Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.307
Filtrar
1.
Front Immunol ; 15: 1379570, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957465

RESUMEN

There is a reciprocal relationship between extracellular matrix (ECM) remodelling and inflammation that could be operating in the progression of severe COVID-19. To explore the immune-driven ECM remodelling in COVID-19, we in this explorative study analysed these interactions in hospitalised COVID-19 patients. RNA sequencing and flow analysis were performed on peripheral blood mononuclear cells. Inflammatory mediators in plasma were measured by ELISA and MSD, and clinical information from hospitalised COVID-19 patients (N=15) at admission was included in the analysis. Further, we reanalysed two publicly available datasets: (1) lung tissue RNA-sequencing dataset (N=5) and (2) proteomics dataset from PBCM. ECM remodelling pathways were enriched in PBMC from COVID-19 patients compared to healthy controls. Patients treated at the intensive care unit (ICU) expressed distinct ECM remodelling gene profiles compared to patients in the hospital ward. Several markers were strongly correlated to immune cell subsets, and the dysregulation in the ICU patients was positively associated with plasma levels of inflammatory cytokines and negatively associated with B-cell activating factors. Finally, our analysis of publicly accessible datasets revealed (i) an augmented ECM remodelling signature in inflamed lung tissue compared to non-inflamed tissue and (ii) proteomics analysis of PBMC from severe COVID-19 patients demonstrated an up-regulation in an ECM remodelling pathway. Our results may suggest the presence of an interaction between ECM remodelling, inflammation, and immune cells, potentially initiating or perpetuating pulmonary pathology in severe COVID-19.


Asunto(s)
COVID-19 , Matriz Extracelular , Leucocitos Mononucleares , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/sangre , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Matriz Extracelular/metabolismo , Masculino , Femenino , Persona de Mediana Edad , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Anciano , Citocinas/sangre , Proteómica/métodos , Pulmón/inmunología , Pulmón/patología , Adulto
2.
Front Immunol ; 15: 1424374, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38966641

RESUMEN

At the beginning of the COVID-19 pandemic those with underlying chronic lung conditions, including tuberculosis (TB), were hypothesized to be at higher risk of severe COVID-19 disease. However, there is inconclusive clinical and preclinical data to confirm the specific risk SARS-CoV-2 poses for the millions of individuals infected with Mycobacterium tuberculosis (M.tb). We and others have found that compared to singly infected mice, mice co-infected with M.tb and SARS-CoV-2 leads to reduced SARS-CoV-2 severity compared to mice infected with SARS-CoV-2 alone. Consequently, there is a large interest in identifying the molecular mechanisms responsible for the reduced SARS-CoV-2 infection severity observed in M.tb and SARS-CoV-2 co-infection. To address this, we conducted a comprehensive characterization of a co-infection model and performed mechanistic in vitro modeling to dynamically assess how the innate immune response induced by M.tb restricts viral replication. Our study has successfully identified several cytokines that induce the upregulation of anti-viral genes in lung epithelial cells, thereby providing protection prior to challenge with SARS-CoV-2. In conclusion, our study offers a comprehensive understanding of the key pathways induced by an existing bacterial infection that effectively restricts SARS-CoV-2 activity and identifies candidate therapeutic targets for SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Coinfección , Inmunidad Innata , Mycobacterium tuberculosis , SARS-CoV-2 , COVID-19/inmunología , Animales , Mycobacterium tuberculosis/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Ratones , Coinfección/inmunología , Humanos , Tuberculosis/inmunología , Tuberculosis/microbiología , Citocinas/metabolismo , Citocinas/inmunología , Modelos Animales de Enfermedad , Índice de Severidad de la Enfermedad , Pulmón/inmunología , Pulmón/virología , Pulmón/microbiología , Pulmón/patología , Replicación Viral , Ratones Endogámicos C57BL , Femenino
3.
Nat Commun ; 15(1): 5606, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961055

RESUMEN

Viral mutations frequently outpace technologies used to detect harmful variants. Given the continual emergence of SARS-CoV-2 variants, platforms that can identify the presence of a virus and its propensity for infection are needed. Our electronic biomembrane sensing platform recreates distinct SARS-CoV-2 host cell entry pathways and reports the progression of entry as electrical signals. We focus on two necessary entry processes mediated by the viral Spike protein: virus binding and membrane fusion, which can be distinguished electrically. We find that closely related variants of concern exhibit distinct fusion signatures that correlate with trends in cell-based infectivity assays, allowing us to report quantitative differences in their fusion characteristics and hence their infectivity potentials. We use SARS-CoV-2 as our prototype, but we anticipate that this platform can extend to other enveloped viruses and cell lines to quantifiably assess virus entry.


Asunto(s)
COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Humanos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , COVID-19/virología , Fusión de Membrana , Sistema Libre de Células , Mutación , Acoplamiento Viral
4.
Front Cell Infect Microbiol ; 14: 1371837, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994005

RESUMEN

Virus receptors determine the tissue tropism of viruses and have a certain relationship with the clinical outcomes caused by viral infection, which is of great importance for the identification of virus receptors to understand the infection mechanism of viruses and to develop entry inhibitor. Proximity labeling (PL) is a new technique for studying protein-protein interactions, but it has not yet been applied to the identification of virus receptors or co-receptors. Here, we attempt to identify co-receptor of SARS-CoV-2 by employing TurboID-catalyzed PL. The membrane protein angiotensin-converting enzyme 2 (ACE2) was employed as a bait and conjugated to TurboID, and a A549 cell line with stable expression of ACE2-TurboID was constructed. SARS-CoV-2 pseudovirus were incubated with ACE2-TurboID stably expressed cell lines in the presence of biotin and ATP, which could initiate the catalytic activity of TurboID and tag adjacent endogenous proteins with biotin. Subsequently, the biotinylated proteins were harvested and identified by mass spectrometry. We identified a membrane protein, AXL, that has been functionally shown to mediate SARS-CoV-2 entry into host cells. Our data suggest that PL could be used to identify co-receptors for virus entry.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Receptores Virales , SARS-CoV-2 , Internalización del Virus , Humanos , Enzima Convertidora de Angiotensina 2/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Células A549 , Receptores Virales/metabolismo , Tirosina Quinasa del Receptor Axl , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , COVID-19/virología , COVID-19/metabolismo , Coloración y Etiquetado/métodos , Células HEK293 , Biotinilación , Mapeo de Interacción de Proteínas , Biotina/metabolismo
7.
J Gen Virol ; 105(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38995681

RESUMEN

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is associated with neurological sequelae including haemorrhage, thrombosis and ischaemic necrosis and encephalitis. However, the mechanism by which this occurs is unclear. Neurological disease associated with COVID-19 has been proposed to occur following direct infection of the central nervous system and/or indirectly by local or systemic immune activation. We evaluated the expression of angiotensin-converting enzyme-2 and transmembrane protease, serine 2 (TMPRSS2) in brain tissue from five healthy human donors and observed low-level expression of these proteins in cells morphologically consistent with astrocytes, neurons and choroidal ependymal cells within the frontal cortex and medulla oblongata. Primary human astrocytes, neurons, choroid plexus epithelial cells and pericytes supported productive SARS-CoV-2 infection with ancestral, Alpha, Delta and Omicron variants. Infected cells supported the full viral life cycle, releasing infectious virus particles. In contrast, primary brain microvascular endothelial cells and microglia were refractory to SARS-CoV-2 infection. These data support a model whereby SARS-CoV-2 can infect human brain cells, and the mechanism of viral entry warrants further investigation.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Astrocitos , COVID-19 , Plexo Coroideo , Células Epiteliales , Neuronas , Pericitos , SARS-CoV-2 , Serina Endopeptidasas , Humanos , Pericitos/virología , SARS-CoV-2/fisiología , Astrocitos/virología , Plexo Coroideo/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Neuronas/virología , COVID-19/virología , COVID-19/patología , Células Epiteliales/virología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Células Cultivadas , Encéfalo/virología , Encéfalo/patología , Sistema Nervioso Central/virología
8.
Int J Mol Sci ; 25(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-39000469

RESUMEN

Coronavirus can cause various diseases, from mild symptoms to the recent severe COVID-19. The coronavirus RNA genome is frequently mutated due to its RNA nature, resulting in many pathogenic and drug-resistant variants. Therefore, many medicines should be prepared to respond to the various coronavirus variants. In this report, we demonstrated that Forsythia viridissima fruit ethanol extract (FVFE) effectively reduces coronavirus replication. We attempted to identify the active compounds and found that actigenin from FVFE effectively reduces human coronavirus replication. Arctigenin treatment can reduce coronavirus protein expression and coronavirus-induced cytotoxicity. These results collectively suggest that arctigenin is a potent natural compound that prevents coronavirus replication.


Asunto(s)
Forsythia , Frutas , Furanos , Lignanos , Extractos Vegetales , Replicación Viral , Forsythia/química , Lignanos/farmacología , Replicación Viral/efectos de los fármacos , Furanos/farmacología , Humanos , Frutas/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Antivirales/farmacología , Antivirales/química , Animales , Chlorocebus aethiops , Células Vero
9.
Sci Rep ; 14(1): 15351, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38961189

RESUMEN

As SARS-CoV-2 continues to spread worldwide, tractable primary airway cell models that recapitulate the cell-intrinsic response to arising viral variants are needed. Here we describe an adult stem cell-derived human airway organoid model overexpressing the ACE2 receptor (ACE2-OE) that supports robust viral replication while maintaining 3D architecture and cellular diversity of the airway epithelium. ACE2-OE organoids were infected with SARS-CoV-2 variants and subjected to single-cell RNA-sequencing. Interferon-lambda was upregulated in cells with low-level infection while the NF-kB inhibitor alpha gene (encoding IkBa) was consistently upregulated in infected cells, and its expression positively correlated with infection levels. Confocal microscopy showed more IkBa expression in infected than bystander cells, but found concurrent nuclear translocation of NF-kB that IkBa usually prevents. Overexpressing a nondegradable IkBa mutant reduced NF-kB translocation and increased viral infection. These data demonstrate the functionality of ACE2-OE organoids in SARS-CoV-2 research and underscore that the strength of the NF-kB feedback loop in infected cells controls viral replication.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Inhibidor NF-kappaB alfa , Organoides , SARS-CoV-2 , Replicación Viral , Humanos , Organoides/virología , Organoides/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , SARS-CoV-2/fisiología , COVID-19/virología , COVID-19/metabolismo , COVID-19/genética , Inhibidor NF-kappaB alfa/metabolismo , Inhibidor NF-kappaB alfa/genética , FN-kappa B/metabolismo
10.
Arch Microbiol ; 206(8): 345, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38976047

RESUMEN

Neurological complications, both acute and chronic, are reported commonly in COVID-19 affected individuals. In this context, the understanding of pathogenesis of SARS-CoV-2 in specific cells of central nervous system (CNS) origin is relevant. The present study explores infection biology of a clinical isolate of SARS-CoV-2 in human cell lines of neural origin such as the glioblastoma (U87-MG), neuroblastoma (SHSY5Y) and microglia (C20). Despite showing clear evidence of infection by immunofluorescence with an anti-spike protein antibody, all the three neural cell lines were observed to be highly restrictive to the replication of the infecting virus. While the U87-MG glioblastoma cells demonstrated no cytopathic effects and a low viral titre with no signs of replication, the SHSY5Y neuroblastoma cells exhibited cytopathic effects with bleb formation but no evidence of viable virus. The C20 microglial cells showed neither signs of cytopathic effects nor viable virus. Ultrastructural studies demonstrated intracellular virions in infected neural cells. The presence of lipid droplets in infected SHSY5Y cells suggested an impact on host cell metabolism. The decrease in viral RNA levels over time in all the neural cell lines suggested restricted viral replication. In conclusion, this study highlights the limited susceptibility of neural cells to SARS-CoV-2 infection. This reduced permissibility of neural cell lines to SARS-CoV-2 may point to their inherent lower expression of receptors that support viral entry in addition to the intracellular factors that potently inhibit viral replication. The study findings prompt further investigation into the mechanisms of SARS-CoV-2 infection of neural cells.


Asunto(s)
COVID-19 , Microglía , Neuroglía , Neuronas , SARS-CoV-2 , Replicación Viral , Humanos , Microglía/virología , SARS-CoV-2/fisiología , SARS-CoV-2/patogenicidad , Neuronas/virología , COVID-19/virología , Neuroglía/virología , Línea Celular Tumoral , Línea Celular , Efecto Citopatogénico Viral , Glicoproteína de la Espiga del Coronavirus/metabolismo , ARN Viral/genética
11.
Int J Mol Sci ; 25(13)2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38999930

RESUMEN

Although SARS-CoV-2 induces mucin hypersecretion in the respiratory tract, hyposalivation/xerostomia has been reported by COVID-19 patients. We evaluate the submandibular gland (SMGs) pathogenesis in SARS-CoV-2-infected K18-hACE2 mice, focusing on the impact of infection on the mucin production and structural integrity of acini, ductal system, myoepithelial cells (MECs) and telocytes. The spike protein, the nucleocapsid protein, hACE2, actin, EGF, TNF-α and IL-1ß were detected by immunofluorescence, and the Egfr and Muc5b expression was evaluated. In the infected animals, significant acinar hypertrophy was observed in contrast to ductal atrophy. Nucleocapsid proteins and/or viral particles were detected in the SMG cells, mainly in the nuclear membrane-derived vesicles, confirming the nuclear role in the viral formation. The acinar cells showed intense TNF-α and IL-1ß immunoexpression, and the EGF-EGFR signaling increased, together with Muc5b upregulation. This finding explains mucin hypersecretion and acinar hypertrophy, which compress the ducts. Dying MECs and actin reduction were also observed, indicating failure of contraction and acinar support, favoring acinar hypertrophy. Viral assembly was found in the dying telocytes, pointing to these intercommunicating cells as viral transmitters in SMGs. Therefore, EGF-EGFR-induced mucin hypersecretion was triggered by SARS-CoV-2 in acinar cells, likely mediated by cytokines. The damage to telocytes and MECs may have favored the acinar hypertrophy, leading to ductal obstruction, explaining xerostomia in COVID-19 patients. Thus, acinar cells, telocytes and MECs may be viral targets, which favor replication and cell-to-cell viral transmission in the SMG, corroborating the high viral load in saliva of infected individuals.


Asunto(s)
COVID-19 , Receptores ErbB , SARS-CoV-2 , Glándula Submandibular , Xerostomía , COVID-19/patología , COVID-19/virología , COVID-19/metabolismo , Animales , Glándula Submandibular/virología , Glándula Submandibular/patología , Glándula Submandibular/metabolismo , SARS-CoV-2/fisiología , Ratones , Xerostomía/etiología , Xerostomía/patología , Xerostomía/virología , Xerostomía/metabolismo , Receptores ErbB/metabolismo , Humanos , Enzima Convertidora de Angiotensina 2/metabolismo , Mucina 5B/metabolismo , Células Acinares/patología , Células Acinares/metabolismo , Células Acinares/virología , Interleucina-1beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Modelos Animales de Enfermedad
12.
J Biomed Sci ; 31(1): 70, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003473

RESUMEN

Coronaviruses employ various strategies for survival, among which the activation of endogenous or exogenous apoptosis stands out, with viral proteins playing a pivotal role. Notably, highly pathogenic coronaviruses such as SARS-CoV-2, SARS-CoV, and MERS-CoV exhibit a greater array of non-structural proteins compared to low-pathogenic strains, facilitating their ability to induce apoptosis via multiple pathways. Moreover, these viral proteins are adept at dampening host immune responses, thereby bolstering viral replication and persistence. This review delves into the intricate interplay between highly pathogenic coronaviruses and apoptosis, systematically elucidating the molecular mechanisms underpinning apoptosis induction by viral proteins. Furthermore, it explores the potential therapeutic avenues stemming from apoptosis inhibition as antiviral agents and the utilization of apoptosis-inducing viral proteins as therapeutic modalities. These insights not only shed light on viral pathogenesis but also offer novel perspectives for cancer therapy.


Asunto(s)
Apoptosis , SARS-CoV-2 , Humanos , SARS-CoV-2/fisiología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , COVID-19/virología
13.
Front Immunol ; 15: 1388517, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39034993

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been demonstrated to limit the host interferon response; however, the underlying mechanism remains unclear. Here, we found that SARS-CoV-2 infection upregulated the E3 ubiquitin ligase Huwe1, which in turn facilitated the degradation of the transcription factor Miz1. The degradation of Miz1 hampered interferon alpha and gamma responses, consequently fostering viral replication and impeding viral clearance. Conversely, silencing or inhibiting Huwe1 enhanced the interferon responses, effectively curbing viral replication. Consistently, overexpressing Miz1 augmented the interferon responses and limited viral replication, whereas silencing Miz1 had the opposite effect. Targeting Huwe1 or overexpressing Miz1 elicited transcriptomic alterations characterized by enriched functions associated with bolstered antiviral response and diminished virus replication. Further study revealed Miz1 exerted epigenetic control over the transcription of specific interferon signaling molecules, which acted as common upstream regulators responsible for the observed transcriptomic changes following Huwe1 or Miz1 targeting. These findings underscore the critical role of the Huwe1-Miz1 axis in governing the host antiviral response, with its dysregulation contributing to the impaired interferon response observed during COVID-19.


Asunto(s)
COVID-19 , Interferón-alfa , Interferón gamma , SARS-CoV-2 , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Replicación Viral , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Humanos , SARS-CoV-2/fisiología , Interferón gamma/metabolismo , COVID-19/inmunología , COVID-19/virología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Interferón-alfa/metabolismo , Animales , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Proteolisis , Células HEK293 , Chlorocebus aethiops , Factores de Transcripción de Tipo Kruppel
14.
Nature ; 631(8021): 645-653, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38987596

RESUMEN

Platelet homeostasis is essential for vascular integrity and immune defence1,2. Although the process of platelet formation by fragmenting megakaryocytes (MKs; thrombopoiesis) has been extensively studied, the cellular and molecular mechanisms required to constantly replenish the pool of MKs by their progenitor cells (megakaryopoiesis) remains unclear3,4. Here we use intravital imaging to track the cellular dynamics of megakaryopoiesis over days. We identify plasmacytoid dendritic cells (pDCs) as homeostatic sensors that monitor the bone marrow for apoptotic MKs and deliver IFNα to the MK niche triggering local on-demand proliferation and maturation of MK progenitors. This pDC-dependent feedback loop is crucial for MK and platelet homeostasis at steady state and under stress. pDCs are best known for their ability to function as vigilant detectors of viral infection5. We show that virus-induced activation of pDCs interferes with their function as homeostatic sensors of megakaryopoiesis. Consequently, activation of pDCs by SARS-CoV-2 leads to excessive megakaryopoiesis. Together, we identify a pDC-dependent homeostatic circuit that involves innate immune sensing and demand-adapted release of inflammatory mediators to maintain homeostasis of the megakaryocytic lineage.


Asunto(s)
COVID-19 , Células Dendríticas , Homeostasis , Megacariocitos , Trombopoyesis , Células Dendríticas/inmunología , Células Dendríticas/citología , Animales , Megacariocitos/citología , Megacariocitos/inmunología , Ratones , COVID-19/inmunología , COVID-19/virología , Masculino , Femenino , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Interferón-alfa/metabolismo , Inmunidad Innata , Plaquetas/inmunología , Plaquetas/citología , Humanos , Apoptosis , Ratones Endogámicos C57BL , Médula Ósea/inmunología , Linaje de la Célula , Proliferación Celular , Retroalimentación Fisiológica
15.
Front Immunol ; 15: 1382524, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39026676

RESUMEN

Introduction: The outbreak of SARS-CoV-2, leading to COVID-19, poses a major global health threat. While specific treatments and vaccines are under development, Traditional Chinese Medicine (TCM) has historically been effective against pandemics, including viral pneumonias. Our study explores the efficacy and mechanisms of Jinhua Qinggan Granules (JHQG) in treating COVID-19. Methods: We analyzed JHQG's components using UHPLC-Q-Exactive-Orbitrap-MS, identifying 73 compounds. Network pharmacology and single-cell RNA sequencing (scRNA-seq) were used to assess JHQG's effects on immune cells from peripheral blood mononuclear cells (PBMCs). Literature review supported the antiviral and anti-inflammatory effects of JHQG. Results: JHQG targets were found to interact with immune cells, including neutrophils, monocytes, plasmablasts, and effector T cells, reducing their overactivation in severe COVID-19. JHQG's modulation of these cells' activity likely contributes to reduced inflammation and improved clinical outcomes. Discussion: Our findings provide insights into JHQG's mechanism of action, highlighting its potential in controlling the inflammatory response in COVID-19 patients. The study supports the use of JHQG as a safe and effective treatment for COVID-19 and similar viral infections, leveraging its ability to modulate immune cell activity and reduce inflammation.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Medicamentos Herbarios Chinos , Farmacología en Red , SARS-CoV-2 , Humanos , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , SARS-CoV-2/efectos de los fármacos , COVID-19/inmunología , COVID-19/virología , Cromatografía Líquida de Alta Presión , Inflamación/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , Antivirales/farmacología , Antivirales/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Espectrometría de Masas/métodos
16.
Proc Natl Acad Sci U S A ; 121(30): e2408109121, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39028694

RESUMEN

The prevalence of "long COVID" is just one of the conundrums highlighting how little we know about the lung's response to viral infection, particularly to syndromecoronavirus-2 (SARS-CoV-2), for which the lung is the point of entry. We used an in vitro human lung system to enable a prospective, unbiased, sequential single-cell level analysis of pulmonary cell responses to infection by multiple SARS-CoV-2 strains. Starting with human induced pluripotent stem cells and emulating lung organogenesis, we generated and infected three-dimensional, multi-cell-type-containing lung organoids (LOs) and gained several unexpected insights. First, SARS-CoV-2 tropism is much broader than previously believed: Many lung cell types are infectable, if not through a canonical receptor-mediated route (e.g., via Angiotensin-converting encyme 2(ACE2)) then via a noncanonical "backdoor" route (via macropinocytosis, a form of endocytosis). Food and Drug Administration (FDA)-approved endocytosis blockers can abrogate such entry, suggesting adjunctive therapies. Regardless of the route of entry, the virus triggers a lung-autonomous, pulmonary epithelial cell-intrinsic, innate immune response involving interferons and cytokine/chemokine production in the absence of hematopoietic derivatives. The virus can spread rapidly throughout human LOs resulting in mitochondrial apoptosis mediated by the prosurvival protein Bcl-xL. This host cytopathic response to the virus may help explain persistent inflammatory signatures in a dysfunctional pulmonary environment of long COVID. The host response to the virus is, in significant part, dependent on pulmonary Surfactant Protein-B, which plays an unanticipated role in signal transduction, viral resistance, dampening of systemic inflammatory cytokine production, and minimizing apoptosis. Exogenous surfactant, in fact, can be broadly therapeutic.


Asunto(s)
COVID-19 , Pulmón , Organoides , SARS-CoV-2 , Internalización del Virus , Humanos , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , COVID-19/inmunología , COVID-19/virología , Pulmón/virología , Pulmón/inmunología , Pulmón/patología , Organoides/virología , Tratamiento Farmacológico de COVID-19 , Células Madre Pluripotentes Inducidas/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Inflamación , Citocinas/metabolismo , Apoptosis
17.
J Med Virol ; 96(7): e29782, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39011762

RESUMEN

Extracellular vesicles (EVs) are shown to be a novel viral transmission model capable of increasing a virus's tropism. According to our earlier research, cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or transfected with envelope protein plasmids generate a novel type of EVs that are micrometer-sized and able to encase virus particles. Here, we showed the capacity of these EVs to invade various animals both in vitro and in vivo independent of the angiotensin-converting enzyme 2 receptor. First, via macropinocytosis, intact EVs produced from Vero E6 (monkey) cells were able to enter cells from a variety of animals, including cats, dogs, bats, hamsters, and minks, and vice versa. Second, when given to zebrafish with cutaneous wounds, the EVs showed favorable stability in aqueous environments and entered the fish. Moreover, infection of wild-type (WT) mice with heterogeneous EVs carrying SARS-CoV-2 particles led to a strong cytokine response and a notable amount of lung damage. Conversely, free viral particles did not infect WT mice. These results highlight the variety of processes behind viral transmission and cross-species evolution by indicating that EVs may be possible vehicles for SARS-CoV-2 spillover and raising risk concerns over EVs' potential for viral gene transfer.


Asunto(s)
COVID-19 , Vesículas Extracelulares , SARS-CoV-2 , Animales , Vesículas Extracelulares/virología , Vesículas Extracelulares/metabolismo , SARS-CoV-2/fisiología , SARS-CoV-2/patogenicidad , SARS-CoV-2/genética , COVID-19/transmisión , COVID-19/virología , Ratones , Chlorocebus aethiops , Células Vero , Humanos , Cricetinae , Proteínas de la Envoltura de Coronavirus/metabolismo , Proteínas de la Envoltura de Coronavirus/genética , Perros , Pez Cebra/virología , Gatos , Quirópteros/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética
18.
Phys Biol ; 21(4)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38949432

RESUMEN

Theoretical analysis of epidemic dynamics has attracted significant attention in the aftermath of the COVID-19 pandemic. In this article, we study dynamic instabilities in a spatiotemporal compartmental epidemic model represented by a stochastic system of coupled partial differential equations (SPDE). Saturation effects in infection spread-anchored in physical considerations-lead to strong nonlinearities in the SPDE. Our goal is to study the onset of dynamic, Turing-type instabilities, and the concomitant emergence of steady-state patterns under the interplay between three critical model parameters-the saturation parameter, the noise intensity, and the transmission rate. Employing a second-order perturbation analysis to investigate stability, we uncover both diffusion-driven and noise-induced instabilities and corresponding self-organized distinct patterns of infection spread in the steady state. We also analyze the effects of the saturation parameter and the transmission rate on the instabilities and the pattern formation. In summary, our results indicate that the nuanced interplay between the three parameters considered has a profound effect on the emergence of dynamical instabilities and therefore on pattern formation in the steady state. Moreover, due to the central role played by the Turing phenomenon in pattern formation in a variety of biological dynamic systems, the results are expected to have broader significance beyond epidemic dynamics.


Asunto(s)
COVID-19 , Dinámicas no Lineales , SARS-CoV-2 , Procesos Estocásticos , COVID-19/epidemiología , COVID-19/transmisión , COVID-19/virología , Humanos , SARS-CoV-2/fisiología , Epidemias , Pandemias , Análisis Espacio-Temporal , Modelos Epidemiológicos
19.
Sci Rep ; 14(1): 16895, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043712

RESUMEN

SARS-CoV-2-contributes to sickness and death in COVID-19 patients partly by inducing a hyper-proinflammatory immune response in the host airway. This hyper-proinflammatory state involves activation of signaling by NFκB, and unexpectedly, ENaC, the epithelial sodium channel. Post-infection inflammation may also contribute to "Long COVID"/PASC. Enhanced signaling by NFκB and ENaC also marks the airway of patients suffering from cystic fibrosis, a life-limiting proinflammatory genetic disease due to inactivating mutations in the CFTR gene. We therefore hypothesized that inflammation in the COVID-19 airway might similarly be due to inhibition of CFTR signaling by SARS-CoV-2 spike protein, and therefore activation of both NFκB and ENaC signaling. We used western blot and electrophysiological techniques, and an organoid model of normal airway epithelia, differentiated on an air-liquid-interface (ALI). We found that CFTR protein expression and CFTR cAMP-activated chloride channel activity were lost when the model epithelium was exposed to SARS-CoV-2 spike proteins. As hypothesized, the absence of CFTR led to activation of both TNFα/NFκB signaling and α and γ ENaC. We had previously shown that the cardiac glycoside drugs digoxin, digitoxin and ouabain blocked interaction of spike protein and ACE2. Consistently, addition of 30 nM concentrations of the cardiac glycoside drugs, prevented loss of both CFTR protein and CFTR channel activity. ACE2 and CFTR were found to co-immunoprecipitate in both basal cells and differentiated epithelia. Thus spike-dependent CFTR loss might involve ACE2 as a bridge between Spike and CFTR. In addition, spike exposure to the epithelia resulted in failure of endosomal recycling to return CFTR to the plasma membrane. Thus, failure of CFTR recovery from endosomal recycling might be a mechanism for spike-dependent loss of CFTR. Finally, we found that authentic SARS-CoV-2 virus infection induced loss of CFTR protein, which was rescued by the cardiac glycoside drugs digitoxin and ouabain. Based on experiments with this organoid model of small airway epithelia, and comparisons with 16HBE14o- and other cell types expressing normal CFTR, we predict that inflammation in the COVID-19 airway may be mediated by inhibition of CFTR signaling by the SARS-CoV-2 spike protein, thus inducing a cystic fibrosis-like clinical phenotype. To our knowledge this is the first time COVID-19 airway inflammation has been experimentally traced in normal subjects to a contribution from SARS-CoV-2 spike-dependent inhibition of CFTR signaling.


Asunto(s)
COVID-19 , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Inflamación , SARS-CoV-2 , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Glicoproteína de la Espiga del Coronavirus/metabolismo , COVID-19/metabolismo , COVID-19/virología , SARS-CoV-2/fisiología , Inflamación/metabolismo , FN-kappa B/metabolismo , Canales Epiteliales de Sodio/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ouabaína/farmacología
20.
Methods Mol Biol ; 2813: 117-123, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38888774

RESUMEN

The emergence of zoonotic viruses like severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2 have significantly impacted global health and economy. The discovery of other viruses in wildlife reservoir species present a threat for future emergence in humans and animals. Therefore, assays that are less reliant on virus-specific information, such as neutralization assays, are crucial to rapidly develop diagnostics, understand virus replication and pathogenicity, and assess the efficacy of therapeutics against newly emerging viruses. Here, we describe the discontinuous median tissue culture infectious dose 50 (TCID50) assay to quantitatively determine the titer of any virus that can produce a visible cytopathic effect in infected cells.


Asunto(s)
Efecto Citopatogénico Viral , Animales , Humanos , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Chlorocebus aethiops , COVID-19/virología , Células Vero , Replicación Viral , Técnicas de Cultivo de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...