Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.731
Filtrar
2.
Influenza Other Respir Viruses ; 18(7): e13348, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38949103

RESUMEN

Understanding the clinical spectrum of SARS-CoV-2 infection, including the asymptomatic fraction, is important as asymptomatic individuals are still able to infect other individuals and contribute to ongoing transmission. The WHO Unity Household transmission investigation (HHTI) protocol provides a platform for the prospective and systematic collection of high-quality clinical, epidemiological, serological and virological data from SARS-CoV-2 confirmed cases and their household contacts. These data can be used to understand key severity and transmissibility parameters-including the asymptomatic proportion-in relation to local epidemic context and help inform public health response. We aimed to estimate the asymptomatic proportion of SARS-CoV-2 Omicron variant infections in Unity-aligned HHTIs. We conducted a systematic review and meta-analysis in alignment with the PRISMA 2020 guidelines and registered our systematic review on PROSPERO (CRD42022378648). We searched EMBASE, Web of Science, MEDLINE and bioRxiv and medRxiv from 1 November 2021 to 22 August 2023. We identified 8368 records, of which 98 underwent full text review. We identified only three studies for data extraction, with substantial variation in study design and corresponding estimates of the asymptomatic proportion. As a result, we did not generate a pooled estimate or I2 metric. The limited number of quality studies that we identified highlights the need for improved preparedness and response capabilities to facilitate robust HHTI implementation, analysis and reporting, to better inform national, regional and global risk assessments and policymaking.


Asunto(s)
Infecciones Asintomáticas , COVID-19 , Composición Familiar , SARS-CoV-2 , Humanos , Infecciones Asintomáticas/epidemiología , COVID-19/epidemiología , COVID-19/transmisión , COVID-19/virología , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación
3.
World J Gastroenterol ; 30(22): 2866-2880, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38947288

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by the highly pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily impacts the respiratory tract and can lead to severe outcomes such as acute respiratory distress syndrome, multiple organ failure, and death. Despite extensive studies on the pathogenicity of SARS-CoV-2, its impact on the hepatobiliary system remains unclear. While liver injury is commonly indicated by reduced albumin and elevated bilirubin and transaminase levels, the exact source of this damage is not fully understood. Proposed mechanisms for injury include direct cytotoxicity, collateral damage from inflammation, drug-induced liver injury, and ischemia/hypoxia. However, evidence often relies on blood tests with liver enzyme abnormalities. In this comprehensive review, we focused solely on the different histopathological manifestations of liver injury in COVID-19 patients, drawing from liver biopsies, complete autopsies, and in vitro liver analyses. We present evidence of the direct impact of SARS-CoV-2 on the liver, substantiated by in vitro observations of viral entry mechanisms and the actual presence of viral particles in liver samples resulting in a variety of cellular changes, including mitochondrial swelling, endoplasmic reticulum dilatation, and hepatocyte apoptosis. Additionally, we describe the diverse liver pathology observed during COVID-19 infection, encompassing necrosis, steatosis, cholestasis, and lobular inflammation. We also discuss the emergence of long-term complications, notably COVID-19-related secondary sclerosing cholangitis. Recognizing the histopathological liver changes occurring during COVID-19 infection is pivotal for improving patient recovery and guiding decision-making.


Asunto(s)
COVID-19 , Hígado , SARS-CoV-2 , Humanos , COVID-19/complicaciones , COVID-19/patología , COVID-19/virología , Hígado/patología , Hígado/virología , SARS-CoV-2/patogenicidad , Hepatopatías/patología , Hepatopatías/virología , Hepatopatías/etiología , Hepatocitos/patología , Hepatocitos/virología
4.
Nat Commun ; 15(1): 5503, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951531

RESUMEN

Proline is widely known as the only proteogenic amino acid with a secondary amine. In addition to its crucial role in protein structure, the secondary amino acid modulates neurotransmission and regulates the kinetics of signaling proteins. To understand the structural basis of proline import, we solved the structure of the proline transporter SIT1 in complex with the COVID-19 viral receptor ACE2 by cryo-electron microscopy. The structure of pipecolate-bound SIT1 reveals the specific sequence requirements for proline transport in the SLC6 family and how this protein excludes amino acids with extended side chains. By comparing apo and substrate-bound SIT1 states, we also identify the structural changes that link substrate release and opening of the cytoplasmic gate and provide an explanation for how a missense mutation in the transporter causes iminoglycinuria.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Microscopía por Crioelectrón , Prolina , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/genética , Prolina/metabolismo , Humanos , SARS-CoV-2/metabolismo , SARS-CoV-2/genética , COVID-19/virología , COVID-19/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/química , Modelos Moleculares
5.
J Med Microbiol ; 73(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38963412

RESUMEN

Introduction. In India, the SARS-CoV-2 Delta wave (2020-2021) faded away with the advent of the Omicron variants (2021-present). Dengue incidences were observed to be less in Southeast Asia during the active years of the pandemic (2020-2021). However, dengue virus type 3 (DV3) cases were increasingly reported in this region (including India) concurrent with the progression of the Omicron waves since 2022.Hypothesis. What could be the reason(s) behind this unusual DV3 surge after an overall dip in dengue incidences in many parts of Southeast Asia?Aim. We, therefore, investigated the current state of cross-reactivity of prevalent (Omicron era) SARS-CoV-2 serums with different DV serotypes and evaluated the impact of such serums on DV neutralization in cell culture.Methodology. Fifty-five COVID-19 serum samples (January-September 2022) and three pre-pandemic archived serum samples from apparently healthy individuals were tested for DV or SARS-CoV-2 IgM/IgG using the lateral flow immunoassays. DV1-4 virus neutralization tests (VNTs) were done with the SARS-CoV-2 antibody (Ab)-positive serums in Huh7 cells. DV3 envelope (env) gene was PCR amplified and sequenced for three archived DV isolates, one from 2017 and two from 2021.Results. SARS-CoV-2 Ab-positive samples constituted 74.5 % of the serums. Of these, 41.5 % were DV cross-reactive and 58.5 % were not. The DV cross-reactive serums neutralized all DV serotypes (DV1-4), as per previous results and this study. The DV non-cross-reactive serums (58.5 %) also cross-neutralized DV1, 2 and 4 but increased DV3 infectivity by means of antibody-dependent enhancement of infection as evident from significantly higher DV3 titres in VNT compared to control serums. The DV3 envelope was identical among the three isolates, including isolate 1 used in VNTs. Our results suggest that DV cross-reactivity of SARS-CoV-2 serums diminished with the shift from Delta to Omicron prevalence. Such COVID-19 serums (DV non-cross-reactive) might have played a major role in causing DV3 surge during the Omicron waves.Conclusion. Patients suspected of dengue or COVID-19 should be subjected to virus/antigen tests and serological tests for both the diseases for definitive diagnosis, prognosis and disease management.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Reacciones Cruzadas , Virus del Dengue , SARS-CoV-2 , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/virología , COVID-19/epidemiología , COVID-19/sangre , COVID-19/inmunología , Anticuerpos Antivirales/sangre , Virus del Dengue/genética , Virus del Dengue/inmunología , Virus del Dengue/clasificación , India/epidemiología , Dengue/virología , Dengue/sangre , Dengue/epidemiología , Dengue/inmunología , Pruebas de Neutralización , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre
6.
Nat Commun ; 15(1): 5577, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38956082

RESUMEN

Recent advances in single-cell immune profiling have enabled the simultaneous measurement of transcriptome and T cell receptor (TCR) sequences, offering great potential for studying immune responses at the cellular level. However, integrating these diverse modalities across datasets is challenging due to their unique data characteristics and technical variations. Here, to address this, we develop the multimodal generative model mvTCR to fuse modality-specific information across transcriptome and TCR into a shared representation. Our analysis demonstrates the added value of multimodal over unimodal approaches to capture antigen specificity. Notably, we use mvTCR to distinguish T cell subpopulations binding to SARS-CoV-2 antigens from bystander cells. Furthermore, when combined with reference mapping approaches, mvTCR can map newly generated datasets to extensive T cell references, facilitating knowledge transfer. In summary, we envision mvTCR to enable a scalable analysis of multimodal immune profiling data and advance our understanding of immune responses.


Asunto(s)
COVID-19 , Receptores de Antígenos de Linfocitos T , SARS-CoV-2 , Análisis de la Célula Individual , Transcriptoma , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Análisis de la Célula Individual/métodos , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/inmunología , COVID-19/virología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Perfilación de la Expresión Génica/métodos , Antígenos Virales/inmunología , Antígenos Virales/genética
7.
Sci Rep ; 14(1): 15058, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38956433

RESUMEN

Since the COVID-19 pandemic, the diversity of clinical manifestations in patients has been a tremendous challenge. It seems that genetic variations, as one of the players, contribute to the variety of symptoms. Genome-wide association studies have demonstrated the influence of certain genomic regions on the disease prognosis. Particularly, a haplotype at 3p21.31 locus, inherited from Neanderthals, showed an association with COVID-19 severity. Despite several studies regarding this haplotype, some key variants are not sufficiently addressed. In the present study, we investigated the association of rs17713054 at 3p21.31 with COVID-19 severity. We analyzed the genotype of 251 Iranian COVID-19 patients (151 patients with asymptomatic to mild form as control and 100 patients with severe to critical symptoms without any comorbidities as case group) using the ARMS-PCR method. Results demonstrated that the A allele confers an almost twofold increased risk for COVID-19 severity (P value = 0.008). The AA genotype also raises the risk by more than 11 times following the recessive model (P value = 0.013). In conclusion, the A allele in rs17713054 was a risk allele in Iranian patients and was independently associated with COVID-19 severity. More studies are beneficial to confirm these findings in other populations and to develop strategies for risk assessment, prevention, and personalized medicine.


Asunto(s)
COVID-19 , Predisposición Genética a la Enfermedad , Hombre de Neandertal , Polimorfismo de Nucleótido Simple , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Humanos , COVID-19/genética , COVID-19/virología , COVID-19/epidemiología , Irán/epidemiología , Hombre de Neandertal/genética , Masculino , Femenino , Persona de Mediana Edad , Animales , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Adulto , Haplotipos , Cromosomas Humanos Par 3/genética , Alelos , Estudio de Asociación del Genoma Completo , Genotipo , Anciano
8.
Sci Adv ; 10(27): eadg3747, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38959314

RESUMEN

Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.


Asunto(s)
Adyuvantes Inmunológicos , Pirimidinas , Receptor Toll-Like 7 , Receptor Toll-Like 8 , Humanos , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/metabolismo , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Receptor Toll-Like 7/agonistas , Pirimidinas/farmacología , Pirimidinas/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Imidazoles/farmacología , Imidazoles/química , Células THP-1 , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , COVID-19/virología , COVID-19/inmunología , FN-kappa B/metabolismo , Femenino , Descubrimiento de Drogas/métodos , Inmunidad Innata/efectos de los fármacos
9.
Reprod Fertil Dev ; 362024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38968399

RESUMEN

Context In recent years, the COVID-19 pandemic became a threat to human health and induced global concern. The SARS-CoV-2 virus causes various disorders in the body's systems, and the reproductive system is no exception. Further, the rate of infertile couples is increasing and part of this is related to male infertility. Aims The aim of the present study was to investigate the impacts of COVID-19 infection history on semen quality in men referred to public and private infertility centres. Methods In this research, patients were divided into two groups: 88 men with a history of COVID-19 (Covid+) and 51 men without (Covid-). After semen collection, sperm parameters, fertilisation rate and oxidative stress were investigated. Key results Sperms with normal morphology and mature chromatin in patients with COVID-19 infection history decreased, and seminal oxidative stress and sperm DNA fragmentation were increased; moreover, the fertilisation rate in the Covid+ group decreased in compare to the Covid- group. Conclusion COVID-19 infection increases oxidative stress in the semen, so has a negative effect on some sperm parameters and fertilisation rate. Implications COVID-19 infection impairs semen quality by increasing in oxidative stress, thus reducing the fertility potential.


Asunto(s)
COVID-19 , Fragmentación del ADN , Infertilidad Masculina , Estrés Oxidativo , Análisis de Semen , Semen , Espermatozoides , Humanos , Masculino , COVID-19/complicaciones , COVID-19/epidemiología , COVID-19/virología , Adulto , Infertilidad Masculina/virología , Infertilidad Masculina/epidemiología , Estrés Oxidativo/fisiología , Espermatozoides/virología , Espermatozoides/patología , Semen/virología , SARS-CoV-2 , Clínicas de Fertilidad , Motilidad Espermática
10.
Sci Rep ; 14(1): 15433, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965354

RESUMEN

The COVID-19 pandemic continues to challenge healthcare systems globally, necessitating advanced tools for clinical decision support. Amidst the complexity of COVID-19 symptomatology and disease severity prediction, there is a critical need for robust decision support systems to aid healthcare professionals in timely and informed decision-making. In response to this pressing demand, we introduce BayesCovid, a novel decision support system integrating Bayesian network models and deep learning techniques. BayesCovid automates data preprocessing and leverages advanced computational methods to unravel intricate patterns in COVID-19 symptom dynamics. By combining Bayesian networks and Bayesian deep learning models, BayesCovid offers a comprehensive solution for uncovering hidden relationships between symptoms and predicting disease severity. Experimental validation demonstrates BayesCovid 's high prediction accuracy (83.52-98.97%). Our work represents a significant stride in addressing the urgent need for clinical decision support systems tailored to the complexities of managing COVID-19 cases. By providing healthcare professionals with actionable insights derived from sophisticated computational analysis, BayesCovid aims to enhance clinical decision-making, optimise resource allocation, and improve patient outcomes in the ongoing battle against the COVID-19 pandemic.


Asunto(s)
Teorema de Bayes , COVID-19 , Aprendizaje Profundo , Pandemias , COVID-19/epidemiología , COVID-19/virología , Humanos , SARS-CoV-2/aislamiento & purificación , Sistemas de Apoyo a Decisiones Clínicas , Inteligencia Artificial
11.
Sci Rep ; 14(1): 15363, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965389

RESUMEN

The therapeutic potential of insect-derived bioactive molecules as anti-SARS-CoV-2 agents has shown promising results. Hymenopteran venoms, notably from Apis mellifera (honeybee) and Vespa orientalis (oriental wasp), were examined for the first time in an in vitro setting for their potential anti-COVID-19 activity. This assessment utilized an immunodiagnostic system to detect the SARS-CoV-2 nucleocapsid antigen titer reduction. Further analyses, including cytotoxicity assays, plaque reduction assays, and in silico docking-based screening, were performed to evaluate the efficacy of the most potent venom. Results indicated that bee and wasp venoms contain bioactive molecules with potential therapeutic effects against SARS-CoV-2.Nevertheless, the wasp venom exhibited superior efficacy compared to bee venom, achieving a 90% maximal (EC90) concentration effect of antigen depletion at 0.184 mg/mL, in contrast to 2.23 mg/mL for bee venom. The cytotoxicity of the wasp venom was assessed on Vero E6 cells 48 h post-treatment using the MTT assay. The CC 50 of the cell growth was 0.16617 mg/mL for Vero E6 cells. The plaque reduction assay of wasp venom revealed 50% inhibition (IC50) at a 0.208 mg/mL concentration. The viral count at 50% inhibition was 2.5 × 104 PFU/mL compared to the initial viral count of 5 × 104 PFU/mL. In silico data for the wasp venom revealed a strong attraction to binding sites on the ACE2 protein, indicating ideal interactions. This substantiates the potential of wasp venom as a promising viral inhibitor against SARS-CoV-2, suggesting its consideration as a prospective natural preventive and curative antiviral drug. In conclusion, hymenopteran venoms, particularly wasp venom, hold promise as a source of potential therapeutic biomolecules against SARS-CoV-2. More research and clinical trials are needed to evaluate these results and investigate their potential for translation into innovative antiviral therapies.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Simulación del Acoplamiento Molecular , SARS-CoV-2 , Venenos de Avispas , Células Vero , SARS-CoV-2/efectos de los fármacos , Chlorocebus aethiops , Animales , Humanos , Antivirales/farmacología , COVID-19/virología , Venenos de Avispas/farmacología , Venenos de Avispas/química , Venenos de Abeja/farmacología , Venenos de Abeja/química , Egipto , Abejas , Avispas
12.
Cell Commun Signal ; 22(1): 349, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965547

RESUMEN

T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.


Asunto(s)
Basigina , COVID-19 , Linfopenia , SARS-CoV-2 , Humanos , Linfopenia/inmunología , Linfopenia/virología , COVID-19/inmunología , COVID-19/virología , COVID-19/patología , SARS-CoV-2/metabolismo , Basigina/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Síndrome de Liberación de Citoquinas/inmunología , Animales
13.
PLoS One ; 19(7): e0306554, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38968178

RESUMEN

The differences of SARS-CoV-2 variants brought the changes of transmission characteristics and clinical manifestations during the prevalence of COVID-19. In order to explore the evolution mechanisms of SARS-CoV-2 variants and the impacts of variant evolution, the classic SIR (Susceptible-Infected-Recovered) compartment model was modified to a generalized SVEIR (Susceptible-Vaccinated-Exposed-Infected-Recovered) compartment model with age-group and varying variants in this study. By using of the SVEIR model and least squares method, the optimal fittings against the surveillance data from Fujian Provincial Center for Disease Control and Prevention were performed for the five epidemics of Fujian Province. The main epidemiological characteristics such as basic reproduction number, effective reproduction number, sensitivity analysis, and cross-variant scenario investigations were extensively investigated during dynamic zero-COVID policy. The study results showed that the infectivities of the variants became fast from wild strain to the Delta variant, further to the Omicron variant. Meanwhile, the cross-variant investigations showed that the average incubation periods were shortened, and that the infection scales quickly enhanced. Further, the risk estimations with the new variants were performed without implements of the non-pharmaceutical interventions, based on the dominant variants XBB.1.9.1 and EG.5. The results of the risk estimations suggested that non-pharmaceutical interventions were necessary on the Chinese mainland for controlling severe infections and deaths, and also that the regular variant monitors were still workable against the aggressive variant evolution and the emergency of new transmission risks in the future.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/transmisión , COVID-19/epidemiología , COVID-19/virología , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , China/epidemiología , Número Básico de Reproducción , Modelos Epidemiológicos , Factores de Edad
14.
J Gene Med ; 26(7): e3710, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38967229

RESUMEN

BACKGROUND: Patients with non-small cell lung cancer (NSCLC) are susceptible to coronavirus disease-2019 (COVID-19), but current treatments are limited. Icariside II (IS), a flavonoid compound derived from the plant epimedin, showed anti-cancer,anti-inflammation and immunoregulation effects. The present study aimed to evaluate the possible effect and underlying mechanisms of IS on NSCLC patients with COVID-19 (NSCLC/COVID-19). METHODS: NSCLC/COVID-19 targets were defined as the common targets of NSCLC (collected from The Cancer Genome Atlas database) and COVID-19 targets (collected from disease database of Genecards, OMIM, and NCBI). The correlations of NSCLC/COVID-19 targets and survival rates in patients with NSCLC were analyzed using the survival R package. Prognostic analyses were performed using univariate and multivariate Cox proportional hazards regression models. Furthermore, the targets in IS treatment of NSCLC/COVID-19 were defined as the overlapping targets of IS (predicted from drug database of TMSCP, HERBs, SwissTarget Prediction) and NSCLC/COVID-19 targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis of these treatment targets were performed aiming to understand the biological process, cellular component, molecular function and signaling pathway. The hub targets were analyzed by a protein-protein interaction network and the binding capacity with IS was characterized by molecular docking. RESULTS: The hub targets for IS in the treatment of NSCLC/COVID-19 includes F2, SELE, MMP1, MMP2, AGTR1 and AGTR2, and the molecular docking results showed that the above target proteins had a good binding degree to IS. Network pharmacology showed that IS might affect the leucocytes migration, inflammation response and active oxygen species metabolic process, as well as regulate the interleukin-17, tumor necrosus factor and hypoxia-inducible factor-1 signaling pathway in NSCLC/COVID-19. CONCLUSIONS: IS may enhance the therapeutic efficacy of current clinical anti-inflammatory and anti-cancer therapy to benefit patients with NSCLC combined with COVID-19.


Asunto(s)
COVID-19 , Carcinoma de Pulmón de Células no Pequeñas , Flavonoides , Neoplasias Pulmonares , Simulación del Acoplamiento Molecular , Farmacología en Red , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , COVID-19/virología , COVID-19/metabolismo , Flavonoides/uso terapéutico , Flavonoides/química , Flavonoides/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Tratamiento Farmacológico de COVID-19 , Mapas de Interacción de Proteínas/efectos de los fármacos , Pronóstico
15.
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
16.
J Exp Med ; 221(9)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-38949638

RESUMEN

Studies during the COVID-19 pandemic showed that children had heightened nasal innate immune responses compared with adults. To evaluate the role of nasal viruses and bacteria in driving these responses, we performed cytokine profiling and comprehensive, symptom-agnostic testing for respiratory viruses and bacterial pathobionts in nasopharyngeal samples from children tested for SARS-CoV-2 in 2021-22 (n = 467). Respiratory viruses and/or pathobionts were highly prevalent (82% of symptomatic and 30% asymptomatic children; 90 and 49% for children <5 years). Virus detection and load correlated with the nasal interferon response biomarker CXCL10, and the previously reported discrepancy between SARS-CoV-2 viral load and nasal interferon response was explained by viral coinfections. Bacterial pathobionts correlated with a distinct proinflammatory response with elevated IL-1ß and TNF but not CXCL10. Furthermore, paired samples from healthy 1-year-olds collected 1-2 wk apart revealed frequent respiratory virus acquisition or clearance, with mucosal immunophenotype changing in parallel. These findings reveal that frequent, dynamic host-pathogen interactions drive nasal innate immune activation in children.


Asunto(s)
COVID-19 , Inmunidad Innata , SARS-CoV-2 , Humanos , Inmunidad Innata/inmunología , Preescolar , Lactante , COVID-19/inmunología , COVID-19/virología , Niño , SARS-CoV-2/inmunología , Femenino , Masculino , Nasofaringe/inmunología , Nasofaringe/virología , Nasofaringe/microbiología , Carga Viral , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Mucosa Nasal/microbiología , Citocinas/metabolismo , Citocinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Adolescente , Nariz/inmunología , Nariz/virología , Nariz/microbiología , Coinfección/inmunología , Coinfección/virología
17.
J Med Virol ; 96(7): e29752, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38949191

RESUMEN

Antiviral signaling, immune response and cell metabolism are dysregulated by SARS-CoV-2, the causative agent of COVID-19. Here, we show that SARS-CoV-2 accessory proteins ORF3a, ORF9b, ORF9c and ORF10 induce a significant mitochondrial and metabolic reprogramming in A549 lung epithelial cells. While ORF9b, ORF9c and ORF10 induced largely overlapping transcriptomes, ORF3a induced a distinct transcriptome, including the downregulation of numerous genes with critical roles in mitochondrial function and morphology. On the other hand, all four ORFs altered mitochondrial dynamics and function, but only ORF3a and ORF9c induced a marked alteration in mitochondrial cristae structure. Genome-Scale Metabolic Models identified both metabolic flux reprogramming features both shared across all accessory proteins and specific for each accessory protein. Notably, a downregulated amino acid metabolism was observed in ORF9b, ORF9c and ORF10, while an upregulated lipid metabolism was distinctly induced by ORF3a. These findings reveal metabolic dependencies and vulnerabilities prompted by SARS-CoV-2 accessory proteins that may be exploited to identify new targets for intervention.


Asunto(s)
COVID-19 , Mitocondrias , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Mitocondrias/metabolismo , COVID-19/metabolismo , COVID-19/virología , COVID-19/patología , Células A549 , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Transcriptoma , Sistemas de Lectura Abierta , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Viroporinas
18.
PLoS One ; 19(7): e0305077, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38985808

RESUMEN

Optimal timing for intubating patients with coronavirus disease 2019 (COVID-19) has been debated throughout the pandemic. Early use of high-flow nasal cannula (HFNC) can help reduce the need for intubation, but delay can result in poorer outcomes. This study examines trends in laboratory parameters and serum severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA levels of patients with COVID-19 in relation to HFNC failure. Patients requiring HFNC within three days of hospitalization between July 1 and September 30, 2021 were enrolled. The primary outcome was HFNC failure (early failure ≤Day 3; late failure ≥Day 4), defined as transfer to intensive care just before/after intubation or in-hospital death. We examined changes in laboratory markers and SARS-CoV2-RNAemia on Days 1, 4, and 7, together with demographic data, oxygenation status, and therapeutic agents. We conducted a univariate logistic regression with the explanatory variables defined as 10% change rate in each laboratory marker from Day 1 to 4. We utilized the log-rank test to assess the differences in HFNC failure rates, stratified based on the presence of SARS-CoV2 RNAemia. Among 122 patients, 17 (13.9%) experienced HFNC failure (early: n = 6, late: n = 11). Seventy-five patients (61.5%) showed an initial SpO2/FiO2 ratio ≤243, equivalent to PaO2/FiO2 ratio ≤200, and the initial SpO2/FiO2 ratio was significantly lower in the failure group (184 vs. 218, p = 0.018). Among the laboratory markers, a 10% increase from Day 1 to 4 of lactate dehydrogenase (LDH) and interleukin (IL)-6 was associated with late failure (Odds ratio [OR]: 1.42, 95% confidence interval [CI]: 1.09-1.89 and OR: 1.04, 95%CI: 1.00-1.19, respectively). Furthermore, in patients with persistent RNAemia on Day 4 or 7, the risk of late HFNC failure was significantly higher (Log-rank test, p<0.01). In conclusion, upward trends in LDH and IL-6 levels and the persistent RNAemia even after treatment were associated with HFNC failure.


Asunto(s)
Biomarcadores , COVID-19 , Terapia por Inhalación de Oxígeno , ARN Viral , SARS-CoV-2 , Humanos , COVID-19/terapia , COVID-19/sangre , COVID-19/virología , Masculino , Femenino , Biomarcadores/sangre , Persona de Mediana Edad , ARN Viral/sangre , Terapia por Inhalación de Oxígeno/métodos , Anciano , L-Lactato Deshidrogenasa/sangre , Insuficiencia del Tratamiento , Resultado del Tratamiento , Cánula
19.
J Infect Dev Ctries ; 18(6): 851-861, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38990988

RESUMEN

INTRODUCTION: In Senegal, molecular diagnosis was widely used for the detection and management of COVID-19 patients. However, genomic surveillance was very limited in the public sector. This study aimed to share the experience of a Senegalese public sector laboratory in response to the COVID-19 pandemic, and to describe the distribution of variants circulating in 2020 and 2021. METHODOLOGY: From July 2020 to December 2021, SARS-CoV-2 qRT-PCR was performed on nasopharyngeal samples from travelers and symptomatic patients at the Bacteriology and Virology Laboratory (LBV) of the Aristide le Dantec University Teaching Hospital. Samples with a cycle threshold (Ct) ≤ 30 were selected for whole-genome sequencing (WGS) using the Nanopore technology. In-house scripts were developed to study the spatial and temporal distribution of SARS-CoV-2 variants in Senegal, using our sequences and those retrieved from the GISAID database. RESULTS: Of 8,207 patients or travelers screened for SARS-CoV-2, 970 (11.8%) were positive and 386 had a Ct ≤ 30. WGS was performed on 133 samples. Concomitantly with high-quality sequences deposited in the GISAID database covering nine cities in Senegal in 2020 and 2021 (n = 1,539), we observed a high circulation of the 20A (B.1, B.1.416 and B.1.620) and 20B (B.1.1.420) lineages in 2020, while most of the samples belonged to Delta variants (AY34 and AY.34.1, 22%) in 2021. CONCLUSIONS: Despite its late involvement, COVID-19 diagnosis was routinely performed in LBV, but genomic characterization remained challenging. The genomic diversity of SARS-CoV-2 strains in Senegal reflected that observed worldwide during the first waves of the pandemic.


Asunto(s)
COVID-19 , Genoma Viral , SARS-CoV-2 , Humanos , Senegal/epidemiología , COVID-19/epidemiología , COVID-19/virología , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Secuenciación Completa del Genoma , Epidemiología Molecular , Nasofaringe/virología , Adulto , Masculino , Femenino , Filogenia , Persona de Mediana Edad
20.
Front Cell Infect Microbiol ; 14: 1394721, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975331

RESUMEN

Since 2019, Coronavirus Disease 2019(COVID-19) has affected millions of people worldwide. Except for acute respiratory distress syndrome, dysgeusis is also a common symptom of COVID-19 that burdens patients for weeks or permanently. However, the mechanisms underlying taste dysfunctions remain unclear. Here, we performed complete autopsies of five patients who died of COVID-19. Integrated tongue samples, including numerous taste buds, salivary glands, vessels, and nerves were collected to map the pathology, distribution, cell tropism, and receptor distribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the tongue. Our results revealed that all patients had moderate lymphocyte infiltration around the salivary glands and in the lamina propria adjacent to the mucosa, and pyknosis in the epithelia of taste buds and salivary glands. This may be because the serous acini, salivary gland ducts, and taste buds are the primary sites of SARS-CoV-2 infection. Multicolor immunofluorescence showed that SARS-CoV-2 readily infects Keratin (KRT)7+ taste receptor cells in taste buds, secretory cells in serous acini, and inner epithelial cells in the ducts. The major receptors, angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine subtype 2 (TMPRSS2), were both abundantly expressed in these cells. Viral antigens and receptor were both rarely detected in vessels and nerves. This indicates that SARS-CoV-2 infection triggers pathological injury in the tongue, and that dysgeusis may be directly related to viral infection and cellular damage.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Autopsia , COVID-19 , SARS-CoV-2 , Serina Endopeptidasas , Lengua , Tropismo Viral , Humanos , COVID-19/patología , COVID-19/virología , SARS-CoV-2/patogenicidad , Lengua/virología , Lengua/patología , Masculino , Enzima Convertidora de Angiotensina 2/metabolismo , Femenino , Persona de Mediana Edad , Serina Endopeptidasas/metabolismo , Glándulas Salivales/virología , Glándulas Salivales/patología , Anciano , Papilas Gustativas/virología , Papilas Gustativas/patología , Receptores Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...