Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.297
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1371837, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38994005

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2 , Receptores Virais , SARS-CoV-2 , Internalização do Vírus , Humanos , Enzima de Conversão de Angiotensina 2/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Células A549 , Receptores Virais/metabolismo , Receptor Tirosina Quinase Axl , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , COVID-19/virologia , COVID-19/metabolismo , Coloração e Rotulagem/métodos , Células HEK293 , Biotinilação , Mapeamento de Interação de Proteínas , Biotina/metabolismo
2.
Int J Mol Sci ; 25(13)2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-39000469

RESUMO

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.


Assuntos
Forsythia , Frutas , Furanos , Lignanas , Extratos Vegetais , Replicação Viral , Forsythia/química , Lignanas/farmacologia , Replicação Viral/efeitos dos fármacos , Furanos/farmacologia , Humanos , Frutas/química , Extratos Vegetais/farmacologia , Extratos Vegetais/química , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/fisiologia , Antivirais/farmacologia , Antivirais/química , Animais , Chlorocebus aethiops , Células Vero
3.
Sci Rep ; 14(1): 15351, 2024 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-38961189

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Inibidor de NF-kappaB alfa , Organoides , SARS-CoV-2 , Replicação Viral , Humanos , Organoides/virologia , Organoides/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , SARS-CoV-2/fisiologia , COVID-19/virologia , COVID-19/metabolismo , COVID-19/genética , Inibidor de NF-kappaB alfa/metabolismo , Inibidor de NF-kappaB alfa/genética , NF-kappa B/metabolismo
4.
Int J Mol Sci ; 25(13)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38999930

RESUMO

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.


Assuntos
COVID-19 , Receptores ErbB , SARS-CoV-2 , Glândula Submandibular , Xerostomia , COVID-19/patologia , COVID-19/virologia , COVID-19/metabolismo , Animais , Glândula Submandibular/virologia , Glândula Submandibular/patologia , Glândula Submandibular/metabolismo , SARS-CoV-2/fisiologia , Camundongos , Xerostomia/etiologia , Xerostomia/patologia , Xerostomia/virologia , Xerostomia/metabolismo , Receptores ErbB/metabolismo , Humanos , Enzima de Conversão de Angiotensina 2/metabolismo , Mucina-5B/metabolismo , Células Acinares/patologia , Células Acinares/metabolismo , Células Acinares/virologia , Interleucina-1beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Modelos Animais de Doenças
7.
J Biomed Sci ; 31(1): 70, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39003473

RESUMO

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.


Assuntos
Apoptose , SARS-CoV-2 , Humanos , SARS-CoV-2/fisiologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , COVID-19/virologia
8.
J Med Virol ; 96(7): e29782, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39011762

RESUMO

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.


Assuntos
COVID-19 , Vesículas Extracelulares , SARS-CoV-2 , Animais , Vesículas Extracelulares/virologia , Vesículas Extracelulares/metabolismo , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , SARS-CoV-2/genética , COVID-19/transmissão , COVID-19/virologia , Camundongos , Chlorocebus aethiops , Células Vero , Humanos , Cricetinae , Proteínas do Envelope de Coronavírus/metabolismo , Proteínas do Envelope de Coronavírus/genética , Cães , Peixe-Zebra/virologia , Gatos , Quirópteros/virologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética
9.
J Gen Virol ; 105(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38995681

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2 , Astrócitos , COVID-19 , Plexo Corióideo , Células Epiteliais , Neurônios , Pericitos , SARS-CoV-2 , Serina Endopeptidases , Humanos , Pericitos/virologia , SARS-CoV-2/fisiologia , Astrócitos/virologia , Plexo Corióideo/virologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Neurônios/virologia , COVID-19/virologia , COVID-19/patologia , Células Epiteliais/virologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Células Cultivadas , Encéfalo/virologia , Encéfalo/patologia , Sistema Nervoso Central/virologia
10.
Nat Commun ; 15(1): 5606, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961055

RESUMO

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.


Assuntos
COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Internalização do Vírus , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , Humanos , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , COVID-19/virologia , Fusão de Membrana , Sistema Livre de Células , Mutação , Ligação Viral
11.
Arch Microbiol ; 206(8): 345, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38976047

RESUMO

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.


Assuntos
COVID-19 , Microglia , Neuroglia , Neurônios , SARS-CoV-2 , Replicação Viral , Humanos , Microglia/virologia , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , Neurônios/virologia , COVID-19/virologia , Neuroglia/virologia , Linhagem Celular Tumoral , Linhagem Celular , Efeito Citopatogênico Viral , Glicoproteína da Espícula de Coronavírus/metabolismo , RNA Viral/genética
12.
Phys Biol ; 21(4)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38949432

RESUMO

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.


Assuntos
COVID-19 , Dinâmica não Linear , SARS-CoV-2 , Processos Estocásticos , COVID-19/epidemiologia , COVID-19/transmissão , COVID-19/virologia , Humanos , SARS-CoV-2/fisiologia , Epidemias , Pandemias , Análise Espaço-Temporal , Modelos Epidemiológicos
13.
Front Immunol ; 15: 1374828, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39026668

RESUMO

Introduction: Innate lymphoid cells (ILCs) are enriched at mucosal surfaces where they respond rapidly to environmental stimuli and contribute to both tissue inflammation and healing. Methods: To gain insight into the role of ILCs in the pathology and recovery from COVID-19 infection, we employed a multi-omics approach consisting of Abseq and targeted mRNA sequencing to respectively probe the surface marker expression, transcriptional profile and heterogeneity of ILCs in peripheral blood of patients with COVID-19 compared with healthy controls. Results: We found that the frequency of ILC1 and ILC2 cells was significantly increased in COVID-19 patients. Moreover, all ILC subsets displayed a significantly higher frequency of CD69-expressing cells, indicating a heightened state of activation. ILC2s from COVID-19 patients had the highest number of significantly differentially expressed (DE) genes. The most notable genes DE in COVID-19 vs healthy participants included a) genes associated with responses to virus infections and b) genes that support ILC self-proliferation, activation and homeostasis. In addition, differential gene regulatory network analysis revealed ILC-specific regulons and their interactions driving the differential gene expression in each ILC. Discussion: Overall, this study provides mechanistic insights into the characteristics of ILC subsets activated during COVID-19 infection.


Assuntos
COVID-19 , Imunidade Inata , Linfócitos , SARS-CoV-2 , Análise de Célula Única , Humanos , COVID-19/imunologia , COVID-19/genética , Imunidade Inata/genética , Linfócitos/imunologia , Linfócitos/metabolismo , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Perfilação da Expressão Gênica , Transcriptoma , Idoso , Redes Reguladoras de Genes , Multiômica
14.
Front Immunol ; 15: 1382524, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39026676

RESUMO

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.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Medicamentos de Ervas Chinesas , Farmacologia em Rede , SARS-CoV-2 , Humanos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , SARS-CoV-2/efeitos dos fármacos , COVID-19/imunologia , COVID-19/virologia , Cromatografia Líquida de Alta Pressão , Inflamação/tratamento farmacológico , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/imunologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Espectrometria de Massas/métodos
15.
Front Immunol ; 15: 1379570, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38957465

RESUMO

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.


Assuntos
COVID-19 , Matriz Extracelular , Leucócitos Mononucleares , SARS-CoV-2 , Humanos , COVID-19/imunologia , COVID-19/sangue , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Matriz Extracelular/metabolismo , Masculino , Feminino , Pessoa de Meia-Idade , SARS-CoV-2/fisiologia , SARS-CoV-2/imunologia , Idoso , Citocinas/sangue , Proteômica/métodos , Pulmão/imunologia , Pulmão/patologia , Adulto
16.
Front Immunol ; 15: 1424374, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38966641

RESUMO

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.


Assuntos
COVID-19 , Coinfecção , Imunidade Inata , Mycobacterium tuberculosis , SARS-CoV-2 , COVID-19/imunologia , Animais , Mycobacterium tuberculosis/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Camundongos , Coinfecção/imunologia , Humanos , Tuberculose/imunologia , Tuberculose/microbiologia , Citocinas/metabolismo , Citocinas/imunologia , Modelos Animais de Doenças , Índice de Gravidade de Doença , Pulmão/imunologia , Pulmão/virologia , Pulmão/microbiologia , Pulmão/patologia , Replicação Viral , Camundongos Endogâmicos C57BL , Feminino
17.
Nature ; 631(8021): 645-653, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38987596

RESUMO

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.


Assuntos
COVID-19 , Células Dendríticas , Homeostase , Megacariócitos , Trombopoese , Células Dendríticas/imunologia , Células Dendríticas/citologia , Animais , Megacariócitos/citologia , Megacariócitos/imunologia , Camundongos , COVID-19/imunologia , COVID-19/virologia , Masculino , Feminino , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Interferon-alfa/metabolismo , Imunidade Inata , Plaquetas/imunologia , Plaquetas/citologia , Humanos , Apoptose , Camundongos Endogâmicos C57BL , Medula Óssea/imunologia , Linhagem da Célula , Proliferação de Células , Retroalimentação Fisiológica
18.
Sci Rep ; 14(1): 13134, 2024 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849411

RESUMO

The pandemic of coronavirus disease 19 (COVID-19), caused by severe respiratory syndrome coronavirus 2 (SARS-CoV-2), had severe repercussions for breast cancer patients. Increasing evidence indicates that SARS-CoV-2 infection may directly impact breast cancer biology, but the effects of SARS-CoV-2 on breast tumor cells are still unknown. Here, we analyzed the molecular events occurring in the MCF7, MDA-MB-231 and HCC1937 breast cancer cell lines, representative of the luminal A, basal B/claudin-low and basal A subtypes, respectively, upon SARS-CoV-2 infection. Viral replication was monitored over time, and gene expression profiling was conducted. We found that MCF7 cells were the most permissive to viral replication. Treatment of MCF7 cells with Tamoxifen reduced the SARS-CoV-2 replication rate, suggesting an involvement of the estrogen receptor in sustaining virus replication in malignant cells. Interestingly, a metagene signature based on genes upregulated by SARS-CoV-2 infection in all three cell lines distinguished a subgroup of premenopausal luminal A breast cancer patients with a poor prognosis. As SARS-CoV-2 still spreads among the population, it is essential to understand the impact of SARS-CoV-2 infection on breast cancer, particularly in premenopausal patients diagnosed with the luminal A subtype, and to assess the long-term impact of COVID-19 on breast cancer outcomes.


Assuntos
Neoplasias da Mama , COVID-19 , SARS-CoV-2 , Tamoxifeno , Replicação Viral , Humanos , Neoplasias da Mama/virologia , Neoplasias da Mama/patologia , COVID-19/virologia , Feminino , SARS-CoV-2/fisiologia , Linhagem Celular Tumoral , Tamoxifeno/farmacologia , Células MCF-7 , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica
19.
Front Immunol ; 15: 1381059, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38855100

RESUMO

Background: Understanding COVID-19 outcomes remains a challenge. While numerous biomarkers have been proposed for severity at admission, limited exploration exists for markers during the infection course, especially for the requirement of oxygen therapy. This study investigates the potential of eosinophil count normalization as a predictor for oxygen weaning during the initial wave of the pandemic. Methods: A retrospective study was conducted between March and April 2020 (first wave) among adults admitted directly to a medicine ward. Biological abnormalities, including lymphocyte count, eosinophil count, and C-reactive protein (CRP), were gathered daily during the first week of admission according to oxygen level. In case of worsening, oxygen level was censored at 15 L/min. The primary aim was to assess whether eosinophil count normalization predicts a subsequent decrease in oxygen requirements. Results: Overall, 132 patients were admitted, with a mean age of 59.0 ± 16.3 years. Of the patients, 72% required oxygen, and 20.5% were admitted to the intensive care unit after a median delay of 48 hours. The median CRP at admission was 79 (26-130) mg/L, whereas the eosinophil count was 10 (0-60)/mm3. Eosinophil count normalization (≥100/mm3) by day 2 correlated significantly with decreased oxygen needs (<2 L) with hazard ratio (HR) = 3.7 [1.1-12.9] (p = 0.04). Likewise, CRP < 80 mg/L was associated with reduced oxygen requirements (p < 0.001). Predictors, including underlying chronic respiratory disease, exhibited a trend toward a negative association (p = 0.06). Conclusion: The study highlights the relationship between eosinophil count and CRP, with implications for predicting oxygen weaning during COVID-19. Further research is warranted to explore the relevance of these biomarkers in other respiratory infections.


Assuntos
COVID-19 , Eosinófilos , Oxigenoterapia , SARS-CoV-2 , Humanos , COVID-19/sangue , COVID-19/imunologia , COVID-19/terapia , COVID-19/epidemiologia , Masculino , Pessoa de Meia-Idade , Feminino , Estudos Retrospectivos , Eosinófilos/imunologia , Idoso , Contagem de Leucócitos , SARS-CoV-2/fisiologia , Adulto , Hospitalização , Biomarcadores/sangue , Proteína C-Reativa/análise , Proteína C-Reativa/metabolismo , Oxigênio/metabolismo , Oxigênio/sangue
20.
Front Immunol ; 15: 1398369, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38835759

RESUMO

Introduction: Although many studies have underscored the importance of T cells, phenotypically and functionally, fewer have studied the functions of myeloid cells in COVID disease. In particular, the potential role of myeloid cells such as monocytes and low-density neutrophils (LDNs) in innate responses and particular in the defense against secondary bacterial infections has been much less documented. Methods: Here, we compared, in a longitudinal study, healthy subjects, idiopathic fibrosis patients, COVID patients who were either hospitalized/moderate (M-) or admitted to ICU (COV-ICU) and patients in ICU hospitalized for other reasons (non-COV-ICU). Results: We show that COVID patients have an increased proportion of low-density neutrophils (LDNs), which produce high levels of proteases (particularly, NE, MMP-8 and MMP-9) (unlike non-COV-ICU patients), which are partly responsible for causing type II alveolar cell damage in co-culture experiments. In addition, we showed that M- and ICU-COVID monocytes had reduced responsiveness towards further live Pseudomonas aeruginosa (PAO1 strain) infection, an important pathogen colonizing COVID patients in ICU, as assessed by an impaired secretion of myeloid cytokines (IL-1, TNF, IL-8,…). By contrast, lymphoid cytokines (in particular type 2/type 3) levels remained high, both basally and post PAO1 infection, as reflected by the unimpaired capacity of T cells to proliferate, when stimulated with anti-CD3/CD28 beads. Discussion: Overall, our results demonstrate that COVID circulatory T cells have a biased type 2/3 phenotype, unconducive to proper anti-viral responses and that myeloid cells have a dual deleterious phenotype, through their LDN-mediated damaging effect on alveolar cells and their impaired responsiveness (monocyte-mediated) towards bacterial pathogens such as P. aeruginosa.


Assuntos
COVID-19 , Monócitos , Neutrófilos , Infecções por Pseudomonas , Pseudomonas aeruginosa , SARS-CoV-2 , Humanos , COVID-19/imunologia , Pseudomonas aeruginosa/imunologia , Monócitos/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Infecções por Pseudomonas/imunologia , Neutrófilos/imunologia , Idoso , Citocinas/metabolismo , Citocinas/imunologia , Adulto , Estudos Longitudinais , Leucócitos Mononucleares/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA