Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nature ; 599(7884): 283-289, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34517409

RESUMEN

Derailed cytokine and immune cell networks account for the organ damage and the clinical severity of COVID-19 (refs. 1-4). Here we show that SARS-CoV-2, like other viruses, evokes cellular senescence as a primary stress response in infected cells. Virus-induced senescence (VIS) is indistinguishable from other forms of cellular senescence and is accompanied by a senescence-associated secretory phenotype (SASP), which comprises pro-inflammatory cytokines, extracellular-matrix-active factors and pro-coagulatory mediators5-7. Patients with COVID-19 displayed markers of senescence in their airway mucosa in situ and increased serum levels of SASP factors. In vitro assays demonstrated macrophage activation with SASP-reminiscent secretion, complement lysis and SASP-amplifying secondary senescence of endothelial cells, which mirrored hallmark features of COVID-19 such as macrophage and neutrophil infiltration, endothelial damage and widespread thrombosis in affected lung tissue1,8,9. Moreover, supernatant from VIS cells, including SARS-CoV-2-induced senescence, induced neutrophil extracellular trap formation and activation of platelets and the clotting cascade. Senolytics such as navitoclax and a combination of dasatinib plus quercetin selectively eliminated VIS cells, mitigated COVID-19-reminiscent lung disease and reduced inflammation in SARS-CoV-2-infected hamsters and mice. Our findings mark VIS as a pathogenic trigger of COVID-19-related cytokine escalation and organ damage, and suggest that senolytic targeting of virus-infected cells is a treatment option against SARS-CoV-2 and perhaps other viral infections.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/patología , COVID-19/virología , Senescencia Celular/efectos de los fármacos , Terapia Molecular Dirigida , SARS-CoV-2/patogenicidad , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Animales , COVID-19/complicaciones , Línea Celular , Cricetinae , Dasatinib/farmacología , Dasatinib/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Quercetina/farmacología , Quercetina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Trombosis/complicaciones , Trombosis/inmunología , Trombosis/metabolismo
2.
J Virol ; 96(1): e0151121, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34668780

RESUMEN

The development of mouse models for coronavirus disease 2019 (COVID-19) has enabled testing of vaccines and therapeutics and defining aspects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis. SARS-CoV-2 disease is severe in K18 transgenic mice (K18-hACE2 Tg) expressing human angiotensin-converting enzyme 2 (hACE2), the SARS-CoV-2 receptor, under an ectopic cytokeratin promoter, with high levels of infection measured in the lung and brain. Here, we evaluated SARS-CoV-2 infection in hACE2 knock-in (KI) mice that express hACE2 under an endogenous promoter in place of murine ACE2 (mACE2). Intranasal inoculation of hACE2 KI mice with SARS-CoV-2 WA1/2020 resulted in substantial viral replication within the upper and lower respiratory tracts with limited spread to extrapulmonary organs. However, SARS-CoV-2-infected hACE2 KI mice did not lose weight and developed limited pathology. Moreover, no significant differences in viral burden were observed in hACE2 KI mice infected with B.1.1.7 or B.1.351 variants compared to the WA1/2020 strain. Because the entry mechanisms of SARS-CoV-2 in mice remain uncertain, we evaluated the impact of the naturally occurring, mouse-adapting N501Y mutation by comparing infection of hACE2 KI, K18-hACE2 Tg, ACE2-deficient, and wild-type C57BL/6 mice. The N501Y mutation minimally affected SARS-CoV-2 infection in hACE2 KI mice but was required for viral replication in wild-type C57BL/6 mice in a mACE2-dependent manner and augmented pathogenesis in the K18-hACE2 Tg mice. Thus, the N501Y mutation likely enhances interactions with mACE2 or hACE2 in vivo. Overall, our study highlights the hACE2 KI mice as a model of mild SARS-CoV-2 infection and disease and clarifies the requirement of the N501Y mutation in mice. IMPORTANCE Mouse models of SARS-CoV-2 pathogenesis have facilitated the rapid evaluation of countermeasures. While the first generation of models developed pneumonia and severe disease after SARS-CoV-2 infection, they relied on ectopic expression of supraphysiological levels of human ACE2 (hACE2). This has raised issues with their relevance to humans, as the hACE2 receptor shows a more restricted expression pattern in the respiratory tract. Here, we evaluated SARS-CoV-2 infection and disease with viruses containing or lacking a key mouse-adapting mutation in the spike gene in hACE2 KI mice, which express hACE2 under an endogenous promoter in place of murine ACE2. While infection of hACE2 KI mice with multiple strains of SARS-CoV-2 including variants of concern resulted in viral replication within the upper and lower respiratory tracts, the animals did not sustain severe lung injury. Thus, hACE2 KI mice serve as a model of mild infection with both ancestral and emerging SARS-CoV-2 variant strains.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/virología , Pulmón/virología , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/patología , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Inflamación , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , Mutación , SARS-CoV-2/genética , Carga Viral , Replicación Viral
3.
Proc Natl Acad Sci U S A ; 117(45): 28344-28354, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33097660

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic that is a serious global health problem. Evasion of IFN-mediated antiviral signaling is a common defense strategy that pathogenic viruses use to replicate and propagate in their host. In this study, we show that SARS-CoV-2 is able to efficiently block STAT1 and STAT2 nuclear translocation in order to impair transcriptional induction of IFN-stimulated genes (ISGs). Our results demonstrate that the viral accessory protein Orf6 exerts this anti-IFN activity. We found that SARS-CoV-2 Orf6 localizes at the nuclear pore complex (NPC) and directly interacts with Nup98-Rae1 via its C-terminal domain to impair docking of cargo-receptor (karyopherin/importin) complex and disrupt nuclear import. In addition, we show that a methionine-to-arginine substitution at residue 58 impairs Orf6 binding to the Nup98-Rae1 complex and abolishes its IFN antagonistic function. All together our data unravel a mechanism of viral antagonism in which a virus hijacks the Nup98-Rae1 complex to overcome the antiviral action of IFN.


Asunto(s)
COVID-19/metabolismo , Interferones/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Animales , Sitios de Unión , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Unión Proteica , Transducción de Señal , Células Vero
4.
J Immunol ; 191(2): 875-83, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23749635

RESUMEN

Four of the five members of the Toll/IL-1R domain-containing adaptor family are required for signaling downstream of TLRs, promoting innate immune responses against different pathogens. However, the role of the fifth member of this family, sterile α and Toll/IL-1R domain-containing 1 (SARM), is unclear. SARM is expressed primarily in the CNS where it is required for axonal death. Studies in Caenorhabditis elegans have also shown a role for SARM in innate immunity. To clarify the role of mammalian SARM in innate immunity, we infected SARM(-/-) mice with a number of bacterial and viral pathogens. SARM(-/-) mice show normal responses to Listeria monocytogenes, Mycobacterium tuberculosis, and influenza virus, but show dramatic protection from death after CNS infection with vesicular stomatitis virus. Protection correlates with reduced CNS injury and cytokine production by nonhematopoietic cells, suggesting that SARM is a positive regulator of cytokine production. Neurons and microglia are the predominant source of cytokines in vivo, supporting a role for SARM as a link between neuronal injury and innate immunity.


Asunto(s)
Proteínas del Dominio Armadillo/metabolismo , Enfermedades Virales del Sistema Nervioso Central/inmunología , Proteínas del Citoesqueleto/metabolismo , Infecciones por Rhabdoviridae/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/inmunología , Células de la Médula Ósea , Células Cultivadas , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/virología , Enfermedades Virales del Sistema Nervioso Central/metabolismo , Citocinas/biosíntesis , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/inmunología , Inmunidad Innata , Virus de la Influenza A/inmunología , Listeria monocytogenes/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Mycobacterium tuberculosis/inmunología , Neuronas/metabolismo , Infecciones por Rhabdoviridae/metabolismo
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167193, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38648902

RESUMEN

SARS-CoV-2 infection can cause severe pneumonia, wherein exacerbated inflammation plays a major role. This is reminiscent of the process commonly termed cytokine storm, a condition dependent on a disproportionated production of cytokines. This state involves the activation of the innate immune response by viral patterns and coincides with the biosynthesis of the biomass required for viral replication, which may overwhelm the capacity of the endoplasmic reticulum and drive the unfolded protein response (UPR). The UPR is a signal transduction pathway composed of three branches that is initiated by a set of sensors: inositol-requiring protein 1 (IRE1), protein kinase RNA-like ER kinase (PERK), and activating transcription factor 6 (ATF6). These sensors control adaptive processes, including the transcriptional regulation of proinflammatory cytokines. Based on this background, the role of the UPR in SARS-CoV-2 replication and the ensuing inflammatory response was investigated using in vivo and in vitro models of infection. Mice and Syrian hamsters infected with SARS-CoV-2 showed a sole activation of the Ire1α-Xbp1 arm of the UPR associated with a robust production of proinflammatory cytokines. Human lung epithelial cells showed the dependence of viral replication on the expression of UPR-target proteins branching on the IRE1α-XBP1 arm and to a lower extent on the PERK route. Likewise, activation of the IRE1α-XBP1 branch by Spike (S) proteins from different variants of concern was a uniform finding. These results show that the IRE1α-XBP1 system enhances viral replication and cytokine expression and may represent a potential therapeutic target in SARS-CoV-2 severe pneumonia.


Asunto(s)
COVID-19 , Endorribonucleasas , Proteínas Serina-Treonina Quinasas , SARS-CoV-2 , Respuesta de Proteína Desplegada , Replicación Viral , Proteína 1 de Unión a la X-Box , Animales , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Proteína 1 de Unión a la X-Box/metabolismo , Proteína 1 de Unión a la X-Box/genética , SARS-CoV-2/metabolismo , Humanos , COVID-19/metabolismo , COVID-19/virología , COVID-19/patología , COVID-19/inmunología , Ratones , Mesocricetus , Transducción de Señal , Ratones Endogámicos C57BL , Citocinas/metabolismo , Femenino
6.
Proc Natl Acad Sci U S A ; 106(15): 6262-7, 2009 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-19332776

RESUMEN

In autoimmune prone murine strains, sequential engagement of the B cell antigen receptor (BCR) on the cell surface and toll-like receptors (TLRs) in late endosomes is necessary and sufficient for secretion of autoantibodies. However, ubiquitous nucleoprotein self-antigens fail to elicit productive TLR activation, and break self-tolerance in anergic DNA-reactive B cells. The mechanisms limiting TLR activation in these cells are largely unknown. Here, we demonstrate that in anergic 3H9/Vkappa8 and Ars/A1 B cells the normal endocytic transit of both the ligated BCR and TLR9 into late endosomes is abrogated. The BCR and TLR9 arrest together just outside late endosomes, indicating that they enter this compartment along a single, regulated endocytic route. Access to late endosomes could be restored by reversing anergy through several methods, including conferring genetic susceptibility to autoimmunity, complementing proximal BCR signaling or by preventing BCR binding to self-antigen. Downstream of the BCR, JNK, which is activated in naive but not anergic B cells, regulated entry into late endosomes. Restoration of BCR and TLR9 endocytic trafficking rescued TLR9 activation by BCR-captured ligands. These results indicate that B cell anergy is reinforced by the exclusion of both TLRs and their BCR captured ligands from subcellular environments necessary for TLR activation.


Asunto(s)
Linfocitos B/inmunología , Anergia Clonal/inmunología , Endocitosis/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Receptor Toll-Like 9/inmunología , Animales , Anticuerpos Antinucleares/genética , Anticuerpos Antinucleares/inmunología , Anticuerpos Antinucleares/metabolismo , Anticuerpos Antifosfolípidos/genética , Anticuerpos Antifosfolípidos/inmunología , Anticuerpos Antifosfolípidos/metabolismo , Antígenos Ly/genética , Antígenos Ly/inmunología , Antígenos Ly/metabolismo , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Bazo/inmunología , Factores de Tiempo , Ubiquitinación
7.
Eur J Immunol ; 40(10): 2692-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20809520

RESUMEN

Crosslinking of Fc γ receptor II B (FcγRIIB) and the BCR by immune complexes (IC) can downregulate antigen-specific B-cell responses. Accordingly, FcγRIIB deficiencies have been associated with B-cell hyperactivity in patients with systemic lupus erythematosus and mouse models of lupus. However, we have previously shown that murine IgG2a-autoreactive AM14 B cells respond robustly to chromatin-associated IC through a mechanism dependent on both the BCR and the endosomal TLR9, despite FcγRIIB coexpression. To further evaluate the potential contribution of FcγRIIB to the regulation of autoreactive B cells, we have now compared the IC-triggered responses of FcγRIIB-deficient and FcγRIIB-sufficient AM14 B cells. We find that FcγRIIB-deficient cells respond significantly better than FcγRIIB-sufficient cells when stimulated with DNA IC that incorporate low-affinity TLR9 ligand (CG-poor dsDNA fragments). AM14 B cells also respond to RNA-associated IC through BCR/TLR7 coengagement, but such BCR/TLR7-dependent responses are normally highly dependent on IFN-α costimulation. However, we now show that AM14 FcγRIIB(-/-) B cells are very effectively activated by RNA IC without supplemental IFN-α priming. These results demonstrate that FcγRIIB can effectively modulate both BCR/TLR9 and BCR/TLR7 endosomal-dependent activation of autoreactive B cells.


Asunto(s)
Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de IgG/inmunología , Receptor Toll-Like 9/inmunología , Animales , Complejo Antígeno-Anticuerpo/inmunología , Autoantígenos/inmunología , Autoinmunidad/inmunología , Comunicación Celular/inmunología , Línea Celular , Activación de Linfocitos , Ratones , Ratones Noqueados , Ratones Transgénicos , Organismos Libres de Patógenos Específicos
8.
J Immunol ; 183(5): 3109-17, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19648272

RESUMEN

Although TLR9 was originally thought to specifically recognize microbial DNA, it is now evident that mammalian DNA can be an effective TLR9 ligand. However, the DNA sequence required for TLR9 activation is controversial, as studies have shown conflicting results depending on the nature of the DNA backbone, the route of DNA uptake, and the cell type being studied. In systemic lupus erythematosus, a major route whereby DNA gains access to intracellular TLR9, and thereby activates dendritic cells (DCs), is through uptake as a DNA-containing immune complex. In this report, we used defined dsDNA fragments with a natural (phosphodiester) backbone and show that unmethylated CpG dinucleotides within dsDNA are required for murine DC TLR9 activation induced by a DNA-containing immune complex. The strongest activation is seen with dsDNA fragments containing optimal CpG motifs (purine-purine-CpG-pyrimidine-pyrimidine) that are common in microbial DNA but rare in mammalian DNA. Importantly, however, activation can also be induced by CpG-rich DNA fragments that lack these optimal CpG motifs and that we show are plentiful in CpG islands within mammalian DNA. No activation is induced by DNA fragments lacking CpG dinucleotides, although this CpG-free DNA can induce DC activation if internalized by liposomal transfection instead of as an immune complex. Overall, the data suggest that the release of CpG-rich DNA from mammalian DNA may contribute to the pathogenesis of autoimmune diseases such as systemic lupus erythematosus and psoriasis in which activation of TLR9 in DCs by self DNA has been implicated in disease pathogenesis.


Asunto(s)
Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/genética , Islas de CpG/inmunología , ADN/química , ADN/fisiología , Células Dendríticas/inmunología , Oligonucleótidos Fosforotioatos/fisiología , Receptor Toll-Like 9/fisiología , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/fisiología , Animales , Complejo Antígeno-Anticuerpo/fisiología , Células Cultivadas , Islas de CpG/genética , ADN/metabolismo , Fragmentación del ADN , Metilación de ADN/inmunología , Células Dendríticas/química , Células Dendríticas/metabolismo , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Oligonucleótidos Fosforotioatos/química , Oligonucleótidos Fosforotioatos/genética , Psoriasis/genética , Psoriasis/inmunología , Psoriasis/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/metabolismo
9.
Vaccines (Basel) ; 9(6)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204268

RESUMEN

Fc-dependent effector functions are an important determinant of the in vivo potency of therapeutic antibodies. Effector function is determined by the combination of FcRs bound by the antibody and the cell expressing the relevant FcRs, leading to antibody-dependent cellular cytotoxicity (ADCC). A number of ADCC assays have been developed; however, they suffer from limitations in terms of throughput, reproducibility, and in vivo relevance. Existing assays measure NK cell-mediated ADCC activity; however, studies suggest that macrophages mediate the effector function of many antibodies in vivo. Here, we report the development of a macrophage-based ADCC assay that relies on luciferase expression in target cells as a measure of live cell number. In the presence of primary mouse macrophages and specific antibodies, loss of luciferase signal serves as a surrogate for ADCC-dependent killing. We show that the assay functions for a variety of mouse and human isotypes with a model antigen/antibody complex in agreement with the known effector function of the isotypes. We also use this assay to measure the activity of a number of influenza-specific antibodies and show that the assay correlates well with the known in vivo effector functions of these antibodies.

10.
Science ; 371(6532): 926-931, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33495306

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteins interact with the eukaryotic translation machinery, and inhibitors of translation have potent antiviral effects. We found that the drug plitidepsin (aplidin), which has limited clinical approval, possesses antiviral activity (90% inhibitory concentration = 0.88 nM) that is more potent than remdesivir against SARS-CoV-2 in vitro by a factor of 27.5, with limited toxicity in cell culture. Through the use of a drug-resistant mutant, we show that the antiviral activity of plitidepsin against SARS-CoV-2 is mediated through inhibition of the known target eEF1A (eukaryotic translation elongation factor 1A). We demonstrate the in vivo efficacy of plitidepsin treatment in two mouse models of SARS-CoV-2 infection with a reduction of viral replication in the lungs by two orders of magnitude using prophylactic treatment. Our results indicate that plitidepsin is a promising therapeutic candidate for COVID-19.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Depsipéptidos/farmacología , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Animales , Antivirales/uso terapéutico , COVID-19/prevención & control , COVID-19/virología , Proteínas de la Nucleocápside de Coronavirus/biosíntesis , Proteínas de la Nucleocápside de Coronavirus/genética , Depsipéptidos/administración & dosificación , Depsipéptidos/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Células HEK293 , Humanos , Pulmón/virología , Ratones Endogámicos C57BL , Mutación , Péptidos Cíclicos , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , ARN Viral/biosíntesis , ARN Viral/genética , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
11.
J Immunol ; 181(9): 5875-84, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941176

RESUMEN

Autoreactive B cells are activated by DNA, chromatin, or chromatin-containing immune complexes (ICs) through a mechanism dependent on dual engagement of the BCR and TLR9. We examined the contribution of endogenous DNA sequence elements to this process. DNA sequence can determine both recognition by the BCR and by TLR9. DNA fragments containing CpG islands, a natural source of unmethylated CpG dinucleotides, promote the activation of DNA-reactive B cells derived from BCR transgenic mice as well as DNA-reactive B cells present in the normal repertoire. ICs containing these CpG island fragments are potent ligands for AM14 IgG2a-reactive B cells. In contrast, ICs containing total mammalian DNA, or DNA fragments lacking immunostimulatory motifs, fail to induce B cell proliferation, indicating that BCR crosslinking alone is insufficient to activate low-affinity autoreactive B cells. Importantly, priming B cells with IFN-alpha lowers the BCR activation threshold and relaxes the selectivity for CpG-containing DNA. Taken together, our findings underscore the importance of endogenous CpG-containing DNAs in the TLR9-dependent activation of autoreactive B cells and further identify an important mechanism through which IFN-alpha can contribute to the pathogenesis of systemic lupus erythematosus.


Asunto(s)
Autoantígenos/metabolismo , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Islas de CpG/inmunología , Interferón-alfa/fisiología , Animales , Células Clonales , Islas de CpG/genética , ADN Bacteriano/inmunología , ADN Bacteriano/metabolismo , Relación Dosis-Respuesta Inmunológica , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos B/metabolismo
12.
Emerg Microbes Infect ; 9(1): 2433-2445, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33073694

RESUMEN

Severe acute respiratory syndrome CoV-2 (SARS-CoV-2) is currently causing a worldwide pandemic with high morbidity and mortality. Development of animal models that recapitulate important aspects of coronavirus disease 2019 (COVID-19) is critical for the evaluation of vaccines and antivirals, and understanding disease pathogenesis. SARS-CoV-2 has been shown to use the same entry receptor as SARS-CoV-1, human angiotensin-converting enzyme 2 (hACE2) [1-3]. Due to amino acid differences between murine and hACE2, inbred mouse strains fail to support high titer viral replication of SARS-CoV-2 virus. Therefore, a number of transgenic and knock-in mouse models, as well as viral vector-mediated hACE2 delivery systems have been developed. Here we compared the K18-hACE2 transgenic model to adenovirus-mediated delivery of hACE2 to the mouse lung. We show that K18-hACE2 mice replicate virus to high titers in the nasal turbinates, lung and brain, with high lethality, and cytokine/chemokine production. In contrast, adenovirus-mediated delivery results in viral replication to lower titers limited to the nasal turbinates and lung, and no clinical signs of infection. The K18-hACE2 model provides a stringent model for testing vaccines and antivirals, whereas the adenovirus delivery system has the flexibility to be used across multiple genetic backgrounds and modified mouse strains.


Asunto(s)
Betacoronavirus/crecimiento & desarrollo , Infecciones por Coronavirus/patología , Peptidil-Dipeptidasa A/genética , Neumonía Viral/patología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/crecimiento & desarrollo , Replicación Viral/genética , Células A549 , Adenoviridae/genética , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/metabolismo , COVID-19 , Línea Celular , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Pandemias , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , SARS-CoV-2 , Células Vero , Acoplamiento Viral
13.
bioRxiv ; 2020 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-32676603

RESUMEN

Severe acute respiratory syndrome CoV-2 (SARS-CoV-2) is currently causing a worldwide pandemic with high morbidity and mortality. Development of animal models that recapitulate important aspects of coronavirus disease 2019 (COVID-19) is critical for the evaluation of vaccines and antivirals, and understanding disease pathogenesis. SARS-CoV-2 has been shown to use the same entry receptor as SARS-CoV-1, human angiotensin-converting enzyme 2 (hACE2)(1-3). Due to amino acid differences between murine and hACE2, inbred mouse strains fail to support high titer viral replication of SARS-CoV-2 virus. Therefore, a number of transgenic and knock-in mouse models, as well as viral vector-mediated hACE2 delivery systems have been developed. Here we compared the K18-hACE2 transgenic model to adenovirus-mediated delivery of hACE2 to the mouse lung. We show that K18-hACE2 mice replicate virus to high titers in both the lung and brain leading to lethality. In contrast, adenovirus-mediated delivery results in viral replication to lower titers limited to the lung, and no clinical signs of infection with a challenge dose of 10 4 plaque forming units. The K18-hACE2 model provides a stringent model for testing the ability of vaccines and antivirals to protect against disease, whereas the adenovirus delivery system has the flexibility to be used across multiple genetic backgrounds and modified mouse strains.

14.
Viruses ; 12(7)2020 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-32635475

RESUMEN

The host tropism of viral infection is determined by a variety of factors, from cell surface receptors to innate immune signaling. Many viruses encode proteins that interfere with host innate immune recognition in order to promote infection. STAT2 is divergent between species and therefore has a role in species restriction of some viruses. To understand the role of STAT2 in human metapneumovirus (HMPV) infection of human and murine tissues, we first infected STAT2-/- mice and found that HMPV could be serially passaged in STAT2-/-, but not WT, mice. We then used in vitro methods to show that HMPV inhibits expression of both STAT1 and STAT2 in human and primate cells, but not in mouse cells. Transfection of the murine form of STAT2 into STAT2-deficient human cells conferred resistance to STAT2 inhibition. Finally, we sought to understand the in vivo role of STAT2 by infecting hSTAT2 knock-in mice with HMPV, and found that mice had increased weight loss, inhibition of type I interferon signaling, and a Th2-polarized cytokine profile compared to WT mice. These results indicate that STAT2 is a target of HMPV in human infection, while the murine version of STAT2 restricts tropism of HMPV for murine cells and tissue.


Asunto(s)
Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/inmunología , Factor de Transcripción STAT2/inmunología , Animales , Femenino , Especificidad del Huésped , Humanos , Inmunidad Innata , Interferones/genética , Interferones/inmunología , Masculino , Metapneumovirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Factor de Transcripción STAT2/genética , Células Th2
15.
Cell Rep ; 31(1): 107498, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268088

RESUMEN

The Toll/IL-1R-domain-containing adaptor protein SARM1 is expressed primarily in the brain, where it mediates axonal degeneration. Roles for SARM1 in TLR signaling, viral infection, inflammasome activation, and chemokine and Xaf1 expression have also been described. Much of the evidence for SARM1 function relies on SARM1-deficient mice generated in 129 ESCs and backcrossed to B6. The Sarm1 gene lies in a gene-rich region encompassing Xaf1 and chemokine loci, which remain 129 in sequence. We therefore generated additional knockout strains on the B6 background, confirming the role of SARM1 in axonal degeneration and WNV infection, but not in VSV or LACV infection, or in chemokine or Xaf1 expression. Sequence variation in proapoptotic Xaf1 between B6 and 129 results in coding changes and distinct splice variants, which may account for phenotypes previously attributed to SARM1. Reevaluation of phenotypes in these strains will be critical for understanding the function of SARM1.


Asunto(s)
Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Axones/metabolismo , Encéfalo/metabolismo , Encefalitis de California/genética , Enfermedades Hereditarias del Ojo , Femenino , Virus La Crosse , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Fenotipo , Receptores de Interleucina-1/metabolismo , Degeneración Retiniana , Transducción de Señal/genética , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Trastornos de la Visión , Fiebre del Nilo Occidental/genética
16.
Methods Mol Biol ; 517: 363-80, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378022

RESUMEN

High titers of autoantibodies reactive with DNA/RNA molecular complexes are characteristic of autoimmune disorders such as systemic lupus erythematosus (SLE). In vitro and in vivo studies have implicated Toll-like receptor 9 (TLR9) and Toll-like receptor 7 (TLR7) in the activation of the corresponding autoantibody producing B cells. Importantly, TLR9/TLR7-deficiency results in the inability of autoreactive B cells to proliferate in response to DNA/RNA-associated autoantigens in vitro, and in marked changes in the autoantibody repertoire of autoimmune-prone mice. Uptake of DNA/RNA-associated autoantigen immune complexes (ICs) also leads to activation of dendritic cells (DCs) through TLR9 and TLR7.The initial studies from our lab involved ICs formed by a mixture of autoantibodies and cell debris released from dying cells in culture. To better understand the nature of the mammalian ligands that can effectively activate TLR7 and TLR9, we have developed a methodology for preparing ICs containing defined DNA fragments that recapitulate the immunostimulatory activity of the previous "black box" ICs. These reagents reveal an important role for nucleic acid sequence, even when the ligand is mammalian DNA.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Linfocitos B/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Receptores Toll-Like/análisis , Receptores Toll-Like/inmunología , Animales , Linfocitos B/citología , Proliferación Celular , Células Cultivadas , Citocinas/análisis , Citocinas/inmunología , ADN/genética , ADN/aislamiento & purificación , Ratones , ARN/genética , ARN/aislamiento & purificación
17.
Cell Rep ; 25(10): 2784-2796.e3, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517866

RESUMEN

Type I and type III interferons (IFNs) are critical for controlling viral infections. However, the precise dynamics of the IFN response have been difficult to define in vivo. Signaling through type I IFN receptors leads to interferon-stimulated response element (ISRE)-dependent gene expression and an antiviral state. As an alternative to tracking IFN, we used an ISRE-dependent reporter mouse to define the cell types, localization, and kinetics of IFN responding cells during influenza virus infection. We find that measurable IFN responses are largely limited to hematopoietic cells, which show a high sensitivity to IFN. Inflammatory monocytes display high basal IFN responses, which are enhanced upon infection and correlate with infection of these cells. We find that inflammatory monocyte development is independent of IFN signaling; however, IFN is critical for chemokine production and recruitment following infection. The data reveal a role for inflammatory monocytes in both basal IFN responses and responses to infection.


Asunto(s)
Genes Reporteros , Interferón Tipo I/metabolismo , Monocitos/patología , Elementos de Respuesta/genética , Alelos , Animales , Antígenos Ly/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inflamación/patología , Pulmón/virología , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Monocitos/metabolismo , Orthomyxoviridae/fisiología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Transducción de Señal
18.
Cell Host Microbe ; 23(5): 672-685.e6, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29746837

RESUMEN

Progress toward understanding Zika virus (ZIKV) pathogenesis is hindered by lack of immunocompetent small animal models, in part because ZIKV fails to effectively antagonize Stat2-dependent interferon (IFN) responses in mice. To address this limitation, we first passaged an African ZIKV strain (ZIKV-Dak-41525) through Rag1-/- mice to obtain a mouse-adapted virus (ZIKV-Dak-MA) that was more virulent than ZIKV-Dak-41525 in mice treated with an anti-Ifnar1 antibody. A G18R substitution in NS4B was the genetic basis for the increased replication, and resulted in decreased IFN-ß production, diminished IFN-stimulated gene expression, and the greater brain infection observed with ZIKV-Dak-MA. To generate a fully immunocompetent mouse model of ZIKV infection, human STAT2 was introduced into the mouse Stat2 locus (hSTAT2 KI). Subcutaneous inoculation of pregnant hSTAT2 KI mice with ZIKV-Dak-MA resulted in spread to the placenta and fetal brain. An immunocompetent mouse model of ZIKV infection may prove valuable for evaluating countermeasures to limit disease.


Asunto(s)
Ratones/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Virus Zika/patogenicidad , Animales , Encéfalo , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Enfermedades Fetales/metabolismo , Enfermedades Fetales/virología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunidad , Transmisión Vertical de Enfermedad Infecciosa , Interferón beta/metabolismo , Interferones/metabolismo , Ratones/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Placenta/metabolismo , Embarazo , Complicaciones Infecciosas del Embarazo/virología , ARN Helicasas/genética , Receptor de Interferón alfa y beta , Factor de Transcripción STAT2/metabolismo , Serina Endopeptidasas/genética , Proteínas no Estructurales Virales/genética , Virus Zika/genética , Infección por el Virus Zika/virología
19.
Nat Commun ; 8(1): 846, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-29018261

RESUMEN

The aim of candidate universal influenza vaccines is to provide broad protection against influenza A and B viruses. Studies have demonstrated that broadly reactive antibodies require Fc-Fc gamma receptor interactions for optimal protection; however, the innate effector cells responsible for mediating this protection remain largely unknown. Here, we examine the roles of alveolar macrophages, natural killer cells, and neutrophils in antibody-mediated protection. We demonstrate that alveolar macrophages play a dominant role in conferring protection provided by both broadly neutralizing and non-neutralizing antibodies in mice. Our data also reveal the potential mechanisms by which alveolar macrophages mediate protection in vivo, namely antibody-induced inflammation and antibody-dependent cellular phagocytosis. This study highlights the importance of innate effector cells in establishing a broad-spectrum antiviral state, as well as providing a better understanding of how multiple arms of the immune system cooperate to achieve an optimal antiviral response following influenza virus infection or immunization.Broadly reactive antibodies that recognize influenza A virus HA can be protective, but the mechanism is not completely understood. Here, He et al. show that the inflammatory response and phagocytosis mediated by the interaction between protective antibodies and macrophages are essential for protection.


Asunto(s)
Anticuerpos Neutralizantes/fisiología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Activación de Macrófagos , Macrófagos Alveolares/fisiología , Células A549 , Animales , Perros , Femenino , Células HEK293 , Hemaglutininas/inmunología , Humanos , Células Asesinas Naturales/fisiología , Células de Riñón Canino Madin Darby , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/fisiología , Infecciones por Orthomyxoviridae/inmunología , Fagocitosis , Receptores de IgG/metabolismo
20.
J Endotoxin Res ; 12(6): 379-84, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17254393

RESUMEN

AM14 B cells are a prototype for those low affinity autoreactive B cells that routinely mature as naïve cells in peripheral lymphoid tissues. These cells express a transgene-encoded receptor specific for IgG2a and can be effectively activated by immune complexes that incorporate either mammalian DNA or mammalian RNA that has been released from dead or dying cells. Activation depends on the ability of the B-cell receptor to deliver antigen to an internal vesicular compartment containing either Toll-like receptor-9 (TLR9) or TLR7. Since TLR9 and TLR7 are thought to recognize microbial DNA and RNA preferentially, it is important to determine under what conditions mammalian DNA and RNA become effective TLR ligands, and whether the determining factor is delivery or structure. This issue has been addressed by using IgG2a mAbs to deliver immune complexes preloaded with defined fragments of DNA or RNA, or by using modified ODNs/ORNs. The data demonstrate that only certain nucleic acid sequences or structures can induce autoreactive B-cell proliferation, even when delivery to the appropriate TLR compartment is facilitated by uptake through the B-cell receptor (BCR).


Asunto(s)
Autoantígenos/inmunología , Linfocitos B/inmunología , ADN/inmunología , ARN/inmunología , Adyuvantes Inmunológicos , Animales , Secuencia de Bases , Células de la Médula Ósea/fisiología , Cartilla de ADN , Inmunoglobulina M , Cinética , Activación de Linfocitos , Macrófagos/inmunología , Mamíferos , Ratones , Ratones Noqueados , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA