Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 752
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Med Virol ; 96(5): e29660, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38727136

RESUMO

During the coronavirus disease 2019 (COVID-19) pandemic, known viral diseases declined in all ages. By using the current situation as a natural experiment, this study aimed to evaluate whether the change in the incidence of Kawasaki disease (KD) during the COVID-19 pandemic varies with age and whether a specific infectious disease mediates the occurrence of KD. Monthly number of KD patients were extracted from the nationwide inpatient database. Segmented regression analysis was conducted on the interrupted time series data. Additionally, causal mediation analysis was performed to examine the role of viral infections in the changes in the number of KD patients. After the first emergency declaration for COVID-19 in Japan, there was an immediate decrease in the number of KD patients per 100 000 population aged between 6 months and 4 years (immediate change = -2.66; 95% confidence interval [CI]: -5.16 to -0.16) and aged 5-15 years (immediate change = -0.26; 95% CI: -0.49 to -0.04). However, no immediate change was observed in patients under 6 months of age. In the causal mediation analysis for each viral infection, it was found that the decrease in the number of patients with KD was mediated by changes in the number of patients with pharyngoconjunctival fever and infectious gastroenteritis. The current results suggest that viral infections may be one of the etiological agents for KD, while they may not be the main cause in early infancy. Specifically, we found that adenovirus infection and gastroenteritis was closely related to the onset of KD in some areas of Japan.


Assuntos
COVID-19 , Síndrome de Linfonodos Mucocutâneos , Humanos , Síndrome de Linfonodos Mucocutâneos/epidemiologia , Síndrome de Linfonodos Mucocutâneos/virologia , COVID-19/epidemiologia , COVID-19/complicações , Pré-Escolar , Japão/epidemiologia , Lactente , Criança , Adolescente , Incidência , Masculino , Feminino , Viroses/epidemiologia , Viroses/complicações , SARS-CoV-2/patogenicidade
2.
ACS Infect Dis ; 10(5): 1431-1457, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38682683

RESUMO

Mucormycosis, a rare but deadly fungal infection, was an epidemic during the COVID-19 pandemic. The rise in cases (COVID-19-associated mucormycosis, CAM) is attributed to excessive steroid and antibiotic use, poor hospital hygiene, and crowded settings. Major contributing factors include diabetes and weakened immune systems. The main manifesting forms of CAM─cutaneous, pulmonary, and the deadliest, rhinocerebral─and disseminated infections elevated mortality rates to 85%. Recent focus lies on small-molecule inhibitors due to their advantages over standard treatments like surgery and liposomal amphotericin B (which carry several long-term adverse effects), offering potential central nervous system penetration, diverse targets, and simpler dosing owing to their small size, rendering the ability to traverse the blood-brain barrier via passive diffusion facilitated by the phospholipid membrane. Adaptation and versatility in mucormycosis are facilitated by a multitude of virulence factors, enabling the pathogen to dynamically respond to various environmental stressors. A comprehensive understanding of these virulence mechanisms is imperative for devising effective therapeutic interventions against this highly opportunistic pathogen that thrives in immunocompromised individuals through its angio-invasive nature. Hence, this Review delineates the principal virulence factors of mucormycosis, the mechanisms it employs to persist in challenging host environments, and the current progress in developing small-molecule inhibitors against them.


Assuntos
Antifúngicos , Inteligência Artificial , COVID-19 , Mucormicose , Fatores de Virulência , Mucormicose/tratamento farmacológico , Humanos , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/metabolismo , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade
3.
Nat Microbiol ; 9(5): 1293-1311, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38622380

RESUMO

Children infected with SARS-CoV-2 rarely progress to respiratory failure. However, the risk of mortality in infected people over 85 years of age remains high. Here we investigate differences in the cellular landscape and function of paediatric (<12 years), adult (30-50 years) and older adult (>70 years) ex vivo cultured nasal epithelial cells in response to infection with SARS-CoV-2. We show that cell tropism of SARS-CoV-2, and expression of ACE2 and TMPRSS2 in nasal epithelial cell subtypes, differ between age groups. While ciliated cells are viral replication centres across all age groups, a distinct goblet inflammatory subtype emerges in infected paediatric cultures and shows high expression of interferon-stimulated genes and incomplete viral replication. In contrast, older adult cultures infected with SARS-CoV-2 show a proportional increase in basaloid-like cells, which facilitate viral spread and are associated with altered epithelial repair pathways. We confirm age-specific induction of these cell types by integrating data from in vivo COVID-19 studies and validate that our in vitro model recapitulates early epithelial responses to SARS-CoV-2 infection.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Células Epiteliais , Mucosa Nasal , SARS-CoV-2 , Serina Endopeptidases , Humanos , COVID-19/virologia , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , SARS-CoV-2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Adulto , Pessoa de Meia-Idade , Idoso , Células Epiteliais/virologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Mucosa Nasal/virologia , Criança , Fatores Etários , Replicação Viral , Pré-Escolar , Tropismo Viral , Masculino , Feminino , Idoso de 80 Anos ou mais , Células Cultivadas , Adolescente , Lactente
4.
Viruses ; 16(4)2024 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-38675974

RESUMO

The Omicron variant of SARS-CoV-2, characterized by multiple subvariants including BA.1, XBB.1.5, EG.5, and JN.1, became the predominant strain in early 2022. Studies indicate that Omicron replicates less efficiently in lung tissue compared to the ancestral strain. However, the infectivity of Omicron in the gastrointestinal tract is not fully defined, despite the fact that 70% of COVID-19 patients experience digestive disease symptoms. Here, using primary human colonoids, we found that, regardless of individual variability, Omicron infects colon cells similarly or less effectively than the ancestral strain or the Delta variant. The variant induced limited type III interferon expression and showed no significant impact on epithelial integrity. Further experiments revealed inefficient cell-to-cell spread and spike protein cleavage in the Omicron spike protein, possibly contributing to its lower infectious particle levels. The findings highlight the variant-specific replication differences in human colonoids, providing insights into the enteric tropism of Omicron and its relevance to long COVID symptoms.


Assuntos
COVID-19 , Colo , Células Epiteliais , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , Colo/virologia , COVID-19/virologia , Células Epiteliais/virologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Replicação Viral , Interferon lambda
5.
Viruses ; 16(4)2024 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-38675991

RESUMO

Tumor necrosis factor (TNF) and interferon-gamma (IFNγ) are important inflammatory mediators in the development of cytokine storm syndrome (CSS). Single nucleotide polymorphisms (SNPs) regulate the expression of these cytokines, making host genetics a key factor in the prognosis of COVID-19. In this study, we investigated the associations of the TNF -308G/A and IFNG +874T/A polymorphisms with COVID-19. We analyzed the frequencies of the two polymorphisms in the control groups (CG: TNF -308G/A, n = 497; IFNG +874T/A, n = 397), a group of patients with COVID-19 (CoV, n = 222) and among the subgroups of patients with nonsevere (n = 150) and severe (n = 72) COVID-19. We found no significant difference between the genotypic and allelic frequencies of TNF -308G/A in the groups analyzed; however, both the frequencies of the high expression genotype (TT) (CoV: 13.51% vs. CG: 6.30%; p = 0.003) and the *T allele (CoV: 33.56% vs. CG: 24. 81%; p = 0.001) of the IFNG +874T/A polymorphism were higher in the COVID-19 group than in the control group, with no differences between the subgroups of patients with nonsevere and severe COVID-19. The *T allele of IFNG +874T/A (rs2430561) is associated with susceptibility to symptomatic COVID-19. These SNPs provided valuables clues about the potential mechanism involved in the susceptibility to developing symptomatic COVID-19.


Assuntos
COVID-19 , Predisposição Genética para Doença , Genótipo , Interferon gama , SARS-CoV-2 , Feminino , Humanos , Masculino , Alelos , COVID-19/genética , COVID-19/virologia , Síndrome da Liberação de Citocina/genética , Frequência do Gene , Interferon gama/genética , Polimorfismo de Nucleotídeo Único , SARS-CoV-2/patogenicidade , Fator de Necrose Tumoral alfa/genética
6.
Cell ; 187(3): 596-608.e17, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38194966

RESUMO

BA.2.86, a recently identified descendant of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron BA.2 sublineage, contains ∼35 mutations in the spike (S) protein and spreads in multiple countries. Here, we investigated whether the virus exhibits altered biological traits, focusing on S protein-driven viral entry. Employing pseudotyped particles, we show that BA.2.86, unlike other Omicron sublineages, enters Calu-3 lung cells with high efficiency and in a serine- but not cysteine-protease-dependent manner. Robust lung cell infection was confirmed with authentic BA.2.86, but the virus exhibited low specific infectivity. Further, BA.2.86 was highly resistant against all therapeutic antibodies tested, efficiently evading neutralization by antibodies induced by non-adapted vaccines. In contrast, BA.2.86 and the currently circulating EG.5.1 sublineage were appreciably neutralized by antibodies induced by the XBB.1.5-adapted vaccine. Collectively, BA.2.86 has regained a trait characteristic of early SARS-CoV-2 lineages, robust lung cell entry, and evades neutralizing antibodies. However, BA.2.86 exhibits low specific infectivity, which might limit transmissibility.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19 , SARS-CoV-2 , Humanos , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Caspases/metabolismo , COVID-19/imunologia , COVID-19/virologia , Pulmão/virologia , SARS-CoV-2/classificação , SARS-CoV-2/genética , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Internalização do Vírus , Glicoproteína da Espícula de Coronavírus/genética
7.
J Virol ; 97(12): e0127623, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-37975674

RESUMO

ABSTRACT: Disease progression during SARS-CoV-2 infection is tightly linked to the fate of lung epithelial cells, with severe cases of COVID-19 characterized by direct injury of the alveolar epithelium and an impairment in its regeneration from progenitor cells. The molecular pathways that govern respiratory epithelial cell death and proliferation during SARS-CoV-2 infection, however, remain unclear. We now report a high-throughput CRISPR screen for host genetic modifiers of the survival and proliferation of SARS-CoV-2-infected Calu-3 respiratory epithelial cells. The top four genes identified in our screen encode components of the same type I interferon (IFN-I) signaling complex­IFNAR1, IFNAR2, JAK1, and TYK2. The fifth gene, ACE2, was an expected control encoding the SARS-CoV-2 viral receptor. Surprisingly, despite the antiviral properties of IFN-I signaling, its disruption in our screen was associated with an increase in Calu-3 cell fitness. We validated this effect and found that IFN-I signaling did not sensitize SARS-CoV-2-infected cultures to cell death but rather inhibited the proliferation of surviving cells after the early peak of viral replication and cytopathic effect. We also found that IFN-I signaling alone, in the absence of viral infection, was sufficient to induce this delayed antiproliferative response in both Calu-3 cells and iPSC-derived type 2 alveolar epithelial cells. Together, these findings highlight a cell autonomous antiproliferative response by respiratory epithelial cells to persistent IFN-I signaling during SARS-CoV-2 infection. This response may contribute to the deficient alveolar regeneration that has been associated with COVID-19 lung injury and represents a promising area for host-targeted therapeutic development.


Assuntos
COVID-19 , Células Epiteliais , Interferon Tipo I , Pulmão , Humanos , COVID-19/imunologia , COVID-19/patologia , COVID-19/virologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Interferon Tipo I/imunologia , Pulmão/patologia , Pulmão/virologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Linhagem Celular , Proliferação de Células
8.
Clin Exp Med ; 23(7): 3251-3264, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37055652

RESUMO

BACKGROUND: Some human polymorphisms of ACE1, ACE2, IFITM3, TMPRSS2 and TNFα genes may have an effect on the susceptibility to SARS-CoV-2 infection and increase the risk to develop severe COVID-19. We conducted a systematic review of current evidence to investigate the association of genetic variants of these genes with the susceptibility to virus infection and patient prognosis. METHODS: We systematically searched Medline, Embase and The Cochrane Library for articles published until May 2022, and included observational studies covering genetic association of ACE1, ACE2, IFITM3, TMPRSS2 and TNFα genes with COVID-19 susceptibility or prognosis. We evaluated the methodological quality of included studies, and pooled data as convenient in meta-analysis (MA). Odds ratio (OR) values and 95% confidence intervals were calculated. RESULTS: We included 35 studies (20 on ACE, 5 each on IFITM3, TMPRSS2, TNFα), enrolling 21,452 participants, of them 9401 were COVID-19 confirmed cases. ACE1 rs4646994 and rs1799752, ACE2 rs2285666, TMPRSS2 rs12329760, IFITM3 rs12252 and TNFα rs1800629 were identifies as common polymorphisms. Our MA showed an association between genetic polymorphisms and susceptibility to SARS-CoV-2 infection for IFITM3 rs12252 CC (OR 5.67) and CT (OR 1.64) genotypes. Furthermore, MA uncovered that both ACE DD (OR 1.27) and IFITM3 CC (OR 2.26) genotypes carriers had a significantly increased risk of developing severe COVID-19. DISCUSSION: These results provide a critical evaluation of genetic polymorphisms as predictors in SARS-CoV-2 infection. ACE1 DD and IFITM3 CC polymorphisms would lead to a genetic predisposition for severe lung injury in patients with COVID-19.


Assuntos
COVID-19 , Humanos , Enzima de Conversão de Angiotensina 2/genética , COVID-19/genética , COVID-19/metabolismo , Proteínas de Membrana/genética , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Polimorfismo Genético , Proteínas de Ligação a RNA/genética , SARS-CoV-2/patogenicidade , Serina Endopeptidases/genética , Fator de Necrose Tumoral alfa/genética
9.
Viruses ; 15(3)2023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36992445

RESUMO

Human coronavirus (HCoV)-NL63 is an important contributor to upper and lower respiratory tract infections, mainly in children, while severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, can cause lower respiratory tract infections, and more severe, respiratory and systemic disease, which leads to fatal consequences in many cases. Using microscopy, immunohistochemistry (IHC), virus-binding assay, reverse transcriptase qPCR (RT-qPCR) assay, and flow cytometry, we compared the characteristics of the susceptibility, replication dynamics, and morphogenesis of HCoV-NL63 and SARS-CoV-2 in monolayer cultures of primary human respiratory epithelial cells (HRECs). Less than 10% HRECs expressed ACE2, and SARS-CoV-2 seemed much more efficient than HCoV-NL63 at infecting the very small proportion of HRECs expressing the ACE2 receptors. Furthermore, SARS-CoV-2 replicated more efficiently than HCoV-NL63 in HREC, which correlates with the cumulative evidence of the differences in their transmissibility.


Assuntos
Coronavirus Humano NL63 , Células Epiteliais , SARS-CoV-2 , Humanos , Enzima de Conversão de Angiotensina 2 , Linhagem Celular , Coronavirus Humano NL63/patogenicidade , COVID-19 , Células Epiteliais/virologia , Infecções Respiratórias , SARS-CoV-2/patogenicidade
10.
Ann Diagn Pathol ; 63: 152102, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36634551

RESUMO

The toll like receptors (TLRs) and RIG-1 are proteins involved in the initial reaction of the innate immune system to infectious diseases and, thus, can provide much information to the surgical pathologist in terms of the molecular dynamics of the infection. The TLRs (TLR1, 2, 3, 4, 7, 8) and RIG-1 distribution as determined by immunohistochemistry was examined in the following diseases: human papillomavirus (n = 30 including 15 squamous intraepithelial lesions (SIL), 5 cancers, and 10 controls); molluscum contagiosum (n = 8 including 4 controls), SARS-CoV2 (n = 52 including 20 mild, 5 fatal, and 27 controls) and reovirus infection as oncolytic therapy. Mild, regressing infection (molluscum contagiosum, mild SARS-CoV2 and low grade SIL) each showed the same pattern: marked up regulation of at least three of the TLRs/RIG-1 with decreased expression of none compared to the controls. Severe infection (fatal SARS-CoV2, and cervical cancer) each showed marked decrease expression in at least three of the TLRs/RIG-1. We recently documented an equivalent marked decrease expression of the TLRs/RIG-1 in the placenta in fatal in utero infections. The reoviral infected tissues showed an overall pattern of marked increase expression of TLRs/RIG-1, consistent with a strong anti-viral response. Thus, the in situ testing of infectious diseases by a panel of these early infectious disease recognition proteins may allow the surgical pathologist to predict the outcome of the disease which, in turn, may assist in the understanding of the role of the TLRs/RIG-1 in determining the fate of a given infectious process.


Assuntos
Doenças Transmissíveis , Proteína DEAD-box 58 , Receptores Toll-Like , Feminino , Humanos , Gravidez , Doenças Transmissíveis/genética , Doenças Transmissíveis/patologia , COVID-19/genética , COVID-19/patologia , Molusco Contagioso/genética , Molusco Contagioso/patologia , RNA Viral , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Receptores Toll-Like/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo
11.
J Med Virol ; 95(1): e28383, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36477795

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global coronavirus disease 2019 (COVID-19) pandemic that has affected the lives of billions of individuals. However, the host-virus interactions still need further investigation to reveal the underling mechanism of SARS-CoV-2 pathogenesis. Here, transcriptomics analysis of SARS-CoV-2 infection highlighted possible correlation between host-associated signaling pathway and virus. In detail, cAMP-protein kinase (PKA) pathway has an essential role in SARS-CoV-2 infection, followed by the interaction between cyclic AMP response element binding protein (CREB) and CREB-binding protein (CBP) could be induced and leading to the enhancement of CREB/CBP transcriptional activity. The replication of Delta and Omicron BA.5 were inhibited by about 49.4% and 44.7% after knockdown of CREB and CBP with small interfering RNAs, respectively. Furthermore, a small organic molecule naphthol AS-E (nAS-E), which targets on the interaction between CREB and CBP, potently inhibited SARS-CoV-2 wild-type (WT) infection with comparable the half-maximal effective concentration (EC50 ) 1.04 µM to Remdesivir 0.57 µM. Compared with WT virus, EC50 in Calu-3 cells against Delta, Omicron BA.2, and Omicron BA.5 were, on average, 1.5-fold, 1.1-fold, and 1.5-fold higher, respectively, nAS-E had a satisfied antiviral effect against Omicron variants. Taken together, our study demonstrated the importance of CREB/CBP induced by cAMP-PKA pathway during SARS-CoV-2 infection, and further provided a novel CREB/CBP interaction therapeutic drug targets for COVID-19.


Assuntos
COVID-19 , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Interações Hospedeiro-Patógeno , Humanos , COVID-19/metabolismo , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Proteína de Ligação a CREB/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia
12.
Arq. ciências saúde UNIPAR ; 26(3): 1427-1436, set-dez. 2022.
Artigo em Português | LILACS | ID: biblio-1414675

RESUMO

A pandemia de COVID-19 se propagou rapidamente pelo mundo, causada pela infecção do novo coronavírus (SARS-CoV-2), que surgiu na China no final de 2019. Apesar da porta de entrada mais comum do agente etiológico ser pelo trato respiratório, evidências demonstram que a doença pode apresentar sintomas extrapulmonares, como os do trato gastrointestinal. Descrever sobre possíveis alterações gastrointestinais ocasionadas em pacientes infectados pelo SARS-CoV-2. Tratou-se de uma revisão bibliográfica, que utilizou artigos científicos disponíveis na íntegra em bases de dados Medical Literature Analysis and Retrieval System Online, Google Acadêmico, Scientific Electronic Library Online, nos meses de abril a outubro de 2021, além de monografias, dissertações, teses e livros. Foram utilizados como descritores as palavras: SARS-CoV-2 e intestino, COVID-19 e intestino, alterações intestinais na COVID-19. Os distúrbios gastrointestinais mais prevalentes são náuseas, vômitos e diarreia e dor abdominal. O papel da microbiota intestinal em influenciar as doenças pulmonares foi bem articulado, devido à existência do eixo intestino-pulmão, a inflamação em um desses órgãos interfere diretamente no perfil inflamatório no outro. Embora ainda não esteja totalmente esclarecido se os sintomas gastrointestinais indicam maior viremia ou um processo fisiopatológico alternativo, observa-se que a presença destes configura um fator de risco para a maior severidade da doença.


The COVID-19 pandemic has spread rapidly around the world, caused by the infection of the new coronavirus (SARS-CoV-2), which emerged in China at the end of 2019. respiratory evidence shows that the disease can present extrapulmonary symptoms, such as those in the gastrointestinal tract. Objective: To describe possible gastrointestinal alterations caused in patients infected by SARS-CoV-2. Methodology: this was a literature review, which used scientific articles available in full in the Medical Literature Analysis and Retrieval System Online (MEDLINE), Academic Google, Scientific Electronic Library Online (SciELO) databases, as well as monographs, dissertations, theses and books. The words used as descriptors were: SARS-CoV-2 and intestine, COVID-19 and intestine, intestinal alterations in COVID-19. Development: The most prevalent gastrointestinal disorders are nausea, vomiting and diarrhea and abdominal pain. The role of the intestinal microbiota in influencing lung diseases was well articulated, due to the existence of the gut- lung axis, inflammation in one of these organs directly interfering with the inflammatory profile in the other. Conclusion: Although it is not yet fully understood whether the gastrointestinal symptoms


La pandemia COVID-19 se ha extendido rápidamente por todo el mundo, causada por la infección del nuevo coronavirus (SARS-CoV-2), que surgió en China a finales de 2019. Las evidencias respiratorias muestran que la enfermedad puede presentar síntomas extrapulmonares, como los del tracto gastrointestinal. Objetivo: Describir las posibles alteraciones gastrointestinales causadas en pacientes infectados por SARS-CoV-2. Metodología: se trató de una revisión bibliográfica, que utilizó artículos científicos disponibles en su totalidad en las bases de datos Medical Literature Analysis and Retrieval System Online (MEDLINE), Academic Google, Scientific Electronic Library Online (SciELO), así como monografías, disertaciones, tesis y libros. Las palabras utilizadas como descriptores fueron: SARS-CoV-2 e intestino, COVID-19 e intestino, alteraciones intestinales en COVID-19. Desarrollo: Las alteraciones gastrointestinales más prevalentes son náuseas, vómitos y diarrea y dolor abdominal. Se articuló bien el papel de la microbiota intestinal en la influencia de las enfermedades pulmonares, debido a la existencia del eje intestino-pulmón, la inflamación en uno de estos órganos interfiere directamente en el perfil inflamatorio del otro. Conclusiones: Aunque aún no se comprenda del todo si los síntomas gastrointestinales indican una mayor viremia o un proceso fisiopatológico alternativo, se observa que su presencia es un factor de riesgo para la mayor gravedad de la enfermedad.


Assuntos
Doenças do Sistema Digestório/patologia , SARS-CoV-2/patogenicidade , Diarreia/diagnóstico , COVID-19/epidemiologia , COVID-19/virologia
14.
Femina ; 50(10): 631-640, out. 30, 2022. ilus
Artigo em Português | LILACS | ID: biblio-1414423

RESUMO

Esta revisão narrativa teve como objetivo avaliar possíveis riscos da associação entre a infecção por SARS-CoV-2 (causa da COVID-19) e as características metabólicas e endócrinas frequentemente encontradas em mulheres com a síndrome dos ovários policísticos (SOP). A COVID-19 é mais grave em indivíduos com obesidade, diabetes mellitus, dislipidemia e hipertensão arterial. Como essas condições são comorbidades comumente associadas à SOP, foi hipotetizado que mulheres com SOP teriam maior risco de adquirir COVID-19 e desenvolver formas clínicas mais graves da doença. Considerando vários estudos epidemiológicos, a presente revisão mostra que mulheres com SOP têm risco 28% a 50% maior de serem infectadas pelo vírus SARS-CoV-2 em todas as idades e que, nessas mulheres, a COVID-19 está associada a maiores taxas de hospitalização, morbidade e mortalidade, especialmente naquelas com alterações no metabolismo de carboidratos e lipídios, hiperandrogenemia e aumento do tecido adiposo visceral. Os mecanismos que explicam o maior risco de infecção por COVID-19 em mulheres com SOP são considerados.(AU)


Assuntos
Humanos , Feminino , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/epidemiologia , SARS-CoV-2/patogenicidade , COVID-19/fisiopatologia , COVID-19/epidemiologia , Deficiência de Vitamina D , Grupos de Risco , Resistência à Insulina , Comorbidade , Fatores de Risco , Bases de Dados Bibliográficas , Hiperandrogenismo , Diabetes Mellitus , Dislipidemias , Hipertensão , Inflamação , Obesidade
15.
Nature ; 611(7935): 346-351, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36130725

RESUMO

Clinical outcomes of severe acute respiratory syndrome 2 (SARS-CoV-2) infection are highly heterogeneous, ranging from asymptomatic infection to lethal coronavirus disease 2019 (COVID-19). The factors underlying this heterogeneity remain insufficiently understood. Genetic association studies have suggested that genetic variants contribute to the heterogeneity of COVID-19 outcomes, but the underlying potential causal mechanisms are insufficiently understood. Here we show that common variants of the apolipoprotein E (APOE) gene, homozygous in approximately 3% of the world's population1 and associated with Alzheimer's disease, atherosclerosis and anti-tumour immunity2-5, affect COVID-19 outcome in a mouse model that recapitulates increased susceptibility conferred by male sex and advanced age. Mice bearing the APOE2 or APOE4 variant exhibited rapid disease progression and poor survival outcomes relative to mice bearing the most prevalent APOE3 allele. APOE2 and APOE4 mice exhibited increased viral loads as well as suppressed adaptive immune responses early after infection. In vitro assays demonstrated increased infection in the presence of APOE2 and APOE4 relative to APOE3, indicating that differential outcomes are mediated by differential effects of APOE variants on both viral infection and antiviral immunity. Consistent with these in vivo findings in mice, our results also show that APOE genotype is associated with survival in patients infected with SARS-CoV-2 in the UK Biobank (candidate variant analysis, P = 2.6 × 10-7). Our findings suggest APOE genotype to partially explain the heterogeneity of COVID-19 outcomes and warrant prospective studies to assess APOE genotyping as a means of identifying patients at high risk for adverse outcomes.


Assuntos
Apolipoproteínas E , COVID-19 , Genética Humana , Camundongos Transgênicos , SARS-CoV-2 , Animais , Humanos , Masculino , Camundongos , Apolipoproteína E2/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , COVID-19/genética , COVID-19/mortalidade , COVID-19/virologia , Camundongos Transgênicos/genética , Camundongos Transgênicos/virologia , Estudos Prospectivos , SARS-CoV-2/patogenicidade , Modelos Animais de Doenças
16.
Epidemiol Infect ; 150: e208, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36128671

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the novel global coronavirus disease 2019 (COVID-19) disease outbreak. Its pathogenesis is mostly located in the respiratory tract. However, other organs are also affected. Hence, realising how such a complex disturbance affects patients after recovery is crucial. Regarding the significance of control of COVID-19-related complications after recovery, the current study was designed to review the cellular and molecular mechanisms linking COVID-19 to significant long-term signs including renal and cardiac complications, cutaneous and neurological manifestations, as well as blood coagulation disorders. This virus can directly influence on the cells through Angiotensin converting enzyme 2 (ACE-2) to induce cytokine storm. Acute release of Interleukin-1 (IL1), IL6 and plasminogen activator inhibitor 1 (PAI-1) have been related to elevating risk of heart failure. Also, inflammatory cytokines like IL-8 and Tumour necrosis factor-α cause the secretion of von Willebrand factor (VWF) from human endothelial cells and then VWF binds to Neutrophil extracellular traps to induce thrombosis. On the other hand, the virus can damage the blood-brain barrier by increasing its permeability and subsequently enters into the central nervous system and the systemic circulation. Furthermore, SARS-induced ACE2-deficiency decreases [des-Arg9]-bradykinin (desArg9-BK) degradation in kidneys to induce inflammation, thrombotic problems, fibrosis and necrosis. Notably, the angiotensin II-angiotensin II type 1 receptor binding causes an increase in aldosterone and mineralocorticoid receptors on the surface of dendritic cells cells, leading to recalling macrophage and monocyte into inflammatory sites of skin. In conclusions, all the pathways play a key role in the pathogenesis of these disturbances. Nevertheless, more investigations are necessary to determine more pathogenetic mechanisms of the virus.


Assuntos
Transtornos da Coagulação Sanguínea , COVID-19 , Cardiopatias , Nefropatias , Doenças do Sistema Nervoso , Síndrome de COVID-19 Pós-Aguda , Dermatopatias , COVID-19/complicações , COVID-19/epidemiologia , Cardiopatias/epidemiologia , Doenças do Sistema Nervoso/epidemiologia , Nefropatias/epidemiologia , Dermatopatias/epidemiologia , SARS-CoV-2/patogenicidade , Síndrome de COVID-19 Pós-Aguda/epidemiologia
17.
Nature ; 609(7928): 754-760, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35940203

RESUMO

Identifying the host genetic factors underlying severe COVID-19 is an emerging challenge1-5. Here we conducted a genome-wide association study (GWAS) involving 2,393 cases of COVID-19 in a cohort of Japanese individuals collected during the initial waves of the pandemic, with 3,289 unaffected controls. We identified a variant on chromosome 5 at 5q35 (rs60200309-A), close to the dedicator of cytokinesis 2 gene (DOCK2), which was associated with severe COVID-19 in patients less than 65 years of age. This risk allele was prevalent in East Asian individuals but rare in Europeans, highlighting the value of genome-wide association studies in non-European populations. RNA-sequencing analysis of 473 bulk peripheral blood samples identified decreased expression of DOCK2 associated with the risk allele in these younger patients. DOCK2 expression was suppressed in patients with severe cases of COVID-19. Single-cell RNA-sequencing analysis (n = 61 individuals) identified cell-type-specific downregulation of DOCK2 and a COVID-19-specific decreasing effect of the risk allele on DOCK2 expression in non-classical monocytes. Immunohistochemistry of lung specimens from patients with severe COVID-19 pneumonia showed suppressed DOCK2 expression. Moreover, inhibition of DOCK2 function with CPYPP increased the severity of pneumonia in a Syrian hamster model of SARS-CoV-2 infection, characterized by weight loss, lung oedema, enhanced viral loads, impaired macrophage recruitment and dysregulated type I interferon responses. We conclude that DOCK2 has an important role in the host immune response to SARS-CoV-2 infection and the development of severe COVID-19, and could be further explored as a potential biomarker and/or therapeutic target.


Assuntos
COVID-19 , Proteínas Ativadoras de GTPase , Estudo de Associação Genômica Ampla , Fatores de Troca do Nucleotídeo Guanina , Interações entre Hospedeiro e Microrganismos , SARS-CoV-2 , Alelos , Animais , COVID-19/complicações , COVID-19/genética , COVID-19/imunologia , COVID-19/fisiopatologia , Modelos Animais de Doenças , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Predisposição Genética para Doença , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Japão , Pulmão/patologia , Macrófagos , Mesocricetus , Pessoa de Meia-Idade , Pneumonia/complicações , Pirazóis/farmacologia , RNA-Seq , SARS-CoV-2/patogenicidade , Carga Viral , Redução de Peso
18.
Cell Host Microbe ; 30(9): 1255-1268.e5, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35931073

RESUMO

SARS-CoV-2 Omicron rapidly outcompeted other variants and currently dominates the COVID-19 pandemic. Its enhanced transmission and immune evasion are thought to be driven by numerous mutations in the Omicron Spike protein. Here, we systematically introduced BA.1 and/or BA.2 Omicron Spike mutations into the ancestral Spike protein and examined the impacts on Spike function, processing, and susceptibility to neutralization. Individual mutations of S371F/L, S375F, and T376A in the ACE2-receptor-binding domain as well as Q954H and N969K in the hinge region 1 impaired infectivity, while changes to G339D, D614G, N764K, and L981F moderately enhanced it. Most mutations in the N-terminal region and receptor-binding domain reduced the sensitivity of the Spike protein to neutralization by sera from individuals vaccinated with the BNT162b2 vaccine and by therapeutic antibodies. Our results represent a systematic functional analysis of Omicron Spike adaptations that have allowed this SARS-CoV-2 variant to dominate the current pandemic.


Assuntos
COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Enzima de Conversão de Angiotensina 2 , Anticorpos Neutralizantes , Anticorpos Antivirais , Vacina BNT162 , Humanos , Pandemias , SARS-CoV-2/genética , SARS-CoV-2/patogenicidade , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Proteínas do Envelope Viral
19.
Nature ; 609(7928): 801-807, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35901960

RESUMO

Anorexia and fasting are host adaptations to acute infection, and induce a metabolic switch towards ketogenesis and the production of ketone bodies, including ß-hydroxybutyrate (BHB)1-6. However, whether ketogenesis metabolically influences the immune response in pulmonary infections remains unclear. Here we show that the production of BHB is impaired in individuals with SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) but not in those with  influenza-induced ARDS. We found that BHB promotes both the survival of and the production of interferon-γ by CD4+ T cells. Applying a metabolic-tracing analysis, we established that BHB provides an alternative carbon source to fuel oxidative phosphorylation (OXPHOS) and the production of bioenergetic amino acids and glutathione, which is important for maintaining the redox balance. T cells from patients with SARS-CoV-2-induced ARDS were exhausted and skewed towards glycolysis, but could be metabolically reprogrammed by BHB to perform OXPHOS, thereby increasing their functionality. Finally, we show in mice that a ketogenic diet and the delivery of BHB as a ketone ester drink restores CD4+ T cell metabolism and function in severe respiratory infections, ultimately reducing the mortality of mice infected with SARS-CoV-2. Altogether, our data reveal that BHB is an alternative source of carbon that promotes T cell responses in pulmonary viral infections, and highlight impaired ketogenesis as a potential confounding factor in severe COVID-19.


Assuntos
COVID-19 , Metabolismo Energético , Cetonas , Síndrome do Desconforto Respiratório , SARS-CoV-2 , Linfócitos T , Ácido 3-Hidroxibutírico/biossíntese , Ácido 3-Hidroxibutírico/metabolismo , Aminoácidos/biossíntese , Aminoácidos/metabolismo , Animais , COVID-19/complicações , COVID-19/imunologia , COVID-19/patologia , Dieta Cetogênica , Ésteres/metabolismo , Glutationa/biossíntese , Glutationa/metabolismo , Glicólise , Interferon gama/biossíntese , Corpos Cetônicos/metabolismo , Cetonas/metabolismo , Camundongos , Orthomyxoviridae/patogenicidade , Oxirredução , Fosforilação Oxidativa , Síndrome do Desconforto Respiratório/complicações , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/virologia , SARS-CoV-2/patogenicidade , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
20.
J Cell Biol ; 221(7)2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35674692

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal pathogen of the ongoing global pandemic of coronavirus disease 2019 (COVID-19). Loss of smell and taste are symptoms of COVID-19, and may be related to cilia dysfunction. Here, we found that the SARS-CoV-2 ORF10 increases the overall E3 ligase activity of the CUL2ZYG11B complex by interacting with ZYG11B. Enhanced CUL2ZYG11B activity by ORF10 causes increased ubiquitination and subsequent proteasome-mediated degradation of an intraflagellar transport (IFT) complex B protein, IFT46, thereby impairing both cilia biogenesis and maintenance. Further, we show that exposure of the respiratory tract of hACE2 mice to SARS-CoV-2 or SARS-CoV-2 ORF10 alone results in cilia-dysfunction-related phenotypes, and the ORF10 expression in primary human nasal epithelial cells (HNECs) also caused a rapid loss of the ciliary layer. Our study demonstrates how SARS-CoV-2 ORF10 hijacks CUL2ZYG11B to eliminate IFT46 and leads to cilia dysfunction, thereby offering a powerful etiopathological explanation for how SARS-CoV-2 causes multiple cilia-dysfunction-related symptoms specific to COVID-19.


Assuntos
Cílios , SARS-CoV-2 , Ubiquitina-Proteína Ligases , Animais , Células Cultivadas , Cílios/metabolismo , Cílios/patologia , Proteínas do Citoesqueleto , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Camundongos , SARS-CoV-2/patogenicidade , Olfato , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA