Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 134(11): e150-e175, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38781298

RESUMO

HIV type 1 (HIV-1) is the causative agent of AIDS. Since the start of the epidemic, HIV/AIDS has been responsible for ≈40 million deaths. Additionally, an estimated 39 million people are currently infected with the virus. HIV-1 primarily infects immune cells, such as CD4+ (cluster of differentiation 4+) T lymphocytes (T cells), and as a consequence, the number of CD4+ T cells progressively declines in people living with HIV. Within a span of ≈10 years, HIV-1 infection leads to the systemic failure of the immune system and progression to AIDS. Fortunately, potent antiviral therapy effectively controls HIV-1 infection and prevents AIDS-related deaths. The efficacy of the current antiviral therapy regimens has transformed the outcome of HIV/AIDS from a death sentence to a chronic disease with a prolonged lifespan of people living with HIV. However, antiviral therapy is not curative, is challenged by virus resistance, can be toxic, and, most importantly, requires lifelong adherence. Furthermore, the improved lifespan has resulted in an increased incidence of non-AIDS-related morbidities in people living with HIV including cardiovascular diseases, renal disease, liver disease, bone disease, cancer, and neurological conditions. In this review, we summarize the current state of knowledge of the cardiovascular comorbidities associated with HIV-1 infection, with a particular focus on hypertension. We also discuss the potential mechanisms known to drive HIV-1-associated hypertension and the knowledge gaps in our understanding of this comorbid condition. Finally, we suggest several directions of future research to better understand the factors, pathways, and mechanisms underlying HIV-1-associated hypertension in the post-antiviral therapy era.


Assuntos
Infecções por HIV , Hipertensão , Humanos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia , Infecções por HIV/complicações , Hipertensão/tratamento farmacológico , Hipertensão/epidemiologia , Fatores de Risco , HIV-1/patogenicidade , Animais
2.
bioRxiv ; 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38562877

RESUMO

HIV-1 integration into the human genome is dependent on 3'-processing of the reverse transcribed viral DNA. Recently, we reported that the cellular Three Prime Repair Exonuclease 1 (TREX1) enhances HIV-1 integration by degrading the unprocessed viral DNA, while the integration-competent 3'-processed DNA remained resistant. Here, we describe the mechanism by which the 3'-processed HIV-1 DNA resists TREX1-mediated degradation. Our kinetic studies revealed that the rate of cleavage (kcat) of the 3'-processed DNA was significantly lower than the unprocessed HIV-1 DNA by TREX1. The efficiency of degradation (kcat/KM) of the 3'-processed DNA was also significantly lower than the unprocessed DNA. Furthermore, the binding affinity (Kd) of TREX1 was markedly lower to the 3'-processed DNA compared to the unprocessed DNA. Molecular docking and dynamics studies revealed distinct conformational binding modes of TREX1 with the 3'-processed and unprocessed HIV-1 DNA. Particularly, the unprocessed DNA was favorably positioned in the active site with polar interactions with the catalytic residues of TREX1. Additionally, a stable complex was formed between TREX1 and the unprocessed DNA compared the 3'-processed DNA. These results pinpoint the biochemical mechanism by which TREX1 preferentially degrades the integration-incompetent HIV-1 DNA and reveal the unique structural and conformational properties of the integration-competent 3'-processed HIV-1 DNA.

3.
mBio ; 15(1): e0021222, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38085100

RESUMO

IMPORTANCE: HIV-1 capsid protein (CA)-independently or by recruiting host factors-mediates several key steps of virus replication in the cytoplasm and nucleus of the target cell. Research in the recent years have established that CA is multifunctional and genetically fragile of all the HIV-1 proteins. Accordingly, CA has emerged as a validated and high priority therapeutic target, and the first CA-targeting antiviral drug was recently approved for treating multi-drug resistant HIV-1 infection. However, development of next generation CA inhibitors depends on a better understanding of CA's known roles, as well as probing of CA's novel roles, in HIV-1 replication. In this timely review, we present an updated overview of the current state of our understanding of CA's multifunctional role in HIV-1 replication-with a special emphasis on CA's newfound post-nuclear roles, highlight the pressing knowledge gaps, and discuss directions for future research.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Humanos , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , HIV-1/genética , HIV-1/metabolismo , Soropositividade para HIV/metabolismo , Replicação Viral/genética , Integração Viral
4.
J Biol Chem ; 300(2): 105605, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38159857

RESUMO

Prolidase (PEPD) is the only hydrolase that cleaves the dipeptides containing C-terminal proline or hydroxyproline-the rate-limiting step in collagen biosynthesis. However, the molecular regulation of prolidase expression remains largely unknown. In this study, we have identified overlapping binding sites for the transcription factors Krüppel-like factor 6 (KLF6) and Specificity protein 1 (Sp1) in the PEPD promoter and demonstrate that KLF6/Sp1 transcriptionally regulate prolidase expression. By cloning the PEPD promoter into a luciferase reporter and through site-directed deletion, we pinpointed the minimal sequences required for KLF6 and Sp1-mediated PEPD promoter-driven transcription. Interestingly, Sp1 inhibition abrogated KLF6-mediated PEPD promoter activity, suggesting that Sp1 is required for the basal expression of prolidase. We further studied the regulation of PEPD by KLF6 and Sp1 during transforming growth factor ß1 (TGF-ß1) signaling, since both KLF6 and Sp1 are key players in TGF-ß1 mediated collagen biosynthesis. Mouse and human fibroblasts exposed to TGF-ß1 resulted in the induction of PEPD transcription and prolidase expression. Inhibition of TGF-ß1 signaling abrogated PEPD promoter-driven transcriptional activity of KLF6 and Sp1. Knock-down of KLF6 as well as Sp1 inhibition also reduced prolidase expression. Chromatin immunoprecipitation assay supported direct binding of KLF6 and Sp1 to the PEPD promoter and this binding was enriched by TGF-ß1 treatment. Finally, immunofluorescence studies showed that KLF6 co-operates with Sp1 in the nucleus to activate prolidase expression and enhance collagen biosynthesis. Collectively, our results identify functional elements of the PEPD promoter for KLF6 and Sp1-mediated transcriptional activation and describe the molecular mechanism of prolidase expression.


Assuntos
Dipeptidases , Fator 6 Semelhante a Kruppel , Transdução de Sinais , Fator de Transcrição Sp1 , Animais , Humanos , Camundongos , Colágeno/metabolismo , Fator 6 Semelhante a Kruppel/genética , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
5.
J Virol ; 97(11): e0073223, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37843371

RESUMO

The HIV-1 genome encodes a small number of proteins with structural, enzymatic, regulatory, and accessory functions. These viral proteins interact with a number of host factors to promote the early and late stages of HIV-1 infection. During the early stages of infection, interactions between the viral proteins and host factors enable HIV-1 to enter the target cell, traverse the cytosol, dock at the nuclear pore, gain access to the nucleus, and integrate into the host genome. Similarly, the viral proteins recruit another set of host factors during the late stages of infection to orchestrate HIV-1 transcription, translation, assembly, and release of progeny virions. Among the host factors implicated in HIV-1 infection, Cyclophilin A (CypA) was identified as the first host factor to be packaged within HIV-1 particles. It is now well established that CypA promotes HIV-1 infection by directly binding to the viral capsid. Mechanistic models to pinpoint CypA's role have spanned from an effect in the producer cell to the early steps of infection in the target cell. In this review, we will describe our understanding of the role(s) of CypA in HIV-1 infection, highlight the current knowledge gaps, and discuss the potential role of this host factor in the post-nuclear entry steps of HIV-1 infection.


Assuntos
Ciclofilina A , Infecções por HIV , HIV-1 , Humanos , Proteínas do Capsídeo/genética , Núcleo Celular/metabolismo , Ciclofilina A/genética , Ciclofilina A/metabolismo , Infecções por HIV/metabolismo , HIV-1/genética , HIV-1/metabolismo , Proteínas Virais/metabolismo , Interações Hospedeiro-Patógeno
6.
J Virol ; 96(18): e0101122, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36094316

RESUMO

HIV-1 DNA is preferentially integrated into chromosomal hot spots by the preintegration complex (PIC). To understand the mechanism, we measured the DNA integration activity of PICs-extracted from infected cells-and intasomes, biochemically assembled PIC substructures using a number of relevant target substrates. We observed that PIC-mediated integration into human chromatin is preferred compared to genomic DNA. Surprisingly, nucleosomes lacking histone modifications were not preferred integration compared to the analogous naked DNA. Nucleosomes containing the trimethylated histone 3 lysine 36 (H3K36me3), an epigenetic mark linked to active transcription, significantly stimulated integration, but the levels remained lower than the naked DNA. Notably, H3K36me3-modified nucleosomes with linker DNA optimally supported integration mediated by the PIC but not by the intasome. Interestingly, optimal intasome-mediated integration required the cellular cofactor LEDGF. Unexpectedly, LEDGF minimally affected PIC-mediated integration into naked DNA but blocked integration into nucleosomes. The block for the PIC-mediated integration was significantly relieved by H3K36me3 modification. Mapping the integration sites in the preferred substrates revealed that specific features of the nucleosome-bound DNA are preferred for integration, whereas integration into naked DNA was random. Finally, biochemical and genetic studies demonstrate that DNA condensation by the H1 protein dramatically reduces integration, providing further evidence that features inherent to the open chromatin are preferred for HIV-1 integration. Collectively, these results identify the optimal target substrate for HIV-1 integration, report a mechanistic link between H3K36me3 and integration preference, and importantly, reveal distinct mechanisms utilized by the PIC for integration compared to the intasomes. IMPORTANCE HIV-1 infection is dependent on integration of the viral DNA into the host chromosomes. The preintegration complex (PIC) containing the viral DNA, the virally encoded integrase (IN) enzyme, and other viral/host factors carries out HIV-1 integration. HIV-1 integration is not dependent on the target DNA sequence, and yet the viral DNA is selectively inserted into specific "hot spots" of human chromosomes. A growing body of literature indicates that structural features of the human chromatin are important for integration targeting. However, the mechanisms that guide the PIC and enable insertion of the PIC-associated viral DNA into specific hot spots of the human chromosomes are not fully understood. In this study, we describe a biochemical mechanism for the preference of the HIV-1 DNA integration into open chromatin. Furthermore, our study defines a direct role for the histone epigenetic mark H3K36me3 in HIV-1 integration preference and identify an optimal substrate for HIV-1 PIC-mediated viral DNA integration.


Assuntos
Cromossomos Humanos , HIV-1 , Código das Histonas , Histonas , Nucleossomos , Integração Viral , Cromatina/metabolismo , Cromossomos Humanos/virologia , DNA Viral/genética , DNA Viral/metabolismo , Infecções por HIV/virologia , Integrase de HIV/genética , Integrase de HIV/metabolismo , HIV-1/genética , Histonas/química , Histonas/metabolismo , Humanos , Lisina/genética , Metilação , Nucleossomos/genética , Nucleossomos/metabolismo , Nucleossomos/virologia , Integração Viral/genética
7.
PNAS Nexus ; 1(2): pgac064, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35719891

RESUMO

HIV-1 replication is durably controlled without antiretroviral therapy (ART) in certain infected individuals called elite controllers (ECs). These individuals express specific human leukocyte antigens (HLA) that tag HIV-infected cells for elimination by presenting viral epitopes to CD8+ cytotoxic T-lymphocytes (CTL). In HIV-infected individuals expressing HLA-B27, CTLs primarily target the viral capsid protein (CA)-derived KK10 epitope. While selection of CA mutation R264K helps HIV-1 escape this potent CTL response, the accompanying fitness cost severely diminishes virus infectivity. Interestingly, selection of a compensatory CA mutation S173A restores HIV-1 replication. However, the molecular mechanism(s) underlying HIV-1 escape from this ART-free virus control by CTLs is not fully understood. Here, we report that the R264K mutation-associated infectivity defect arises primarily from impaired HIV-1 DNA integration, which is restored by the S173A mutation. Unexpectedly, the integration defect of the R264K variant was also restored upon depletion of the host cyclophilin A. These findings reveal a nuclear crosstalk between CA and HIV-1 integration as well as identify a previously unknown role of cyclophilin A in viral DNA integration. Finally, our study identifies a novel immune escape mechanism of an HIV-1 variant escaping a CA-directed CTL response.

8.
Front Mol Biosci ; 8: 723003, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34532344

RESUMO

Prolidase (peptidase D), encoded by the PEPD gene, is a ubiquitously expressed cytosolic metalloproteinase, the only enzyme capable of cleaving imidodipeptides containing C-terminal proline or hydroxyproline. Prolidase catalyzes the rate-limiting step during collagen recycling and is essential in protein metabolism, collagen turnover, and matrix remodeling. Prolidase, therefore plays a crucial role in several physiological processes such as wound healing, inflammation, angiogenesis, cell proliferation, and carcinogenesis. Accordingly, mutations leading to loss of prolidase catalytic activity result in prolidase deficiency a rare autosomal recessive metabolic disorder characterized by defective wound healing. In addition, alterations in prolidase enzyme activity have been documented in numerous pathological conditions, making prolidase a useful biochemical marker to measure disease severity. Furthermore, recent studies underscore the importance of a non-enzymatic role of prolidase in cell regulation and infectious disease. This review aims to provide comprehensive information on prolidase, from its discovery to its role in health and disease, while addressing the current knowledge gaps.

9.
J Virol ; 95(17): e0055521, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34105995

RESUMO

Three prime repair exonuclease 1 (TREX1) is the most abundant 3'→5' exonuclease in mammalian cells. It has been suggested that TREX1 degrades HIV-1 DNA to enable the virus to evade the innate immune system. However, the exact role of TREX1 during early steps of HIV-1 infection is not clearly understood. In this study, we report that HIV-1 infection is associated with upregulation, perinuclear accumulation, and nuclear localization of TREX1. However, TREX1 overexpression did not affect reverse transcription or nuclear entry of the virus. Surprisingly, HIV-1 DNA integration was increased in TREX1-overexpressing cells, suggesting a role of the exonuclease in the post-nuclear entry step of infection. Accordingly, preintegration complexes (PICs) extracted from TREX1-overexpressing cells retained higher levels of DNA integration activity. TREX1 depletion resulted in reduced levels of proviral integration, and PICs formed in TREX1-depleted cells retained lower DNA integration activity. Addition of purified TREX1 to PICs also enhanced DNA integration activity, suggesting that TREX1 promotes HIV-1 integration by stimulating PIC activity. To understand the mechanism, we measured TREX1 exonuclease activity on substrates containing viral DNA ends. These studies revealed that TREX1 preferentially degrades the unprocessed viral DNA, but the integration-competent 3'-processed viral DNA remains resistant to degradation. Finally, we observed that TREX1 addition stimulates the activity of HIV-1 intasomes assembled with the unprocessed viral DNA but not that of intasomes containing the 3'-processed viral DNA. These biochemical analyses provide a mechanism by which TREX1 directly promotes HIV-1 integration. Collectively, our study demonstrates that HIV-1 infection upregulates TREX1 to facilitate viral DNA integration. IMPORTANCE Productive HIV-1 infection is dependent on a number of cellular factors. Therefore, a clear understanding of how the virus exploits the cellular machinery will identify new targets for inhibiting HIV-1 infection. The three prime repair exonuclease 1 (TREX1) is the most active cellular exonuclease in mammalian cells. It has been reported that TREX1 prevents accumulation of HIV-1 DNA and enables the virus to evade the host innate immune response. Here, we show that HIV-1 infection results in the upregulation, perinuclear accumulation, and nuclear localization of TREX1. We also provide evidence that TREX1 promotes HIV-1 integration by preferentially degrading viral DNAs that are incompatible with chromosomal insertion. These observations identify a novel role of TREX1 in a post-nuclear entry step of HIV-1 infection.


Assuntos
DNA Viral/metabolismo , Exodesoxirribonucleases/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Imunidade Inata/imunologia , Fosfoproteínas/metabolismo , Integração Viral , Replicação Viral , Núcleo Celular , DNA Viral/genética , Exodesoxirribonucleases/genética , Células HEK293 , Infecções por HIV/genética , Células HeLa , Humanos , Fosfoproteínas/genética
10.
Sci Rep ; 11(1): 1422, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446840

RESUMO

Methamphetamine (METH) is a highly addictive psychostimulant that causes long-lasting effects in the brain and increases the risk of developing neurodegenerative diseases. The cellular and molecular effects of METH in the brain are functionally linked to alterations in glutamate levels. Despite the well-documented effects of METH on glutamate neurotransmission, the underlying mechanism by which METH alters glutamate levels is not clearly understood. In this study, we report an essential role of proline biosynthesis in maintaining METH-induced glutamate homeostasis. We observed that acute METH exposure resulted in the induction of proline biosynthetic enzymes in both undifferentiated and differentiated neuronal cells. Proline level was also increased in these cells after METH exposure. Surprisingly, METH treatment did not increase glutamate levels nor caused neuronal excitotoxicity. However, METH exposure resulted in a significant upregulation of pyrroline-5-carboxylate synthase (P5CS), the key enzyme that catalyzes synthesis of proline from glutamate. Interestingly, depletion of P5CS by CRISPR/Cas9 resulted in a significant increase in glutamate levels upon METH exposure. METH exposure also increased glutamate levels in P5CS-deficient proline-auxotropic cells. Conversely, restoration of P5CS expression in P5CS-deficient cells abrogated the effect of METH on glutamate levels. Consistent with these findings, P5CS expression was significantly enhanced in the cortical brain region of mice administered with METH and in the slices of cortical brain tissues treated with METH. Collectively, these results uncover a key role of P5CS for the molecular effects of METH and highlight that excess glutamate can be sequestered for proline biosynthesis as a protective mechanism to maintain glutamate homeostasis during drug exposure.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Córtex Cerebral/metabolismo , Ácido Glutâmico/metabolismo , Homeostase/efeitos dos fármacos , Metanfetamina/toxicidade , Prolina/biossíntese , Doença Aguda , Aldeído Desidrogenase/metabolismo , Animais , Células CHO , Cricetulus , Humanos , Masculino , Camundongos , Neurônios/metabolismo
11.
J Prosthet Dent ; 126(3): 393-397, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32951870

RESUMO

STATEMENT OF PROBLEM: How different methods of recording posterior palatal seal affect removable complete-denture retention and oral health quality of life is unclear. PURPOSE: The purpose of this clinical study was to compare the retention and oral health quality of life (OHIP-14) between conventional and arbitrary posterior palatal seal techniques in participants with removable complete dentures. MATERIAL AND METHODS: Edentulous patients were recruited according to definitive criteria. The participants were randomly divided into conventional and arbitrary seal. After the delivery of the denture, the retention was evaluated with a force gauge dynamometer and at 3, 6, 9, and 12 months. Denture satisfaction was evaluated with the OHIP-14 questionnaire. Data were statistically analyzed by using the t test and repeated measure ANOVA (α=.05). RESULTS: The mean ±standard deviation values (N) for conventional seal at 0, 3, 6, 9, and 12 months by dynamometer in the anterior region ranged from 4.73 ±0.78 to 4.90 ±0.81 and in the posterior region between 5.07 ±0.84 and 5.31 ±0.99. Dynamometer values for arbitrary seal in the anterior region were from 4.56 ±0.77 to 4.88 ±0.81, and in the posterior, it varied between 4.74 ±0.74 and 5.15 ±0.81. Force gauge values (N) for conventional and arbitrary seal were in the range of 18.35 ±2.84 to 20.69 ±3.89. The general mean ±SD OHIP-14 was higher for the conventional seal at 3.12 ±0.25 than for the arbitrary seal at 2.73 ±0.23 The difference between the conventional and arbitrary seal techniques was not statistically significant (P>.05) CONCLUSIONS: No significant difference in complete denture retention was detected between the 2 posterior palatal seal techniques. Oral health quality of life was higher with the conventional seal technique.


Assuntos
Boca Edêntula , Qualidade de Vida , Retenção de Dentadura , Prótese Total , Humanos , Saúde Bucal , Satisfação do Paciente
12.
Contemp Clin Dent ; 11(2): 116-120, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33110322

RESUMO

BACKGROUND: Clinical studies have established mastication as a stress relaxation behavior in humans. Absence of teeth compromises mastication, increasing psychologic stress in individuals depicted by many physiologic changes in body. Quantitative level of psychologic stress bio-markers serve as indicators of underlying physical ailment. Lesser literatures are available in determining the role of alpha amylase stress bio marker in partially edentulous clinical situations. AIM: The purpose of this clinical study was to evaluate the levels of salivary alpha-amylase (sAA) stress biomarker in partially edentulous subjects before and after restoration with fixed dental prosthesis. MATERIAL AND METHODS: Forty partially edentulous patients with missing mandibular first molar were selected for this study. Two questionnaires, state trait anxiety inventory (STAI) and perceived stress scale (PSS) was used to evaluate stress and anxiety levels of participants. The recruited participants were treated with metal ceramic fixed dental prosthesis (FDP). A visual analog scale (VAS) was used to determine the patient satisfaction .Unstimulated salivary samples were collected preoperative, 3rd and 6th month post FDP placement. Level of sAA was estimated. Data obtained in the form of mean ± SD was subjected to statistical analysis using paired sample t-test (α=.05). RESULTS: The salivary alpha amylase level was highest with mean of 36.73 µM/min/mg ptn before restoration with FDP. In the third month after prosthesis placement, the enzyme values decreased to16.62 µM/min/mg ptn and least value of 8.58 µM/min/mg ptn was detected in sixth month (P < 0.05). CONCLUSION: The salivary alpha amylase stress biomarker decreased after tooth replacement with FDP.

13.
J Oral Biol Craniofac Res ; 10(4): 523-528, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32874882

RESUMO

PURPOSE: The study evaluated the clinical efficacy of Polyether ether ketone (PEEK) fixed dental prosthesis (FDP) over a period of one year using modified Ryges criteria and California Dental Assessment system. METHODS: The group consisted of 20 patients restored with posterior three-unit PEEK FDP. Patient recall and clinical examination of the restorations were done at interval of 0, 3, 6, 9 and 12 months. Clinical examination for evaluation of longevity of restorations was done using modified Ryges criteria and California dental assessment system on chipping of the veneered composite, discoloration at the marginal areas, and marginal adaptation, retention, periodontal health and hygiene of PEEK FDP. Radiographic assessment was done after 12 months. Statistical analysis were done using Friedman test. RESULTS: 95% of the patients had maintained the restoration of PEEK FDP without fracture during the study period. 5% patient reported with de-cementation of fixed dental prosthesis. 10% of the PEEK FDP shows marginal discoloration. However, no significant changes were observed during the periodic time interval evaluation in marginal adaptation, oral hygiene status and periodontal health. (p < 0.5). CONCLUSION: PEEK FDP had satisfactory clinical efficacy and acceptable clinical outcome during the observation period of 12 months. No significant radiological changes were observed at the end of 12 months.

14.
Sci Rep ; 10(1): 11197, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32641757

RESUMO

MiR-124 is a highly expressed miRNA in the brain and regulates genes involved in neuronal function. We report that miR-124 post-transcriptionally regulates PARP-1. We have identified a highly conserved binding site of miR-124 in the 3'-untranslated region (3'UTR) of Parp-1 mRNA. We demonstrate that miR-124 directly binds to the Parp-1 3'UTR and mutations in the seed sequences abrogate binding between the two RNA molecules. Luciferase reporter assay revealed that miR-124 post-transcriptionally regulates Parp-1 3'UTR activity in a dopaminergic neuronal cell model. Interestingly, the binding region of miR-124 in Parp-1 3'UTR overlapped with the target sequence of miR-125b, another post-transcriptional regulator of Parp-1. Our results from titration and pull-down studies revealed that miR-124 binds to Parp-1 3'UTR with greater affinity and confers a dominant post-transcriptional inhibition compared to miR-125b. Interestingly, acute or chronic cocaine exposure downregulated miR-124 levels concomitant with upregulation of PARP-1 protein in dopaminergic-like neuronal cells in culture. Levels of miR-124 were also downregulated upon acute or chronic cocaine exposure in the mouse nucleus accumbens (NAc)-a key reward region of brain. Time-course studies revealed that cocaine treatment persistently downregulated miR-124 in NAc. Consistent with this finding, miR-124 expression was also significantly reduced in the NAc of animals conditioned for cocaine place preference. Collectively, these studies identify Parp-1 as a direct target of miR-124 in neuronal cells, establish miR-124 as a cocaine-regulated miRNA in the mouse NAc, and highlight a novel pathway underlying the molecular effects of cocaine.


Assuntos
Cocaína/farmacologia , MicroRNAs/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/genética , Regiões 3' não Traduzidas/genética , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , MicroRNAs/genética , Modelos Animais , Mutação , Núcleo Accumbens/citologia , Núcleo Accumbens/metabolismo
15.
J Biol Chem ; 295(15): 5081-5094, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32152226

RESUMO

Cleavage and polyadenylation specificity factor 6 (CPSF6) is a cellular protein involved in mRNA processing. Emerging evidence suggests that CPSF6 also plays key roles in HIV-1 infection, specifically during nuclear import and integration targeting. However, the cellular and molecular mechanisms that regulate CPSF6 expression are largely unknown. In this study, we report a post-transcriptional mechanism that regulates CPSF6 via the cellular microRNA miR-125b. An in silico analysis revealed that the 3'UTR of CPSF6 contains a miR-125b-binding site that is conserved across several mammalian species. Because miRNAs repress protein expression, we tested the effects of miR-125b expression on CPSF6 levels in miR-125b knockdown and over-expression experiments, revealing that miR-125b and CPSF6 levels are inversely correlated. To determine whether miR-125b post-transcriptionally regulates CPSF6, we introduced the 3'UTR of CPSF6 mRNA into a luciferase reporter and found that miR-125b negatively regulates CPSF6 3'UTR-driven luciferase activity. Accordingly, mutations in the miR-125b seed sequence abrogated the regulatory effect of the miRNA on the CPSF6 3'UTR. Finally, pulldown experiments demonstrated that miR-125b physically interacts with CPSF6 3'UTR. Interestingly, HIV-1 infection down-regulated miR-125b expression concurrent with up-regulation of CPSF6. Notably, miR-125b down-regulation in infected cells was not due to reduced pri-miRNA or pre-miRNA levels. However, miR-125b down-regulation depended on HIV-1 reverse transcription but not viral DNA integration. These findings establish a post-transcriptional mechanism that controls CPSF6 expression and highlight a novel function of miR-125b during HIV-host interaction.


Assuntos
Regiões 3' não Traduzidas/genética , Capsídeo/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , MicroRNAs/genética , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Sítios de Ligação , Infecções por HIV/genética , Infecções por HIV/metabolismo , Humanos , MicroRNAs/metabolismo , Mutação , Integração Viral , Fatores de Poliadenilação e Clivagem de mRNA/química , Fatores de Poliadenilação e Clivagem de mRNA/genética
16.
J Life Sci (Westlake Village) ; 1(1): 4-37, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31468033

RESUMO

The human immunodeficiency virus (HIV) infection of the immune cells expressing the cluster of differentiation 4 cell surface glycoprotein (CD4+ cells) causes progressive decline of the immune system and leads to the acquired immunodeficiency syndrome (AIDS). The ongoing global HIV/AIDS pandemic has already claimed over 35 million lives. Even after 37 years into the epidemic, neither a cure is available for the 37 million people living with HIV (PLHIV) nor is a vaccine discovered to avert the millions of new HIV infections that continue to occur each year. If left untreated, HIV infection typically progresses to AIDS and, ultimately, causes death in a majority of PLHIV. The recommended combination antiretroviral therapy (cART) suppresses virus replication and viremia, prevents or delays progression to AIDS, reduces transmission rates, and lowers HIV-associated mortality and morbidity. However, because cART does not eliminate HIV, and an enduring pool of infected resting memory CD4+ T cells (latent HIV reservoir) is established early on, any interruption to cART leads to a relapse of viremia and disease progression. Hence, strict adherence to a life-long cART regimen is mandatory for managing HIV infection in PLHIV. The HIV-1-specific cytotoxic T cells expressing the CD8 glycoprotein (CD8+ CTL) limit the virus replication in vivo by recognizing the viral antigens presented by human leukocyte antigen (HLA) class I molecules on the infected cell surface and killing those cells. Nevertheless, CTLs fail to durably control HIV-1 replication and disease progression in the absence of cART. Intriguingly, <1% of cART-naive HIV-infected individuals called elite controllers/HIV controllers (HCs) exhibit the core features that define a HIV-1 "functional cure" outcome in the absence of cART: durable viral suppression to below the limit of detection, long-term non-progression to AIDS, and absence of viral transmission. Robust HIV-1-specific CTL responses and prevalence of protective HLA alleles associated with enduring HIV-1 control have been linked to the HC phenotype. An understanding of the molecular mechanisms underlying the CTL-mediated suppression of HIV-1 replication and disease progression in HCs carrying specific protective HLA alleles may yield promising insights towards advancing the research on HIV cure and prophylactic HIV vaccine.

17.
Sci Rep ; 9(1): 2567, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30796241

RESUMO

Cocaine use is associated with breach in the blood brain barrier (BBB) and increased HIV-1 neuro-invasion. We show that the cellular enzyme "Prolidase" plays a key role in cocaine-induced disruption of the BBB. We established a barrier model to mimic the BBB by culturing human brain microvascular endothelial cells (HBMECs) in transwell inserts. In this model, cocaine treatment enhanced permeability of FITC-dextran suggesting a breach in the barrier. Interestingly, cocaine treatment increased the activity of matrix metallo-proteinases that initiate degradation of the BBB-associated collagen. Cocaine exposure also induced prolidase expression and activity in HBMECs. Prolidase catalyzes the final and rate-limiting step of collagen degradation during BBB remodeling. Knock-down of prolidase abrogated cocaine-mediated increased permeability suggesting a direct role of prolidase in BBB breach. To decipher the mechanism by which cocaine regulates prolidase, we probed the inducible nitric oxide synthase (iNOS) mediated phosphorylation of prolidase since mRNA levels of the protein were not altered upon cocaine treatment. We observed increased iNOS expression concurrent with increased prolidase phosphorylation in cocaine treated cells. Subsequently, inhibition of iNOS decreased prolidase phosphorylation and reduced cocaine-mediated permeability. Finally, cocaine treatment increased transmigration of monocytic cells through the HBMEC barrier. Knock-down of prolidase reduced cocaine-mediated monocyte transmigration, establishing a key role of prolidase in cocaine-induced breach in endothelial cell barrier.


Assuntos
Barreira Hematoencefálica/enzimologia , Cocaína/efeitos adversos , Dipeptidases/biossíntese , Células Endoteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Microvasos/enzimologia , Barreira Hematoencefálica/lesões , Barreira Hematoencefálica/patologia , Cocaína/farmacologia , Células Endoteliais/patologia , Humanos , Microvasos/lesões , Microvasos/patologia , Monócitos/metabolismo , Monócitos/patologia , Óxido Nítrico Sintase Tipo II/biossíntese , Células THP-1 , Migração Transendotelial e Transepitelial/efeitos dos fármacos
18.
J Prosthodont ; 28(1): e68-e73, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29086458

RESUMO

PURPOSE: To evaluate the shade matching capabilities in natural dentitions using Vita Toothguide 3D-Master and an intraoral digital spectrophotometer (Vita Easyshade Advance 4.0) in various light sources. MATERIALS AND METHODS: Participants between 20 and 40 years old with natural, unrestored right maxillary central incisors, no history of bleaching, orthodontic treatment, or malocclusion and no rotations were included. According to their shades, subjects were randomly selected and grouped into A1, A2, and A3. A total of 100 participants (50 male and 50 female) in each group were chosen for this study. Shade selection was made between 10 am and 2 pm for all light sources. The same examiner selected the shade of natural teeth with Vita Toothguide 3D-Master under natural light within 2 minutes. Once the Vita Toothguide 3D-Masterwas matched with the maxillary right central incisor, the L*, a*, and b* values, chroma, and hue were recorded with Vita Easyshade Advance 4.0 by placing it on the shade tab under the same light source. The values were statistically analyzed using one-way ANOVA and Tukey's HSD post hoc test with SPSS v22.0 software. RESULTS: The mean ∆E*ab values for shades A1, A2, and A3 for groups 1, 2, and 3 were statistically significantly different from each other (p < 0.001). CONCLUSION: The intraoral digital spectrophotometer showed statistically significant differences in shade matching compared to Vita Toothguide 3D-Master. Incandescent light showed more accurate shade matching than the filtered LED, LED, and daylight.


Assuntos
Pigmentação em Prótese/métodos , Dente/anatomia & histologia , Adulto , Cor , Feminino , Humanos , Incisivo/anatomia & histologia , Luz , Masculino , Espectrofotometria/métodos , Adulto Jovem
19.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30567984

RESUMO

The HIV-1 capsid protein (CA) facilitates reverse transcription and nuclear entry of the virus. However, CA's role in post-nuclear entry steps remains speculative. We describe a direct link between CA and integration by employing the capsid inhibitor PF74 as a probe coupled with the biochemical analysis of HIV-1 preintegration complexes (PICs) isolated from acutely infected cells. At a low micromolar concentration, PF74 potently inhibited HIV-1 infection without affecting reverse transcription. Surprisingly, PF74 markedly reduced proviral integration owing to inhibition of nuclear entry and/or integration. However, a 2-fold reduction in nuclear entry by PF74 did not quantitatively correlate with the level of antiviral activity. Titration of PF74 against the integrase inhibitor raltegravir showed an additive antiviral effect that is dependent on a block at the post-nuclear entry step. PF74's inhibitory effect was not due to the formation of defective viral DNA ends or a delay in integration, suggesting that the compound inhibits PIC-associated integration activity. Unexpectedly, PICs recovered from cells infected in the presence of PF74 exhibited elevated integration activity. PF74's effect on PIC activity is CA specific since the compound did not increase the integration activity of PICs of a PF74-resistant HIV-1 CA mutant. Sucrose gradient-based fractionation studies revealed that PICs assembled in the presence of PF74 contained lower levels of CA, suggesting a negative association between CA and PIC-associated integration activity. Finally, the addition of a CA-specific antibody or PF74 inhibited PIC-associated integration activity. Collectively, our results demonstrate that PF74's targeting of PIC-associated CA results in impaired HIV-1 integration.IMPORTANCE Antiretroviral therapy (ART) that uses various combinations of small molecule inhibitors has been highly effective in controlling HIV. However, the drugs used in the ART regimen are expensive, cause side effects, and face viral resistance. The HIV-1 CA plays critical roles in the virus life cycle and is an attractive therapeutic target. While currently there is no CA-based therapy, highly potent CA-specific inhibitors are being developed as a new class of antivirals. Efforts to develop a CA-targeted therapy can be aided through a clear understanding of the role of CA in HIV-1 infection. CA is well established to coordinate reverse transcription and nuclear entry of the virus. However, the role of CA in post-nuclear entry steps of HIV-1 infection is poorly understood. We show that a CA-specific drug PF74 inhibits HIV-1 integration revealing a novel role of this multifunctional viral protein in a post-nuclear entry step of HIV-1 infection.


Assuntos
Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Indóis/farmacologia , Fenilalanina/análogos & derivados , Fármacos Anti-HIV , Capsídeo/efeitos dos fármacos , Proteínas do Capsídeo/genética , Linhagem Celular , DNA Viral/genética , Células HEK293 , Soropositividade para HIV/genética , HIV-1/genética , Humanos , Fenilalanina/farmacologia , Transcrição Reversa/genética , Integração Viral/genética , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
20.
mBio ; 9(5)2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30327442

RESUMO

Late in the HIV-1 replication cycle, the viral structural protein Gag is targeted to virus assembly sites at the plasma membrane of infected cells. The capsid (CA) domain of Gag plays a critical role in the formation of the hexameric Gag lattice in the immature virion, and, during particle release, CA is cleaved from the Gag precursor by the viral protease and forms the conical core of the mature virion. A highly conserved Pro-Pro-Ile-Pro (PPIP) motif (CA residues 122 to 125) [PPIP(122-125)] in a loop connecting CA helices 6 and 7 resides at a 3-fold axis formed by neighboring hexamers in the immature Gag lattice. In this study, we characterized the role of this PPIP(122-125) loop in HIV-1 assembly and maturation. While mutations P123A and P125A were relatively well tolerated, mutation of P122 and I124 significantly impaired virus release, caused Gag processing defects, and abolished infectivity. X-ray crystallography indicated that the P122A and I124A mutations induce subtle changes in the structure of the mature CA lattice which were permissive for in vitro assembly of CA tubes. Transmission electron microscopy and cryo-electron tomography demonstrated that the P122A and I124A mutations induce severe structural defects in the immature Gag lattice and abrogate conical core formation. Propagation of the P122A and I124A mutants in T-cell lines led to the selection of compensatory mutations within CA. Our findings demonstrate that the CA PPIP(122-125) loop comprises a structural element critical for the formation of the immature Gag lattice.IMPORTANCE Capsid (CA) plays multiple roles in the HIV-1 replication cycle. CA-CA domain interactions are responsible for multimerization of the Gag polyprotein at virus assembly sites, and in the mature virion, CA monomers assemble into a conical core that encapsidates the viral RNA genome. Multiple CA regions that contribute to the assembly and release of HIV-1 particles have been mapped and investigated. Here, we identified and characterized a Pro-rich loop in CA that is important for the formation of the immature Gag lattice. Changes in this region disrupt viral production and abrogate the formation of infectious, mature virions. Propagation of the mutants in culture led to the selection of second-site compensatory mutations within CA. These results expand our knowledge of the assembly and maturation steps in the viral replication cycle and may be relevant for development of antiviral drugs targeting CA.


Assuntos
Proteínas do Capsídeo/química , HIV-1/química , Domínios Proteicos , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química , Motivos de Aminoácidos , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Cristalografia por Raios X , Tomografia com Microscopia Eletrônica , Células HEK293 , HIV-1/genética , Células HeLa , Humanos , Modelos Moleculares , Mutação , Estrutura Secundária de Proteína , Linfócitos T/virologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA