Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol ; 97(8): e0054023, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37504571

RESUMEN

Epstein-Barr virus (EBV) is a human oncogenic γ-herpesvirus that establishes persistent infection in more than 90% of the world's population. EBV has two life cycles, latency and lytic replication. Reactivation of EBV from latency to the lytic cycle is initiated and controlled by two viral immediate-early transcription factors, Zta and Rta, encoded by BZLF1 and BRLF1, respectively. In this study, we found that IQGAP2 expression was elevated in EBV-infected B cells and identified Rta as a viral gene responsible for the IQGAP2 upregulation in both B cells and nasopharyngeal carcinoma cell lines. Mechanistically, we showed that Rta increases IQGAP2 expression through direct binding to the Rta-responsive element in the IQGAP2 promoter. We also demonstrated the direct interaction between Rta and IQGAP2 as well as their colocalization in the nucleus. Functionally, we showed that the induced IQGAP2 is required for the Rta-mediated Rta promoter activation in the EBV lytic cycle progression and may influence lymphoblastoid cell line clumping morphology through regulating E-cadherin expression. IMPORTANCE Elevated levels of antibodies against EBV lytic proteins and increased EBV DNA copy numbers in the sera have been reported in patients suffering from Burkitt's lymphoma, Hodgkin's lymphoma, and nasopharyngeal carcinoma, indicating that EBV lytic cycle progression may play an important role in the pathogenesis of EBV-associated diseases and highlighting the need for a more complete mechanistic understanding of the EBV lytic cycle. Rta acts as an essential transcriptional activator to induce lytic gene expression and thus trigger EBV reactivation. In this study, scaffolding protein IQGAP2 was found to be upregulated prominently following EBV infection via the direct binding of Rta to the RRE in the IQGAP2 promoter but not in response to other biological stimuli. Importantly, IQGAP2 was demonstrated to interact with Rta and promote the EBV lytic cycle progression. Suppression of IQGAP2 was also found to decrease E-cadherin expression and affect the clumping morphology of lymphoblastoid cell lines.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Proteínas Inmediatas-Precoces , Neoplasias Nasofaríngeas , Humanos , Infecciones por Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Proteínas Virales/genética , Proteínas Virales/metabolismo , Activación Viral
2.
J Virol ; 97(4): e0010223, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37022164

RESUMEN

Whether and how a local virus infection affects the hematopoietic system in the bone marrow is largely unknown, unlike with systemic infection. In this study, we showed that influenza A virus (IAV) infection leads to demand-adapted monopoiesis in the bone marrow. The beta interferon (IFN-ß) promoter stimulator 1 (IPS-1)-type I IFN-IFN-α receptor 1 (IFNAR1) axis-mediated signaling was found to induce the emergency expansion of the granulocyte-monocyte progenitor (GMP) population and upregulate the expression of the macrophage colony-stimulating factor receptor (M-CSFR) on bipotent GMPs and monocyte progenitors via the signal transducer and activator of transcription 1 (STAT1), leading to a scaled-back proportion of granulocyte progenitors. To further address the influence of demand-adapted monopoiesis on IAV-induced secondary bacterial infection, IAV-infected wild-type (WT) and Stat1-/- mice were challenged with Streptococcus pneumoniae. Compared with WT mice, Stat1-/- mice did not demonstrate demand-adapted monopoiesis, had more infiltrating granulocytes, and were able to effectively eliminate the bacterial infection. IMPORTANCE Our findings show that influenza A virus infection induces type I interferon (IFN)-mediated emergency hematopoiesis to expand the GMP population in the bone marrow. The type I IFN-STAT1 axis was identified as being involved in mediating the viral-infection-driven demand-adapted monopoiesis by upregulating M-CSFR expression in the GMP population. As secondary bacterial infections often manifest during a viral infection and can lead to severe or even fatal clinical complications, we further assessed the impact of the observed monopoiesis on bacterial clearance. Our results suggest that the resulting decrease in the proportion of granulocytes may play a role in diminishing the IAV-infected host's ability to effectively clear secondary bacterial infection. Our findings not only provide a more complete picture of the modulatory functions of type I IFN but also highlight the need for a more comprehensive understanding of potential changes in hematopoiesis during local infections to better inform clinical interventions.


Asunto(s)
Interferón Tipo I , Infecciones por Orthomyxoviridae , Receptor de Factor Estimulante de Colonias de Macrófagos , Factor de Transcripción STAT1 , Regulación hacia Arriba , Animales , Humanos , Ratones , Virus de la Influenza A/inmunología , Interferón Tipo I/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Infecciones por Orthomyxoviridae/inmunología , Hematopoyesis/inmunología , Células Progenitoras de Granulocitos y Macrófagos/inmunología , Streptococcus pneumoniae/inmunología , Infecciones Neumocócicas/inmunología
3.
Science ; 377(6603): eabq1841, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35699621

RESUMEN

The Omicron, or Pango lineage B.1.1.529, variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) carries multiple spike mutations with high transmissibility and partial neutralizing antibody (nAb) escape. Vaccinated individuals show protection against severe disease, often attributed to primed cellular immunity. We investigated T and B cell immunity against B.1.1.529 in triple BioNTech BNT162b2 messenger RNA-vaccinated health care workers (HCWs) with different SARS-CoV-2 infection histories. B and T cell immunity against previous variants of concern was enhanced in triple-vaccinated individuals, but the magnitude of T and B cell responses against B.1.1.529 spike protein was reduced. Immune imprinting by infection with the earlier B.1.1.7 (Alpha) variant resulted in less durable binding antibody against B.1.1.529. Previously infection-naïve HCWs who became infected during the B.1.1.529 wave showed enhanced immunity against earlier variants but reduced nAb potency and T cell responses against B.1.1.529 itself. Previous Wuhan Hu-1 infection abrogated T cell recognition and any enhanced cross-reactive neutralizing immunity on infection with B.1.1.529.


Asunto(s)
Linfocitos B , Vacuna BNT162 , COVID-19 , Inmunización Secundaria , SARS-CoV-2 , Linfocitos T , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Vacuna BNT162/inmunología , Vacuna BNT162/uso terapéutico , COVID-19/inmunología , COVID-19/prevención & control , Reacciones Cruzadas , Humanos , Ratones , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T/inmunología
4.
Nat Commun ; 13(1): 1379, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35296643

RESUMEN

Anti tumour necrosis factor (anti-TNF) drugs increase the risk of serious respiratory infection and impair protective immunity following pneumococcal and influenza vaccination. Here we report SARS-CoV-2 vaccine-induced immune responses and breakthrough infections in patients with inflammatory bowel disease, who are treated either with the anti-TNF antibody, infliximab, or with vedolizumab targeting a gut-specific anti-integrin that does not impair systemic immunity. Geometric mean [SD] anti-S RBD antibody concentrations are lower and half-lives shorter in patients treated with infliximab than vedolizumab, following two doses of BNT162b2 (566.7 U/mL [6.2] vs 4555.3 U/mL [5.4], p <0.0001; 26.8 days [95% CI 26.2 - 27.5] vs 47.6 days [45.5 - 49.8], p <0.0001); similar results are also observed with ChAdOx1 nCoV-19 vaccination (184.7 U/mL [5.0] vs 784.0 U/mL [3.5], p <0.0001; 35.9 days [34.9 - 36.8] vs 58.0 days [55.0 - 61.3], p value < 0.0001). One fifth of patients fail to mount a T cell response in both treatment groups. Breakthrough SARS-CoV-2 infections are more frequent (5.8% (201/3441) vs 3.9% (66/1682), p = 0.0039) in patients treated with infliximab than vedolizumab, and the risk of breakthrough SARS-CoV-2 infection is predicted by peak anti-S RBD antibody concentration after two vaccine doses. Irrespective of the treatments, higher, more sustained antibody levels are observed in patients with a history of SARS-CoV-2 infection prior to vaccination. Our results thus suggest that adapted vaccination schedules may be required to induce immunity in at-risk, anti-TNF-treated patients.


Asunto(s)
COVID-19 , Enfermedades Inflamatorias del Intestino , Vacunas Virales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Vacuna BNT162 , Vacunas contra la COVID-19 , ChAdOx1 nCoV-19 , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Infliximab/uso terapéutico , SARS-CoV-2 , Linfocitos T , Inhibidores del Factor de Necrosis Tumoral
5.
Immunology ; 166(1): 68-77, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35156709

RESUMEN

SARS-CoV-2 infection results in different outcomes ranging from asymptomatic infection to mild or severe disease and death. Reasons for this diversity of outcome include differences in challenge dose, age, gender, comorbidity and host genomic variation. Human leukocyte antigen (HLA) polymorphisms may influence immune response and disease outcome. We investigated the association of HLAII alleles with case definition symptomatic COVID-19, virus-specific antibody and T-cell immunity. A total of 1364 UK healthcare workers (HCWs) were recruited during the first UK SARS-CoV-2 wave and analysed longitudinally, encompassing regular PCR screening for infection, symptom reporting, imputation of HLAII genotype and analysis for antibody and T-cell responses to nucleoprotein (N) and spike (S). Of 272 (20%) HCW who seroconverted, the presence of HLA-DRB1*13:02 was associated with a 6·7-fold increased risk of case definition symptomatic COVID-19. In terms of immune responsiveness, HLA-DRB1*15:02 was associated with lower nucleocapsid T-cell responses. There was no association between DRB1 alleles and anti-spike antibody titres after two COVID vaccine doses. However, HLA DRB1*15:01 was associated with increased spike T-cell responses following both first and second dose vaccination. Trial registration: NCT04318314 and ISRCTN15677965.


Asunto(s)
COVID-19 , Anticuerpos Antivirales , COVID-19/genética , Vacunas contra la COVID-19 , Cadenas HLA-DRB1/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , SARS-CoV-2
6.
Science ; 375(6577): 183-192, 2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-34855510

RESUMEN

The impact of the initial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infecting strain on downstream immunity to heterologous variants of concern (VOCs) is unknown. Studying a longitudinal healthcare worker cohort, we found that after three antigen exposures (infection plus two vaccine doses), S1 antibody, memory B cells, and heterologous neutralization of B.1.351, P.1, and B.1.617.2 plateaued, whereas B.1.1.7 neutralization and spike T cell responses increased. Serology using the Wuhan Hu-1 spike receptor binding domain poorly predicted neutralizing immunity against VOCs. Neutralization potency against VOCs changed with heterologous virus encounter and number of antigen exposures. Neutralization potency fell differentially depending on targeted VOCs over the 5 months from the second vaccine dose. Heterologous combinations of spike encountered during infection and vaccination shape subsequent cross-protection against VOC, with implications for future-proof next-generation vaccines.


Asunto(s)
Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/virología , SARS-CoV-2/inmunología , Adulto , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Vacuna BNT162/administración & dosificación , Vacunas contra la COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Protección Cruzada , Femenino , Personal de Salud , Humanos , Estudios Longitudinales , Masculino , Células B de Memoria/inmunología , Mutación , Fosfoproteínas/inmunología , Dominios Proteicos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T/inmunología , Vacunación , Potencia de la Vacuna
7.
Ying Yong Sheng Tai Xue Bao ; 31(2): 407-416, 2020 Feb.
Artículo en Chino | MEDLINE | ID: mdl-32476332

RESUMEN

Given the importance of undergrowth vegetation to plantation ecosystem, this study analyzed the effects of three kinds of understory management measures, including understory preservation, understory removal, and interplanting, on the soil bacterial diversity, community structure and relative abundance under large diameter timber plantation of Cunninghamia lanceolata using high-throughput sequencing technology. The relationship between soil physical and chemical properties and bacterial community diversity were analyzed. The results showed that Chao1, Ace and Shannon indices of soil bacterial communities of understory preservation were higher than those of understory removal and interplanting. Actinobacteria, Acidobacteria and Chloroflexi were the dominant bacteria groups in the soil of C. lanceolata plantation. Compared with understory removal and interplanting, the relative abundance of Proteobacteria, Planctomycetes, Firmicutes and Verrucomicrobia in the soil of understory preservation was relatively high, while that of Actinobacteria, Acidobacteria and Chloroflexi was relatively low. There were significant differences in the relative abundance of Firmicutes, Planctomycetes, Verrucomicrobia, Parcubacteria and Actinobacteria among three understory management measures. The contents of moisture, total nitrogen, total phosphorus, hydrolyzed nitrogen and available phosphorus in the soil were important factors affecting soil bacterial community structure. Soil bacterial diversity indices had significant positive correlation with the contents of total nitrogen, total phosphorus, total potassium, hydrolyzed nitrogen and available potassium in the soil.


Asunto(s)
Cunninghamia , China , Ecosistema , Nitrógeno , Suelo , Microbiología del Suelo
8.
Front Cell Infect Microbiol ; 10: 578259, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33585265

RESUMEN

Human cytomegalovirus (HCMV) is an important pathogen in immunocompromised individuals and neonates, and a paradigm for viral immune evasion. We previously developed a quantitative proteomic approach that identified 133 proteins degraded during the early phase of HCMV infection, including known and novel antiviral factors. The majority were rescued from degradation by MG132, which is known to inhibit lysosomal cathepsins in addition to the proteasome. Global definition of the precise mechanisms of host protein degradation is important both to improve our understanding of viral biology, and to inform novel antiviral therapeutic strategies. We therefore developed and optimized a multiplexed comparative proteomic analysis using the selective proteasome inhibitor bortezomib in addition to MG132, to provide a global mechanistic view of protein degradation. Of proteins rescued from degradation by MG132, 34-47 proteins were also rescued by bortezomib, suggesting both that the predominant mechanism of protein degradation employed by HCMV is via the proteasome, and that alternative pathways for degradation are nevertheless important. Our approach and data will enable improved mechanistic understanding of HCMV and other viruses, and provide a shortlist of candidate restriction factors for further analysis.


Asunto(s)
Citomegalovirus , Proteómica , Humanos , Evasión Inmune , Recién Nacido , Proteínas , Proteolisis
9.
J Virol ; 94(4)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31776277

RESUMEN

The strongest evidence of the oncogenicity of Epstein-Barr virus (EBV) in vitro is its ability to immortalize human primary B lymphocytes into lymphoblastoid cell lines (LCLs). Yet the underlying mechanisms explaining how the virus tempers the growth program of the host cells have not been fully elucidated. The mitogen-activated protein kinases (MAPKs) are implicated in many cellular processes and are constitutively activated in LCLs. We questioned the expression and regulation of the dual-specificity phosphatases (DUSPs), the main negative regulator of MAPKs, during EBV infection and immortalization. Thirteen DUSPs, including 10 typical and 3 atypical types of DUSPs, were tested. Most of them were downregulated after EBV infection. Here, a role of viral oncogene latent membrane protein 1 (LMP1) in limiting DUSP6 and DUSP8 expression was identified. Using MAPK inhibitors, we found that LMP1 activates extracellular signal-regulated kinase (ERK) or p38 to repress the expression of DUSP6 and DUSP8, with corresponding substrate specificity. Morphologically, overexpression of DUSP6 and DUSP8 attenuates the ability of EBV-immortalized LCL cells to clump together. Mechanistically, apoptosis induced by restoring DUSP6 and DUSP8 in LCLs indicated a novel mechanism for LMP1 to provide a survival signal during EBV immortalization. Collectively, this report provides the first description of the interplay between EBV genes and DUSPs and contributes considerably to the interpretation of MAPK regulation in EBV immortalization.IMPORTANCE Infections by the ubiquitous Epstein-Barr virus (EBV) are associated with a wide spectrum of lymphomas and carcinomas. It has been well documented that activation levels of MAPKs are found in cancer cells to translate various external or intrinsic stimuli into cellular responses. Physiologically, the dual-specificity phosphates (DUSPs) exhibit great ability in regulating MAPK activities with respect to their capability of dephosphorylating MAPKs. In this study, we found that DUSPs were generally downregulated after EBV infection. EBV oncogenic latent membrane protein 1 (LMP1) suppressed DUSP6 and DUSP8 expression via MAPK pathway. In this way, LMP1-mediated MAPK activation was a continuous process. Furthermore, DUSP downregulation was found to contribute greatly to prevent apoptosis of EBV-infected cells. To sum up, this report sheds light on a novel molecular mechanism explaining how EBV maintains the unlimited proliferation status of the immortalized cells and provides a new link to understand EBV-induced B cell survival.


Asunto(s)
Fosfatasas de Especificidad Dual/genética , Herpesvirus Humano 4/metabolismo , Proteínas de la Matriz Viral/metabolismo , Apoptosis/genética , Linfocitos B/virología , Línea Celular Tumoral , Fosfatasas de Especificidad Dual/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genes Virales/genética , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Cultivo Primario de Células , Proteínas de la Matriz Viral/fisiología , Proteínas Virales/metabolismo , Latencia del Virus/genética , Latencia del Virus/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Ying Yong Sheng Tai Xue Bao ; 30(6): 1901-1910, 2019 Jun.
Artículo en Chino | MEDLINE | ID: mdl-31257762

RESUMEN

Taking evergreen broad-leaved forest in mid-subtropical areas, and its converted Phoebe bournei, Phyllostachys heterocycla and Cunninghamia lanceolata plantations as research objects, microbial biomass carbon (MBC) and nitrogen (MBN) in the surface (0-10 cm) and deep soil layer (40-60 cm) were measured by chloroform fumigation and extraction method, with their seasonal dynamics and the relationships with soil physicochemical properties in four types of forests being investigated. The results showed that the MBC and MBN in the surface soil was the highest in the evergreen broad-leaved forest, followed by P. bournei, P. heterocycla and C. lanceolata plantations, with that in the former three being significantly higher than in C. lanceolata plantion. There was no significant difference in the MBC and MBN contents in the deep soil layer among the four types of forests, while those in surface soil were significantly higher than in the deep soil layer. The MBC and MBN contents showed obvious seasonal dynamics, with highest values in summer and lowest in winter presenting a single peak change pattern. MBC and MBN had significantly positive correlations with soil total carbon (TC), total nitrogen (TN) and temperature, but significantly negative correlation with soil bulk density. The conversion of evergreen broad-leaved forest to the three plantation resulted in lower MBC and MBN in the surface soil to some degree, with C. lanceolata plantation being the first to be affected, but little change occurred in the deep soil layer. The quantity and quality of litter, contents of TC, TN and soil temperature were the key factors driving the differences of MBC and MBN contents and their seasonal dynamics of the four types of forests.


Asunto(s)
Carbono , Bosques , Nitrógeno , Microbiología del Suelo , Biomasa , China , Estaciones del Año , Suelo
11.
Cell Host Microbe ; 24(3): 447-460.e11, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30122656

RESUMEN

Human cytomegalovirus (HCMV) is an important pathogen with multiple immune evasion strategies, including virally facilitated degradation of host antiviral restriction factors. Here, we describe a multiplexed approach to discover proteins with innate immune function on the basis of active degradation by the proteasome or lysosome during early-phase HCMV infection. Using three orthogonal proteomic/transcriptomic screens to quantify protein degradation, with high confidence we identified 35 proteins enriched in antiviral restriction factors. A final screen employed a comprehensive panel of viral mutants to predict viral genes that target >250 human proteins. This approach revealed that helicase-like transcription factor (HLTF), a DNA helicase important in DNA repair, potently inhibits early viral gene expression but is rapidly degraded during infection. The functionally unknown HCMV protein UL145 facilitates HLTF degradation by recruiting the Cullin4 E3 ligase complex. Our approach and data will enable further identifications of innate pathways targeted by HCMV and other viruses.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Proteínas/química , Proteínas Virales/química , Citomegalovirus/genética , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Humanos , Evasión Inmune , Estabilidad Proteica , Proteínas/genética , Proteínas/inmunología , Proteómica , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
12.
Science ; 359(6371): 48-55, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29302006

RESUMEN

Plasmodium vivax shows a strict host tropism for reticulocytes. We identified transferrin receptor 1 (TfR1) as the receptor for P. vivax reticulocyte-binding protein 2b (PvRBP2b). We determined the structure of the N-terminal domain of PvRBP2b involved in red blood cell binding, elucidating the molecular basis for TfR1 recognition. We validated TfR1 as the biological target of PvRBP2b engagement by means of TfR1 expression knockdown analysis. TfR1 mutant cells deficient in PvRBP2b binding were refractory to invasion of P. vivax but not to invasion of P. falciparum Using Brazilian and Thai clinical isolates, we show that PvRBP2b monoclonal antibodies that inhibit reticulocyte binding also block P. vivax entry into reticulocytes. These data show that TfR1-PvRBP2b invasion pathway is critical for the recognition of reticulocytes during P. vivax invasion.


Asunto(s)
Antígenos CD/metabolismo , Malaria Vivax/metabolismo , Malaria Vivax/parasitología , Proteínas de la Membrana/química , Plasmodium vivax/patogenicidad , Proteínas Protozoarias/química , Receptores de Transferrina/metabolismo , Reticulocitos/parasitología , Antígenos CD/genética , Cristalografía por Rayos X , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Parásitos , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Plasmodium vivax/metabolismo , Dominios Proteicos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/ultraestructura , Receptores de Transferrina/genética
13.
Proc Natl Acad Sci U S A ; 114(44): E9356-E9365, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29078358

RESUMEN

During malaria blood-stage infections, Plasmodium parasites interact with the RBC surface to enable invasion followed by intracellular proliferation. Critical factors involved in invasion have been identified using biochemical and genetic approaches including specific knockdowns of genes of interest from primary CD34+ hematopoietic stem cells (cRBCs). Here we report the development of a robust in vitro culture system to produce RBCs that allow the generation of gene knockouts via CRISPR/Cas9 using the immortal JK-1 erythroleukemia line. JK-1 cells spontaneously differentiate, generating cells at different stages of erythropoiesis, including terminally differentiated nucleated RBCs that we term "jkRBCs." A screen of small-molecule epigenetic regulators identified several bromodomain-specific inhibitors that promote differentiation and enable production of synchronous populations of jkRBCs. Global surface proteomic profiling revealed that jkRBCs express all known Pfalciparum host receptors in a similar fashion to cRBCs and that multiple Pfalciparum strains invade jkRBCs at comparable levels to cRBCs and RBCs. Using CRISPR/Cas9, we deleted two host factors, basigin (BSG) and CD44, for which no natural nulls exist. BSG interacts with the parasite ligand Rh5, a prominent vaccine candidate. A BSG knockout was completely refractory to parasite invasion in a strain-transcendent manner, confirming the essential role for BSG during invasion. CD44 was recently identified in an RNAi screen of blood group genes as a host factor for invasion, and we show that CD44 knockout results in strain-transcendent reduction in invasion. Furthermore, we demonstrate a functional interaction between these two determinants in mediating Pfalciparum erythrocyte invasion.


Asunto(s)
Sistemas CRISPR-Cas/genética , Eritrocitos/metabolismo , Eritrocitos/parasitología , Plasmodium falciparum/genética , Antígenos de Protozoos/metabolismo , Basigina/metabolismo , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/fisiología , Epigénesis Genética/fisiología , Técnicas de Inactivación de Genes/métodos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/parasitología , Interacciones Huésped-Parásitos/fisiología , Humanos , Receptores de Hialuranos/metabolismo , Células K562 , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/parasitología , Ligandos , Malaria/parasitología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Proteómica/métodos , Proteínas Protozoarias/metabolismo
14.
Blood ; 128(12): 1578-89, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27338098

RESUMEN

Epstein-Barr virus (EBV), an oncogenic human virus, is associated with several lymphoproliferative disorders, including Burkitt lymphoma, Hodgkin disease, diffuse large B-cell lymphoma (DLBCL), and posttransplant lymphoproliferative disorder (PTLD). In vitro, EBV transforms primary B cells into lymphoblastoid cell lines (LCLs). Recently, several studies have shown that receptor tyrosine kinases (RTKs) play important roles in EBV-associated neoplasia. However, details of the involvement of RTKs in EBV-regulated B-cell neoplasia and malignancies remain largely unclear. Here, we found that erythropoietin-producing hepatocellular receptor A4 (EphA4), which belongs to the largest RTK Eph family, was downregulated in primary B cells post-EBV infection at the transcriptional and translational levels. Overexpression and knockdown experiments confirmed that EBV-encoded latent membrane protein 1 (LMP1) was responsible for this EphA4 suppression. Mechanistically, LMP1 triggered the extracellular signal-regulated kinase (ERK) pathway and promoted Sp1 to suppress EphA4 promoter activity. Functionally, overexpression of EphA4 prevented LCLs from proliferation. Pathologically, the expression of EphA4 was detected in EBV(-) tonsils but not in EBV(+) PTLD. In addition, an inverse correlation of EphA4 expression and EBV presence was verified by immunochemical staining of EBV(+) and EBV(-) DLBCL, suggesting EBV infection was associated with reduced EphA4 expression. Analysis of a public data set showed that lower EphA4 expression was correlated with a poor survival rate of DLBCL patients. Our findings provide a novel mechanism by which EphA4 can be regulated by an oncogenic LMP1 protein and explore its possible function in B cells. The results provide new insights into the role of EphA4 in EBV(+) PTLD and DLBCL.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Linfoma de Células B Grandes Difuso/mortalidad , Trastornos Linfoproliferativos/mortalidad , Receptor EphA4/metabolismo , Proteínas de la Matriz Viral/metabolismo , Células Cultivadas , Regulación hacia Abajo , Infecciones por Virus de Epstein-Barr/virología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Herpesvirus Humano 4 , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/virología , Trastornos Linfoproliferativos/metabolismo , Trastornos Linfoproliferativos/virología , Pronóstico , Receptor EphA4/genética , Transducción de Señal , Tasa de Supervivencia , Proteínas de la Matriz Viral/genética
15.
J Virol ; 89(11): 5968-80, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25810549

RESUMEN

UNLABELLED: Epstein-Barr virus (EBV), an oncogenic herpesvirus, has the potential to immortalize primary B cells into lymphoblastoid cell lines (LCLs) in vitro. During immortalization, several EBV products induce cytokines or chemokines, and most of these are required for the proliferation of LCLs. Interleukin-32 (IL-32), a recently discovered proinflammatory cytokine, is upregulated after EBV infection, and this upregulation is detectable in all LCLs tested. EBV latent membrane protein 1 (LMP1) is responsible for inducing IL-32 expression at the mRNA and protein levels. Mechanistically, we showed that this LMP1 induction is provided by the p65 subunit of NF-κB, which binds to and activates the IL-32 promoter. Furthermore, the short hairpin RNA (shRNA)-mediated depletion of endogenous LMP1 and p65 in LCLs suppressed IL-32 expression, further suggesting that LMP1 is the key factor that stimulates IL-32 in LCLs via the NF-κB p65 pathway. Functionally, knockdown of IL-32 in LCLs elicits viral reactivation and affects cytokine expression, but it has no impact on cell proliferation and apoptosis. Of note, we reveal the mechanism whereby IL-32 is involved in the maintenance of EBV viral latency by inactivation of Zta promoter activity. This atypical cytoplasmic IL-32 hijacks the Zta activator protein kinase Cδ (PKCδ) and inhibits its translocation from the cytoplasm to the nucleus, where PKCδ binds to the Zta promoter and activates lytic cycle progression. These novel findings reveal that IL-32 is involved in the maintenance of EBV latency in LCLs. This finding may provide new information to explain how EBV maintains latency, in addition to viral chromatin structure and epigenetic modification. IMPORTANCE: EBV persists in two states, latency and lytic replication, which is a unique characteristic of human infections. So far, little is known about how herpesviruses maintain latency in particular tissues or cell types. EBV is an excellent model to study this question because more than 90% of people are latently infected. EBV can immortalize primary B cells into lymphoblastoid cell lines in vitro. Expression of IL-32, a novel atypical cytoplasmic proinflammatory cytokine, increased after infection. The expression of IL-32 was controlled by LMP1. In investigating the regulatory mechanism, we demonstrated that the p65 subunit of NF-κB is required for this upregulation. Of note, the important biological activity of IL-32 was to trap protein kinase Cδ in the cytoplasm and prevent it from binding to the Zta promoter, which is the key event for EBV reaction. So, the expression of LMP1-induced IL-32 plays a role in the maintenance of EBV latency.


Asunto(s)
Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Interleucinas/biosíntesis , Proteína Quinasa C-delta/metabolismo , Proteínas de la Matriz Viral/metabolismo , Latencia del Virus , Linfocitos B/virología , Células Cultivadas , Herpesviridae , Humanos , Factor de Transcripción ReIA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...