Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
EMBO Rep ; 23(10): e54322, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35999696

RESUMEN

The emergence of SARS-CoV-2 variants has exacerbated the COVID-19 global health crisis. Thus far, all variants carry mutations in the spike glycoprotein, which is a critical determinant of viral transmission being responsible for attachment, receptor engagement and membrane fusion, and an important target of immunity. Variants frequently bear truncations of flexible loops in the N-terminal domain (NTD) of spike; the functional importance of these modifications has remained poorly characterised. We demonstrate that NTD deletions are important for efficient entry by the Alpha and Omicron variants and that this correlates with spike stability. Phylogenetic analysis reveals extensive NTD loop length polymorphisms across the sarbecoviruses, setting an evolutionary precedent for loop remodelling. Guided by these analyses, we demonstrate that variations in NTD loop length, alone, are sufficient to modulate virus entry. We propose that variations in NTD loop length act to fine-tune spike; this may provide a mechanism for SARS-CoV-2 to navigate a complex selection landscape encompassing optimisation of essential functionality, immune-driven antigenic variation and ongoing adaptation to a new host.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/genética , Humanos , Filogenia , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
2.
J Gen Virol ; 104(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37310000

RESUMEN

Human cytomegalovirus (HCMV) remains an important cause of mortality in immune-compromised transplant patients and following congenital infection. Such is the burden, an effective vaccine strategy is considered to be of the highest priority. The most successful vaccines to date have focused on generating immune responses against glycoprotein B (gB) - a protein essential for HCMV fusion and entry. We have previously reported that an important component of the humoral immune response elicited by gB/MF59 vaccination of patients awaiting transplant is the induction of non-neutralizing antibodies that target cell-associated virus with little evidence of concomitant classical neutralizing antibodies. Here we report that a modified neutralization assay that promotes prolonged binding of HCMV to the cell surface reveals the presence of neutralizing antibodies in sera taken from gB-vaccinated patients that cannot be detected using standard assays. We go on to show that this is not a general feature of gB-neutralizing antibodies, suggesting that specific antibody responses induced by vaccination could be important. Although we can find no evidence that these neutralizing antibody responses are a correlate of protection in vivo in transplant recipients their identification demonstrates the utility of the approach in identifying these responses. We hypothesize that further characterization has the potential to aid the identification of functions within gB that are important during the entry process and could potentially improve future vaccine strategies directed against gB if they prove to be effective against HCMV at higher concentrations.


Asunto(s)
Anticuerpos Neutralizantes , Vacunas , Humanos , Citomegalovirus , Temperatura , Vacunación
3.
Immunology ; 162(3): 314-327, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33283275

RESUMEN

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that is potentially pathogenic in immunosuppressed individuals and pregnant females during primary infection. The HCMV envelope glycoprotein B (gB) facilitates viral entry into all cell types and induces a potent immune response. AD-2 epitope is a highly conserved linear neutralizing epitope of gB and a critical target for antibodies; however, only 50% of sero-positive individuals make IgG antibodies to this site and IgA responses have not been fully investigated. This study aimed to compare IgG and IgA responses against gB and the AD-2 epitope in naturally exposed individuals and those receiving a recombinant gB/MF59 adjuvant vaccine. Thus, vaccination of sero-positive individuals improved pre-existing gB-specific IgA and IgG levels and induced de novo gB-specific IgA and IgG responses in sero-negative recipients. Pre-existing AD-2 IgG and IgA responses were boosted with vaccination, but de novo AD-2 responses were not detected. Naturally exposed individuals had dominant IgG responses towards gB and AD-2 compared with weaker and variable IgA responses, although a significant IgA binding response to AD-2 was observed within human breastmilk samples. All antibodies binding AD-2 contained kappa light chains, whereas balanced kappa/lambda light chain usage was found for those binding to gB. V region-matched AD-2-specific recombinant IgG and IgA bound both to gB and to AD-2 and neutralized HCMV infection in vitro. Overall, these results indicate that although human IgG responses dominate, IgA class antibodies against AD-2 are a significant component of human milk, which may function to protect neonates from HCMV.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/prevención & control , Citomegalovirus/inmunología , Epítopos , Inmunogenicidad Vacunal , Inmunoglobulina A/sangre , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Animales , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Línea Celular Tumoral , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/sangre , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Células HEK293 , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Leche Humana/inmunología , Leche Humana/virología , Polisorbatos/administración & dosificación , Unión Proteica , Escualeno/administración & dosificación , Vacunación , Proteínas del Envoltorio Viral/metabolismo , Vacunas Virales/inmunología
4.
Proc Natl Acad Sci U S A ; 115(24): 6273-6278, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29686064

RESUMEN

Human cytomegalovirus (HCMV) is an important pathogen in transplant patients and in congenital infection. Previously, we demonstrated that vaccination with a recombinant viral glycoprotein B (gB)/MF59 adjuvant formulation before solid organ transplant reduced viral load parameters post transplant. Reduced posttransplant viremia was directly correlated with antibody titers against gB consistent with a humoral response against gB being important. Here we show that sera from the vaccinated seronegative patients displayed little evidence of a neutralizing antibody response against cell-free HCMV in vitro. Additionally, sera from seronegative vaccine recipients had minimal effect on the replication of a strain of HCMV engineered to be cell-associated in a viral spread assay. Furthermore, although natural infection can induce antibody-dependent cellular cytotoxicity (ADCC) responses, serological analysis of seronegative vaccinees again presented no evidence of a substantial ADCC-promoting antibody response being generated de novo. Finally, analyses for responses against major antigenic domains of gB following vaccination were variable, and their pattern was distinct compared with natural infection. Taken together, these data argue that the protective effect elicited by the gB vaccine is via a mechanism of action in seronegative vaccinees that cannot be explained by neutralization or the induction of ADCC. More generally, these data, which are derived from a human challenge model that demonstrated that the gB vaccine is protective, highlight the need for more sophisticated analyses of new HCMV vaccines over and above the quantification of an ability to induce potent neutralizing antibody responses in vitro.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Viremia/inmunología , Adyuvantes Inmunológicos/farmacología , Humanos , Vacunación/métodos , Carga Viral/inmunología
5.
J Infect Dis ; 221(Suppl 1): S45-S59, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134477

RESUMEN

Human cytomegalovirus (HCMV) is the most common infectious cause of infant birth defects and an etiology of significant morbidity and mortality in solid organ and hematopoietic stem cell transplant recipients. There is tremendous interest in developing a vaccine or immunotherapeutic to reduce the burden of HCMV-associated disease, yet after nearly a half-century of research and development in this field we remain without such an intervention. Defining immune correlates of protection is a process that enables targeted vaccine/immunotherapeutic discovery and informed evaluation of clinical performance. Outcomes in the HCMV field have previously been measured against a variety of clinical end points, including virus acquisition, systemic replication, and progression to disease. Herein we review immune correlates of protection against each of these end points in turn, showing that control of HCMV likely depends on a combination of innate immune factors, antibodies, and T-cell responses. Furthermore, protective immune responses are heterogeneous, with no single immune parameter predicting protection against all clinical outcomes and stages of HCMV infection. A detailed understanding of protective immune responses for a given clinical end point will inform immunogen selection and guide preclinical and clinical evaluation of vaccines or immunotherapeutics to prevent HCMV-mediated congenital and transplant disease.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Citomegalovirus/inmunología , Resistencia a la Enfermedad/inmunología , Interacciones Huésped-Patógeno/inmunología , Replicación Viral/inmunología , Infecciones por Citomegalovirus/epidemiología , Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Humanos , Inmunidad Mucosa , Incidencia , Vacunación , Viremia , Esparcimiento de Virus
6.
J Biol Chem ; 294(35): 12901-12910, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31273084

RESUMEN

Human cytomegalovirus (HCMV) latency and reactivation rely on a complex interplay between cellular differentiation, cell signaling pathways, and viral gene functions. HCMV reactivation in dendritic cells (DCs) is triggered by IL-6 and extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase signaling. However, activation of the same pathway fails to reactivate HCMV in other myeloid cell types, despite this signaling axis being active in those cells. We hypothesized that IL-6-induced ERK activation initiates the changes in chromatin structure required for viral reactivation but that a concomitant signal is necessary to complete the changes in chromatin structure required for gene expression to occur. Using a differential phosphoproteomics approach in cells that do or do not support IL-6-induced viral reactivation, we identified the concomitant activation of an Src family kinase (SFK), hematopoietic cell kinase (HCK), specifically in DCs in response to IL-6. Pharmacological and genetic inhibition of HCK activity indicated that HCK is required for HCMV reactivation. Furthermore, the HCK/SFK activity was linked to recruitment of the monocytic leukemia zinc finger protein (MOZ) histone acetyltransferase to the viral promoter, which promoted histone acetylation after ERK-mediated histone phosphorylation. Importantly, pharmacological and genetic inhibition of MOZ activity prevented reactivation. These results provide an explanation for the selective activation of viral gene expression in DCs by IL-6, dependent on concomitant SFK and ERK signaling. They also reveal a previously unreported role for SFK activity in the regulation of chromatin structure at promoters in eukaryotic cells via MOZ histone acetyltransferase activity.


Asunto(s)
Citomegalovirus/genética , Citomegalovirus/fisiología , Histona Acetiltransferasas/metabolismo , Regiones Promotoras Genéticas/genética , Activación Viral/genética , Familia-src Quinasas/metabolismo , Células Cultivadas , Humanos , Dedos de Zinc
7.
J Gen Virol ; 101(6): 635-644, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375946

RESUMEN

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.


Asunto(s)
Citomegalovirus/genética , Células Dendríticas/virología , Genes Inmediatos-Precoces/genética , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas/genética , Células Madre/virología , Transcripción Genética/genética , Cromatina/genética , Infecciones por Citomegalovirus/virología , Regulación Viral de la Expresión Génica/genética , Humanos , Macrófagos/virología , Monocitos/virología , Células THP-1/virología , Activación Viral/genética , Latencia del Virus/genética
8.
Biochem Soc Trans ; 48(2): 667-675, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32311019

RESUMEN

Primary infection with human cytomegalovirus (HCMV) is usually asymptomatic and leads to the establishment of lifelong latent infection. A major site of latency are the CD34+ hematopoietic progenitor cells. Importantly, normal cellular differentiation of CD34+ cells to a macrophage or dendritic cell phenotype is concomitant with viral reactivation. Molecular studies of HCMV latency have shown that the latent viral genome is associated with histone proteins and that specific post-translational modifications of these histones correlates with the transcriptional activity of the genome arguing that expression of key viral genes that dictate latency and reactivation are subject to the rules of the histone code hypothesis postulated for the regulation of eukaryotic gene expression. Finally, many studies now point to a key role for multiple signaling pathways to provide the cue for HCMV reactivation. The challenge now is to understand the complex interplay between cell identity, transcriptional regulation and cell signaling that occurs to promote reactivation and, additionally, how HCMV may further manipulate these events to support reactivation. Understanding how HCMV utilizes these pathways to drive HCMV reactivation will provide new insight into the mechanisms that govern viral and host gene expression and, potentially, illuminate new, host-directed, therapeutic opportunities to support our attempts to control this important medical pathogen of immune-compromised individuals.


Asunto(s)
Citomegalovirus/fisiología , Transducción de Señal , Familia-src Quinasas/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Epigénesis Genética , Regulación Viral de la Expresión Génica , Genoma Viral , Células Madre Hematopoyéticas/citología , Histonas/metabolismo , Humanos , Inflamación , Interleucina-6/metabolismo , Macrófagos/citología , Fenotipo , Procesamiento Proteico-Postraduccional , Proteínas Virales/metabolismo
9.
Rev Med Virol ; 28(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29148608

RESUMEN

In this review article, we consider results suggesting that transmission of human cytomegalovirus (HCMV) from a donor of a solid organ to an immunologically naive individual can be reduced. Two randomized controlled trials have been conducted recently, one of active immunization of recipients pretransplant and another of passive immunization with monoclonal antibodies specific for HCMV given at the time of transplant. Although the available data are encouraging-providing evidence of a reduction in the incidence of HCMV viraemia-they fall short of what would be required to prove definitively that transmission has been completely prevented. Here, we reflect on these studies and propose a set of 5 criteria, which, if satisfied in the future, could be taken as proof that active and/or passive immunization against HCMV effectively interrupts transmission of virus from the donor. We suggest that these criteria are considered when designing future randomized controlled trials.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/transmisión , Citomegalovirus , Trasplante de Órganos/efectos adversos , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Ensayos Clínicos como Asunto , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/etiología , Vacunas contra Citomegalovirus/inmunología , Humanos , Inmunización Pasiva , Proyectos de Investigación , Donantes de Tejidos , Receptores de Trasplantes , Vacunación
10.
J Infect Dis ; 217(12): 1907-1917, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29528415

RESUMEN

The human cytomegalovirus (HCMV) virion envelope protein glycoprotein B (gB) is essential for viral entry and represents a major target for humoral responses following infection. Previously, a phase 2 placebo-controlled clinical trial conducted in solid organ transplant candidates demonstrated that vaccination with gB plus MF59 adjuvant significantly increased gB enzyme-linked immunosorbent assay (ELISA) antibody levels whose titer correlated directly with protection against posttransplant viremia. The aim of the current study was to investigate in more detail this protective humoral response in vaccinated seropositive transplant recipients. We focused on 4 key antigenic domains (AD) of gB (AD1, AD2, AD4, and AD5), measuring antibody levels in patient sera and correlating these with posttransplant HCMV viremia. Vaccination of seropositive patients significantly boosted preexisting antibody levels against the immunodominant region AD1 as well as against AD2, AD4, and AD5. A decreased incidence of viremia correlated with higher antibody levels against AD2 but not with antibody levels against the other 3 ADs. Overall, these data support the hypothesis that antibodies against AD2 are a major component of the immune protection of seropositives seen following vaccination with gB/MF59 vaccine and identify a correlate of protective immunity in allograft patients.


Asunto(s)
Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Epítopos/inmunología , Inmunidad Humoral/inmunología , Escualeno/inmunología , Proteínas del Envoltorio Viral/inmunología , Viremia/inmunología , Adyuvantes Inmunológicos/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Humanos , Polisorbatos , Vacunación/métodos , Internalización del Virus
11.
J Gen Virol ; 98(4): 754-768, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28100301

RESUMEN

To identify new compounds with anti-human cytomegalovirus (HCMV) activity and new anti-HCMV targets, we developed a high-throughput strategy to screen a GlaxoSmithKline Published Kinase Inhibitor Set. This collection contains a range of extensively characterized compounds grouped into chemical families (chemotypes). From our screen, we identified compounds within chemotypes that impede HCMV protein production and identified kinase proteins associated with inhibition of HCMV protein production that are potential novel anti-HCMV targets. We focused our study on a top 'hit' in our screen, SB-734117, which we found inhibits productive replication of several HCMV strains. Kinase selectivity data indicated that SB-734117 exhibited polypharmacology and was an inhibitor of several proteins from the AGC and CMCG kinase groups. Using Western blotting, we found that SB-734711 inhibited accumulation of HCMV immediate-early proteins, phosphorylation of cellular proteins involved in immediate-early protein production (cAMP response element-binding protein and histone H3) and histone H3 lysine 36 trimethylation (H3K36me3). Therefore, we identified SB-734117 as a novel anti-HCMV compound and found that inhibition of AGC and CMCG kinase proteins during productive HCMV replication was associated with inhibition of viral protein production and prevented post-translational modification of cellular factors associated with viral protein production.


Asunto(s)
Antivirales/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citomegalovirus/efectos de los fármacos , Histonas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Western Blotting , Citomegalovirus/fisiología , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos
12.
Rev Med Virol ; 26(2): 75-89, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26572645

RESUMEN

Human cytomegalovirus (HCMV) infection remains a major cause of morbidity in patient populations. In certain clinical settings, it is the reactivation of the pre-existing latent infection in the host that poses the health risk. The prevailing view of HCMV latency was that the virus was essentially quiescent in myeloid progenitor cells and that terminal differentiation resulted in the initiation of the lytic lifecycle and reactivation of infectious virus. However, our understanding of HCMV latency and reactivation at the molecular level has been greatly enhanced through recent advancements in systems biology approaches to perform global analyses of both experimental and natural latency. These approaches, in concert with more classical reductionist experimentation, are furnishing researchers with new concepts in cytomegalovirus latency and suggest that latent infection is far more active than first thought. In this review, we will focus on new studies that suggest that distinct sites of cellular latency could exist in the human host, which, when coupled with recent observations that report different transcriptional programmes within cells of the myeloid lineage, argues for multiple latent phenotypes that could impact differently on the biology of this virus in vivo. Finally, we will also consider how the biology of the host cell where the latent infection persists further contributes to the concept of a spectrum of latent phenotypes in multiple cell types that can be exploited by the virus.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Activación Viral/genética , Latencia del Virus/genética , Citomegalovirus/patogenicidad , Regulación Viral de la Expresión Génica , Humanos
13.
PLoS Pathog ; 10(6): e1004195, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945302

RESUMEN

The devastating clinical consequences associated with human cytomegalovirus (HCMV) infection and reactivation underscores the importance of understanding triggers of HCMV reactivation in dendritic cells (DC). Here we show that ERK-mediated reactivation is dependent on the mitogen and stress activated kinase (MSK) family. Furthermore, this MSK mediated response is dependent on CREB binding to the viral major immediate early promoter (MIEP). Specifically, CREB binding to the MIEP provides the target for MSK recruitment. Importantly, MSK mediated phosphorylation of histone H3 is required to promote histone de-methylation and the subsequent exit of HCMV from latency. Taken together, these data suggest that CREB binding to the MIEP is necessary for the recruitment of the kinase activity of MSKs to initiate the chromatin remodelling at the MIEP required for reactivation. Thus the importance of CREB during HCMV reactivation is to promote chromatin modifications conducive for viral gene expression as well as acting as a classical transcription factor. Clearly, specific inhibition of this interaction between CREB and MSKs could provide a strategy for therapeutic intervention.


Asunto(s)
Antígenos Virales/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Inmediatas-Precoces/genética , MAP Quinasa Quinasa 1/metabolismo , Transactivadores/genética , Antígenos Virales/biosíntesis , Sitios de Unión , Células Cultivadas , Citomegalovirus/genética , Infecciones por Citomegalovirus/patología , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/virología , Regulación Viral de la Expresión Génica/genética , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Sistema de Señalización de MAP Quinasas , Ácido Ocadaico/farmacología , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Transactivadores/biosíntesis , Activación Viral/fisiología , Latencia del Virus/genética
14.
Proc Natl Acad Sci U S A ; 109(2): 588-93, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22203987

RESUMEN

The ability of human CMV (HCMV) to enter and establish a latent infection in myeloid cells is crucial for survival and transmission in the human population. Initial pathogen binding and entry triggers a number of antiviral responses, including the activation of proapoptotic cell death pathways, which must be countered during latency establishment. However, mechanisms responsible for a prosurvival state in myeloid cells upon latent HCMV infection remain completely undefined. We hypothesized that the cellular antiapoptotic machinery must be initially activated by HCMV to promote early survival events upon entry. Here we show that HCMV transiently protects nonpermissive myeloid cells from chemical and virus entry induced cell death by up-regulating a key myeloid cell survival gene, myeloid cell leukemia (MCL)-1 protein. The induction of MCL-1 expression was independent of viral gene expression but dependent on activation of the ERK-MAPK pathway by viral glycoprotein B. Inhibition of ERK-MAPK signaling, inhibition of HCMV fusion, antibody-mediated neutralization of glycoprotein B signaling or expression of a shRNA against MCL-1 all correlated with increased cell death in response to virus infection or chemical stimulation. Finally we show that activation of ERK-MAPK signaling impacts on long-term latency and reactivation in hematopoietic cells. Thus, HCMV primes myeloid cells for from the initial virus-cell encounter. Given the importance of ERK and MCL-1 for myeloid cell survival, the successful establishment of HCMV latency in myeloid progenitors begins at the point of virus entry.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Citomegalovirus/metabolismo , Regulación de la Expresión Génica/fisiología , Células Mieloides/virología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/fisiología , Internalización del Virus , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Butadienos/farmacología , Cartilla de ADN/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Vectores Genéticos , Humanos , Imidazoles/farmacología , Lentivirus , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Células Mieloides/metabolismo , Nitrilos/farmacología , Piridinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas del Envoltorio Viral/metabolismo
15.
J Gen Virol ; 95(Pt 10): 2260-2266, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24986086

RESUMEN

Human cytomegalovirus (HCMV) infection and reactivation are a major cause of morbidity in immune-suppressed patients. Interestingly, epidemiological studies have shown that patients administered the mammalian target of rapamycin (mTOR) inhibitor, sirolimus (rapamycin), exhibit more favourable outcomes, suggestive of activity against HCMV in vivo. Given its relative lack of activity against lytic infection, it is postulated that rapamycin inhibits HCMV reactivation. Here, we showed that rapamycin administered acutely or chronically has little impact on induction of immediate early (IE) gene expression in experimentally latent dendritic cells or cells from naturally latent individuals. Furthermore, we extended these observations to include other inhibitors of mTORC1 and mTORC 2, which similarly have minimal effects on induction of IE gene expression from latency. Taken together, these data suggest that favourable outcomes associated with sirolimus are attributable to indirect effects that influence HCMV reactivation, rather than a direct mechanistic action against HCMV itself.


Asunto(s)
Antivirales/farmacología , Citomegalovirus/efectos de los fármacos , Citomegalovirus/fisiología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Sirolimus/farmacología , Activación Viral/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos
16.
J Virol ; 87(19): 10660-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23885077

RESUMEN

Primary infection with human cytomegalovirus (HCMV) is generally asymptomatic in healthy individuals and results in a lifelong infection of the host. In contrast, in immunosuppressed transplant recipients and late-stage AIDS patients, HCMV infection and reactivation can result in severe disease or death. In vivo, latency is established in bone marrow CD34(+) progenitor cells with reactivation linked with their differentiation to macrophages and dendritic cells (DCs). However, previous analyses have relied on ex vivo differentiation of myeloid progenitor cells to DCs in culture. Here, we now report on the isolation and analysis of circulating blood myeloid DCs, resulting from natural differentiation in vivo, from healthy HCMV-seropositive carriers. We show that these in vivo-differentiated circulating DCs are fully permissive for HCMV and exhibit a phenotype similar to that of monocyte-derived DCs routinely used for in vitro studies of HCMV. Importantly, we also show that these DCs from healthy HCMV-seropositive donors carry HCMV genomes and, significantly, are typically positive for viral immediate-early (IE) gene expression, in contrast to circulating monocytes, which carry genomes with an absence of IE expression. Finally, we show that HCMV reactivation from these circulating DCs is enhanced by inflammatory stimuli. Overall, these data argue that the differentiation in vivo of myeloid progenitors to circulating DCs promotes the reactivation of HCMV lytic gene expression in healthy individuals, thereby providing valuable confirmation of studies performed using in vitro generation of DCs from myeloid precursors to study HCMV reactivation.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/patogenicidad , Células Dendríticas/virología , Activación Viral , Latencia del Virus , Células Cultivadas , Inmunoprecipitación de Cromatina , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , ADN Viral/genética , Células Dendríticas/metabolismo , Células Dendríticas/patología , Fibroblastos/citología , Fibroblastos/virología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Prepucio/citología , Prepucio/virología , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Estudios Seroepidemiológicos
17.
J Virol ; 86(16): 8507-15, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22647696

RESUMEN

Studies from a number of laboratories have shown that the myeloid lineage is prominent in human cytomegalovirus (HCMV) latency, reactivation, dissemination, and pathogenesis. Existing as a latent infection in CD34(+) progenitors and circulating CD14(+) monocytes, reactivation is observed upon differentiation to mature macrophage or dendritic cell (DC) phenotypes. Langerhans' cells (LCs) are a subset of periphery resident DCs that represent a DC population likely to encounter HCMV early during primary infection. Furthermore, we have previously shown that CD34(+) derived LCs are a site of HCMV reactivation ex vivo. Accordingly, we have utilized healthy-donor CD34(+) cells to study latency and reactivation of HCMV in LCs. However, the increasing difficulty acquiring healthy-donor CD34(+) cells--particularly from seropositive donors due to the screening regimens used--led us to investigate the use of CD14(+) monocytes to generate LCs. We show here that CD14(+) monocytes cultured with transforming growth factor ß generate Langerin-positive DCs (MoLCs). Consistent with observations using CD34(+) derived LCs, only mature MoLCs were permissive for HCMV infection. The lytic infection of mature MoLCs is productive and results in a marked inhibition in the capacity of these cells to promote T cell proliferation. Pertinently, differentiation of experimentally latent monocytes to the MoLC phenotype promotes reactivation in a maturation and interleukin-6 (IL-6)-dependent manner. Intriguingly, however, IL-6-mediated effects were restricted to mature LCs, in contrast to observations with classical CD14(+) derived DCs. Consequently, elucidation of the molecular basis behind the differential response of the two DC subsets should further our understanding of the fundamental mechanisms important for reactivation.


Asunto(s)
Citomegalovirus/fisiología , Células Dendríticas/inmunología , Células Dendríticas/virología , Activación Viral , Latencia del Virus , Antígenos CD34/análisis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Dendríticas/química , Humanos , Receptores de Lipopolisacáridos/análisis , Monocitos/química , Monocitos/inmunología , Monocitos/virología , Virología/métodos
18.
J Virol ; 85(23): 12750-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21937636

RESUMEN

Human cytomegalovirus (HCMV) remains a major cause of viral disease in immunosuppressed transplant patients. The ability of HCMV to establish lifelong infection in humans and reactivate with devastating clinical consequences underscores the importance of understanding the triggers of HCMV reactivation in mature myeloid cells. Dendritic cell (DC) differentiation is concomitant with the activation of cellular signaling pathways and inflammatory gene expression and also HCMV reactivation. Here, we show a major role for interleukin-6 (IL-6) through extracellular signal-regulated kinase-mitogen-activated protein kinase (ERK-MAPK) signaling upon DC differentiation to promote HCMV reactivation. IL-6 drives reactivation by transcriptional upregulation of the major immediate-early (IE) genes, resulting in efficient progression of the virus life cycle and, ultimately, higher titers of infectious virus. Furthermore, the interception of IL-6 signaling with biological inhibitors significantly abrogated HCMV reactivation from experimental latency. Crucially, using cells derived from healthy seropositive donors, we observed a key role for IL-6 during reactivation from natural latency ex vivo in interstitial DCs. Clinically, HCMV reactivation occurs in highly inflammatory environments (i.e., transplantation); thus, the implications of this study could potentially provide novel approaches for therapeutic intervention.


Asunto(s)
Infecciones por Citomegalovirus/virología , Células Dendríticas/virología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Interleucina-6/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Activación Viral/fisiología , Western Blotting , Diferenciación Celular , Células Cultivadas , Citomegalovirus , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Células Dendríticas/metabolismo , Regulación Viral de la Expresión Génica , Genes Inmediatos-Precoces , Humanos , Interleucina-6/antagonistas & inhibidores , Monocitos/citología , Monocitos/metabolismo , Monocitos/virología , Células Mieloides/citología , Células Mieloides/metabolismo , Células Mieloides/virología , ARN Mensajero/genética , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Activación Transcripcional , Latencia del Virus
19.
Front Immunol ; 13: 1083230, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591233

RESUMEN

Human cytomegalovirus (HCMV) infection and periodic reactivation is, generally, well controlled by adaptative immune responses in the healthy. In older people, overt HCMV disease is rarely seen despite the association of HCMV with increased risk of mortality; evidence from studies of unwell aged populations suggest that HCMV seropositivity is an important co-morbidity factor. HCMV genomes have been detected in urine from older donors, suggesting that the immune response prevents systemic disease but possibly immunomodulation due to lifelong viral carriage may alter its efficacy at peripheral tissue sites. Previously we have demonstrated that there were no age-related expansions of T cell responses to HCMV or increase in latent viral carriage with age and these T cells produced anti-viral cytokines and viremia was very rarely detected. To investigate the efficacy of anti-HCMV responses with increasing age, we used an in vitro Viral Dissemination Assay (VDA) using autologous dermal fibroblasts to determine the anti-viral effector capacity of total PBMC, as well as important subsets (T cells, NK cells). In parallel we assessed components of the humoral response (antibody neutralization) and combined this with qPCR detection of HCMV in blood, saliva and urine in a cohort of young and old donors. Consistent with previous studies, we again show HCMV specific cIL-10, IFNγ and TNFα T cell responses to peptides did not show an age-related defect. However, assessment of direct anti-viral cellular and antibody-mediated adaptive immune responses using the VDA shows that older donors are significantly less able to control viral dissemination in an in vitro assay compared to young donors. Corroborating this observation, we detected viral genomes in saliva samples only from older donors, these donors had a defect in cellular control of viral spread in our in vitro assay. Phenotyping of fibroblasts used in this study shows expression of a number of checkpoint inhibitor ligands which may contribute to the defects observed. The potential to therapeutically intervene in checkpoint inhibitor pathways to prevent HCMV reactivation in the unwell aged is an exciting avenue to explore.


Asunto(s)
Antivirales , Citomegalovirus , Anciano , Humanos , Leucocitos Mononucleares , Inmunidad Adaptativa , Replicación Viral
20.
J Infect ; 82(5): 170-177, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33753152

RESUMEN

OBJECTIVES: To assess whether a commercially available CE-IVD, ELISA-based surrogate neutralisation assay (cPass, Genscript) provides a genuine measure of SARS-CoV-2 neutralisation by human sera, and further to establish whether measuring responses against the RBD of S was a diagnostically useful proxy for responses against the whole S protein. METHODS: Serum samples from 30 patients were assayed for anti-NP responses, for 'neutralisation' by the surrogate neutralisation assay and for neutralisation by SARS-CoV-2 S pseudotyped virus assays utilising two target cell lines. Correlation between assays was measured using linear regression. RESULTS: The responses observed within the surrogate neutralisation assay demonstrated an extremely strong, highly significant positive correlation with those observed in both pseudotyped virus assays. CONCLUSIONS: The tested ELISA-based surrogate assay provides an immunologically useful measure of functional immune responses in a much quicker and highly automatable fashion. It also reinforces that detection of anti-RBD neutralising antibodies alone is a powerful measure of the capacity to neutralise viral infection.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Ensayo de Inmunoadsorción Enzimática , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA