Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 291
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(26): e2319322121, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38900789

RESUMEN

Thymocyte selection-associated high-mobility group box (TOX) is a transcription factor that is crucial for T cell exhaustion during chronic antigenic stimulation, but its role in inflammation is poorly understood. Here, we report that TOX extracellularly mediates drastic inflammation upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection by binding to the cell surface receptor for advanced glycation end-products (RAGE). In various diseases, including COVID-19, TOX release was highly detectable in association with disease severity, contributing to lung fibroproliferative acute respiratory distress syndrome (ARDS). Recombinant TOX-induced blood vessel rupture, similar to a clinical signature in patients experiencing a cytokine storm, further exacerbating respiratory function impairment. In contrast, disruption of TOX function by a neutralizing antibody and genetic removal of RAGE diminished TOX-mediated deleterious effects. Altogether, our results suggest an insight into TOX function as an inflammatory mediator and propose the TOX-RAGE axis as a potential target for treating severe patients with pulmonary infection and mitigating lung fibroproliferative ARDS.


Asunto(s)
COVID-19 , Receptor para Productos Finales de Glicación Avanzada , SARS-CoV-2 , Humanos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/patología , COVID-19/complicaciones , COVID-19/virología , Animales , Ratones , Inflamación/metabolismo , Inflamación/patología , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Masculino , Pulmón/patología , Pulmón/metabolismo , Pulmón/inmunología , Femenino
2.
Cell Mol Life Sci ; 81(1): 405, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39284944

RESUMEN

Emerging evidence indicates that activation of complement system leading to the formation of the membrane attack complex (MAC) plays a detrimental role in COVID-19. However, their pathogenic roles have never been experimentally investigated before. We used three knock out mice strains (1. C3-/-; 2. C7-/-; and 3. Cd59ab-/-) to evaluate the role of complement in severe COVID-19 pathogenesis. C3 deficient mice lack a key common component of all three complement activation pathways and are unable to generate C3 and C5 convertases. C7 deficient mice lack a complement protein needed for MAC formation. Cd59ab deficient mice lack an important inhibitor of MAC formation. We also used anti-C5 antibody to block and evaluate the therapeutic potential of inhibiting MAC formation. We demonstrate that inhibition of complement activation (in C3-/-) and MAC formation (in C3-/-. C7-/-, and anti-C5 antibody) attenuates severe COVID-19; whereas enhancement of MAC formation (Cd59ab-/-) accelerates severe COVID-19. The degree of MAC but not C3 deposits in the lungs of C3-/-, C7-/- mice, and Cd59ab-/- mice as compared to their control mice is associated with the attenuation or acceleration of SARS-CoV-2-induced disease. Further, the lack of terminal complement activation for the formation of MAC in C7 deficient mice protects endothelial dysfunction, which is associated with the attenuation of diseases and pathologic changes. Our results demonstrated the causative effect of MAC in severe COVID-19 and indicate a potential avenue for modulating the complement system and MAC formation in the treatment of severe COVID-19.


Asunto(s)
Antígenos CD59 , COVID-19 , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento , Ratones Noqueados , SARS-CoV-2 , Animales , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Activación de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Ratones , SARS-CoV-2/inmunología , Antígenos CD59/metabolismo , Antígenos CD59/genética , Antígenos CD59/inmunología , Complemento C3/inmunología , Complemento C3/metabolismo , Complemento C3/genética , Ratones Endogámicos C57BL , Humanos , Complemento C5/inmunología , Complemento C5/metabolismo , Complemento C5/antagonistas & inhibidores , Modelos Animales de Enfermedad
3.
J Infect Dis ; 229(5): 1372-1381, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38109685

RESUMEN

BACKGROUND: Altered mediators of airway tissue remodeling such as matrix metalloproteinases (MMPs) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may contribute to morbidity in coronavirus disease 2019 (COVID-19); however, the differential impact of SARS-CoV-2 variants of concern (VOCs) on MMPs is unknown. METHODS: Using both in vitro human airway cell culture model and in vivo transgenic mouse model of SARS-CoV-2 infection, we studied the differential effect of SARS-CoV-2 VOCs on expression of key MMPs and inflammatory mediators in airway cells and tissues. RESULTS: The most consistent findings with all SARS-CoV-2 variants in infected compared to uninfected human bronchial epithelial cell air-liquid interface cultures were the SARS-CoV-2-induced increases in MMP-12 and tissue inhibitor of MMPs. Infection with both SARS-CoV-2 wild type and SARS-CoV-2 Delta variant over 3 days postinfection (dpi) and with Beta variant over 7 dpi increased lung tissue levels of MMP-9 compared to uninfected mice. Overall, SARS-CoV-2 variants had differential dose-dependent impact on secretion of MMP-1, MMP-2, MMP-9, and MMP-12 that varied at the protein versus the gene level and in the early noninflammatory compared to late inflammatory phase of infection. CONCLUSIONS: We provide novel mechanistic insight that the differential impact of SARS-CoV-2 variants on severity of COVID-19 may partially be attributed to unique changes in MMPs.


Asunto(s)
COVID-19 , Pulmón , Metaloproteinasa 12 de la Matriz , Ratones Transgénicos , SARS-CoV-2 , Animales , COVID-19/virología , COVID-19/patología , COVID-19/metabolismo , Humanos , Ratones , Pulmón/virología , Pulmón/patología , Metaloproteinasa 12 de la Matriz/metabolismo , Metaloproteinasa 12 de la Matriz/genética , Modelos Animales de Enfermedad , Remodelación de las Vías Aéreas (Respiratorias) , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Células Epiteliales/virología
4.
BMC Genomics ; 25(1): 482, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38750426

RESUMEN

BACKGROUND: The severity of COVID-19 is influenced by various factors including the presence of respiratory diseases. Studies have indicated a potential relationship between asthma and COVID-19 severity. OBJECTIVE: This study aimed to conduct a genome-wide association study (GWAS) to identify genetic and clinical variants associated with the severity of COVID-19, both among patients with and without asthma. METHODS: We analyzed data from 2131 samples sourced from the Biobanque québécoise de la COVID-19 (BQC19), with 1499 samples from patients who tested positive for COVID-19. Among these, 1110 exhibited mild-to-moderate symptoms, 389 had severe symptoms, and 58 had asthma. We conducted a comparative analysis of clinical data from individuals in these three groups and GWAS using a logistic regression model. Phenotypic data analysis resulted in the refined covariates integrated into logistic models for genetic studies. RESULTS: Considering a significance threshold of 1 × 10-6, seven genetic variants were associated with severe COVID-19. These variants were located proximal to five genes: sodium voltage-gated channel alpha subunit 1 (SCN10A), desmoplakin (DSP), RP1 axonemal microtubule associated (RP1), IGF like family member 1 (IGFL1), and docking protein 5 (DOK5). The GWAS comparing individuals with severe COVID-19 with asthma to those without asthma revealed four genetic variants in transmembrane protein with EGF like and two follistatin like domains 2 (TMEFF2) and huntingtin interacting protein-1 (HIP1) genes. CONCLUSION: This study provides significant insights into the genetic profiles of patients with severe forms of the disease, whether accompanied by asthma or not. These findings enhance our comprehension of the genetic factors that affect COVID-19 severity. KEY MESSAGES: Seven genetic variants were associated with the severe form of COVID-19; Four genetic variants were associated with the severe form of COVID-19 in individuals with comorbid asthma; These findings help define the genetic component of the severe form of COVID-19 in relation to asthma as a comorbidity.


Asunto(s)
Asma , COVID-19 , Comorbilidad , Estudio de Asociación del Genoma Completo , SARS-CoV-2 , Humanos , COVID-19/genética , Asma/genética , Asma/complicaciones , Masculino , Femenino , Persona de Mediana Edad , SARS-CoV-2/genética , Adulto , Índice de Severidad de la Enfermedad , Estudios de Cohortes , Polimorfismo de Nucleótido Simple , Anciano , Genómica/métodos , Predisposición Genética a la Enfermedad
5.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34535795

RESUMEN

Whether risk genes of severe coronavirus disease 2019 (COVID-19) from genome-wide association study could play their regulatory roles by interacting with host genes that were interacted with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) proteins was worthy of exploration. In this study, we implemented a network-based approach by developing a user-friendly software Network Calculator (https://github.com/Haoxiang-Qi/Network-Calculator.git). By using Network Calculator, we identified a network composed of 13 risk genes and 28 SARS-CoV-2 interacted host genes that had the highest network proximity with each other, with a hub gene HNRNPK identified. Among these genes, 14 of them were identified to be differentially expressed in RNA-seq data from severe COVID-19 cases. Besides, by expression enrichment analysis in single-cell RNA-seq data, compared with mild COVID-19, these genes were significantly enriched in macrophage, T cell and epithelial cell for severe COVID-19. Meanwhile, 74 pathways were significantly enriched. Our analysis provided insights for the underlying genetic etiology of severe COVID-19 from the perspective of network biology.


Asunto(s)
COVID-19 , RNA-Seq , SARS-CoV-2 , Proteínas Virales , COVID-19/genética , COVID-19/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Gravedad del Paciente , Factores de Riesgo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
6.
J Transl Med ; 22(1): 698, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075394

RESUMEN

BACKGROUND: Severe COVID-19 infection has been associated with the development of pulmonary fibrosis, a condition that significantly affects patient prognosis. Understanding the underlying cellular communication mechanisms contributing to this fibrotic process is crucial. OBJECTIVE: In this study, we aimed to investigate the role of the TNFSF12-TNFRSF12A pathway in mediating communication between alveolar macrophages and fibroblasts, and its implications for the development of pulmonary fibrosis in severe COVID-19 patients. METHODS: We conducted single-cell RNA sequencing (scRNA-seq) analysis using lung tissue samples from severe COVID-19 patients and healthy controls. The data was processed, analyzed, and cell types were annotated. We focused on the communication between alveolar macrophages and fibroblasts and identified key signaling pathways. In vitro experiments were performed to validate our findings, including the impact of TNFRSF12A silencing on fibrosis reversal. RESULTS: Our analysis revealed that in severe COVID-19 patients, alveolar macrophages communicate with fibroblasts primarily through the TNFSF12-TNFRSF12A pathway. This communication pathway promotes fibroblast proliferation and expression of fibrotic factors. Importantly, silencing TNFRSF12A effectively reversed the pro-proliferative and pro-fibrotic effects of alveolar macrophages. CONCLUSION: The TNFSF12-TNFRSF12A pathway plays a central role in alveolar macrophage-fibroblast communication and contributes to pulmonary fibrosis in severe COVID-19 patients. Silencing TNFRSF12A represents a potential therapeutic strategy for mitigating fibrosis in severe COVID-19 lung disease.


Asunto(s)
COVID-19 , Fibroblastos , Macrófagos Alveolares , Fibrosis Pulmonar , Transducción de Señal , Receptor de TWEAK , Humanos , COVID-19/complicaciones , COVID-19/patología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/complicaciones , Receptor de TWEAK/metabolismo , Receptor de TWEAK/genética , Citocina TWEAK/metabolismo , Comunicación Celular , Masculino , SARS-CoV-2 , Femenino , Persona de Mediana Edad , Proliferación Celular , Pulmón/patología , Índice de Severidad de la Enfermedad
7.
Hum Genomics ; 17(1): 17, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36859360

RESUMEN

BACKGROUND: Genome-wide association studies have identified numerous human host genetic risk variants that play a substantial role in the host immune response to SARS-CoV-2. Although these genetic risk variants significantly increase the severity of COVID-19, their influence on body systems is poorly understood. Therefore, we aim to interpret the biological mechanisms and pathways associated with the genetic risk factors and immune responses in severe COVID-19. We perform a deep analysis of previously identified risk variants and infer the hidden interactions between their molecular networks through disease mapping and the similarity of the molecular functions between constructed networks. RESULTS: We designed a four-stage computational workflow for systematic genetic analysis of the risk variants. We integrated the molecular profiles of the risk factors with associated diseases, then constructed protein-protein interaction networks. We identified 24 protein-protein interaction networks with 939 interactions derived from 109 filtered risk variants in 60 risk genes and 56 proteins. The majority of molecular functions, interactions and pathways are involved in immune responses; several interactions and pathways are related to the metabolic and cardiovascular systems, which could lead to multi-organ complications and dysfunction. CONCLUSIONS: This study highlights the importance of analyzing molecular interactions and pathways to understand the heterogeneous susceptibility of the host immune response to SARS-CoV-2. We propose new insights into pathogenicity analysis of infections by including genetic risk information as essential factors to predict future complications during and after infection. This approach may assist more precise clinical decisions and accurate treatment plans to reduce COVID-19 complications.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Estudio de Asociación del Genoma Completo , Mapas de Interacción de Proteínas , Factores de Riesgo
8.
Virol J ; 21(1): 74, 2024 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-38532424

RESUMEN

BACKGROUND: To investigate the efficacy of different doses of corticosteroids in treating severe coronavirus disease 2019 (COVID-19) pneumonia. METHODS: Between May 01, 2023, and June 20, 2023, 48 patients with severe COVID-19 pneumonia were treated at the Department of Respiratory and Critical Care Medicine of Jinan Fourth People's Hospital. The observation group (21 patients) received standard care and high-dose corticosteroids, (high-dose group). The control group (27 patients) received standard care and low-dose corticosteroids (low-dose group). We collected baseline data and recorded inflammatory marker levels after 3 days of treatment, body temperature recovery time, length of stay, and 28-day all-cause mortality. The results of outpatient follow-up were recorded after 1 month. RESULTS: There were no significant differences in 28-day mortality and length of stay. The number of days it took for body temperature to return to normal in the high-dose group was less than in the low-dose group. The high-dose group had significantly more reduced inflammatory factors (C-reactive protein (CRP), interleukin-6 (IL-6). A total of 20 discharged patients were given 8-16 mg of methylprednisolone, depending on chest computed tomography (CT) and clinical symptoms after 1 month; in all discharged patients using oral corticosteroids, CT features improved. CONCLUSION: High-dose corticosteroids had a significantly positive effect on the reduction of inflammatory factors and shortening body temperature recovery time. In the treatment of severe COVID-19 pneumonia, early administration of high-dose, short-course corticosteroids should be implemented.


Asunto(s)
COVID-19 , Neumonía , Humanos , SARS-CoV-2 , Corticoesteroides , Metilprednisolona
9.
Eur J Clin Pharmacol ; 80(10): 1523-1530, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38926166

RESUMEN

OBJECTIVE: To systematically evaluate the efficacy of subcutaneous tocilizumab in the treatment of patients with severe COVID-19 and provide evidence for the rational use of subcutaneous tocilizumab in patients with severe COVID-19. METHODS: This meta-analysis was carried out in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. We searched the Cochrane Library, PubMed, Embase, CNKI, SinoMed, and Wanfang Medical Network electronic databases up to 11 January 2023 to identify relevant studies. To obtain the most recent clinical studies of subcutaneous injection of tocilizumab for the treatment of patients with severe COVID-19, we also searched the preprint platforms medRxiv and ChinaXiv. Furthermore, we searched ClinicalTrials.gov for relevant unpublished studies. The studies were screened based on the PICOS principle. The included studies were classified and evaluated for quality based on research type. The RevMan 5.3 software was used to conduct the meta-analysis, and a descriptive analysis was performed to examine relevant outcome indicators. RESULTS: Five observational studies were obtained, involving a total of 498 patients (240 patients in the subcutaneous injection group and 258 patients in the intravenous injection group). All of the studies were of the highest quality. The meta-analysis of the included studies revealed that the mortality rate of patients who received subcutaneous tocilizumab to treat COVID-19 was not significantly higher than that of the intravenous injection group [23.3% (45/193) vs. 18.4% (39/212), RD = 0.06, 95% CI = - 0.01 ~ 0.13, P = 0.11]. Furthermore, there was no significant difference in the proportion of patients requiring mechanical ventilation between the two groups [24.5% (35/143) vs. 22% (35/159), RD = 0.03, 95% CI = - 0.07 ~ 0.12, P = 0.56]. CONCLUSIONS: The meta-analyses do not provide evidence that subcutaneous and intravenous tocilizumab formulations for the treatment of severe COVID-19 infection differ regarding their effectiveness. Considering that the meta-analyses cannot replace an appropriately powered non-inferiority study, subcutaneous formulations still need to be used with caution and only when intravenous formulations are in short supply. At present, there is a lack of randomized controlled trials of subcutaneous injection of tocilizumab for the treatment of severe COVID-19, and more clinical research should be conducted.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Tratamiento Farmacológico de COVID-19 , Humanos , Administración Intravenosa , Anticuerpos Monoclonales Humanizados/administración & dosificación , COVID-19/diagnóstico , COVID-19/mortalidad , Tratamiento Farmacológico de COVID-19/métodos , Tratamiento Farmacológico de COVID-19/estadística & datos numéricos , Inyecciones Subcutáneas , Resultado del Tratamiento
10.
Blood Purif ; 53(10): 793-803, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39102799

RESUMEN

INTRODUCTION: Hemoadsorption, an extracorporeal apheresis technique, is reportedly used in severe COVID-19 patients. However, limited evidence from randomized clinical trials supports this practice. METHODS: In this single-center study, severe COVID-19 patients requiring ICU admission were randomly assigned (1:1) to receive HA-330 hemoadsorption in combination with standard treatment or standard therapy alone. Both groups received tocilizumab intravenously if their clinical conditions worsened within 24-48 h. The primary outcome was mortality from any cause within 28 days after randomization. Secondary outcomes included mechanical ventilator-free days, daily C-reactive protein levels, oxygenation (defined by PaO2/FiO2 ratio), daily sequential organ failure assessment score, and severity score of lung infiltration on chest X-rays (CXR RALE score). RESULTS: A total of 28 patients underwent randomization, with 14 (50%) receiving HA-330 hemoadsorption. Only 9 out of 14 patients (64.3%) in the control group experienced clinical worsening and were subsequently administered intravenous tocilizumab. At 28 days, the mortality rate was significantly lower in the intervention group (28.57% vs. 78.57%, p = 0.021), with a hazard ratio of death of 0.26 (95% CI = 0.08-0.81; p = 0.021). All of secondary outcomes were comparable in both groups. CONCLUSION: Based on our pilot randomized trial, the early application of HA-330 hemoadsorption in patients with severe COVID-19 may establish a favorable outcome in term of mortality. These data provide the initial proof of concept for conducting a large-scale study in the future.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/terapia , COVID-19/complicaciones , COVID-19/sangre , COVID-19/mortalidad , Masculino , Persona de Mediana Edad , Femenino , Proyectos Piloto , Anciano , SARS-CoV-2/aislamiento & purificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Resultado del Tratamiento , Índice de Severidad de la Enfermedad
11.
Int J Mol Sci ; 25(14)2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39062917

RESUMEN

Determining the genetic contribution of susceptibility to severe SARS-CoV-2 infection outcomes is important for public health measures and individualized treatment. Through intense research on this topic, several hundred genes have been implicated as possibly contributing to the severe infection phenotype(s); however, the findings are complex and appear to be population-dependent. We aimed to determine the contribution of human rare genetic variants associated with a severe outcome of SARS-CoV-2 infections and their burden in the Slovenian population. A panel of 517 genes associated with severe SARS-CoV-2 infection were obtained by combining an extensive review of the literature, target genes identified by the COVID-19 Host Genetic Initiative, and the curated Research COVID-19 associated genes from PanelApp, England Genomics. Whole genome sequencing was performed using PCR-free WGS on DNA from 60 patients hospitalized due to severe COVID-19 disease, and the identified rare genomic variants were analyzed and classified according to the ACMG criteria. Background prevalence in the general Slovenian population was determined by comparison with sequencing data from 8025 individuals included in the Slovenian genomic database (SGDB). Results show that several rare pathogenic/likely pathogenic genomic variants in genes CFTR, MASP2, MEFV, TNFRSF13B, and RNASEL likely contribute to the severe infection outcomes in our patient cohort. These results represent an insight into the Slovenian genomic diversity associated with a severe COVID-19 outcome.


Asunto(s)
COVID-19 , Predisposición Genética a la Enfermedad , SARS-CoV-2 , Humanos , COVID-19/genética , COVID-19/epidemiología , COVID-19/virología , Eslovenia/epidemiología , SARS-CoV-2/genética , Masculino , Femenino , Persona de Mediana Edad , Anciano , Secuenciación Completa del Genoma , Variación Genética , Adulto , Genómica/métodos , Pandemias , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Betacoronavirus/genética
12.
J Infect Dis ; 227(8): 961-969, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36415904

RESUMEN

BACKGROUND: We assessed coronavirus disease 2019 (COVID-19) vaccination impact on illness severity among adults hospitalized with COVID-19, August 2021-March 2022. METHODS: We evaluated differences in intensive care unit (ICU) admission, in-hospital death, and length of stay among vaccinated (2 or 3 mRNA vaccine doses) versus unvaccinated patients aged ≥18 years hospitalized for ≥24 hours with COVID-19-like illness and positive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) molecular testing. We calculated odds ratios (ORs) for ICU admission and death and subdistribution hazard ratios (SHR) for time to hospital discharge adjusted for age, geographic region, calendar time, and local virus circulation. RESULTS: We included 27 149 SARS-CoV-2-positive hospitalizations. During both Delta- and Omicron-predominant periods, protection against ICU admission was strongest among 3-dose vaccinees compared with unvaccinated patients (Delta OR, 0.52 [95% CI, .28-.96]; Omicron OR, 0.69 [95% CI, .54-.87]). During both periods, risk of in-hospital death was lower among vaccinated compared with unvaccinated patients but ORs overlapped across vaccination strata. We observed SHR >1 across all vaccination strata in both periods indicating faster discharge for vaccinated patients. CONCLUSIONS: COVID-19 vaccination was associated with lower rates of ICU admission and in-hospital death in both Delta and Omicron periods compared with being unvaccinated.


Asunto(s)
COVID-19 , Humanos , Adulto , Adolescente , COVID-19/prevención & control , SARS-CoV-2 , Vacunas contra la COVID-19 , Mortalidad Hospitalaria , Vacunas de ARNm
13.
J Phys Ther Sci ; 36(3): 142-150, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38434997

RESUMEN

[Purpose] This study aimed to identify the factors and cutoffs associated with walking independence in patients with severe COVID-19 pneumonia. [Participants and Methods] In total, 112 patients with COVID-19 pneumonia (98 males and 14 females) who were hospitalized between March 2020 and August 2021 and underwent physiotherapy during mechanical ventilation were included in the study. Attributes, respiratory function, physical function, and bed-withdrawal status were compared between two groups of patients, who were classified according to their ability to walk independently at discharge. The independent variables were reduced to four components by principal component analysis. Logistic regression analysis was performed with walking independence at discharge as the dependent variable. Receiver operating characteristic curves for the extracted factors were drawn, and cutoff values were calculated. [Results] At discharge, 76 patients were able to walk independently, while 36 were not. The logistic regression analysis was adjusted according to age and mechanical ventilation time. Cutoffs were an age of 56 years and a ventilation period of 7.5 days. [Conclusion] In cases of patients with severe COVID-19 pneumonia who required ventilators, age and mechanical ventilation time were associated with ambulatory independence at discharge, indicating the importance of reducing the ventilation period by providing respiratory physiotherapy, including expectoration, positioning, and weaning.

14.
Neurobiol Dis ; 182: 106147, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37178811

RESUMEN

Coronavirus disease of 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has sparked a global pandemic with severe complications and high morbidity rate. Neurological symptoms in COVID-19 patients, and neurological sequelae post COVID-19 recovery have been extensively reported. Yet, neurological molecular signature and signaling pathways that are affected in the central nervous system (CNS) of COVID-19 severe patients remain still unknown and need to be identified. Plasma samples from 49 severe COVID-19 patients, 50 mild COVID-19 patients, and 40 healthy controls were subjected to Olink proteomics analysis of 184 CNS-enriched proteins. By using a multi-approach bioinformatics analysis, we identified a 34-neurological protein signature for COVID-19 severity and unveiled dysregulated neurological pathways in severe cases. Here, we identified a new neurological protein signature for severe COVID-19 that was validated in different independent cohorts using blood and postmortem brain samples and shown to correlate with neurological diseases and pharmacological drugs. This protein signature could potentially aid the development of prognostic and diagnostic tools for neurological complications in post-COVID-19 convalescent patients with long term neurological sequelae.


Asunto(s)
COVID-19 , Enfermedades del Sistema Nervioso , Humanos , COVID-19/complicaciones , SARS-CoV-2 , Enfermedades del Sistema Nervioso/etiología , Sistema Nervioso Central , Encéfalo
15.
Immunogenetics ; 75(3): 295-307, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36752852

RESUMEN

Infection with SARS-CoV-2 causes wide range of disease severities from asymptomatic to life-threatening disease. Understanding the contribution of immunological traits in immunity against SARS-CoV-2 and in protection against severe COVID-19 could result in effective measures to prevent development of severe disease. While the role of cytokines and antibodies has been thoroughly studied, this is not the case for T cells. In this review, the association between T cells and COVID-19 disease severity and protection upon reexposure is discussed. While infiltration of overactivated cytotoxic T cells might be harmful in the infected tissue, fast responding T cells are important in the protection against severe COVID-19. This protection could even be viable in the long term as long-living memory T cells seem to be stabilized and mutations do not appear to have a large impact on T cell responses. Thus, after vaccination and infections, memory T cells should be able to help prevent onset of severe disease for most cases. Considering this, it would be useful to add N or M proteins in vaccinations, alongside the S protein which is currently used, as this results in a broader T cell response.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Vacunación , Linfocitos T Citotóxicos , Anticuerpos Antivirales
16.
J Med Virol ; 95(12): e29318, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38112106

RESUMEN

To examine the effectiveness of azvudine and nirmatrelvir-ritonavir in treating hospitalized patients with moderate-to-severe COVID-19. We emulated a target trial with a multicenter retrospective cohort of hospitalized adults with moderate-to-severe COVID-19 without contraindications for azvudine or nirmatrelvir-ritonavir between December 01, 2022 and January 19, 2023 (during the Omicron BA.5.2 variant wave). Exposures included treatment with azvudine or nirmatrelvir-ritonavir for 5 days versus no antiviral treatment during hospitalization. Primary composite outcome (all-cause death and initiation of invasive mechanical ventilation), and their separate events were evaluated. Of the 1154 patients, 27.2% were severe cases. In the intent-to-treat analyses, azvudine reduced all-cause death (Hazard ratio [HR]: 0.31; 95% CI: 0.12-0.78), and its composite with invasive mechanical ventilation (HR: 0.47; 95% CI: 0.24-0.92). Nirmatrelvir-ritonavir reduced invasive mechanical ventilation (HR: 0.42; 95% CI: 0.17-1.05), and its composite with all-cause death (HR: 0.38; 95% CI: 0.18-0.81). The study did not identify credible subgroup effects. The per-protocol analyses and all sensitivity analyses confirmed the robustness of the findings. Both azvudine and nirmatrelvir-ritonavir improved the prognosis of hospitalized adults with moderate-to-severe COVID-19.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Ritonavir , Adulto , Humanos , Antivirales/uso terapéutico , Estudios Retrospectivos , Ritonavir/uso terapéutico
17.
Artículo en Inglés | MEDLINE | ID: mdl-37632774

RESUMEN

OBJECTIVE: To describe the clinical efficacy of tixagevimab/cilgavimab in pre-exposure prophylaxis in patients at risk of severe COVID-19 and unresponsive to vaccination (anti-SARS-CoV-2 antibodies <260 BAU/mL) in rheumatology. METHODS: In this multicenter observational study we included patients with autoimmune or inflammatory diseases who received a pre-exposure prophylaxis by tixagevimab/cilgavimab between December 2021 and August 2022. The endpoint was incidence and severity of COVID-19. RESULTS: Tixagevimab/cilgavimab was administered to 115 patients, median age 62 years (52-71), with chronic arthritis (n = 53), connective tissue disease (n = 38) or vasculitis (n = 11). Main background immunosuppressants were rituximab (n = 98), corticosteroids (n = 62, median dose 5mg, CI95% 5-8 mg) and methotrexate (n = 48). During a median follow-up of 128 days (93-173), COVID-19 occurred in 23/115patients (20%), Omicron identified for the 8 genotyped patients. During study period, the average weekly incidence was 1071/100.000 inhabitants in Ile-de-France vs. 588/100.000 in our patients. Patients who received a 2-injections regimen had a lower risk of infection than with a single injection (16/49, 33% vs. 5/64, 8%, p = 0.0012). The COVID-19+ patients did not differ from uninfected patients concerning age, comorbidities, underlying rheumatic disease, immunosuppressant. All COVID-19 were non-severe. The tolerance of injections was excellent. CONCLUSION: In a population with autoimmune or inflammatory diseases at risk of severe COVID-19 unresponsive to vaccination, pre-exposure prophylaxis by tixagevimab/cilgavimab was associated with lower incidence of COVID-19 and no severe infection to report.

18.
Cancer Control ; 30: 10732748231205864, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37817417

RESUMEN

OBJECTIVES: This study aims to describe the clinical outcomes of combination therapy with sarilumab and baricitinib for severe novel Coronavirus-19 (COVID-19) infection in cancer patients. With this study, we aim to evaluate the role of expanded immunotherapy for severely ill patients with COVID-19 respiratory infections with limited options. The secondary objective is to assess the safety of combination therapy with sarilumab and baricitinib for severe COVID-19 infection. METHODS: This was a retrospective cohort study of patients admitted to Moffitt Cancer Center with COVID-19 infection between January 2020 and April 2022. Our research received a waiver to sign consent by the patients according to our institutional IRB because it was free of any risk for the patients and respected the patient's privacy. Following the Institutional IRB approval and relevant Equator guidelines, we collected information on patients with severe COVID-19 infection and received sarilumab and baricitinib. We evaluated the survival rate and safety of combination therapy. All the patient's information was de-identified to protect their information according to Health Insurance Portability and Accountability Act (HIPAA). RESULTS: Four patients were included in the data analysis. Two survived, and two of them died (Table 1). All the patients that survived were previously vaccinated. Among the two patients who died, one was vaccinated, and the other was unvaccinated. All the patients tolerated the combination therapy well, and none of the patients who survived developed secondary infections or COVID-19-associated complications beyond 12 months of discharge. CONCLUSION: Our study explores the potential safe combination use of different immune modulators targeting multiple pathways of the inflammatory cascade for severe and refractory COVID-19 respiratory infections in high-risk oncology patients. The small number of patients in our observational study was a limitation. A larger sample of patients will be needed to conclude more precisely the efficacy of the combination therapy of sarilumab and baricitinib for refractory cases of severe COVID-19 respiratory infection. Moreover, exploring other cytokine release signaling pathway targets may be the key to significantly reducing inflammation and further pulmonary fibrosis with chronic unbearable respiratory sequela.


Asunto(s)
COVID-19 , Neoplasias , Humanos , COVID-19/complicaciones , Estudios Retrospectivos , Resultado del Tratamiento , Tratamiento Farmacológico de COVID-19 , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico
19.
Inflamm Res ; 72(5): 929-932, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36988640

RESUMEN

The blood levels of neutrophils are associated with the severity of COVID -19. However, their role in the pulmonary environment during COVID -19 severity is not clear. Here, we found a decrease in the neutrophil count in BAL (bronchoalveolar lavage) in non-survivors and in older patients (> 60 years). In addition, we have shown that older patients have higher serum concentration of CXCL8 and increased IL-10 expression by neutrophils.


Asunto(s)
COVID-19 , Neutrófilos , Humanos , Anciano , Líquido del Lavado Bronquioalveolar , Pulmón , Pronóstico
20.
J Thromb Thrombolysis ; 55(1): 51-59, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36371754

RESUMEN

Only a percentage of COVID-19 patients develop thrombotic complications. We hypothesized that genetic profiles may explain part of the inter-individual differences. Our goal was to evaluate the genotypic distribution of targeted DNA polymorphisms in COVID-19 patients complicated (PE+) or not (PE-) by pulmonary embolism. We designed a retrospective observational study enrolling N = 94 consecutive patients suffering severe COVID-19 with pulmonary embolism (PE+, N = 47) or not (PE-, N = 47) during hospitalization. A panel of N = 13 prothrombotic DNA polymorphisms (FV R506Q and H1299R, FII G20210A, MTHFR C677T and A1298C, CBS 844ins68, PAI-1 4G/5G, GPIIIa HPA-1 a/b, ACE I/D, AGT T9543C, ATR-1 A1166C, FGB - 455G > A, FXIII103G > T) and N = 2 lipid metabolism-related DNA polymorphisms (APOE T 112C and T158C) were investigated using Reverse Dot Blot technique. Then, we investigated possible associations between genotypic subclasses and demographic, clinical, and laboratory parameters including age, obesity, smoking, pro-inflammatory cytokines, drug therapy, and biomarkers of thrombotic risk such as D-dimer (DD). We found that 58.7% of PE+ had homozygous mutant D/D genotype at ACE I/D locus vs. PE- (40.4%) and 87% of PE+ had homozygous mutant C/C genotype at APOE T158C locus vs. PE- (68.1%). In PE+ group, DD levels were significantly higher in D/D and I/D genotypes at ACE I/D locus (P = 0.00066 and P = 0.00023, respectively) and in C/C and T/C genotypes at APOE T158C locus (P = 1.6e-06 and P = 0.0012, respectively) than PE- group. For the first time, we showed significant associations between higher DD levels and ACE I/D and APOE T158C polymorphisms in PE+ vs. PE- patients suggesting potential useful biomarkers of poor clinical outcome.


Asunto(s)
COVID-19 , Embolia Pulmonar , Trombosis , Humanos , COVID-19/complicaciones , COVID-19/genética , Embolia Pulmonar/genética , Biomarcadores , Apolipoproteínas E , ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA