Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 161(3): 674-690, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25910214

RESUMEN

Modulation of protein function is used to intervene in cellular processes but is often done indirectly by means of introducing DNA or mRNA encoding the effector protein. Thus far, direct intracellular delivery of proteins has remained challenging. We developed a method termed iTOP, for induced transduction by osmocytosis and propanebetaine, in which a combination of NaCl hypertonicity-induced macropinocytosis and a transduction compound (propanebetaine) induces the highly efficient transduction of proteins into a wide variety of primary cells. We demonstrate that iTOP is a useful tool in systems in which transient cell manipulation drives permanent cellular changes. As an example, we demonstrate that iTOP can mediate the delivery of recombinant Cas9 protein and short guide RNA, driving efficient gene targeting in a non-integrative manner.


Asunto(s)
Técnicas Citológicas , Proteínas , Células Cultivadas , Células Madre Embrionarias , Marcación de Gen , Humanos , ARN , Transducción Genética
2.
Cell ; 158(2): 449-461, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25036638

RESUMEN

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) holds enormous promise for regenerative medicine. To elucidate endogenous barriers limiting this process, we systematically dissected human cellular reprogramming by combining a genome-wide RNAi screen, innovative computational methods, extensive single-hit validation, and mechanistic investigation of relevant pathways and networks. We identify reprogramming barriers, including genes involved in transcription, chromatin regulation, ubiquitination, dephosphorylation, vesicular transport, and cell adhesion. Specific a disintegrin and metalloproteinase (ADAM) proteins inhibit reprogramming, and the disintegrin domain of ADAM29 is necessary and sufficient for this function. Clathrin-mediated endocytosis can be targeted with small molecules and opposes reprogramming by positively regulating TGF-ß signaling. Genetic interaction studies of endocytosis or ubiquitination reveal that barrier pathways can act in linear, parallel, or feedforward loop architectures to antagonize reprogramming. These results provide a global view of barriers to human cellular reprogramming.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Proteínas ADAM/metabolismo , Adhesión Celular , Células Madre Embrionarias/metabolismo , Endocitosis , Humanos , Ubiquitina/metabolismo
3.
Cell ; 152(4): 909-22, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23394947

RESUMEN

Genetic interaction (GI) maps, comprising pairwise measures of how strongly the function of one gene depends on the presence of a second, have enabled the systematic exploration of gene function in microorganisms. Here, we present a two-stage strategy to construct high-density GI maps in mammalian cells. First, we use ultracomplex pooled shRNA libraries (25 shRNAs/gene) to identify high-confidence hit genes for a given phenotype and effective shRNAs. We then construct double-shRNA libraries from these to systematically measure GIs between hits. A GI map focused on ricin susceptibility broadly recapitulates known pathways and provides many unexpected insights. These include a noncanonical role for COPI, a previously uncharacterized protein complex affecting toxin clearance, a specialized role for the ribosomal protein RPS25, and functionally distinct mammalian TRAPP complexes. The ability to rapidly generate mammalian GI maps provides a potentially transformative tool for defining gene function and designing combination therapies based on synergistic pairs.


Asunto(s)
Transporte Biológico , Epistasis Genética , Ricina/toxicidad , Atorvastatina , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteína Coat de Complejo I/metabolismo , Retículo Endoplásmico/metabolismo , Ácidos Heptanoicos/farmacología , Humanos , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Pirroles/farmacología , ARN Interferente Pequeño , Proteínas Ribosómicas/metabolismo , Proteínas de Transporte Vesicular/metabolismo
4.
Mol Cell ; 79(5): 768-781.e7, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32738194

RESUMEN

Misfolded proteins in the endoplasmic reticulum (ER) are degraded by ER-associated degradation (ERAD). Although ERAD components involved in degradation of luminal substrates are well characterized, much less is known about quality control of membrane proteins. Here, we analyzed the degradation pathways of two short-lived ER membrane model proteins in mammalian cells. Using a CRISPR-Cas9 genome-wide library screen, we identified an ERAD branch required for quality control of a subset of membrane proteins. Using biochemical and mass spectrometry approaches, we showed that this ERAD branch is defined by an ER membrane complex consisting of the ubiquitin ligase RNF185, the ubiquitin-like domain containing proteins TMUB1/2 and TMEM259/Membralin, a poorly characterized protein. This complex cooperates with cytosolic ubiquitin ligase UBE3C and p97 ATPase in degrading their membrane substrates. Our data reveal that ERAD branches have remarkable specificity for their membrane substrates, suggesting that multiple, perhaps combinatorial, determinants are involved in substrate selection.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Sistema Enzimático del Citocromo P-450/metabolismo , Células HEK293 , Células HeLa , Humanos , Dominios Proteicos , Pliegue de Proteína , Proteolisis , Proteínas de Saccharomyces cerevisiae/metabolismo , Esterol 14-Desmetilasa/metabolismo
5.
Cancer Immunol Immunother ; 73(1): 16, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38236251

RESUMEN

Collagen expression and structure in the tumour microenvironment are associated with tumour development and therapy response. Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a widely expressed inhibitory collagen receptor. LAIR-2 is a soluble homologue of LAIR-1 that competes for collagen binding. Multiple studies in mice implicate blockade of LAIR-1:collagen interaction in cancer as a promising therapeutic strategy. Here, we investigated the role of LAIR-1 in anti-tumour responses. We show that although LAIR-1 inhibits activation, proliferation, and cytokine production of mouse T cells in vitro, tumour outgrowth in LAIR-1-deficient mice did not differ from wild type mice in several in vivo tumour models. Furthermore, treatment with NC410, a LAIR-2-Fc fusion protein, did not result in increased tumour clearance in tested immunocompetent mice, which contrasts with previous data in humanized mouse models. This discrepancy may be explained by our finding that NC410 blocks human LAIR-1:collagen interaction more effectively than mouse LAIR-1:collagen interaction. Despite the lack of therapeutic impact of NC410 monotherapy, mice treated with a combination of NC410 and anti-programmed death-ligand 1 did show reduced tumour burden and increased survival. Using LAIR-1-deficient mice, we showed that this effect seemed to be dependent on the presence of LAIR-1. Taken together, our data demonstrate that the absence of LAIR-1 signalling alone is not sufficient to control tumour growth in multiple immunocompetent mouse models. However, combined targeting of LAIR-1 and PD-L1 results in increased tumour control. Thus, additional targeting of the LAIR-1:collagen pathway with NC410 is a promising approach to treating tumours where conventional immunotherapy is ineffective.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Animales , Humanos , Ratones , Colágeno , Modelos Animales de Enfermedad , Leucocitos , Ligandos , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
6.
Rev Med Virol ; 32(3): e2284, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34543489

RESUMEN

Respiratory syncytial virus (RSV) is a major health problem. A better understanding of the geographical and temporal dynamics of RSV circulation will assist in tracking resistance against therapeutics currently under development. Since 2015, the field of RSV molecular epidemiology has evolved rapidly with around 20-30 published articles per year. The objective of this systematic review is to identify knowledge gaps in recent RSV genetic literature to guide global molecular epidemiology research. We included 78 studies published between 2015 and 2020 describing 12,998 RSV sequences of which 8,233 (63%) have been uploaded to GenBank. Seventeen (22%) studies were performed in low- and middle-income countries (LMICs), and seven (9%) studies sequenced whole-genomes. Although most reported polymorphisms for monoclonal antibodies in clinical development (nirsevimab, MK-1654) have not been tested for resistance in neutralisation essays, known resistance was detected at low levels for the nirsevimab and palivizumab binding site. High resistance was found for the suptavumab binding site. We present the first literature review of an enormous amount of RSV genetic data. The need for global monitoring of RSV molecular epidemiology becomes increasingly important in evaluating the effectiveness of monoclonal antibody candidates as they reach their final stages of clinical development. We have identified the following three knowledge gaps: whole-genome data to study global RSV evolution, data from LMICs and data from global surveillance programs.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Anticuerpos Monoclonales/uso terapéutico , Antivirales/uso terapéutico , Humanos , Palivizumab/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/epidemiología , Virus Sincitial Respiratorio Humano/genética
8.
EMBO J ; 37(8)2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29496741

RESUMEN

Negative regulation of immune pathways is essential to achieve resolution of immune responses and to avoid excess inflammation. DNA stimulates type I IFN expression through the DNA sensor cGAS, the second messenger cGAMP, and the adaptor molecule STING Here, we report that STING degradation following activation of the pathway occurs through autophagy and is mediated by p62/SQSTM1, which is phosphorylated by TBK1 to direct ubiquitinated STING to autophagosomes. Degradation of STING was impaired in p62-deficient cells, which responded with elevated IFN production to foreign DNA and DNA pathogens. In the absence of p62, STING failed to traffic to autophagy-associated vesicles. Thus, DNA sensing induces the cGAS-STING pathway to activate TBK1, which phosphorylates IRF3 to induce IFN expression, but also phosphorylates p62 to stimulate STING degradation and attenuation of the response.


Asunto(s)
Nucleotidiltransferasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteína Sequestosoma-1/fisiología , Animales , Autofagia , Línea Celular , ADN/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
9.
Eur J Immunol ; 50(8): 1209-1219, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32198923

RESUMEN

Ankylosing spondylitis (AS) is associated with autoantibody production to class II MHC-associated invariant chain peptide, CD74/CLIP. In this study, we considered that anti-CD74/CLIP autoantibodies present in sera from AS might recognize CD74 degradation products that accumulate upon deficiency of the enzyme signal peptide peptidase-like 2A (SPPL2a). We analyzed monocytes from healthy controls (n = 42), psoriatic arthritis (n = 25), rheumatoid arthritis (n = 16), and AS patients (n = 15) for SPPL2a enzyme activity and complemented the experiments using SPPL2a-sufficient and -deficient THP-1 cells. We found defects in SPPL2a function and CD74 processing in a subset of AS patients, which culminated in CD74 and HLA class II display at the cell surface. These findings were verified in SPPL2a-deficient THP-1 cells, which showed expedited expression of MHC class II, total CD74 and CD74 N-terminal degradation products at the plasma membrane upon receipt of an inflammatory trigger. Furthermore, we observed that IgG anti-CD74/CLIP autoantibodies recognize CD74 N-terminal degradation products that accumulate upon SPPL2a defect. In conclusion, reduced activity of SPPL2a protease in monocytes from AS predisposes to endosomal accumulation of CD74 and CD74 N-terminal fragments, which, upon IFN-γ-exposure, is deposited at the plasma membrane and can be recognized by anti-CD74/CLIP autoantibodies.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Ácido Aspártico Endopeptidasas/fisiología , Autoanticuerpos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Proteolisis , Espondilitis Anquilosante/inmunología , Adulto , Anciano , Antígenos de Diferenciación de Linfocitos B/metabolismo , Femenino , Antígenos HLA-DR/análisis , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunoglobulina G/inmunología , Interferón gamma/farmacología , Masculino , Persona de Mediana Edad , Células THP-1
10.
J Clin Microbiol ; 59(1)2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33087438

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among infants and young children, resulting in annual epidemics worldwide. INFORM-RSV is a multiyear clinical study designed to describe the global molecular epidemiology of RSV in children under 5 years of age by monitoring temporal and geographical evolution of current circulating RSV strains, F protein antigenic sites, and their relationships with clinical features of RSV disease. During the pilot season (2017-2018), 410 RSV G-F gene sequences were obtained from 476 RSV-positive nasal samples collected from 8 countries (United Kingdom, Spain, The Netherlands, Finland, Japan, Brazil, South Africa, and Australia). RSV B (all BA9 genotype) predominated over RSV A (all ON1 genotype) globally (69.0% versus 31.0%) and in all countries except South Africa. Geographic clustering patterns highlighted wide transmission and continued evolution with viral spread. Most RSV strains were from infants of <1 year of age (81.2%), males (56.3%), and patients hospitalized for >24 h (70.5%), with no differences in subtype distribution. Compared to 2013 reference sequences, variations at F protein antigenic sites were observed for both RSV A and B strains, with high-frequency polymorphisms at antigenic site Ø (I206M/Q209R) and site V (L172Q/S173L/K191R) in RSV B strains. The INFORM-RSV 2017-2018 pilot season establishes an important molecular baseline of RSV strain distribution and sequence variability with which to track the emergence of new strains and provide an early warning system of neutralization escape variants that may impact transmission or the effectiveness of vaccines and MAbs under development.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Australia , Brasil , Niño , Preescolar , Finlandia , Genotipo , Humanos , Lactante , Japón , Masculino , Epidemiología Molecular , Países Bajos , Filogenia , Infecciones por Virus Sincitial Respiratorio/epidemiología , Virus Sincitial Respiratorio Humano/genética , Sudáfrica , España , Reino Unido
11.
J Virol ; 93(13)2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30996093

RESUMEN

Vaccinia virus is a promising viral vaccine and gene delivery candidate and has historically been used as a model to study poxvirus-host cell interactions. We employed a genome-wide insertional mutagenesis approach in human haploid cells to identify host factors crucial for vaccinia virus infection. A library of mutagenized HAP1 cells was exposed to modified vaccinia virus Ankara (MVA). Deep-sequencing analysis of virus-resistant cells identified host factors involved in heparan sulfate synthesis, Golgi organization, and vesicular protein trafficking. We validated EXT1, TM9SF2, and TMED10 (TMP21/p23/p24δ) as important host factors for vaccinia virus infection. The critical roles of EXT1 in heparan sulfate synthesis and vaccinia virus infection were confirmed. TM9SF2 was validated as a player mediating heparan sulfate expression, explaining its contribution to vaccinia virus infection. In addition, TMED10 was found to be crucial for virus-induced plasma membrane blebbing and phosphatidylserine-induced macropinocytosis, presumably by regulating the cell surface expression of the TAM receptor Axl.IMPORTANCE Poxviruses are large DNA viruses that can infect a wide range of host species. A number of these viruses are clinically important to humans, including variola virus (smallpox) and vaccinia virus. Since the eradication of smallpox, zoonotic infections with monkeypox virus and cowpox virus are emerging. Additionally, poxviruses can be engineered to specifically target cancer cells and are used as a vaccine vector against tuberculosis, influenza, and coronaviruses. Poxviruses rely on host factors for most stages of their life cycle, including attachment to the cell and entry. These host factors are crucial for virus infectivity and host cell tropism. We used a genome-wide knockout library of host cells to identify host factors necessary for vaccinia virus infection. We confirm a dominant role for heparin sulfate in mediating virus attachment. Additionally, we show that TMED10, previously not implicated in virus infections, facilitates virus uptake by modulating the cellular response to phosphatidylserine.


Asunto(s)
Haploidia , Heparitina Sulfato/genética , Heparitina Sulfato/aislamiento & purificación , Pinocitosis/fisiología , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Vaccinia/virología , Proteínas de Transporte Vesicular/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Virus de la Viruela Vacuna/genética , Virus ADN , Técnicas de Inactivación de Genes , Pruebas Genéticas , Aparato de Golgi , Células HEK293 , Células HeLa , Heparitina Sulfato/metabolismo , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana , Monkeypox virus/genética , N-Acetilglucosaminiltransferasas , Fosfatidilserinas/metabolismo , Poxviridae/genética , Acoplamiento Viral
12.
PLoS Pathog ; 14(1): e1006823, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29304174

RESUMEN

Herpesviruses are large DNA viruses which depend on many nuclear functions, and therefore on host transport factors to ensure specific nuclear import of viral and host components. While some import cargoes bind directly to certain transport factors, most recruit importin ß1 via importin α. We identified importin α1 in a small targeted siRNA screen to be important for herpes simplex virus (HSV-1) gene expression. Production of infectious virions was delayed in the absence of importin α1, but not in cells lacking importin α3 or importin α4. While nuclear targeting of the incoming capsids, of the HSV-1 transcription activator VP16, and of the viral genomes were not affected, the nuclear import of the HSV-1 proteins ICP4 and ICP0, required for efficient viral transcription, and of ICP8 and pUL42, necessary for DNA replication, were reduced. Furthermore, quantitative electron microscopy showed that fibroblasts lacking importin α1 contained overall fewer nuclear capsids, but an increased proportion of mature nuclear capsids indicating that capsid formation and capsid egress into the cytoplasm were impaired. In neurons, importin α1 was also not required for nuclear targeting of incoming capsids, but for nuclear import of ICP4 and for the formation of nuclear capsid assembly compartments. Our data suggest that importin α1 is specifically required for the nuclear localization of several important HSV1 proteins, capsid assembly, and capsid egress into the cytoplasm, and may become rate limiting in situ upon infection at low multiplicity or in terminally differentiated cells such as neurons.


Asunto(s)
Proteínas de la Cápside/metabolismo , Núcleo Celular/metabolismo , Fibroblastos/virología , Herpesvirus Humano 1/fisiología , Neuronas/virología , Ensamble de Virus/genética , alfa Carioferinas/fisiología , Transporte Activo de Núcleo Celular/genética , Animales , Cápside/metabolismo , Línea Celular , Núcleo Celular/virología , Cricetinae , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Herpesvirus Humano 1/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , alfa Carioferinas/genética
13.
BMC Infect Dis ; 20(1): 450, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32591017

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) is a global cause of severe respiratory morbidity and mortality in infants. While preventive and therapeutic interventions are being developed, including antivirals, vaccines and monoclonal antibodies, little is known about the global molecular epidemiology of RSV. INFORM is a prospective, multicenter, global clinical study performed by ReSViNET to investigate the worldwide molecular diversity of RSV isolates collected from children less than 5 years of age. METHODS: The INFORM study is performed in 17 countries spanning all inhabited continents and will provide insight into the molecular epidemiology of circulating RSV strains worldwide. Sequencing of > 4000 RSV-positive respiratory samples is planned to detect temporal and geographical molecular patterns on a molecular level over five consecutive years. Additionally, RSV will be cultured from a subset of samples to study the functional implications of specific mutations in the viral genome including viral fitness and susceptibility to different monoclonal antibodies. DISCUSSION: The sequencing and functional results will be used to investigate susceptibility and resistance to novel RSV preventive or therapeutic interventions. Finally, a repository of globally collected RSV strains and a database of RSV sequences will be created.


Asunto(s)
Genoma Viral , Epidemiología Molecular/métodos , Polimorfismo Genético , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/genética , Anticuerpos Monoclonales/uso terapéutico , Antivirales/efectos adversos , Antivirales/uso terapéutico , Preescolar , Farmacorresistencia Bacteriana/genética , Femenino , Genotipo , Humanos , Inmunización Pasiva , Lactante , Recién Nacido , Masculino , Estudios Prospectivos , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Proc Natl Acad Sci U S A ; 114(8): E1413-E1421, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28174275

RESUMEN

Fibroblasts are a main player in the tumor-inhibitory microenvironment. Upon tumor initiation and progression, fibroblasts can lose their tumor-inhibitory capacity and promote tumor growth. The molecular mechanisms that underlie this switch have not been defined completely. Previously, we identified four proteins overexpressed in cancer-associated fibroblasts and linked to Rho GTPase signaling. Here, we show that knocking out the Ras homolog family member A (RhoA) gene in normal fibroblasts decreased their tumor-inhibitory capacity, as judged by neighbor suppression in vitro and accompanied by promotion of tumor growth in vivo. This also induced PC3 cancer cell motility and increased colony size in 2D cultures. RhoA knockout in fibroblasts induced vimentin intermediate filament reorganization, accompanied by reduced contractile force and increased stiffness of cells. There was also loss of wide F-actin stress fibers and large focal adhesions. In addition, we observed a significant loss of α-smooth muscle actin, which indicates a difference between RhoA knockout fibroblasts and classic cancer-associated fibroblasts. In 3D collagen matrix, RhoA knockout reduced fibroblast branching and meshwork formation and resulted in more compactly clustered tumor-cell colonies in coculture with PC3 cells, which might boost tumor stem-like properties. Coculturing RhoA knockout fibroblasts and PC3 cells induced expression of proinflammatory genes in both. Inflammatory mediators may induce tumor cell stemness. Network enrichment analysis of transcriptomic changes, however, revealed that the Rho signaling pathway per se was significantly triggered only after coculturing with tumor cells. Taken together, our findings in vivo and in vitro indicate that Rho signaling governs the inhibitory effects by fibroblasts on tumor-cell growth.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular/fisiología , Neoplasias/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Cultivadas , Colágeno/metabolismo , Femenino , Adhesiones Focales/metabolismo , Células HEK293 , Humanos , Ratones , Ratones SCID , Transducción de Señal/fisiología , Fibras de Estrés/metabolismo , Quinasas Asociadas a rho/metabolismo
15.
J Cell Sci ; 130(17): 2883-2892, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28743740

RESUMEN

Misfolded endoplasmic reticulum (ER) proteins are dislocated towards the cytosol and degraded by the ubiquitin-proteasome system in a process called ER-associated protein degradation (ERAD). During infection with human cytomegalovirus (HCMV), the viral US2 protein targets HLA class I molecules (HLA-I) for degradation via ERAD to avoid elimination by the immune system. US2-mediated degradation of HLA-I serves as a paradigm of ERAD and has facilitated the identification of TRC8 (also known as RNF139) as an E3 ubiquitin ligase. No specific E2 enzymes had previously been described for cooperation with TRC8. In this study, we used a lentiviral CRISPR/Cas9 library targeting all known human E2 enzymes to assess their involvement in US2-mediated HLA-I downregulation. We identified multiple E2 enzymes involved in this process, of which UBE2G2 was crucial for the degradation of various immunoreceptors. UBE2J2, on the other hand, counteracted US2-induced ERAD by downregulating TRC8 expression. These findings indicate the complexity of cellular quality control mechanisms, which are elegantly exploited by HCMV to elude the immune system.


Asunto(s)
Citomegalovirus/metabolismo , Regulación hacia Abajo , Receptores Inmunológicos/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Sistemas CRISPR-Cas/genética , Pruebas Genéticas , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Modelos Biológicos , Proteolisis , Receptores de Superficie Celular/metabolismo , Células U937 , Regulación hacia Arriba
16.
J Immunol ; 198(10): 4062-4073, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28416598

RESUMEN

Type I IFNs play critical roles in orchestrating the antiviral defense by inducing direct antiviral activities and shaping the adaptive immune response. Viruses have evolved numerous strategies to specifically interfere with IFN production or its downstream mediators, thereby allowing successful infection of the host to occur. The prototypic human gammaherpesvirus EBV, which is associated with infectious mononucleosis and malignant tumors, harbors many immune-evasion proteins that manipulate the adaptive and innate immune systems. In addition to proteins, the virus encodes >40 mature microRNAs for which the functions remain largely unknown. In this article, we identify EBV-encoded miR-BART16 as a novel viral immune-evasion factor that interferes with the type I IFN signaling pathway. miR-BART16 directly targets CREB-binding protein, a key transcriptional coactivator in IFN signaling, thereby inducing CREB-binding protein downregulation in EBV-transformed B cells and gastric carcinoma cells. miR-BART16 abrogates the production of IFN-stimulated genes in response to IFN-α stimulation and it inhibits the antiproliferative effect of IFN-α on latently infected BL cells. By obstructing the type I IFN-induced antiviral response, miR-BART16 provides a means to facilitate the establishment of latent EBV infection and enhance viral replication.


Asunto(s)
Herpesvirus Humano 4/genética , Interferón Tipo I/metabolismo , MicroARNs/metabolismo , ARN Viral/metabolismo , Transducción de Señal , Proteína de Unión a CREB/metabolismo , Línea Celular , Herpesvirus Humano 4/inmunología , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Interferón Tipo I/inmunología , MicroARNs/genética , ARN Viral/genética , Replicación Viral
17.
J Gen Virol ; 99(6): 790-804, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29676720

RESUMEN

Poxviruses comprise a group of large dsDNA viruses that include members relevant to human and animal health, such as variola virus, monkeypox virus, cowpox virus and vaccinia virus (VACV). Poxviruses are remarkable for their unique replication cycle, which is restricted to the cytoplasm of infected cells. The independence from the host nucleus requires poxviruses to encode most of the enzymes involved in DNA replication, transcription and processing. Here, we use the CRISPR/Cas9 genome engineering system to induce DNA damage to VACV (strain Western Reserve) genomes. We show that targeting CRISPR/Cas9 to essential viral genes limits virus replication efficiently. Although VACV is a strictly cytoplasmic pathogen, we observed extensive viral genome editing at the target site; this is reminiscent of a non-homologous end-joining DNA repair mechanism. This pathway was not dependent on the viral DNA ligase, but critically involved the cellular DNA ligase IV. Our data show that DNA ligase IV can act outside of the nucleus to allow repair of dsDNA breaks in poxvirus genomes. This pathway might contribute to the introduction of mutations within the genome of poxviruses and may thereby promote the evolution of these viruses.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN Ligasa (ATP)/metabolismo , Reparación del ADN , Genoma Viral , Interacciones Microbiota-Huesped/genética , Virus Vaccinia/genética , Sistemas CRISPR-Cas , Línea Celular Tumoral , Citosol/metabolismo , Citosol/virología , ADN Ligasa (ATP)/genética , Replicación del ADN , ADN Viral/genética , Células HEK293 , Humanos , Mutagénesis , Virus Vaccinia/fisiología , Replicación Viral/genética
18.
PLoS Pathog ; 12(6): e1005701, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27362483

RESUMEN

Herpesviruses infect the majority of the human population and can cause significant morbidity and mortality. Herpes simplex virus (HSV) type 1 causes cold sores and herpes simplex keratitis, whereas HSV-2 is responsible for genital herpes. Human cytomegalovirus (HCMV) is the most common viral cause of congenital defects and is responsible for serious disease in immuno-compromised individuals. Epstein-Barr virus (EBV) is associated with infectious mononucleosis and a broad range of malignancies, including Burkitt's lymphoma, nasopharyngeal carcinoma, Hodgkin's disease, and post-transplant lymphomas. Herpesviruses persist in their host for life by establishing a latent infection that is interrupted by periodic reactivation events during which replication occurs. Current antiviral drug treatments target the clinical manifestations of this productive stage, but they are ineffective at eliminating these viruses from the infected host. Here, we set out to combat both productive and latent herpesvirus infections by exploiting the CRISPR/Cas9 system to target viral genetic elements important for virus fitness. We show effective abrogation of HCMV and HSV-1 replication by targeting gRNAs to essential viral genes. Simultaneous targeting of HSV-1 with multiple gRNAs completely abolished the production of infectious particles from human cells. Using the same approach, EBV can be almost completely cleared from latently infected EBV-transformed human tumor cells. Our studies indicate that the CRISPR/Cas9 system can be effectively targeted to herpesvirus genomes as a potent prophylactic and therapeutic anti-viral strategy that may be used to impair viral replication and clear latent virus infection.


Asunto(s)
Sistemas CRISPR-Cas/genética , Citomegalovirus/genética , Edición Génica/métodos , Genoma Viral , Infecciones por Herpesviridae/genética , Herpesviridae/genética , Línea Celular , Herpesvirus Humano 1 , Humanos , Reacción en Cadena de la Polimerasa , Latencia del Virus/genética
19.
PLoS Pathog ; 12(4): e1005550, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27077376

RESUMEN

Cell-mediated immunity plays a key role in host control of viral infection. This is exemplified by life-threatening reactivations of e.g. herpesviruses in individuals with impaired T-cell and/or iNKT cell responses. To allow lifelong persistence and virus production in the face of primed immunity, herpesviruses exploit immune evasion strategies. These include a reduction in viral antigen expression during latency and a number of escape mechanisms that target antigen presentation pathways. Given the plethora of foreign antigens expressed in virus-producing cells, herpesviruses are conceivably most vulnerable to elimination by cell-mediated immunity during the replicative phase of infection. Here, we show that a prototypic herpesvirus, Epstein-Barr virus (EBV), encodes a novel, broadly acting immunoevasin, gp150, that is expressed during the late phase of viral replication. In particular, EBV gp150 inhibits antigen presentation by HLA class I, HLA class II, and the non-classical, lipid-presenting CD1d molecules. The mechanism of gp150-mediated T-cell escape does not depend on degradation of the antigen-presenting molecules nor does it require gp150's cytoplasmic tail. Through its abundant glycosylation, gp150 creates a shield that impedes surface presentation of antigen. This is an unprecedented immune evasion mechanism for herpesviruses. In view of its likely broader target range, gp150 could additionally have an impact beyond escape of T cell activation. Importantly, B cells infected with a gp150-null mutant EBV displayed rescued levels of surface antigen presentation by HLA class I, HLA class II, and CD1d, supporting an important role for iNKT cells next to classical T cells in fighting EBV infection. At the same time, our results indicate that EBV gp150 prolongs the timespan for producing viral offspring at the most vulnerable stage of the viral life cycle.


Asunto(s)
Presentación de Antígeno/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Evasión Inmune/inmunología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas Virales/inmunología , Western Blotting , Citometría de Flujo , Herpesvirus Humano 4/inmunología , Humanos , Microscopía Confocal , Linfocitos T/inmunología , Transducción Genética
20.
Cell Microbiol ; 19(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27860066

RESUMEN

Over 90% of the adult population is infected with one or multiple herpesviruses. These viruses are characterized by their ability to establish latency, where the host is unable to clear the invader from infected cells resulting in a lifelong infection. Herpesviruses cause a wide variety of (recurrent) diseases such as cold sores, shingles, congenital defects and several malignancies. Although the productive phase of a herpesvirus infection can often be efficiently limited by nucleoside analogs, these drugs are ineffective during a latent herpesvirus infection and are therefore unable to clear herpesviruses from the human host. Advances in genome engineering using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 facilitates virus research and may hold potential to treat or cure previously incurable herpesvirus infections by directly targeting these viruses within infected cells. Here, we review recent applications of the CRISPR/Cas9 system for herpesviral research and discuss the therapeutic potential of the system to treat, or even cure, productive and latent herpesviral infections.


Asunto(s)
Terapia Biológica/métodos , Sistemas CRISPR-Cas , Infecciones por Herpesviridae/terapia , Herpesviridae/patogenicidad , Interacciones Huésped-Patógeno , Latencia del Virus , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA