Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genome ; 64(4): 400-415, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33197212

RESUMEN

In the absence of a vaccine, the treatment of SARS-CoV2 has focused on eliminating the virus with antivirals or mitigating the cytokine storm syndrome (CSS) that leads to the most common cause of death: respiratory failure. Herein we discuss the mechanisms of antiviral treatments for SARS-CoV2 and treatment strategies for the CSS. Antivirals that have shown in vitro activity against SARS-CoV2, or the closely related SARS-CoV1 and MERS-CoV, are compared on the enzymatic level and by potency in cells. For treatment of the CSS, we discuss medications that reduce the effects or expression of cytokines involved in the CSS with an emphasis on those that reduce IL-6 because of its central role in the development of the CSS. We show that some of the medications covered influence the activity or expression of enzymes involved in epigenetic processes and specifically those that add or remove modifications to histones or DNA. Where available, the latest clinical data showing the efficacy of the medications is presented. With respect to their mechanisms, we explain why some medications are successful, why others have failed, and why some untested medications may yet prove useful.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/virología , Citocinas , Epigénesis Genética , Expresión Génica , Humanos , Interleucina-6 , SARS-CoV-2/efectos de los fármacos
2.
IUBMB Life ; 72(11): 2313-2330, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32918855

RESUMEN

SARS-CoV-2, the causing agent of the ongoing COVID-19 pandemic, is a beta-coronavirus which has 80% genetic homology with SARS-CoV, but displays increased virulence and transmissibility. Initially, SARS-CoV-2 was considered a respiratory virus generally causing a mild disease, only severe and fatal in the elderly and individuals with underlying conditions. Severe illnesses and fatalities were attributed to a cytokine storm, an excessive response from the host immune system. However, with the number of infections over 10 millions and still soaring, the insidious and stealthy nature of the virus has emerged, as it causes a vast array of diverse unexpected symptoms among infected individuals, including the young and healthy. It has become evident that besides infecting the respiratory tract, SARS-CoV-2 can affect many organs, possibly through the infection of the endothelium. This review presents an overview of our learning curve with the novel virus emergence, transmission, pathology, biological properties and host-interactions. It also briefly describes remedial measures taken until an effective vaccine is available, that is non-pharmaceutical interventions to reduce the viral spread and the repurposing of existing drugs, approved or in development for other conditions to eliminate the virus or mitigate the cytokine storm.


Asunto(s)
COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Genoma Viral , Interacciones Huésped-Patógeno/inmunología , SARS-CoV-2/patogenicidad , Antiinflamatorios/uso terapéutico , Anticoagulantes/uso terapéutico , Antivirales/uso terapéutico , COVID-19/virología , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/virología , Reposicionamiento de Medicamentos/métodos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Factores Inmunológicos/uso terapéutico , Inflamación , Máscaras , Distanciamiento Físico , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Síndrome Respiratorio Agudo Grave , Tratamiento Farmacológico de COVID-19
3.
IUBMB Life ; 72(11): 2331-2354, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32936531

RESUMEN

The SARS-CoV-2 makes its way into the cell via the ACE2 receptor and the proteolytic action of TMPRSS2. In response to the SARS-CoV-2 infection, the innate immune response is the first line of defense, triggering multiple signaling pathways to produce interferons, pro-inflammatory cytokines and chemokines, and initiating the adaptive immune response against the virus. Unsurprisingly, the virus has developed strategies to evade detection, which can result in delayed, excessive activation of the innate immune system. The response elicited by the host depends on multiple factors, including health status, age, and sex. An overactive innate immune response can lead to a cytokine storm, inflammation, and vascular disruption, leading to the vast array of symptoms exhibited by COVID-19 patients. What is known about the expression and epigenetic regulation of the ACE2 gene and the various players in the host response are explored in this review.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Epigénesis Genética , Interacciones Huésped-Patógeno/inmunología , Serina Endopeptidasas/genética , Glicoproteína de la Espiga del Coronavirus/genética , Enzima Convertidora de Angiotensina 2/inmunología , COVID-19/genética , COVID-19/virología , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/virología , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata , Interferones/genética , Interferones/inmunología , Receptores Virales/genética , Receptores Virales/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Serina Endopeptidasas/inmunología , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/inmunología , Internalización del Virus , Replicación Viral
4.
Virol J ; 16(1): 42, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940169

RESUMEN

BACKGROUND: HIV integrase (IN) and its cellular cofactors, including lens-epithelium-derived growth factor (LEDGF/p75), Ku70, p300, and Rad52, are subject to small ubiquitin-like modifier (SUMO) modification. In addition to covalent SUMOylation, SUMO paralogs can also noncovalently bind proteins through SUMO-interacting motifs (SIMs). However, little is known about whether HIV IN contains SIMs and the roles of these motifs. RESULTS: We searched for the amino acid sequence of HIV IN and investigated three putative SIMs of IN: SIM1 72VILV75, SIM2 200IVDI203 and SIM3 257IKVV260. Our mutational analysis showed that 200IVDI203 and 257IKVV260 are two bona fide SIMs that mediate IN-SUMO noncovalent interactions. Additionally, a cell-based SUMOylation assay revealed that IN SIMs negatively regulate the SUMOylation of IN, as well as the interaction between IN and SUMO E2 conjugation enzyme Ubc9. Conversely, IN SIMs are required for its interactions with LEDGF/p75 but not with Ku70. Furthermore, our study reveals that SIM2 and SIM3 are required for the nuclear localization of IN. Finally, we investigated the impact of IN SIM2 and SIM3 on HIV single cycle replication in CD4+ C8166 T cells, and the results showed that viruses carrying IN SIM mutants are replication defective at the steps of the early viral life cycle, including reverse transcription, nuclear import and integration. CONCLUSION: Our data suggested that the INSIM-SUMO interaction constitutes a new regulatory mechanism of IN functions and might be important for HIV-1 replication.


Asunto(s)
Integrasa de VIH/metabolismo , VIH-1/fisiología , Proteína SUMO-1/metabolismo , Sumoilación , Replicación Viral , Secuencias de Aminoácidos , Células HEK293 , Integrasa de VIH/genética , VIH-1/enzimología , Humanos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Biochim Biophys Acta ; 1849(8): 1104-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25934542

RESUMEN

Nonsense-mediated mRNA decay (NMD), an mRNA surveillance mechanism, eliminates premature termination codon-containing (PTC⁺) transcripts. For instance, it maintains the homeostasis of splicing factors and degrades aberrant transcripts of human genetic disease genes. Here we examine the inhibitory effect on the NMD pathway and consequent increase of PTC+ transcripts by the dietary compound curcumin. We have found that several PTC⁺ transcripts including that of serine/arginine-rich splicing factor 1 (SRSF1) were specifically increased in cells by curcumin. We also observed a similar curcumin effect on the PTC⁺ mutant transcript from a Tay-Sachs-causing HEXA allele or from a beta-globin reporter gene. The curcumin effect was accompanied by significantly reduced expression of the NMD factors UPF1, 2, 3A and 3B. Consistently, in chromatin immunoprecipitation assays, curcumin specifically reduced the occupancy of acetyl-histone H3 and RNA polymerase II at the promoter region (-376 to -247nt) of human UPF1, in a time- and dosage-dependent way. Importantly, knocking down UPF1 abolished or substantially reduced the difference of PTC(+) transcript levels between control and curcumin-treated cells. The disrupted curcumin effect was efficiently rescued by expression of exogenous Myc-UPF1 in the knockdown cells. Together, our data demonstrate that a group of PTC⁺ transcripts are stabilized by a dietary compound curcumin through the inhibition of UPF factor expression and the NMD pathway.


Asunto(s)
Codón sin Sentido/genética , Curcumina/farmacología , Degradación de ARNm Mediada por Codón sin Sentido/efectos de los fármacos , ARN Mensajero/metabolismo , Terminación de la Transcripción Genética/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Proteínas Nucleares/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Factores de Empalme Serina-Arginina , Enfermedad de Tay-Sachs/genética , Enfermedad de Tay-Sachs/metabolismo , Cadena alfa de beta-Hexosaminidasa/genética , Cadena alfa de beta-Hexosaminidasa/metabolismo
6.
J Control Release ; 366: 849-863, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38176469

RESUMEN

Women are more susceptible to HIV transmission through unprotected heterosexual intercourse due to biological and social vulnerabilities. Intravaginal delivery of siRNAs targeting viral genes, host genes, or in combination has shown promising outcomes against HSV, HPV and HIV. Therefore, in this study, we designed, developed and evaluated a pH-sensitive RNAi-based combination nanomicrobide for the prevention/reduction of vaginal transmission of HIV. The nanomicrobide was composed of siRNA-PEI encapsulated PLGA-PEG nanoparticles (siRNA NP) loaded in a HEC gel dosage form with siRNA targeting host gene CCR5 and the viral gene Nef as a dual preventive strategy. Knocking down CCR5, a co-receptor for HIV could prevent HIV from attaching to and entering host cells and knocking down Nef could reactivate autophagy that was inhibited by Nef to improve the elimination of intracellular virus that escaped the first line of defense. The siRNA NP showed a desirable particle size and zeta potential for intravaginal delivery and a pH-dependent release profile whereby low amounts of siRNA was released under acidic vaginal conditions (vaginal fluid simulant; VFS, pH 4.2) (6.0 ± 0.4% released over 15 days) but significantly higher amounts of siRNA was released under neutral pH conditions (phosphate buffered saline; PBS, pH 7.4) (22.9 ± 0.4% released over 15 days). The CCR5-Nef-specific siRNA NP efficiently knocked down CCR5 and Nef protein expression by 43% and 63%, respectively, reactivated Nef-blocked autophagy and inhibited the replication of HIV in vitro (71.8% reduction in p24 expression). After being formulated into a gel dosage form, siRNA NP could be readily released from the gel, penetrate the vaginal epithelial layer, get taken up into the target cells and knockdown Nef and CCR5 without causing cytotoxicity in a vaginal mucosal co-culture model. Functionalization of siRNA NP with anti-CD4 antibody and loaded into a 0.5% HEC gel improved vaginal distribution and uptake of siRNA in a mouse model with distribution of siRNA restricted to the reproductive tract without any unwanted systemic uptake. The 0.5% HEC gel loaded with siRNA NP-(m)CD4 significantly downregulated approximately 40% of CCR5 protein in the lower vagina and 36% of CCR5 protein in the upper vaginal and cervical region. In contrast, 0.5% HEC gel loaded with siRNA NP-IgG did not result in significant gene knockdown.


Asunto(s)
Infecciones por VIH , Vagina , Animales , Femenino , Humanos , Ratones , Autofagia , Linfocitos T CD4-Positivos , Infecciones por VIH/prevención & control , Infecciones por VIH/transmisión , Concentración de Iones de Hidrógeno , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Vagina/virología
7.
Blood ; 117(9): 2649-57, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21200019

RESUMEN

Not all individuals exposed to HIV become infected. Understanding why these HIV-exposed seronegative individuals remain uninfected will help inform the development of preventative measures against HIV infection. Interferon regulatory factor-1 (IRF1) plays a critical role both in host antiviral immunity and in HIV-1 replication. This study examined IRF1 expression regulation in the ex vivo peripheral blood mononuclear cells of HIV-exposed seronegative commercial sex workers who can be epidemiologically defined as relatively resistant to HIV infection (HIV-R), versus HIV-uninfected, susceptible controls (HIV-S). Whereas HIV-susceptible individuals demonstrated a biphasic, prolonged increase in IRF1 expression after interferon-γ stimulation, HIV-R individuals demonstrated a robust, but transient response. We also found that the IRF1 promoter in HIV-R was primed by increased basal histone deacetylase-2 binding, independently of transcription regulators, STAT1 and nuclear factor-κB/p65, implicating an epigenetic silencing mechanism. Interestingly, the transitory IRF1 response in HIV-R was sufficient in comparable regulation of interleukin-12 and interleukin-4 expression compared with the HIV-susceptible controls. This is the first study characterizing IRF1 responsiveness in individuals who demonstrate altered susceptibility to HIV infection. These data suggest that transitory IRF1 responsiveness in HIV-R may be one of the key contributors to the altered susceptibility to HIV infection during the early stages of primary HIV infection.


Asunto(s)
Susceptibilidad a Enfermedades , Epigénesis Genética , Infecciones por VIH/genética , Seronegatividad para VIH/genética , Factor 1 Regulador del Interferón/genética , Acetilación/efectos de los fármacos , Estudios de Casos y Controles , Regulación hacia Abajo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Sitios Genéticos , Infecciones por VIH/inmunología , Histona Desacetilasa 2/metabolismo , Histonas/metabolismo , Humanos , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/farmacología , Cinética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción ReIA/metabolismo
8.
Nat Genet ; 36(5): 502-6, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15098035

RESUMEN

Considerable knowledge has been gained from temporal analyses of molecular events culminating in gene activation, but technical hurdles have hindered comparable studies of gene silencing. Here we describe the temporal assembly of silent chromatin at the mouse terminal transferase gene (Dntt), which is silenced and repositioned to pericentromeric heterochromatin during thymocyte maturation. Silencing was nucleated at the Dntt promoter by the ordered deacetylation of histone H3 at Lys9 (H3-Lys9), loss of methylation at H3-Lys4 and acquisition of methylation at H3-Lys9, followed by bidirectional spreading of each event. Deacetylation at H3-Lys9 coincided with pericentromeric repositioning, and neither of these early events required de novo protein synthesis. CpG methylation increased primarily in mature T cells that had left the thymus. A transformed thymocyte line supported reversible inactivation of Dntt without repositioning. In these cells, histone modification changes were nucleated at the promoter but did not spread. These results provide a foundation for elucidating the mechanisms of silent chromatin assembly during development.


Asunto(s)
Cromatina/genética , ADN Nucleotidilexotransferasa/genética , Timo/crecimiento & desarrollo , Acetilación , Animales , Carcinógenos/farmacología , Islas de CpG , Cicloheximida/farmacología , Metilación de ADN , ADN Nucleotidilexotransferasa/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Histona Desacetilasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ionomicina/farmacología , Ionóforos/farmacología , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/fisiología , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Timo/fisiología
9.
Tuberculosis (Edinb) ; 143: 102409, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37729851

RESUMEN

Type I interferon (IFN)-induced genes have the potential for distinguishing active tuberculosis (ATB) from latent TB infection (LTBI) and healthy controls (HC), monitoring treatment, and detection of individuals at risk of progression to active disease. We examined the differential effects of IFN-α, IFN-ß and Mycobacterium tuberculosis whole cell lysate (Mtb WCL) stimulation on the expression of selected IFN-stimulated genes in peripheral blood mononuclear cells from individuals with either LTBI, ATB, and healthy controls. Stimulation with IFN-α and IFN-ß induced a higher expression of the interrogated genes while Mtb WCL stimulation induced expression similar to that observed at baseline, with the exception of IL-1A and IL-1B genes that were downregulated. The expression of IFN-α-induced FCGR1A gene, IFN-ß-induced FCGR1A, FCGR1B, and SOCS3 genes, and Mtb WCL-induced IFI44, IFI44L, IFIT1, and IFITM3 genes differed significantly between LTBI and ATB. These findings suggest stimulation-driven gene expression patterns could potentially discriminate LTBI and ATB. Mechanistic studies are necessary to define the processes through which distinct type I IFNs and downstream ISGs determine infection outcomes and identify potential host-directed therapeutic strategies.


Asunto(s)
Interferón Tipo I , Tuberculosis Latente , Mycobacterium tuberculosis , Tuberculosis , Humanos , Mycobacterium tuberculosis/genética , Tuberculosis Latente/diagnóstico , Tuberculosis Latente/genética , Interferón Tipo I/genética , Leucocitos Mononucleares , Antígenos Bacterianos/genética , Tuberculosis/diagnóstico , Tuberculosis/genética , Proteínas de la Membrana , Proteínas de Unión al ARN
10.
Mucosal Immunol ; 16(3): 341-356, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37121385

RESUMEN

Neutrophil recruitment and activation within the female genital tract are often associated with tissue inflammation, loss of vaginal epithelial barrier integrity, and increased risk for sexually transmitted infections, such as HIV-1. However, the direct role of neutrophils on vaginal epithelial barrier function during genital inflammation in vivo remains unclear. Using complementary proteome and immunological analyses, we show high neutrophil influx into the lower female genital tract in response to physiological surges in progesterone, stimulating distinct stromal, immunological, and metabolic signaling pathways. However, despite the release of extracellular matrix-modifying proteases and inflammatory mediators, neutrophils contributed little to physiological mucosal remodeling events such as epithelial shedding or re-epithelialization during transition from diestrus to estrus phase. In contrast, the presence of bacterial vaginosis-associated bacteria resulted in a rapid and sustained neutrophil recruitment, resulting in vaginal epithelial barrier leakage and decreased cell-cell junction protein expression in vivo. Thus, neutrophils are important mucosal sentinels that rapidly respond to various biological cues within the female genital tract, dictating the magnitude and duration of the ensuing inflammatory response at steady state and during disease processes.


Asunto(s)
Neutrófilos , Enfermedades de Transmisión Sexual , Femenino , Humanos , Inflamación , Genitales Femeninos , Vagina , Bacterias
11.
AIDS Res Hum Retroviruses ; 38(2): 111-126, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34465136

RESUMEN

Resting CD4+ T cells are primary targets of early HIV infection events in vivo, but do not readily support HIV replication in vitro. This barrier to infection can be overcome by exposing resting CD4+ T cells to endothelial cells (ECs). ECs line blood vessels and direct T cell trafficking into inflamed tissues. Cell trafficking pathways have been shown to have overlapping roles in facilitating HIV replication, but their relevance to EC-mediated enhancement of HIV susceptibility in resting CD4+ T cells has not previously been examined. We characterized the phenotype of primary human resting CD4+ T cells that became productively infected with HIV when cocultured with primary human blood and lymphatic ECs. The infected CD4+ T cells were primarily central memory cells enriched for high expression of the integrins LFA-1 and VLA-4. ICAM-1 and VCAM-1, the cognate ligands for LFA-1 and VLA-4, respectively, were expressed by the ECs in the coculture. Blocking LFA-1 and VLA-4 on resting CD4+ T cells inhibited infection by 65.4%-96.9%, indicating that engagement of these integrins facilitates EC-mediated enhancement of productive HIV infection in resting CD4+ T cells. The demonstration that ECs influence cellular HIV susceptibility of resting memory CD4+ T cells through cell trafficking pathways engaged during the transmigration of T cells into tissues highlights the physiological relevance of these findings for HIV acquisition and opportunities for intervention.


Asunto(s)
Células Endoteliales , Infecciones por VIH , Linfocitos T CD4-Positivos/metabolismo , Adhesión Celular , Células Endoteliales/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Linfocitos T , Molécula 1 de Adhesión Celular Vascular
12.
Viruses ; 14(3)2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35336878

RESUMEN

Interferon (IFN) -stimulated genes (ISGs) are critical effectors of IFN response to viral infection, but whether ISG expression is a correlate of protection against HIV infection remains elusive. A well-characterized subcohort of Kenyan female sex workers, who, despite being repeatedly exposed to HIV-1 remain seronegative (HESN), exhibit reduced baseline systemic and mucosal immune activation. This study tested the hypothesis that regulation of ISGs in the cells of HESN potentiates a robust antiviral response against HIV. Transcriptional profile of a panel of ISGs with antiviral function in PBMC and isolated CD4+ T cells from HESN and non-HESN sex worker controls were defined following exogenous IFN-stimulation using relative RT-qPCR. This study identified a unique profile of proinflammatory and proapoptotic ISGs with robust but transient responses to exogenous IFN-γ and IFN-α2 in HESN cells. In contrast, the non-HESN cells had a strong and prolonged proinflammatory ISG profile at baseline and following IFN challenge. Potential mechanisms may include augmented bystander apoptosis due to increased TRAIL expression (16-fold), in non-HESN cells. The study also identified two negative regulators of ISG induction associated with the HESN phenotype. Robust upregulation of SOCS-1 and IRF-1, in addition to HDM2, could contribute to the strict regulation of proinflammatory and proapoptotic ISGs in HESN cells. As reducing IRF-1 in the non-HESN cells resulted in the identified HESN ISG profile, and decreased HIV susceptibility, the unique HESN ISG profile could be a correlate of protection against HIV infection.


Asunto(s)
Infecciones por VIH , Trabajadores Sexuales , Antivirales , Femenino , Humanos , Inflamación , Kenia , Leucocitos Mononucleares , Fenotipo
13.
Nat Commun ; 13(1): 3357, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35688805

RESUMEN

Vaccines against SARS-CoV-2 have shown high efficacy in clinical trials, yet a full immunologic characterization of these vaccines, particularly within the human upper respiratory tract, is less well known. Here, we enumerate and phenotype T cells in nasal mucosa and blood using flow cytometry before and after vaccination with the Pfizer-BioNTech COVID-19 vaccine (n = 21). Tissue-resident memory (Trm) CD8+ T cells expressing CD69+CD103+ increase in number ~12 days following the first and second doses, by 0.31 and 0.43 log10 cells per swab respectively (p = 0.058 and p = 0.009 in adjusted linear mixed models). CD69+CD103+CD8+ T cells in the blood decrease post-vaccination. Similar increases in nasal CD8+CD69+CD103- T cells are observed, particularly following the second dose. CD4+ cells co-expressing CCR6 and CD161 are also increased in abundance following both doses. Stimulation of nasal CD8+ T cells with SARS-CoV-2 spike peptides elevates expression of CD107a at 2- and 6-months (p = 0.0096) post second vaccine dose, with a subset of donors also expressing increased cytokines. These data suggest that nasal T cells may be induced and contribute to the protective immunity afforded by this vaccine.


Asunto(s)
Linfocitos T CD8-positivos , COVID-19 , Vacuna BNT162 , Linfocitos T CD4-Positivos , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Memoria Inmunológica , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Mucosa Nasal , ARN Mensajero , Receptores CCR6 , SARS-CoV-2 , Vacunación
14.
Front Cell Infect Microbiol ; 11: 790627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34976864

RESUMEN

The microbiome, the collection of microbial species at a site or compartment, has been an underappreciated realm of human health up until the last decade. Mounting evidence suggests the microbiome has a critical role in regulating the female genital tract (FGT) mucosa's function as a barrier against sexually transmitted infections (STIs) and pathogens. In this review, we provide the most recent experimental systems and studies for analyzing the interplay between the microbiome and host cells and soluble factors with an influence on barrier function. Key components, such as microbial diversity, soluble factors secreted by host and microbe, as well as host immune system, all contribute to both the physical and immunologic aspects of the FGT mucosal barrier. Current gaps in what is known about the effects of the microbiome on FGT mucosal barrier function are compared and contrasted with the literature of the gut and respiratory mucosa. This review article presents evidence supporting that the vaginal microbiome, directly and indirectly, contributes to how well the FGT protects against infection.


Asunto(s)
Infecciones por VIH , Microbiota , Enfermedades de Transmisión Sexual , Femenino , Genitales Femeninos , Humanos , Vagina
15.
Front Immunol ; 12: 612807, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34163462

RESUMEN

Since being identified as a key receptor for SARS-CoV-2, Angiotensin converting enzyme 2 (ACE2) has been studied as one of the potential targets for the development of preventative and/or treatment options. Tissue expression of ACE2 and the amino acids interacting with the spike protein of SARS-CoV-2 have been mapped. Furthermore, the recombinant soluble extracellular domain of ACE2 is already in phase 2 trials as a treatment for SARS-CoV-2 infection. Most studies have continued to focus on the ACE2 extracellular domain, which is known to play key roles in the renin angiotensin system and in amino acid uptake. However, few also found ACE2 to have an immune-modulatory function and its intracellular tail may be one of the signaling molecules in regulating cellular activation. The implication of its immune-modulatory role in preventing the cytokine-storm, observed in severe COVID-19 disease outcomes requires further investigation. This review focuses on the regulated proteolytic cleavage of ACE2 upon binding to inducer(s), such as the spike protein of SARS-CoV, the potential of cleaved ACE2 intracellular subdomain in regulating cellular function, and the ACE2's immune-modulatory function. This knowledge is critical for targeting ACE2 levels for developing prophylactic treatment or preventative measures in SARS-CoV infections.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/patología , Membrana Celular/metabolismo , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/química , COVID-19/metabolismo , COVID-19/virología , Humanos , Inmunomodulación , Estructura Terciaria de Proteína , Proteolisis , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo
16.
J Virol ; 82(15): 7515-23, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18508904

RESUMEN

Mammalian orthoreoviruses (reoviruses) are ubiquitous viral agents that infect cells in respiratory and enteric tracts. The frequency and nature of human cellular immunoregulatory responses against reovirus are unknown. Here we establish systems to detect and quantify reovirus-induced cytokine and chemokine recall responses using primary cultures of virus-infected peripheral blood mononuclear cells (PBMC) and two widely used reovirus serotypes, type 1 Lang (T1L) and type 3 Dearing (T3D) reexposure in vitro. In cultures from 44 healthy adults, reovirus induced exceptionally strong CD4 and CD8 T-cell-dependent gamma interferon (IFN-gamma) recall responses concomitant with intense interleukin 10 (IL-10) production. These responses were elicited independently of viral replication. Surprisingly, paired analyses of subject responses to these two common serotypes revealed that while both elicit intense Th1-dominated immunity, median T3D-driven responses were 2.2-fold weaker (P = 0.0004) than those elicited by T1L. Recall responses evoked by these viral serotypes differed markedly in their mechanism of regulation. T3D IL-10 and IFN-gamma responses were CD4 and CD8 dependent and blocked by interfering with CD86 costimulation but were CD80 independent. T1L responses were consistently CD28 and CD80/86 independent. Thus, despite extensive genetic and morphological similarities between reovirus serotypes, the nature and intensity of the human recall responses as well as the control mechanisms regulating them are clearly distinct.


Asunto(s)
Memoria Inmunológica , Orthoreovirus de los Mamíferos/inmunología , Adolescente , Adulto , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Antígenos CD28/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Humanos , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Leucocitos Mononucleares/inmunología , Persona de Mediana Edad , Subgrupos de Linfocitos T/inmunología
17.
J Allergy Clin Immunol ; 121(1): 57-63.e3, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17980413

RESUMEN

BACKGROUND: Multiple biologic factors influence maintenance of immunologic responsiveness. Here, we studied whether epigenetics has a regulatory function in maintaining pre-established T(H)1-like and T(H)2-like immunity in human beings. OBJECTIVE: We focused on delineating the role of endogenous histone deacetylase (HDAC) activity in regulating cytokine recall responses. METHODS: Using RT-PCR and ELISA, the effect of increasing cellular acetylation on T(H)1/T(H)2 cytokine expression was systematically examined in 58 children by inhibiting HDAC activity with trichostatin A. RESULTS: Phytohemagglutinin activation selectively stimulates antigen-experienced CD45RO+ T cells, eliciting recall cytokine responses. Trichostatin A reduced HDAC activity by approximately 1/3. The resulting cellular hyperacetylation led to increased T(H)2-associated (IL-13, 139%; IL-5, 168%; P < .0001) and reduced T(H)1-associated recall responses (IFN-gamma, 76%; CXCL10, 47%; P < .0001). IL-2 and IL-10 production were reduced 25% to 55% (P < .0001). These alterations in T(H)2-associated and T(H)1-associated recall responses were associated with increased expression of Gata-3 and sphingosine kinase 1, a T(H)1-negative regulator, independent of T-bet expression. Overall, inhibition of endogenous HDAC activity shifted T(H)1:T(H)2 ratios by 3-fold to 8-fold (P < or = .0001), skewing recall responses toward a more T(H)2-like phenotype, independent of the stimulus used. CONCLUSION: Endogenous HDAC activity plays a crucial role in maintaining the balance of pre-established T(H)1-like and T(H)2-like responses, inhibiting excessive T(H)2 immunity.


Asunto(s)
Citocinas/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Estudios de Casos y Controles , Niño , Estudios de Cohortes , Citocinas/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor de Transcripción GATA3/metabolismo , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/farmacología , Antígenos Comunes de Leucocito/metabolismo , Activación de Linfocitos , Masculino , Fitohemaglutininas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología
18.
Front Immunol ; 10: 144, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30787929

RESUMEN

In the context of HIV sexual transmission at the genital mucosa, initial interactions between the virus and the mucosal immunity determine the outcome of the exposure. Hence, these interactions have been deeply explored in attempts to undercover potential targets for developing preventative strategies. The knowledge gained has led to propose a hypothetical model for mucosal HIV transmission. Subsequent research studies on this topic further revealed new mechanisms and identified new host-HIV interactions. This review aims at integrating these findings to inform better and update the current model of HIV transmission. At the earliest stage of virus exposure, the epithelial integrity and the presence of antiviral factors are critical in preventing viral entry to the submucosa. However, the virus has been shown to enter to the submucosa in the presence of physical abrasion or via epithelial transmigration using paracellular passage or transcytosis mechanisms. The efficiency of these processes is greater with cell-associated viral inoculums and can be influenced by the presence of viral and immune factors, and by the structure of the exposed epithelium. Once the virus reaches the submucosa, dendritic cells and fibroblasts, as recently described, have been shown in vitro of being capable of facilitating the transfer of viral particles to susceptible cells, leading to viral dissemination, most likely in a trans-infection manner. The presence of activated CD4+ T cells in submucosa increases the probability of infection, where the predominant microbiota could be implicated through the modulation of an inflammatory microenvironment. Other factors such as genital fluids and hormones could also play an essential role in HIV transmission. Here, we review the most recent evidence described for mucosal HIV-transmission contributing with the understanding of this phenomenon.


Asunto(s)
Infecciones por VIH/transmisión , Membrana Mucosa/virología , Animales , Genitales/inmunología , Infecciones por VIH/inmunología , Humanos , Inmunidad Mucosa , Membrana Mucosa/inmunología
19.
PLoS One ; 14(9): e0222878, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31550271

RESUMEN

INTRODUCTION: Mucosal immune activation, in the context of sexual transmission of HIV-1 infection, is crucial, as the increased presence of activated T cells enhance susceptibility to infection. In this regard, it has been proposed that immunomodulatory compounds capable of modulating immune activation, such as Vitamin D (VitD) may reduce HIV-1 transmission and might be used as a safe and cost-effective strategy for prevention. Considering this, we examined the in vitro effect of the treatment of peripheral blood mononuclear cells (PBMCs) with the active form of VitD, calcitriol, on cellular activation, function and susceptibility of CD4+ T cells to HIV-1 infection. METHODS: We treated PBMCs from healthy HIV unexposed individuals (Co-HC) and frequently exposed, HIV-1 seronegative individuals (HESNs) from Colombia and from healthy non-exposed individuals from Canada (Ca-HC) with calcitriol and performed in vitro HIV-1 infection assays using X4- and R5-tropic HIV-1 strains respectively. In addition, we evaluated the activation and function of T cells and the expression of viral co-receptors, and select antiviral genes following calcitriol treatment. RESULTS: Calcitriol reduced the frequency of infected CD4+ T cells and the number of viral particles per cell, for both, X4- and R5-tropic viruses tested in the Co-HC and the Ca-HC, respectively, but not in HESNs. Furthermore, in the Co-HC, calcitriol reduced the frequency of polyclonally activated T cells expressing the activation markers HLA-DR and CD38, and those HLA-DR+CD38-, whereas increased the subpopulation HLA-DR-CD38+. Calcitriol treatment also decreased production of granzyme, IL-2 and MIP-1ß by T cells and increased the transcriptional expression of the inhibitor of NF-kB and the antiviral genes cathelicidin (CAMP) and APOBEC3G in PBMCs from Co-HC. CONCLUSION: Our in vitro findings suggest that VitD treatment could reduce HIV-1 transmission through a specific modulation of the activation levels and function of T cells, and the production of antiviral factors. In conclusion, VitD remains as an interesting potential strategy to prevent HIV-1 transmission that should be further explored.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Calcitriol/administración & dosificación , Infecciones por VIH/prevención & control , Activación de Linfocitos/efectos de los fármacos , Vitaminas/administración & dosificación , Desaminasa APOBEC-3G/inmunología , Desaminasa APOBEC-3G/metabolismo , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/transmisión , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/patogenicidad , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Inmunidad Mucosa/efectos de los fármacos , Masculino , Cultivo Primario de Células , Catelicidinas
20.
Front Immunol ; 8: 751, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28713376

RESUMEN

Protein N-myristoylation is a cotranslational lipidic modification specific to the alpha-amino group of an N-terminal glycine residue of many eukaryotic and viral proteins. The ubiquitous eukaryotic enzyme, N-myristoyltransferase, catalyzes the myristoylation process. Precisely, attachment of a myristoyl group increases specific protein-protein interactions leading to subcellular localization of myristoylated proteins with its signaling partners. The birth of the field of myristoylation, a little over three decades ago, has led to the understanding of the significance of protein myristoylation in regulating cellular signaling pathways in several biological processes especially in carcinogenesis and more recently immune function. This review discusses myristoylation as a prerequisite step in initiating many immune cell signaling cascades. In particular, we discuss the hitherto unappreciated implication of myristoylation during myelopoiesis, innate immune response, lymphopoiesis for T cells, and the formation of the immunological synapse. Furthermore, we discuss the role of myristoylation in inducing the virological synapse during human immunodeficiency virus infection as well as its clinical implication. This review aims to summarize existing knowledge in the field and to highlight gaps in our understanding of the role of myristoylation in immune function so as to further investigate into the dynamics of myristoylation-dependent immune regulation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA