7A are comprehensively investigated in neuronal cells by applying CUT&Tag-seq, RNA-seq and mice models. Knockdown of Kdm7a in N2A cells result in the alteration of histone modifications near transcription start sites (TSSs) and the expression changes of a large number of genes. In particular, the expression of immediate early genes (IEGs), a series of genes maintaining the function of the nervous system and associating with neurological disorders, are significantly decreased upon Kdm7a knockdown. Furthermore, in vivo knockdown of Kdm7a in dentate gyrus (DG) neuron of mice hippocampus, via Adeno-associated virus (AAV)-based stereotaxic microinjection, led to a significant decrease of the expression of c-Fos, a marker of neuron activity. Behavior assays in mice further revealed that Kdm7a knockdown in hippocampus repress neuron activity, which leading to impairment of emotion and memory. Collectively, the study reveals that KDM7A affects neuron functions by regulating IEGs, which may provide new clues for understanding epigenetic mechanisms in neurological disorders.


Assuntos
Histona Desmetilases com o Domínio Jumonji , Doenças do Sistema Nervoso , Camundongos , Animais , Histona Desmetilases com o Domínio Jumonji/genética , Lisina/genética , Genes Precoces/genética , Neurônios/metabolismo , Mamíferos/metabolismo
2.
Nat Commun ; 13(1): 7476, 2022 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463234

RESUMO

Growth factor-induced, ERK-mediated induction of immediate-early genes (IEGs) is crucial for cell growth and tumorigenesis. Although IEG expression is mainly regulated at the level of transcription elongation by RNA polymerase-II (Pol-II) promoter-proximal pausing and its release, the role of ERK in this process remains unknown. Here, we identified negative elongation factor (NELF)-A as an ERK substrate. Upon growth factor stimulation, ERK phosphorylates NELF-A, which dissociates NELF from paused Pol-II at the promoter-proximal regions of IEGs, allowing Pol-II to resume elongation and produce full-length transcripts. Furthermore, we found that in cancer cells, PP2A efficiently dephosphorylates NELF-A, thereby preventing aberrant IEG expression induced by ERK-activating oncogenes. However, when PP2A inhibitor proteins are overexpressed, as is frequently observed in cancers, decreased PP2A activity combined with oncogene-mediated ERK activation conspire to induce NELF-A phosphorylation and IEG upregulation, resulting in tumor progression. Our data delineate previously unexplored roles of ERK and PP2A inhibitor proteins in carcinogenesis.


Assuntos
Carcinogênese , Genes Precoces , RNA Polimerase II , Humanos , Carcinogênese/genética , Carcinogênese/metabolismo , Genes Precoces/genética , Genes Precoces/fisiologia , Fosforilação , RNA Polimerase II/genética , RNA Polimerase II/metabolismo
3.
Cell Rep ; 38(6): 110332, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35139389

RESUMO

Endothelial cells (ECs) are phenotypically heterogeneous, mainly due to their dynamic response to the tissue microenvironment. Vascular endothelial cell growth factor (VEGF), the best-known angiogenic factor, activates calcium-nuclear factor of activated T cells (NFAT) signaling following acute angiogenic gene transcription. Here, we evaluate the global mapping of VEGF-mediated dynamic transcriptional events, focusing on major histone-code profiles using chromatin immunoprecipitation sequencing (ChIP-seq). Remarkably, the gene loci of immediate-early angiogenic transcription factors (TFs) exclusively acquire bivalent H3K4me3-H3K27me3 double-positive histone marks after the VEGF stimulus. Moreover, NFAT-associated Pax transactivation domain-interacting protein (PTIP) directs bivalently marked TF genes transcription through a limited polymerase II running. The non-canonical polycomb1 variant PRC1.3 specifically binds to and allows the transactivation of PRC2-enriched bivalent angiogenic TFs until conventional PRC1-mediated gene silencing is achieved. Knockdown of these genes abrogates post-natal aberrant neovessel formation via the selective inhibition of indispensable bivalent angiogenic TF gene transcription. Collectively, the reported dynamic histone mark landscape may uncover the importance of immediate-early genes and the development of advanced anti-angiogenic strategies.


Assuntos
Indutores da Angiogênese/metabolismo , Genes Precoces/genética , Histonas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Imunoprecipitação da Cromatina , Sequenciamento de Cromatina por Imunoprecipitação , Células Endoteliais/metabolismo , Epigênese Genética/genética , Inativação Gênica/fisiologia , Humanos , Camundongos , Neovascularização Patológica/genética , Regiões Promotoras Genéticas/genética
4.
PLoS One ; 17(1): e0253406, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35025862

RESUMO

Early-life adversity (ELA) causes long-lasting structural and functional changes to the brain, rendering affected individuals vulnerable to the development of psychopathologies later in life. Immediate-early genes (IEGs) provide a potential marker for the observed alterations, bridging the gap between activity-regulated transcription and long-lasting effects on brain structure and function. Several heterogeneous studies have used IEGs to identify differences in cellular activity after ELA; systematically investigating the literature is therefore crucial for comprehensive conclusions. Here, we performed a systematic review on 39 pre-clinical studies in rodents to study the effects of ELA (alteration of maternal care) on IEG expression. Females and IEGs other than cFos were investigated in only a handful of publications. We meta-analyzed publications investigating specifically cFos expression. ELA increased cFos expression after an acute stressor only if the animals (control and ELA) had experienced additional hits. At rest, ELA increased cFos expression irrespective of other life events, suggesting that ELA creates a phenotype similar to naïve, acutely stressed animals. We present a conceptual theoretical framework to interpret the unexpected results. Overall, ELA likely alters IEG expression across the brain, especially in interaction with other negative life events. The present review highlights current knowledge gaps and provides guidance to aid the design of future studies.


Assuntos
Expressão Gênica/fisiologia , Genes Precoces/genética , Estresse Psicológico , Animais , Encéfalo/metabolismo , Proteínas do Citoesqueleto/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas c-fos/genética , Roedores
5.
Drug Metab Pers Ther ; 37(2): 191-200, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34881530

RESUMO

OBJECTIVES: This study investigates the influence of Cnestis ferruginea (CF) on kainic acid (KA)-induced immediate early genes (IEGs) associated with hippocampal sclerosis in temporal lobe epilepsy (TLE) in mice. METHODS: Animals were randomly divided into preventive treatment; vehicle (10 mL/kg, p.o.) or CF (400 mg/kg, p.o.) for three consecutive days before KA (5 mg/kg, i.p.) on days 4 and 5. In the reversal model, KA (5 mg/kg, i.p.) was administered on days 1 and 2 before CF (400 mg/kg) administration on days 3-5. Animals were euthanized on day 5, 6 h after KA exposure in preventive model and 1 h after CF administration in reversal model to estimate markers of IEGs. RESULTS: KA upregulated the expression of c-Fos protein by 3.32-, 9.45-, 8.13-, and 8.66-fold in the hippocampal CA1, CA2, CA3, and DG regions, respectively. Also, KA elevated inducible nitric oxide synthase protein expression by 10.9-, 10.6-, 9.78-, and 9.51-fold. Besides, mRNA expression of brain-derived neurotrophic factors and heat shock protein was increased by 2.38- and 1.39-fold, respectively, after exposure to KA which were attenuated by CF. CONCLUSIONS: CF attenuated KA-induced IEGs and could be used as an adjunct in TLE.


Assuntos
Connaraceae , Epilepsia do Lobo Temporal , Animais , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/genética , Genes Precoces/genética , Humanos , Ácido Caínico , Camundongos , Extratos Vegetais/farmacologia
6.
Proc Natl Acad Sci U S A ; 118(52)2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34930843

RESUMO

Immediate-early gene (IEG) expression has been used to identify small neural ensembles linked to a particular experience, based on the principle that a selective subset of activated neurons will encode specific memories or behavioral responses. The majority of these studies have focused on "engrams" in higher-order brain areas where more abstract or convergent sensory information is represented, such as the hippocampus, prefrontal cortex, or amygdala. In primary sensory cortex, IEG expression can label neurons that are responsive to specific sensory stimuli, but experience-dependent shaping of neural ensembles marked by IEG expression has not been demonstrated. Here, we use a fosGFP transgenic mouse to longitudinally monitor in vivo expression of the activity-dependent gene c-fos in superficial layers (L2/3) of primary somatosensory cortex (S1) during a whisker-dependent learning task. We find that sensory association training does not detectably alter fosGFP expression in L2/3 neurons. Although training broadly enhances thalamocortical synaptic strength in pyramidal neurons, we find that synapses onto fosGFP+ neurons are not selectively increased by training; rather, synaptic strengthening is concentrated in fosGFP- neurons. Taken together, these data indicate that expression of the IEG reporter fosGFP does not facilitate identification of a learning-specific engram in L2/3 in barrel cortex during whisker-dependent sensory association learning.


Assuntos
Aprendizagem por Associação/fisiologia , Memória/fisiologia , Plasticidade Neuronal , Proteínas Proto-Oncogênicas c-fos , Córtex Somatossensorial , Animais , Feminino , Genes Precoces/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Córtex Somatossensorial/metabolismo , Córtex Somatossensorial/fisiologia
7.
Nat Commun ; 12(1): 4436, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34290252

RESUMO

Latent human cytomegalovirus (HCMV) infection is characterized by limited gene expression, making latent HCMV infections refractory to current treatments targeting viral replication. However, reactivation of latent HCMV in immunosuppressed solid organ and stem cell transplant patients often results in morbidity. Here, we report the killing of latently infected cells via a virus-specific nanobody (VUN100bv) that partially inhibits signaling of the viral receptor US28. VUN100bv reactivates immediate early gene expression in latently infected cells without inducing virus production. This allows recognition and killing of latently infected monocytes by autologous cytotoxic T lymphocytes from HCMV-seropositive individuals, which could serve as a therapy to reduce the HCMV latent reservoir of transplant patients.


Assuntos
Citomegalovirus/efeitos dos fármacos , Anticorpos de Domínio Único/farmacologia , Linfócitos T Citotóxicos/imunologia , Latência Viral/efeitos dos fármacos , Células Cultivadas , Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Expressão Gênica/efeitos dos fármacos , Genes Precoces/genética , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/virologia , Receptores de Quimiocinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Domínio Único/metabolismo , Proteínas Virais/metabolismo , Ativação Viral/efeitos dos fármacos
8.
J Gen Virol ; 102(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34042564

RESUMO

Viral latency is an active process during which the host cell environment is optimized for latent carriage and reactivation. This requires control of both viral and host gene promoters and enhancers often at the level of chromatin, and several viruses co-opt the chromatin organiser CTCF to control gene expression during latency. While CTCF has a role in the latencies of alpha- and gamma-herpesviruses, it was not known whether CTCF played a role in the latency of the beta-herpesvirus human cytomegalovirus (HCMV). Here, we show that HCMV latency is associated with increased CTCF expression and CTCF binding to the viral major lytic promoter, the major immediate early promoter (MIEP). This increase in CTCF binding is dependent on the virally encoded G protein coupled receptor, US28, and contributes to suppression of MIEP-driven transcription, a hallmark of latency. Furthermore, we show that latency-associated upregulation of CTCF represses expression of the neutrophil chemoattractants S100A8 and S100A9 which we have previously shown are downregulated during HCMV latency. As with downregulation of the MIEP, CTCF binding to the enhancer region of S100A8/A9 drives their suppression, again in a US28-dependent manner. Taken together, we identify CTCF upregulation as an important mechanism for optimizing latent carriage of HCMV at both the levels of viral and cellular gene expression.


Assuntos
Fator de Ligação a CCCTC/metabolismo , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Receptores de Quimiocinas/metabolismo , Proteínas Virais/metabolismo , Latência Viral , Fator de Ligação a CCCTC/genética , Calgranulina A/genética , Calgranulina B/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Genes Precoces/genética , Interações Hospedeiro-Patógeno , Humanos , Monócitos/virologia , Regiões Promotoras Genéticas
9.
PLoS Pathog ; 17(4): e1009560, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33930088

RESUMO

Herpes-Simplex Virus 1 (HSV-1) infects most humans when they are young, sometimes with fatal consequences. Gene expression occurs in a temporal order upon lytic HSV-1 infection: immediate early (IE) genes are expressed, then early (E) genes, followed by late (L) genes. During this infection cycle, the HSV-1 genome has the potential for exposure to APOBEC3 (A3) proteins, a family of cytidine deaminases that cause C>U mutations on single-stranded DNA (ssDNA), often resulting in a C>T transition. We developed a computational model for the mutational pressure of A3 on the lytic cycle of HSV-1 to determine which viral kinetic gene class is most vulnerable to A3 mutations. Using in silico stochastic methods, we simulated the infectious cycle under varying intensities of A3 mutational pressure. We found that the IE and E genes are more vulnerable to A3 than L genes. We validated this model by analyzing the A3 evolutionary footprints in 25 HSV-1 isolates. We find that IE and E genes have evolved to underrepresent A3 hotspot motifs more so than L genes, consistent with greater selection pressure on IE and E genes. We extend this model to two-step infections, such as those of polyomavirus, and find that the same pattern holds for over 25 human Polyomavirus (HPyVs) genomes. Genes expressed earlier during infection are more vulnerable to mutations than those expressed later.


Assuntos
Desaminases APOBEC/fisiologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/genética , Mutagênese/genética , Polyomavirus/fisiologia , Algoritmos , Regulação Viral da Expressão Gênica , Genes Precoces/genética , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Interações Hospedeiro-Patógeno/genética , Humanos , Modelos Teóricos , Mutação , Polyomavirus/genética , Polyomavirus/patogenicidade , Infecções por Polyomavirus/genética , Infecções por Polyomavirus/virologia , Replicação Viral/genética
10.
Neurobiol Learn Mem ; 180: 107401, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33581315

RESUMO

As the incidence of anxiety disorders is more prevalent in females, comparing the neural underpinnings of anxiety in males and females is imperative. The bed nucleus of the stria terminalis (BNST) contributes to long-lasting, anxiety-like states including the expression of context fear conditioning. Currently, there is conflicting evidence as to which nuclei of the BNST contribute to these behaviors. The anterolateral portion of the BNST (BNST-AL) located dorsal to the anterior commissure and lateral to the stria terminalis sends robust projections to the central nucleus of the amygdala (CE). Here we asked whether the BNST-AL is active during the expression of context fear conditioning in both male and female rats. At the cellular level, the expression of context fear produced upregulation of the immediate-early gene ARC in the BNST-AL as well as an upregulation of ARC specifically in neurons projecting to the CE, as labeled by the retrograde tracer Fluorogold infused into the CE. However, this pattern of ARC expression was observed in male rats only. Excitotoxic lesions of the BNST reduced context fear expression in both sexes, suggesting that a different set of BNST subnuclei may be recruited by the expression of fear and anxiety-like behaviors in females. Overall, our data highlight the involvement of the BNST-AL in fear expression in males, and suggest that subnuclei of the BNST may be functionally different in male and female rats.


Assuntos
Tonsila do Cerebelo/fisiologia , Núcleo Central da Amígdala/fisiologia , Condicionamento Clássico/fisiologia , Medo , Núcleos Septais/fisiologia , Tonsila do Cerebelo/metabolismo , Animais , Núcleo Central da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Proteínas do Citoesqueleto/genética , Feminino , Genes Precoces/genética , Masculino , Proteínas do Tecido Nervoso/genética , Vias Neurais/metabolismo , Vias Neurais/fisiologia , Ratos , Núcleos Septais/metabolismo , Caracteres Sexuais , Fatores Sexuais
11.
Neurobiol Learn Mem ; 179: 107388, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33482320

RESUMO

The article reviews our studies of contextual fear conditioning (CFC) in rats during a period of development---Postnatal Day (PND) 17-33---that represents the late-infant, juvenile, and early-adolescent stages. These studies seek to acquire 'systems level' knowledge of brain and memory development and apply it to a rodent model of Fetal Alcohol Spectrum Disorder (FASD). This rodent model focuses on alcohol exposure from PND4-9, a period of brain development equivalent to the human third trimester, when neocortex, hippocampus, and cerebellum are especially vulnerable to adverse effects of alcohol. Our research emphasizes a variant of CFC, termed the Context Preexposure Facilitation Effect (CPFE, Fanselow, 1990), in which context representations incidentally learned on one occasion are retrieved and associated with immediate shock on a subsequent occasion. These representations can be encoded at the earliest developmental stage but seem not to be retained or retrieved until the juvenile period. This is associated with developmental differences in context-elicited expression, in prefrontal cortex, hippocampus, and amygdala, of immediate early genes (IEGs) that are implicated in long-term memory. Loss-of-function studies establish a functional role for these regions as soon as the CPFE emerges during ontogeny. In our rodent model of FASD, the CPFE is much more sensitive to alcohol dose than other commonly used cognitive tasks. This impairment can be reversed by acute administration during behavioral testing of drugs that enhance cholinergic function. This effect is associated with normalized IEG expression in prefrontal cortex during incidental context learning. In summary, our findings suggest that long-term memory of incidentally-learned context representations depends on prefrontal-hippocampal circuitry that is important both for the normative development of context conditioning and for its disruption by developmental alcohol exposure.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/crescimento & desenvolvimento , Condicionamento Clássico/fisiologia , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Memória , Aprendizagem Espacial/fisiologia , Animais , Encéfalo/fisiologia , Modelos Animais de Doenças , Medo , Transtornos do Espectro Alcoólico Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Precoces/genética , Crescimento e Desenvolvimento , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Aprendizagem , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/fisiologia , Ratos
12.
Zoolog Sci ; 37(5): 391-398, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32972079

RESUMO

In this study, we used the immediate early gene, egr-1, as a marker for neural activation and examined whether egr-1 expression is affected in brain regions associated with the social behavioral network (SBN) when social rank is determined and changed in male medaka fish (Oryzias latipes). Based on the behavioral contest protocol used in this study, we obtained four types of males: social ascending, social descending, dominant, and subordinate. In some brain regions associated with the SBN, we detected higher egr-1 expression in ascending and descending males than in dominant and subordinate males. Social-rank stable males (i.e., dominant and subordinate male fish) showed a similar level of egr-1 expression as the control male fish, which were housed without social stimulus of encountering another conspecific. These findings suggested that the transitioning of social rank could enhance neural activity in some brain regions associated with the SBN in male medaka. The use of medaka fish has many advantages in various fields of research such as genetics, developmental biology, environmental biology, and behavioral neurology. The findings of this study would contribute to future research exploring the roles of the SBN regions in regulating physiological and behavioral events associated with social-rank transition.


Assuntos
Comportamento Animal/fisiologia , Comportamento Competitivo/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes Precoces/fisiologia , Oryzias/fisiologia , Animais , Encéfalo/metabolismo , Genes Precoces/genética , Masculino
13.
Proc Natl Acad Sci U S A ; 117(34): 20860-20867, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32788362

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that latently infects hematopoietic cells and has the ability to reactivate when triggered by immunological stress. This reactivation causes significant morbidity and mortality in immune-deficient patients, who are unable to control viral dissemination. While a competent immune system helps prevent clinically detectable viremia, a portrait of the factors that induce reactivation following the proper cues remains incomplete. Our understanding of the complex molecular mechanisms underlying latency and reactivation continues to evolve. We previously showed the HCMV-encoded G protein-coupled receptor US28 is expressed during latency and facilitates latent infection by attenuating the activator protein-1 (AP-1) transcription factor subunit, c-fos, expression and activity. We now show AP-1 is a critical component for HCMV reactivation. Pharmacological inhibition of c-fos significantly attenuates viral reactivation. In agreement, infection with a virus in which we disrupted the proximal AP-1 binding site in the major immediate early (MIE) enhancer results in inefficient reactivation compared to WT. Concomitantly, AP-1 recruitment to the MIE enhancer is significantly decreased following reactivation of the mutant virus. Furthermore, AP-1 is critical for derepression of MIE-driven transcripts and downstream early and late genes, while immediate early genes from other loci remain unaffected. Our data also reveal MIE transcripts driven from the MIE promoter, the distal promoter, and the internal promoter, iP2, are dependent upon AP-1 recruitment, while iP1-driven transcripts are AP-1-independent. Collectively, our data demonstrate AP-1 binding to and activation of the MIE enhancer is a key molecular process controlling reactivation from latency.


Assuntos
Citomegalovirus/genética , Fator de Transcrição AP-1/metabolismo , Ativação Viral/genética , Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Genes Precoces/genética , Humanos , Proteínas Imediatamente Precoces/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Fator de Transcrição AP-1/genética , Ativação Transcricional/genética , Latência Viral/genética
14.
Antiviral Res ; 183: 104867, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32755660

RESUMO

Due to diverse pathogenic potentials, there is a growing need for anti-HCMV agents. In this study, we show that treatment with DAPT, a γ-secretase inhibitor (GSI), impairs HCMV replication as assessed by a progeny assay based on immunostaining. This effect is not limited to DAPT because other GSIs with different structures and distinct mechanisms of action also exhibit a similar level of inhibitory effects on HCMV viral production, indicating that γ-secretase activity is required for efficient HCMV replication. Western blot and qPCR analyses reveal that DAPT does not interfere with the viral entry process, but reduces expression of the immediate early protein IE1 at the transcriptional level. Furthermore, we exclude the possible involvement of Notch signaling pathway during HCMV replication by showing that expression of the dominant-negative form of MAML1, which disrupts the transactivational ability of Notch intracellular domain (NICD), does not reduce viral particle formation, and that NICD cannot rescue the DAPT-treated outcomes. Taken together, these findings indicate that γ-secretase activity plays an important role in a key step of the HCMV life cycle and γ-secretase inhibition could potentially be used as a novel preventive and therapeutic strategy against HCMV infection and HCMV-related diseases.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Diaminas/farmacologia , Genes Precoces/genética , Tiazóis/farmacologia , Transcrição Gênica/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Citomegalovirus/enzimologia , Citomegalovirus/genética , Fibroblastos/virologia , Prepúcio do Pênis/citologia , Regulação Viral da Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/metabolismo , Masculino , Transdução de Sinais/efeitos dos fármacos , Internalização do Vírus
15.
Proc Natl Acad Sci U S A ; 117(31): 18764-18770, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32694203

RESUMO

Human progenitor cells (HPCs) support human cytomegalovirus (HCMV) latency, and their differentiation along the myeloid lineage triggers cellular cues that drive reactivation. A key step during HCMV reactivation in latently infected HPCs is reexpression of viral major immediate early (MIE) genes. We recently determined that the major immediate early promoter (MIEP), which is primarily responsible for MIE gene expression during lytic replication, remains silent during reactivation. Instead, alternative promoters in the MIE locus are induced by reactivation stimuli. Here, we find that forkhead family (FOXO) transcription factors are critical for activation of alternative MIE promoters during HCMV reactivation, as mutating FOXO binding sites in alternative MIE promoters decreased HCMV IE gene expression upon reactivation and significantly decreased the production of infectious virus from latently infected primary CD34+ HPCs. These findings establish a mechanistic link by which infected cells sense environmental cues to regulate latency and reactivation, and emphasize the role of contextual activation of alternative MIE promoters as the primary drivers of reactivation.


Assuntos
Citomegalovirus , Fatores de Transcrição Forkhead/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Virais/metabolismo , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Genes Precoces/genética , Células HeLa , Humanos , Latência Viral
16.
BMC Microbiol ; 20(1): 227, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32723300

RESUMO

BACKGROUND: Wogonin, a natural flavonoid-like chemical compound, exhibits anti-inflammatory, antitumor, antiviral, neuroprotective, and anxiolytic effects by modulating a variety of cellular signaling pathways including PI3K-Akt, p53, nuclear factor κB (NF-κB), mitogen-activated protein kinase (MAPK) pathways. In this study, its antiviral effect against herpes simplex virus (HSV) type 1 and 2 (HSV-1 and HSV-2) replication was investigated. RESULTS: Wogonin suppressed HSV-2-induced cytopathic effect (CPE) and reduced viral mRNA transcription, viral protein synthesis, and infectious virion particle titers in a dose-dependent manner. A time-of-drug-addition assay demonstrated that wogonin acted as a postentry viral inhibitor. Wogonin also significantly reduced HSV-induced NF-κB and MAPK pathway activation, which has previously been demonstrated to be important for viral replication. CONCLUSIONS: Our results suggest that the anti-herpes effect of wogonin may be mediated by modulation of cellular NF-κB and JNK/p38 MAPK pathways and imply that wogonin may be useful as an anti-HSV agent.


Assuntos
Antivirais/farmacologia , Flavanonas/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Aciclovir/farmacologia , Linhagem Celular , Efeito Citopatogênico Viral/efeitos dos fármacos , Sinergismo Farmacológico , Expressão Gênica/efeitos dos fármacos , Genes Precoces/genética , Humanos , Transdução de Sinais/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
17.
Sci Rep ; 10(1): 7887, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398802

RESUMO

Honey bees and bumble bees belong to the same family (Apidae) and their workers exhibit a division of labor, but the style of division of labor differs between species. The molecular and neural bases of the species-specific social behaviors of Apidae workers have not been analyzed. Here, we focused on two immediate early genes, hormone receptor 38 (HR38) and early growth response gene-1 (Egr1), and late-upregulated ecdysone receptor (EcR), all of which are upregulated by foraging flight and expressed preferentially in the small-type Kenyon cells of the mushroom bodies (MBs) in the honey bee brain. Gene expression analyses in Bombus ignitus revealed that HR38 and Egr1, but not EcR, exhibited an immediate early response during awakening from CO2 anesthesia. Both premature mRNA for HR38 and mature mRNA for Egr1 were induced during foraging flight, and mRNAs for HR38 and Egr1 were sparsely detected inside the whole MB calyces. In contrast, EcR expression was higher in forager brains than in nurse bees and was expressed preferentially in the small-type Kenyon cells inside the MBs. Our findings suggest that Kenyon cells are active during foraging flight and that the function of late-upregulated EcR in the brain is conserved among these Apidae species.


Assuntos
Abelhas/genética , Encéfalo/metabolismo , Comportamento Alimentar/fisiologia , Voo Animal/fisiologia , Genes Precoces/genética , Proteínas de Insetos/genética , Animais , Abelhas/fisiologia , Encéfalo/fisiologia , Mapeamento Encefálico , Regulação da Expressão Gênica , Hibridização In Situ , Corpos Pedunculados/metabolismo , Corpos Pedunculados/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Gen Virol ; 101(6): 635-644, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32375946

RESUMO

Human cytomegalovirus latency and reactivation is a major source of morbidity in immune-suppressed patient populations. Lifelong latent infections are established in CD34+progenitor cells in the bone marrow, which are hallmarked by a lack of major lytic gene expression, genome replication and virus production. A number of studies have shown that inhibition of the major immediate early promoter (MIEP) - the promoter that regulates immediate early (IE) gene expression - is important for the establishment of latency and that, by extension, reactivation requires reversal of this repression of the MIEP. The identification of novel promoters (termed ip1 and ip2) downstream of the MIEP that can drive IE gene expression has led to speculation over the precise role of the MIEP in reactivation. In this study we show that IE transcripts arise from both the MIEP and ip2 promoter in the THP1 cell macrophage cell line and also CD14+monocytes stimulated with phorbol ester. In contrast, we show that in in vitro generated dendritic cells or macrophages that support HCMV reactivation IE transcripts arise predominantly from the MIEP and not the intronic promoters. Furthermore, inhibition of histone modifying enzyme activity confirms the view that the MIEP is predominantly regulated by the activity of cellular chromatin. Finally, we observe that ip2-derived IE transcription is cycloheximide-sensitive in reactivating DCs, behaviour consistent with an early gene designation. Taken together, these data argue that MIEP activity is still important for HCMV reactivation but ip2 activity could play cell-type-specific roles in reactivation.


Assuntos
Citomegalovirus/genética , Células Dendríticas/virologia , Genes Precoces/genética , Proteínas Imediatamente Precoces/genética , Regiões Promotoras Genéticas/genética , Células-Tronco/virologia , Transcrição Gênica/genética , Cromatina/genética , Infecções por Citomegalovirus/virologia , Regulação Viral da Expressão Gênica/genética , Humanos , Macrófagos/virologia , Monócitos/virologia , Células THP-1/virologia , Ativação Viral/genética , Latência Viral/genética
19.
Sci Rep ; 10(1): 6325, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286359

RESUMO

Forskolin promotes neuronal differentiation of PC12 cells via the PKA-CREB-dependent signaling pathway. Activation of PKA by forskolin phosphorylates CREB, which then binds to CRE sites in numerous gene promoters. However, it is unclear which gene contains the CRE sites responsible for forskolin-induced neuronal differentiation. In this study, we investigated how an immediate early gene, nur77, which has CRE sites in the promoter region, contributes to the early stage of differentiation of forskolin-treated PC12 cells. After treatment with forskolin, expression of Nur77 was upregulated within 1 hr. In addition, knockdown of nur77 inhibited neurite outgrowth induced by forskolin. We also revealed that the specific four CRE sites near the transcriptional start site (TSS) of nur77 were strongly associated with phosphorylated CREB within 1 hr after treatment with forskolin. To analyze the roles of these four sites, reporter assays using the nur77 promoter region were performed. The results showed that nur77 expression was mediated through three of the CRE sites, -242, -222, and -78, and that -78, the nearest of the three to the TSS of nur77, was particularly important. An analysis of neuronal markers controlled by Nur77 after A-CREB-Nur77-Synapsin1 signaling pathway plays a pivotal role in differentiation of forskolin-induced PC12 cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colforsina/farmacologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Ativação Transcricional/efeitos dos fármacos , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Técnicas de Silenciamento de Genes , Genes Precoces/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Células PC12 , Regiões Promotoras Genéticas/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Sinapsinas/metabolismo
20.
Biochem Biophys Res Commun ; 526(3): 574-579, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32247609

RESUMO

Genetic engineering for neuronal cell activity labeling and neuronal cell activity modulation are invaluable for elucidating the underlying characteristics of the brain and neurons. In this study, ferritin fusion protein (FFP) was combined with Tet expression construct under a modified immediate-early gene (IEG) Arc/Arg3.1 promoter so-called SARE-ArcMin. This expression system is a neuronal activity-dependent expression module for nano-ferritin, a radio/magnetic wave-sensitive protein well-accepted as a potential recombinant neuronal actuator. The system was characterized in transcriptional and translational levels in human neuroblastoma SH-SY5Y cells. The mRNA and protein expression levels of nano-ferritin were significant in the activated neurons suggesting that the activity dependent expression patterns of the ferritin also acted as a neuronal cell activation indicator. The system sufficed the need for precise neuronal cell activity specific expression and demonstrated a platform that suggested the use of the nano-ferritin for the study of neuronal cells.


Assuntos
Ferritinas/genética , Neurônios/metabolismo , Regiões Promotoras Genéticas , Expressão Gênica , Genes Precoces/genética , Engenharia Genética , Vetores Genéticos/genética , Humanos , Campos Magnéticos , Modelos Moleculares , Plasticidade Neuronal , Neurônios/citologia , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...