Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Annu Rev Immunol ; 42(1): 551-584, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38941604

RESUMO

Poxviruses have evolved a wide array of mechanisms to evade the immune response, and we provide an overview of the different immunomodulatory strategies. Poxviruses prevent the recognition of viral DNA that triggers the immune responses and inhibit signaling pathways within the infected cell. A unique feature of poxviruses is the production of secreted proteins that mimic cytokines and cytokine receptors, acting as decoy receptors to neutralize the activity of cytokines and chemokines. The capacity of these proteins to evade cellular immune responses by inhibiting cytokine activation is complemented by poxviruses' strategies to block natural killer cells and cytotoxic T cells, often through interfering with antigen presentation pathways. Mechanisms that target complement activation are also encoded by poxviruses. Virus-encoded proteins that target immune molecules and pathways play a major role in immune modulation, and their contribution to viral pathogenesis, facilitating virus replication or preventing immunopathology, is discussed.


Assuntos
Evasão da Resposta Imune , Infecções por Poxviridae , Poxviridae , Humanos , Poxviridae/imunologia , Poxviridae/fisiologia , Animais , Infecções por Poxviridae/imunologia , Citocinas/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Proteínas Virais/imunologia , Apresentação de Antígeno/imunologia , Interações Hospedeiro-Patógeno/imunologia
2.
PLoS Pathog ; 19(1): e1011136, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36716344

RESUMO

African swine fever virus (ASFV) causes a devastating hemorrhagic disease with worldwide circulation and no widely available therapeutic prevention. The infectious particle has a multilayered architecture that is articulated upon an endoplasmic reticulum (ER)-derived inner envelope. This membrane acts as docking platform for the assembly of the outer icosahedral capsid and the underlying core shell, a bridging layer required for the formation of the central genome-containing nucleoid. While the details of outer capsid assembly are relatively well understood, those of core formation remain unclear. Here we report the functional characterization of pEP84R, a transmembrane polypeptide embedded in the inner envelope that surrounds the viral core. Using an ASFV recombinant inducibly expressing the EP84R gene, we show that absence of pEP84R results in the formation of non-infectious core-less icosahedral particles displaying a significant DNA-packaging defect. Concomitantly, aberrant core shell-like structures formed by co-assembly of viral polyproteins pp220 and pp62 are mistargeted to non-ER membranes, as also occurs when these are co-expressed in the absence of other viral proteins. Interestingly, co-expression of both polyproteins with pEP84R led to the formation of ER-targeted core shell-like assemblies and co-immunoprecipitation assays showed that pEP84R binds to the N-terminal region of pp220. Altogether, these results indicate that pEP84R plays a crucial role in core assembly by targeting the core shell polyproteins to the inner viral envelope, which enables subsequent genome packaging and nucleoid formation. These findings unveil a key regulatory mechanism for ASFV morphogenesis and identify a relevant novel target for the development of therapeutic tools against this re-emerging threat.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Animais , Suínos , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/metabolismo , Montagem de Vírus , Proteínas Virais/genética , Proteínas Virais/metabolismo , Poliproteínas/metabolismo , Proteínas de Membrana
3.
PLoS Pathog ; 12(4): e1005595, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27110717

RESUMO

African swine fever virus (ASFV) is a nucleocytoplasmic large DNA virus (NCLDV) that causes a highly lethal disease in domestic pigs. As other NCLDVs, the extracellular form of ASFV possesses a multilayered structure consisting of a genome-containing nucleoid successively wrapped by a thick protein core shell, an inner lipid membrane, an icosahedral protein capsid and an outer lipid envelope. This structural complexity suggests an intricate mechanism of internalization in order to deliver the virus genome into the cytoplasm. By using flow cytometry in combination with pharmacological entry inhibitors, as well as fluorescence and electron microscopy approaches, we have dissected the entry and uncoating pathway used by ASFV to infect the macrophage, its natural host cell. We found that purified extracellular ASFV is internalized by both constitutive macropinocytosis and clathrin-mediated endocytosis. Once inside the cell, ASFV particles move from early endosomes or macropinosomes to late, multivesicular endosomes where they become uncoated. Virus uncoating requires acidic pH and involves the disruption of the outer membrane as well as of the protein capsid. As a consequence, the inner viral membrane becomes exposed and fuses with the limiting endosomal membrane to release the viral core into the cytosol. Interestingly, virus fusion is dependent on virus protein pE248R, a transmembrane polypeptide of the inner envelope that shares sequence similarity with some members of the poxviral entry/fusion complex. Collective evidence supports an entry model for ASFV that might also explain the uncoating of other multienveloped icosahedral NCLDVs.


Assuntos
Vírus da Febre Suína Africana/patogenicidade , Febre Suína Africana/virologia , Internalização do Vírus , Desenvelopamento do Vírus/fisiologia , Animais , Western Blotting , Capsídeo/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Endocitose , Endossomos/ultraestrutura , Endossomos/virologia , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Microscopia Eletrônica , Microscopia de Fluorescência , Corpos Multivesiculares/metabolismo , Corpos Multivesiculares/ultraestrutura , Suínos , Proteínas do Envelope Viral/metabolismo
4.
J Virol ; 86(3): 1758-67, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22114329

RESUMO

The integrity of the cholesterol biosynthesis pathway is required for efficient African swine fever virus (ASFV) infection. Incorporation of prenyl groups into Rho GTPases plays a key role in several stages of ASFV infection, since both geranylgeranyl and farnesyl pyrophosphates are required at different infection steps. We found that Rho GTPase inhibition impaired virus morphogenesis and resulted in an abnormal viral factory size with the accumulation of envelope precursors and immature virions. Furthermore, abundant defective virions reached the plasma membrane, and filopodia formation in exocytosis was abrogated. Rac1 was activated at early ASFV infection stages, coincident with microtubule acetylation, a process that stabilizes microtubules for virus transport. Rac1 inhibition did not affect the viral entry step itself but impaired subsequent virus production. We found that specific Rac1 inhibition impaired viral induced microtubule acetylation and viral intracellular transport. These findings highlight that viral infection is the result of a carefully orchestrated modulation of Rho family GTPase activity within the host cell; this modulation results critical for virus morphogenesis and in turn, triggers cytoskeleton remodeling, such as microtubule stabilization for viral transport during early infection.


Assuntos
Vírus da Febre Suína Africana/patogenicidade , Febre Suína Africana/metabolismo , Colesterol/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Chlorocebus aethiops , Suínos , Células Vero
5.
Methods Mol Biol ; 2597: 121-129, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36374418

RESUMO

Viruses encode secreted proteins that bind chemokines to modulate their activity. Viral proteins may simultaneously interact with glycosaminoglycans allowing these proteins to be anchored at the cell surface to increase their anti-chemokine activity in the proximity of infection. Here we describe methodology to evaluate the interaction of viral secreted proteins with cell-surface glycosaminoglycans by immunofluorescence and detection by flow cytometry or microscopy. These methods could be equally applied to other chemokine binding proteins that do not have viral origin.


Assuntos
Proteínas de Transporte , Glicosaminoglicanos , Glicosaminoglicanos/metabolismo , Proteínas de Transporte/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Citometria de Fluxo , Quimiocinas/metabolismo , Ligação Proteica , Proteínas Virais/metabolismo
6.
Water Res ; 231: 119621, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36693290

RESUMO

Besides nasopharyngeal swabs, monkeypox virus (MPXV) DNA has been detected in a variety of samples such as saliva, semen, urine and fecal samples. Using the environmental surveillance network previously developed in Spain for the routine wastewater surveillance of SARS-CoV-2 (VATar COVID-19), we have analyzed the presence of MPXV DNA in wastewater from different areas of Spain. Samples (n = 312) from 24 different wastewater treatment plants were obtained between May 9 (week 19 of 2022) and August 4 (week 31 of 2022). Following concentration of viral particles by a validated aluminum adsorption-precipitation method, a qPCR procedure allowed us to detect MPXV DNA in 56 wastewater samples collected from May 16 to August 4, 2022, with values ranging between 2.2 × 103 to 8.7 × 104 genome copies (gc)/L. This study shows that MPXV DNA can be reproducibly detected by qPCR in longitudinal samples collected from different Spanish wastewater treatment plants. According to data from the National Epidemiological Surveillance Network (RENAVE) in Spain a total of 6,119 cases have been confirmed as of August 19, 2022. However, and based on the wastewater data, the reported clinical cases seem to be underestimated and asymptomatic infections may be more frequent than expected.


Assuntos
COVID-19 , Monkeypox virus , Humanos , SARS-CoV-2 , Águas Residuárias , Vigilância Epidemiológica Baseada em Águas Residuárias , DNA , RNA Viral
7.
Lancet Microbe ; 4(1): e21-e28, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36436538

RESUMO

BACKGROUND: The transmission of monkeypox virus occurs through direct contact, but transmission through saliva or exhaled droplets and aerosols has not yet been investigated. We aimed to assess the presence of monkeypox virus DNA and infectious virus in saliva samples and droplets and aerosols exhaled from patients infected with monkeypox virus. METHODS: We did a cross-sectional study in patients with monkeypox confirmed by PCR who attended two health centres in Madrid, Spain. For each patient, we collected samples of saliva, exhaled droplets within a mask, and aerosols captured by air filtration through newly developed nanofiber filters. We evaluated the presence of monkeypox virus in the samples by viral DNA detection by quantitative PCR (qPCR) and isolation of infectious viruses in cell cultures. FINDINGS: Between May 18 and July 15, 2022, 44 patients with symptomatic monkeypox attended two health centres in Madrid and were included in the study. All were cisgender men, with a median age of 35·0 years (IQR 11·3). We identified high loads of monkeypox virus DNA by qPCR in 35 (85%) of 41 saliva samples. Infectious monkeypox virus was recovered from 22 (67%) of 33 saliva samples positive for monkeypox virus DNA. We also found a significant association between the number of affected cutaneous areas or general symptoms and the viral load present in saliva samples. Droplets exhaled from patients with monkeypox, detected inside a mask, contained monkeypox virus DNA in 32 (71%) of 45 samples, with two of the 32 positive samples showing the presence of the infectious virus. Monkeypox virus DNA in aerosols, collected from the medical consultation room, were detected in 27 (64%) of 42 samples, despite patients wearing an FFP2 mask during the visit. Infectious virus was not recovered from aerosol samples. High levels of monkeypox virus DNA were identified in aerosols collected from a hospital isolation room housing a patient with monkeypox. INTERPRETATION: The identification of high viable monkeypox virus loads in saliva in most patients with monkeypox and the finding of monkeypox virus DNA in droplets and aerosols warrants further epidemiological studies to evaluate the potential relevance of the respiratory route of infection in the 2022 monkeypox virus outbreak. FUNDING: EU, Consejo Superior de Investigaciones Científicas, and Ciberinfec.


Assuntos
Monkeypox virus , Mpox , Masculino , Humanos , Criança , Monkeypox virus/genética , Mpox/diagnóstico , Estudos Transversais , Saliva , Espanha/epidemiologia , Aerossóis , DNA
8.
Artigo em Inglês | MEDLINE | ID: mdl-36612897

RESUMO

The COVID-19 pandemic highlighted the dangers of airborne pathogen transmission. SARS-CoV-2 is known to be transmitted through aerosols; however, little is known about the dynamics of these aerosols in real environments, the conditions, and the minimum viral load required for infection. Efficiently measuring and capturing pathogens present in the air would help to understand the infection process. Air samplers usually take several hours to obtain an air sample. In this work a fast (1-2 min) method for capturing bioaerosols into a liquid medium has been tested in hospital rooms with COVID-19 patients. This fast sampling allows detecting transient levels of aerosols in the air. SARS-CoV-2 RNA is detected in aerosols from several hospital rooms at different levels. Interestingly, there are sudden boosts of the SARS-CoV-2 load in the air, suggesting that SARS-CoV-2 could be released abundantly at certain moments. These results show that the distribution of SARS-CoV-2-containing aerosols is not homogeneous in the hospital room. This technology is a fast and effective tool for capturing airborne matter in a very short time, which allows for fast decision-making any kind of hazard in the air is detected. It is also useful for a better understanding of aerosols dynamics.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Pandemias , RNA Viral , Aerossóis e Gotículas Respiratórios , Hospitais
9.
J Virol ; 84(4): 2100-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19939916

RESUMO

African swine fever virus (ASFV) is a large DNA virus that enters host cells after receptor-mediated endocytosis and depends on acidic cellular compartments for productive infection. The exact cellular mechanism, however, is largely unknown. In order to dissect ASFV entry, we have analyzed the major endocytic routes using specific inhibitors and dominant negative mutants and analyzed the consequences for ASFV entry into host cells. Our results indicate that ASFV entry into host cells takes place by clathrin-mediated endocytosis which requires dynamin GTPase activity. Also, the clathrin-coated pit component Eps15 was identified as a relevant cellular factor during infection. The presence of cholesterol in cellular membranes, but not lipid rafts or caveolae, was found to be essential for a productive ASFV infection. In contrast, inhibitors of the Na(+)/H(+) ion channels and actin polymerization inhibition did not significantly modify ASFV infection, suggesting that macropinocytosis does not represent the main entry route for ASFV. These results suggest a dynamin-dependent and clathrin-mediated endocytic pathway of ASFV entry for the cell types and viral strains analyzed.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Vírus da Febre Suína Africana/patogenicidade , Clatrina/fisiologia , Dinaminas/fisiologia , Internalização do Vírus , Actinas/antagonistas & inibidores , Vírus da Febre Suína Africana/efeitos dos fármacos , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Cavéolas/efeitos dos fármacos , Cavéolas/fisiologia , Linhagem Celular , Chlorocebus aethiops , Clorpromazina/farmacologia , Colesterol/metabolismo , Dinaminas/antagonistas & inibidores , Dinaminas/genética , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Canais Iônicos/antagonistas & inibidores , Mutação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Suínos , Transfecção , Transferrina/metabolismo , Células Vero , Internalização do Vírus/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia
10.
J Virol ; 84(20): 10792-801, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20686048

RESUMO

Several viruses target the microtubular motor system in early stages of the viral life cycle. African swine fever virus (ASFV) protein p54 hijacks the microtubule-dependent transport by interaction with a dynein light chain (DYNLL1/DLC8). This was shown to be a high-affinity interaction, and the residues gradually disappearing were mapped on DLC8 to define a putative p54 binding surface by nuclear magnetic resonance (NMR) spectroscopy. The potential of short peptides targeting the binding domain to disrupt this high-affinity protein-protein interaction was assayed, and a short peptide sequence was shown to bind and compete with viral protein binding to dynein. Given the complexity and number of proteins involved in cellular transport, the prevention of this viral-DLC8 interaction might not be relevant for successful viral infection. Thus, we tested the capacity of these peptides to interfere with viral infection by disrupting dynein interaction with viral p54. Using this approach, we report on short peptides that inhibit viral growth.


Assuntos
Vírus da Febre Suína Africana/efeitos dos fármacos , Antivirais/farmacologia , Dineínas/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Estruturais Virais/efeitos dos fármacos , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/patogenicidade , Vírus da Febre Suína Africana/fisiologia , Sequência de Aminoácidos , Animais , Antivirais/química , Ligação Competitiva , Chlorocebus aethiops , Dineínas/química , Dineínas/genética , Dineínas/fisiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Técnicas In Vitro , Modelos Moleculares , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/efeitos dos fármacos , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/fisiologia , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Peptídeos/genética , Domínios e Motivos de Interação entre Proteínas , Homologia de Sequência de Aminoácidos , Sus scrofa , Células Vero , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/fisiologia
11.
Pathogens ; 10(8)2021 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-34451529

RESUMO

Tumour necrosis factor (TNF) is an inflammatory cytokine produced in response to viral infections that promotes the recruitment and activation of leukocytes to sites of infection. This TNF-based host response is essential to limit virus spreading, thus poxviruses have evolutionarily adopted diverse molecular mechanisms to counteract TNF antiviral action. These include the expression of poxvirus-encoded soluble receptors or proteins able to bind and neutralize TNF and other members of the TNF ligand superfamily, acting as decoy receptors. This article reviews in detail the various TNF decoy receptors identified to date in the genomes from different poxvirus species, with a special focus on their impact on poxvirus pathogenesis and their potential use as therapeutic molecules.

12.
Curr Opin Immunol ; 66: 50-56, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32408109

RESUMO

Poxviruses and herpesviruses encode secreted versions of cytokine receptors as a unique strategy to evade the host immune response. Recent advances in the field have shown the great impact of some of these proteins in immune modulation and viral pathogenesis, and have uncovered unique properties of these viral proteins not found in the cellular counterparts. These modifications inspired by viruses lead to improved immune modulatory activity of the soluble cytokine receptors, information that has been used to develop more efficient therapeutics to treat inflammatory conditions.


Assuntos
Citocinas/imunologia , Herpesviridae/imunologia , Poxviridae/imunologia , Proteínas Virais/imunologia , Animais , Humanos
13.
mBio ; 11(4)2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32788374

RESUMO

African swine fever virus (ASFV) is a complex nucleocytoplasmic large DNA virus (NCLDV) causing a lethal hemorrhagic disease that currently threatens the global pig industry. Despite its relevance in the infectious cycle, very little is known about the internalization of ASFV in the host cell. Here, we report the characterization of ASFV protein pE199L, a cysteine-rich structural polypeptide with similarity to proteins A16, G9, and J5 of the entry fusion complex (EFC) of poxviruses. Using biochemical and immunomicroscopic approaches, we found that, like the corresponding poxviral proteins, pE199L localizes to the inner viral envelope and behaves as an integral transmembrane polypeptide with cytosolic intramolecular disulfide bonds. Using an ASFV recombinant that inducibly expresses the E199L gene, we found that protein pE199L is not required for virus assembly and egress or for virus-cell binding and endocytosis but is required for membrane fusion and core penetration. Interestingly, similar results have been previously reported for ASFV protein pE248R, an inner membrane virion component related to the poxviral L1 and F9 EFC proteins. Taken together, these findings indicate that ASFV entry relies on a form of fusion machinery comprising proteins pE248R and pE199L that displays some similarities to the unconventional fusion apparatus of poxviruses. Also, these results provide novel targets for the development of strategies that block the first stages of ASFV replication.IMPORTANCE African swine fever virus (ASFV) causes a highly lethal swine disease that is currently present in many countries of Eastern Europe, the Russian Federation, and Southeast Asia, severely affecting the pig industry. Despite extensive research, effective vaccines or antiviral strategies are still lacking and relevant gaps in knowledge of the fundamental biology of the viral infection cycle exist. In this study, we identified pE199L, a protein of the inner viral membrane that is required for virus entry. More specifically, pE199L is necessary for the fusion event that leads to the penetration of the genome-containing core in the host cell. Our results significantly increase our knowledge of the process of internalization of African swine fever virus, which may instruct future research on antiviral strategies.


Assuntos
Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/fisiologia , Fusão de Membrana , Proteínas Virais/metabolismo , Internalização do Vírus , Vírus da Febre Suína Africana/metabolismo , Animais , Chlorocebus aethiops , Endocitose , Suínos , Células Vero , Proteínas Virais/genética
14.
Sci Adv ; 6(38)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32948585

RESUMO

Cells contain numerous immune sensors to detect virus infection. The cyclic GMP-AMP (cGAMP) synthase (cGAS) recognizes cytosolic DNA and activates innate immune responses via stimulator of interferon genes (STING), but the impact of DNA sensing pathways on host protective responses has not been fully defined. We demonstrate that cGAS/STING activation is required to resist lethal poxvirus infection. We identified viral Schlafen (vSlfn) as the main STING inhibitor, and ectromelia virus was severely attenuated in the absence of vSlfn. Both vSlfn-mediated virulence and STING inhibitory activity were mapped to the recently discovered poxin cGAMP nuclease domain. Animals were protected from subcutaneous, respiratory, and intravenous infection in the absence of vSlfn, and interferon was the main antiviral protective mechanism controlled by the DNA sensing pathway. Our findings support the idea that manipulation of DNA sensing is an efficient therapeutic strategy in diseases triggered by viral infection or tissue damage-mediated release of self-DNA.


Assuntos
Proteínas de Membrana , Viroses , Animais , DNA , Interferons , Proteínas de Membrana/metabolismo , Nucleotídeos Cíclicos , Nucleotidiltransferases/metabolismo
15.
J Clin Med ; 9(4)2020 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32244308

RESUMO

Soluble receptors of cytokines are able to modify cytokine activities and therefore the immune system, and some have intrinsic biological activities without mediation from their cytokines. The soluble interferon beta (IFN-ß) receptor is generated through alternative splicing of IFNAR2 and has both agonist and antagonist properties for IFN-ß, but its role is unknown. We previously demonstrated that a recombinant human soluble IFN-ß receptor showed intrinsic therapeutic efficacy in a mouse model of multiple sclerosis. Here we evaluate the potential biological activities of recombinant sIFNAR2 without the mediation of IFN-ß in human cells. Recombinant sIFNAR2 down-regulated the production of IL-17 and IFN-É£ and reduced the cell proliferation rate. Moreover, it showed a strong antiviral activity, fully protecting the cell monolayer after being infected by the virus. Specific inhibitors completely abrogated the antiviral activity of IFN-ß, but not that of the recombinant sIFNAR2, and there was no activation of the JAK-STAT signaling pathway. Consequently, r-sIFNAR2 exerts immunomodulatory, antiproliferative and antiviral activities without IFN-ß mediation, and could be a promising treatment against viral infections and immune-mediated diseases.

16.
Science ; 368(6497): 1371-1376, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32439659

RESUMO

The effect of immunometabolism on age-associated diseases remains uncertain. In this work, we show that T cells with dysfunctional mitochondria owing to mitochondrial transcription factor A (TFAM) deficiency act as accelerators of senescence. In mice, these cells instigate multiple aging-related features, including metabolic, cognitive, physical, and cardiovascular alterations, which together result in premature death. T cell metabolic failure induces the accumulation of circulating cytokines, which resembles the chronic inflammation that is characteristic of aging ("inflammaging"). This cytokine storm itself acts as a systemic inducer of senescence. Blocking tumor necrosis factor-α signaling or preventing senescence with nicotinamide adenine dinucleotide precursors partially rescues premature aging in mice with Tfam-deficient T cells. Thus, T cells can regulate organismal fitness and life span, which highlights the importance of tight immunometabolic control in both aging and the onset of age-associated diseases.


Assuntos
Senilidade Prematura/imunologia , Proteínas de Ligação a DNA/deficiência , Mitocôndrias/metabolismo , Proteínas Mitocondriais/deficiência , Multimorbidade , Linfócitos T/metabolismo , Fatores de Transcrição/deficiência , Senilidade Prematura/genética , Senilidade Prematura/prevenção & controle , Animais , Síndrome da Liberação de Citocina/imunologia , Proteínas de Ligação a DNA/genética , Feminino , Deleção de Genes , Inflamação/genética , Inflamação/imunologia , Longevidade , Masculino , Camundongos , Camundongos Mutantes , Proteínas Mitocondriais/genética , NAD/administração & dosagem , NAD/farmacologia , Aptidão Física , Linfócitos T/ultraestrutura , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores
17.
Nat Commun ; 11(1): 4938, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33009401

RESUMO

Antiviral strategies to inhibit Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) and the pathogenic consequences of COVID-19 are urgently required. Here, we demonstrate that the NRF2 antioxidant gene expression pathway is suppressed in biopsies obtained from COVID-19 patients. Further, we uncover that NRF2 agonists 4-octyl-itaconate (4-OI) and the clinically approved dimethyl fumarate (DMF) induce a cellular antiviral program that potently inhibits replication of SARS-CoV2 across cell lines. The inhibitory effect of 4-OI and DMF extends to the replication of several other pathogenic viruses including Herpes Simplex Virus-1 and-2, Vaccinia virus, and Zika virus through a type I interferon (IFN)-independent mechanism. In addition, 4-OI and DMF limit host inflammatory responses to SARS-CoV2 infection associated with airway COVID-19 pathology. In conclusion, NRF2 agonists 4-OI and DMF induce a distinct IFN-independent antiviral program that is broadly effective in limiting virus replication and in suppressing the pro-inflammatory responses of human pathogenic viruses, including SARS-CoV2.


Assuntos
Anti-Inflamatórios/farmacologia , Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Fumarato de Dimetilo/agonistas , Fator 2 Relacionado a NF-E2/metabolismo , Pneumonia Viral/tratamento farmacológico , Succinatos/agonistas , Adulto , Antioxidantes/farmacologia , Betacoronavirus/metabolismo , COVID-19 , Infecções por Coronavirus/virologia , Fumarato de Dimetilo/farmacologia , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Interferon Tipo I , Pulmão/patologia , Masculino , Fator 2 Relacionado a NF-E2/genética , Pandemias , Pneumonia Viral/virologia , SARS-CoV-2 , Transdução de Sinais/efeitos dos fármacos , Succinatos/farmacologia , Replicação Viral/efeitos dos fármacos
19.
FEBS Lett ; 582(23-24): 3275-80, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18775702

RESUMO

Heterogeneous nuclear ribonucleoprotein K (hnRNP-K) was identified as interacting cellular protein with the abundant immediate early protein p30 from African swine fever virus (ASFV) in a macrophage cDNA library screening. The interacting regions of hnRNP-K with p30 were established within residues 35-197, which represent KH1 and KH2 domains responsible for RNA binding. Colocalization of hnRNP-K and p30 was observed mainly in the nucleus, but not in the cytoplasm of infected cells and infection modified hnRNP-K subcellular distribution and decreased the incorporation of 5-fluorouridine into nascent RNA. Since similar effects were observed in cells transiently expressing p30, this interaction provides new insights into p30 function and could represent a possible additional mechanism by which ASFV downregulates host cell mRNA translation.


Assuntos
Febre Suína Africana/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Animais , Núcleo Celular/metabolismo , Chlorocebus aethiops , Biblioteca Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Macrófagos/metabolismo , Fosfoproteínas/genética , Biossíntese de Proteínas , RNA Mensageiro/genética , Suínos/metabolismo , Suínos/virologia , Técnicas do Sistema de Duplo-Híbrido , Células Vero , Proteínas Virais/genética
20.
F1000Res ; 72018.
Artigo em Inglês | MEDLINE | ID: mdl-29946427

RESUMO

Poxviruses encode a set of secreted proteins that bind cytokines and chemokines as a strategy to modulate host defense mechanisms. These viral proteins mimic the activity of host cytokine decoy receptors but have unique properties that may enhance their activity. Here, we describe the ability of poxvirus cytokine receptors to attach to the cell surface after secretion from infected cells, and we discuss the advantages that this property may confer to these viral immunomodulatory proteins.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA