Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Front Immunol ; 14: 1162211, 2023.
Artículo en Inglés | MEDLINE | ID: covidwho-20231099

RESUMEN

Spatiotemporal separation of cellular components is vital to ensure biochemical processes. Membrane-bound organelles such as mitochondria and nuclei play a major role in isolating intracellular components, while membraneless organelles (MLOs) are accumulatively uncovered via liquid-liquid phase separation (LLPS) to mediate cellular spatiotemporal organization. MLOs orchestrate various key cellular processes, including protein localization, supramolecular assembly, gene expression, and signal transduction. During viral infection, LLPS not only participates in viral replication but also contributes to host antiviral immune responses. Therefore, a more comprehensive understanding of the roles of LLPS in virus infection may open up new avenues for treating viral infectious diseases. In this review, we focus on the antiviral defense mechanisms of LLPS in innate immunity and discuss the involvement of LLPS during viral replication and immune evasion escape, as well as the strategy of targeting LLPS to treat viral infectious diseases.


Asunto(s)
Antivirales , Núcleo Celular , Inmunidad
2.
Cells ; 12(1)2022 12 23.
Artículo en Inglés | MEDLINE | ID: covidwho-2199809

RESUMEN

The year 2021 marked the 10th anniversary of the publication of Cells [...].


Asunto(s)
Aniversarios y Eventos Especiales , Núcleo Celular
3.
J Virol ; 96(22): e0099722, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: covidwho-2097918

RESUMEN

Modification of the hepatitis C virus (HCV) positive-strand RNA genome by N6-methyladenosine (m6A) regulates the viral life cycle. This life cycle takes place solely in the cytoplasm, while m6A addition on cellular mRNA takes place in the nucleus. Thus, the mechanisms by which m6A is deposited on the viral RNA have been unclear. In this work, we find that m6A modification of HCV RNA by the m6A-methyltransferase proteins methyltransferase-like 3 and 14 (METTL3 and METTL14) is regulated by Wilms' tumor 1-associating protein (WTAP). WTAP, a predominantly nuclear protein, is an essential member of the cellular mRNA m6A-methyltransferase complex and known to target METTL3 to mRNA. We found that HCV infection induces localization of WTAP to the cytoplasm. Importantly, we found that WTAP is required for both METTL3 interaction with HCV RNA and m6A modification across the viral RNA genome. Further, we found that WTAP, like METTL3 and METTL14, negatively regulates the production of infectious HCV virions, a process that we have previously shown is regulated by m6A. Excitingly, WTAP regulation of both HCV RNA m6A modification and virion production was independent of its ability to localize to the nucleus. Together, these results reveal that WTAP is critical for HCV RNA m6A modification by METTL3 and METTL14 in the cytoplasm. IMPORTANCE Positive-strand RNA viruses such as HCV represent a significant global health burden. Previous work has described that HCV RNA contains the RNA modification m6A and how this modification regulates viral infection. Yet, how this modification is targeted to HCV RNA has remained unclear due to the incompatibility of the nuclear cellular processes that drive m6A modification with the cytoplasmic HCV life cycle. In this study, we present evidence for how m6A modification is targeted to HCV RNA in the cytoplasm by a mechanism in which WTAP recruits the m6A-methyltransferase METTL3 to HCV RNA. This targeting strategy for m6A modification of cytoplasmic RNA viruses is likely relevant for other m6A-modified positive-strand RNA viruses with cytoplasmic life cycles such as enterovirus 71 and SARS-CoV-2 and provides an exciting new target for potential antiviral therapies.


Asunto(s)
Proteínas de Ciclo Celular , Hepatitis C , Metiltransferasas , Factores de Empalme de ARN , Humanos , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatitis C/genética , Hepatitis C/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Mensajero/genética , ARN Viral/genética , ARN Viral/metabolismo
4.
J Virol ; 96(17): e0077222, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1992939

RESUMEN

Bats are reservoirs for diverse coronaviruses, including swine acute diarrhea syndrome coronavirus (SADS-CoV). SADS-CoV was first identified in diarrheal piglets in 2017. As a novel alphacoronavirus, SADS-CoV shares ~95% identity with bat alphacoronavirus HKU2. SADS-CoV has been reported to have broad cell tropism and inherent potential to cross host species barriers for dissemination. Thus far, no effective antiviral drugs or vaccines are available to treat infections with SADS-CoV. Therefore, knowledge of the protein-coding gene set and a subcellular localization map of SADS-CoV proteins are fundamental first steps in this endeavor. Here, all SADS-CoV genes were cloned separately into Flag-tagged plasmids, and the subcellular localizations of viral proteins, with the exception of nsp11, were detected using confocal microscopy techniques. As a result, nsp1, nsp3-N, nsp4, nsp5, nsp7, nsp8, nsp9, nsp10, nsp14, and nsp15 were localized in the cytoplasm and nuclear spaces, and these viral proteins may perform specific functions in the nucleus. All structural and accessory proteins were mainly localized in the cytoplasm. NS7a and membrane protein M colocalized with the Golgi compartment, and they may regulate the assembly of SADS-CoV virions. Maturation of SADS-CoV may occur in the late endosomes, during which envelope protein E is involved in the assembly and release of the virus. In summary, the present study demonstrates for the first time the location of all the viral proteins of SADS-CoV. These fundamental studies of SADS-CoV will promote studies of basic virology of SADS-CoV and support preventive strategies for animals with infection of SADS-CoV. IMPORTANCE SADS-CoV is the first documented spillover of a bat coronavirus that causes severe diseases in domestic animals. Our study is an in-depth annotation of the newly discovered swine coronavirus SADS-CoV genome and viral protein expression. Systematic subcellular localization of SADS-CoV proteins can have dramatic significance in revealing viral protein biological functions in the subcellular locations. Furthermore, our study promote understanding the fundamental science behind the novel swine coronavirus to pave the way for treatments and cures.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Enfermedades de los Porcinos , Proteínas Virales , Alphacoronavirus/genética , Animales , Núcleo Celular/virología , Quirópteros , Infecciones por Coronavirus/veterinaria , Endosomas/virología , Aparato de Golgi/virología , Porcinos , Enfermedades de los Porcinos/virología , Proteínas Virales/genética
5.
Genome Biol ; 23(1): 55, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1785167

RESUMEN

BACKGROUND: Multiplexing of samples in single-cell RNA-seq studies allows a significant reduction of the experimental costs, straightforward identification of doublets, increased cell throughput, and reduction of sample-specific batch effects. Recently published multiplexing techniques using oligo-conjugated antibodies or -lipids allow barcoding sample-specific cells, a process called "hashing." RESULTS: Here, we compare the hashing performance of TotalSeq-A and -C antibodies, custom synthesized lipids and MULTI-seq lipid hashes in four cell lines, both for single-cell RNA-seq and single-nucleus RNA-seq. We also compare TotalSeq-B antibodies with CellPlex reagents (10x Genomics) on human PBMCs and TotalSeq-B with different lipids on primary mouse tissues. Hashing efficiency was evaluated using the intrinsic genetic variation of the cell lines and mouse strains. Antibody hashing was further evaluated on clinical samples using PBMCs from healthy and SARS-CoV-2 infected patients, where we demonstrate a more affordable approach for large single-cell sequencing clinical studies, while simultaneously reducing batch effects. CONCLUSIONS: Benchmarking of different hashing strategies and computational pipelines indicates that correct demultiplexing can be achieved with both lipid- and antibody-hashed human cells and nuclei, with MULTISeqDemux as the preferred demultiplexing function and antibody-based hashing as the most efficient protocol on cells. On nuclei datasets, lipid hashing delivers the best results. Lipid hashing also outperforms antibodies on cells isolated from mouse brain. However, antibodies demonstrate better results on tissues like spleen or lung.


Asunto(s)
COVID-19/sangre , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Anticuerpos/química , Estudios de Casos y Controles , Línea Celular Tumoral , Núcleo Celular/química , Humanos , Lípidos/química , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/química , Neutrófilos/inmunología , Neutrófilos/virología
6.
Life Sci Alliance ; 5(5)2022 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1675573

RESUMEN

Acute kidney injury is associated with mortality in COVID-19 patients. However, host cell changes underlying infection of renal cells with SARS-CoV-2 remain unknown and prevent understanding of the molecular mechanisms that may contribute to renal pathology. Here, we carried out quantitative translatome and whole-cell proteomics analyses of primary renal proximal and distal tubular epithelial cells derived from human donors infected with SARS-CoV-2 or MERS-CoV to disseminate virus and cell type-specific changes over time. Our findings revealed shared pathways modified upon infection with both viruses, as well as SARS-CoV-2-specific host cell modulation driving key changes in innate immune activation and cellular protein quality control. Notably, MERS-CoV infection-induced specific changes in mitochondrial biology that were not observed in response to SARS-CoV-2 infection. Furthermore, we identified extensive modulation in pathways associated with kidney failure that changed in a virus- and cell type-specific manner. In summary, we provide an overview of the effects of SARS-CoV-2 or MERS-CoV infection on primary renal epithelial cells revealing key pathways that may be essential for viral replication.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/virología , Riñón , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Proteoma , Proteómica , SARS-CoV-2/fisiología , Biomarcadores , COVID-19/metabolismo , COVID-19/virología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Biología Computacional/métodos , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Túbulos Renales Distales , Túbulos Renales Proximales , Mitocondrias/genética , Mitocondrias/metabolismo , Cultivo Primario de Células , Proteómica/métodos , Replicación Viral
7.
Nat Commun ; 12(1): 4502, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: covidwho-1550282

RESUMEN

Cells in many tissues, such as bone, muscle, and placenta, fuse into syncytia to acquire new functions and transcriptional programs. While it is known that fused cells are specialized, it is unclear whether cell-fusion itself contributes to programmatic-changes that generate the new cellular state. Here, we address this by employing a fusogen-mediated, cell-fusion system to create syncytia from undifferentiated cells. RNA-Seq analysis reveals VSV-G-induced cell fusion precedes transcriptional changes. To gain mechanistic insights, we measure the plasma membrane surface area after cell-fusion and observe it diminishes through increases in endocytosis. Consequently, glucose transporters internalize, and cytoplasmic glucose and ATP transiently decrease. This reduced energetic state activates AMPK, which inhibits YAP1, causing transcriptional-reprogramming and cell-cycle arrest. Impairing either endocytosis or AMPK activity prevents YAP1 inhibition and cell-cycle arrest after fusion. Together, these data demonstrate plasma membrane diminishment upon cell-fusion causes transient nutrient stress that may promote transcriptional-reprogramming independent from extrinsic cues.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Transporte Biológico , Fusión Celular , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Células Gigantes/metabolismo , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Ratones , RNA-Seq/métodos , Transducción de Señal/genética , Factores de Transcripción/genética , Proteínas del Envoltorio Viral/genética , Proteínas Señalizadoras YAP
8.
J Cell Physiol ; 237(2): 1521-1531, 2022 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1490820

RESUMEN

Mechanical forces can modulate the immune response, mostly described as promoting the activation of immune cells, but the role and mechanism of pathological levels of mechanical stress in lymphocyte activation have not been focused on before. By an ex vivo experimental approach, we observed that mechanical stressing of murine spleen lymphocytes with 50 mmHg for 3 h induced the nuclear localization of NFAT1, increased C-Jun, and increased the expression of early activation marker CD69 in resting CD8+ cells. Interestingly, 50 mmHg mechanical stressing induced the nuclear localization of NFAT1; but conversely decreased C-Jun and inhibited the expression of CD69 in lymphocytes under lipopolysaccharide or phorbol 12-myristate 13-acetate/ionomycin stimulation. Additionally, we observed similar changes trends when comparing RNA-seq data of hypertensive and normotensive COVID-19 patients. Our results indicate a biphasic effect of mechanical stress on lymphocyte activation, which provides insight into the variety of immune responses in pathologies involving elevated mechanical stress.


Asunto(s)
Activación de Linfocitos/inmunología , Estrés Mecánico , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , COVID-19/complicaciones , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Comorbilidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Canales Iónicos/metabolismo , Lectinas Tipo C/metabolismo , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Masculino , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología
11.
PLoS One ; 16(6): e0253089, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1282298

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a devastating global pandemic, infecting over 43 million people and claiming over 1 million lives, with these numbers increasing daily. Therefore, there is urgent need to understand the molecular mechanisms governing SARS-CoV-2 pathogenesis, immune evasion, and disease progression. Here, we show that SARS-CoV-2 can block IRF3 and NF-κB activation early during virus infection. We also identify that the SARS-CoV-2 viral proteins NSP1 and NSP13 can block interferon activation via distinct mechanisms. NSP1 antagonizes interferon signaling by suppressing host mRNA translation, while NSP13 downregulates interferon and NF-κB promoter signaling by limiting TBK1 and IRF3 activation, as phospho-TBK1 and phospho-IRF3 protein levels are reduced with increasing levels of NSP13 protein expression. NSP13 can also reduce NF-κB activation by both limiting NF-κB phosphorylation and nuclear translocation. Last, we also show that NSP13 binds to TBK1 and downregulates IFIT1 protein expression. Collectively, these data illustrate that SARS-CoV-2 bypasses multiple innate immune activation pathways through distinct mechanisms.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , COVID-19/inmunología , Núcleo Celular/inmunología , Factor 3 Regulador del Interferón/inmunología , Proteínas de Unión al ARN/inmunología , SARS-CoV-2/inmunología , Transducción de Señal/inmunología , Proteínas no Estructurales Virales/inmunología , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , COVID-19/genética , Núcleo Celular/genética , Células HeLa , Humanos , Factor 3 Regulador del Interferón/genética , FN-kappa B/genética , FN-kappa B/inmunología , Fosforilación/genética , Fosforilación/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas de Unión al ARN/genética , SARS-CoV-2/genética , Transducción de Señal/genética , Proteínas no Estructurales Virales/genética
12.
Nature ; 595(7868): 565-571, 2021 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1275939

RESUMEN

Although SARS-CoV-2 primarily targets the respiratory system, patients with and survivors of COVID-19 can suffer neurological symptoms1-3. However, an unbiased understanding of the cellular and molecular processes that are affected in the brains of patients with COVID-19 is missing. Here we profile 65,309 single-nucleus transcriptomes from 30 frontal cortex and choroid plexus samples across 14 control individuals (including 1 patient with terminal influenza) and 8 patients with COVID-19. Although our systematic analysis yields no molecular traces of SARS-CoV-2 in the brain, we observe broad cellular perturbations indicating that barrier cells of the choroid plexus sense and relay peripheral inflammation into the brain and show that peripheral T cells infiltrate the parenchyma. We discover microglia and astrocyte subpopulations associated with COVID-19 that share features with pathological cell states that have previously been reported in human neurodegenerative disease4-6. Synaptic signalling of upper-layer excitatory neurons-which are evolutionarily expanded in humans7 and linked to cognitive function8-is preferentially affected in COVID-19. Across cell types, perturbations associated with COVID-19 overlap with those found in chronic brain disorders and reside in genetic variants associated with cognition, schizophrenia and depression. Our findings and public dataset provide a molecular framework to understand current observations of COVID-19-related neurological disease, and any such disease that may emerge at a later date.


Asunto(s)
Astrocitos/patología , Encéfalo/patología , COVID-19/diagnóstico , COVID-19/patología , Plexo Coroideo/patología , Microglía/patología , Neuronas/patología , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Encéfalo/fisiopatología , Encéfalo/virología , COVID-19/genética , COVID-19/fisiopatología , Núcleo Celular/genética , Plexo Coroideo/metabolismo , Plexo Coroideo/fisiopatología , Plexo Coroideo/virología , Femenino , Humanos , Inflamación/virología , Masculino , Persona de Mediana Edad , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/patogenicidad , Análisis de la Célula Individual , Transcriptoma , Replicación Viral
13.
Pharmacol Res Perspect ; 9(4): e00798, 2021 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1269136

RESUMEN

Therapeutic regimens for the COVID-19 pandemics remain unmet. In this line, repurposing of existing drugs against known or predicted SARS-CoV-2 protein actions have been advanced, while natural products have also been tested. Here, we propose that p-cymene, a natural monoterpene, can act as a potential novel agent for the treatment of SARS-CoV-2-induced COVID-19 and other RNA-virus-induced diseases (influenza, rabies, Ebola). We show by extensive molecular simulations that SARS-CoV-2 C-terminal structured domain contains a nuclear localization signal (NLS), like SARS-CoV, on which p-cymene binds with low micromolar affinity, impairing nuclear translocation of this protein and inhibiting viral replication, as verified by preliminary in vitro experiments. A similar mechanism may occur in other RNA-viruses (influenza, rabies and Ebola), also verified in vitro for influenza, by interaction of p-cymene with viral nucleoproteins, and structural modification of their NLS site, weakening its interaction with importin A. This common mechanism of action renders therefore p-cymene as a possible antiviral, alone, or in combination with other agents, in a broad spectrum of RNA viruses, from SARS-CoV-2 to influenza A infections.


Asunto(s)
Antivirales/farmacología , Cimenos/farmacología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Proteínas de la Nucleocápside/metabolismo , SARS-CoV-2/fisiología , Animales , Antivirales/química , Núcleo Celular/metabolismo , Núcleo Celular/virología , Chlorocebus aethiops , Cimenos/química , Perros , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Células de Riñón Canino Madin Darby , Modelos Moleculares , Simulación de Dinámica Molecular , Señales de Localización Nuclear , Proteínas de la Nucleocápside/química , Conformación Proteica , Dominios Proteicos , Transporte de Proteínas , SARS-CoV-2/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
14.
Cells ; 10(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1259432

RESUMEN

The host nucleocytoplasmic trafficking system is often hijacked by viruses to accomplish their replication and to suppress the host immune response. Viruses encode many factors that interact with the host nuclear transport receptors (NTRs) and the nucleoporins of the nuclear pore complex (NPC) to access the host nucleus. In this review, we discuss the viral factors and the host factors involved in the nuclear import and export of viral components. As nucleocytoplasmic shuttling is vital for the replication of many viruses, we also review several drugs that target the host nuclear transport machinery and discuss their feasibility for use in antiviral treatment.


Asunto(s)
Núcleo Celular/metabolismo , Núcleo Celular/virología , SARS-CoV-2/fisiología , Fenómenos Fisiológicos de los Virus , Replicación Viral/fisiología , Transporte Activo de Núcleo Celular/fisiología , COVID-19/metabolismo , COVID-19/virología , Interacciones Huésped-Patógeno/fisiología , Humanos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Internalización del Virus , Virus/patogenicidad
16.
Int J Biol Sci ; 17(6): 1547-1554, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1206441

RESUMEN

Suppression of type I interferon (IFN) response is one pathological outcome of the infection of highly pathogenic human coronaviruses. To effect this, severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2 encode multiple IFN antagonists. In this study, we reported on the IFN antagonism of SARS-CoV-2 main protease NSP5. NSP5 proteins of both SARS-CoV and SARS-CoV-2 counteracted Sendai virus-induced IFN production. NSP5 variants G15S and K90R commonly seen in circulating strains of SARS-CoV-2 retained the IFN-antagonizing property. The suppressive effect of NSP5 on IFN-ß gene transcription induced by RIG-I, MAVS, TBK1 and IKKϵ suggested that NSP5 likely acts at a step downstream of IRF3 phosphorylation in the cytoplasm. NSP5 did not influence steady-state expression or phosphorylation of IRF3, suggesting that IRF3, regardless of its phosphorylation state, might not be the substrate of NSP5 protease. However, nuclear translocation of phosphorylated IRF3 was severely compromised in NSP5-expressing cells. Taken together, our work revealed a new mechanism by which NSP5 proteins encoded by SARS-CoV and SARS-CoV-2 antagonize IFN production by retaining phosphorylated IRF3 in the cytoplasm. Our findings have implications in rational design and development of antiviral agents against SARS-CoV-2.


Asunto(s)
Núcleo Celular/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/biosíntesis , SARS-CoV-2/enzimología , Animales , COVID-19/virología , Chlorocebus aethiops , Humanos , Fosforilación , Transporte de Proteínas , Células Vero
17.
mBio ; 12(2)2021 04 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1183285

RESUMEN

RNA viruses that replicate in the cytoplasm often disrupt nucleocytoplasmic transport to preferentially translate their own transcripts and prevent host antiviral responses. The Sarbecovirus accessory protein ORF6 has previously been shown to be a major inhibitor of interferon production in both severe acute respiratory syndrome coronavirus (SARS-CoV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we show SARS-CoV-2-infected cells display an elevated level of nuclear mRNA accumulation compared to mock-infected cells. We demonstrate that ORF6 is responsible for this nuclear imprisonment of host mRNA, and using a cotransfected reporter assay, we show this nuclear retention of mRNA blocks expression of newly transcribed mRNAs. ORF6's nuclear entrapment of host mRNA is associated with its ability to copurify with the mRNA export factors, Rae1 and Nup98. These protein-protein interactions map to the C terminus of ORF6 and can be abolished by a single amino acid mutation in Met58. Overexpression of Rae1 restores reporter expression in the presence of SARS-CoV-2 ORF6. SARS-CoV ORF6 also interacts with Rae1 and Nup98. However, SARS-CoV-2 ORF6 more strongly copurifies with Rae1 and Nup98 and results in significantly reduced expression of reporter proteins compared to SARS-CoV ORF6, a potential mechanism for the delayed symptom onset and presymptomatic transmission uniquely associated with the SARS-CoV-2 pandemic. We also show that both SARS-CoV and SARS-CoV-2 ORF6 block nuclear import of a broad range of host proteins. Together, these data support a model in which ORF6 clogs the nuclear pore through its interactions with Rae1 and Nup98 to prevent both nuclear import and export, rendering host cells incapable of responding to SARS-CoV-2 infection.IMPORTANCE SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19), is an RNA virus with a large genome that encodes multiple accessory proteins. While these accessory proteins are not required for growth in vitro, they can contribute to the pathogenicity of the virus. We demonstrate that SARS-CoV-2-infected cells accumulate poly(A) mRNA in the nucleus, which is attributed to the accessory protein ORF6. Nuclear entrapment of mRNA and reduced expression of newly transcribed reporter proteins are associated with ORF6's interactions with the mRNA export proteins Rae1 and Nup98. SARS-CoV ORF6 also shows the same interactions with Rae1 and Nup98. However, SARS-CoV-2 ORF6 more strongly represses reporter expression and copurifies with Rae1 and Nup98 compared to SARS-CoV ORF6. Both SARS-CoV ORF6 and SARS-CoV-2 ORF6 block nuclear import of a wide range of host factors through interactions with Rae1 and Nup98. Together, our results suggest ORF6's disruption of nucleocytoplasmic transport prevents infected cells from responding to the invading virus.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , SARS-CoV-2/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Sitios de Unión , COVID-19/metabolismo , COVID-19/virología , Línea Celular , Regulación de la Expresión Génica , Humanos , Mutación , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Unión Proteica , ARN Mensajero/metabolismo , SARS-CoV-2/genética , Proteínas Virales/química , Proteínas Virales/genética
18.
Cell Mol Immunol ; 18(4): 945-953, 2021 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1104474

RESUMEN

SARS-CoV-2 is the pathogenic agent of COVID-19, which has evolved into a global pandemic. Compared with some other respiratory RNA viruses, SARS-CoV-2 is a poor inducer of type I interferon (IFN). Here, we report that SARS-CoV-2 nsp12, the viral RNA-dependent RNA polymerase (RdRp), suppresses host antiviral responses. SARS-CoV-2 nsp12 attenuated Sendai virus (SeV)- or poly(I:C)-induced IFN-ß promoter activation in a dose-dependent manner. It also inhibited IFN promoter activation triggered by RIG-I, MDA5, MAVS, and IRF3 overexpression. Nsp12 did not impair IRF3 phosphorylation but suppressed the nuclear translocation of IRF3. Mutational analyses suggested that this suppression was not dependent on the polymerase activity of nsp12. Given these findings, our study reveals that SARS-CoV-2 RdRp can antagonize host antiviral innate immunity and thus provides insights into viral pathogenesis.


Asunto(s)
COVID-19/metabolismo , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , SARS-CoV-2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Núcleo Celular/metabolismo , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Interferón Tipo I/genética , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Mutación , Fosforilación , Regiones Promotoras Genéticas , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , SARS-CoV-2/enzimología , Virus Sendai/metabolismo
19.
PLoS Pathog ; 17(2): e1009207, 2021 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1063225

RESUMEN

The recent Coronavirus Disease 2019 pandemic has once again reminded us the importance of understanding infectious diseases. One important but understudied area in infectious disease research is the role of nuclear architecture or the physical arrangement of the genome in the nucleus in controlling gene regulation and pathogenicity. Recent advances in research methods, such as Genome-wide chromosome conformation capture using high-throughput sequencing (Hi-C), have allowed for easier analysis of nuclear architecture and chromosomal reorganization in both the infectious disease agents themselves as well as in their host cells. This review will discuss broadly on what is known about nuclear architecture in infectious disease, with an emphasis on chromosomal reorganization, and briefly discuss what steps are required next in the field.


Asunto(s)
Núcleo Celular/genética , Cromatina/metabolismo , Enfermedades Transmisibles/genética , Animales , COVID-19/genética , COVID-19/metabolismo , Núcleo Celular/metabolismo , Cromatina/genética , Cromosomas/genética , Cromosomas/metabolismo , Enfermedades Transmisibles/metabolismo , Regulación de la Expresión Génica , Humanos
20.
Biochem Biophys Res Commun ; 536: 59-66, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: covidwho-971357

RESUMEN

The novel human betacoronavirus SARS-CoV-2 has caused an unprecedented pandemic in the 21st century. Several studies have revealed interactions between SARS-CoV-2 viral proteins and host nucleoporins, yet their functions are largely unknown. Here, we demonstrate that the open-reading frame 6 (ORF6) of SARS-CoV-2 can directly manipulate localization and functions of nucleoporins. We found that ORF6 protein disrupted nuclear rim staining of nucleoporins RAE1 and NUP98. Consequently, this disruption caused aberrant nucleocytoplasmic trafficking and led to nuclear accumulation of mRNA transporters such as hnRNPA1. Ultimately, host cell nucleus size was reduced and cell growth was halted.


Asunto(s)
Tamaño del Núcleo Celular , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/virología , Células HEK293 , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Humanos , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA