Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Ann Med ; 56(1): 2313062, 2024 12.
Article in English | MEDLINE | ID: mdl-38354691

ABSTRACT

BACKGROUND: The effectiveness of nirmatrelvir-ritonavir has mainly been shown in non-hospitalized patients with mild-to-moderate coronavirus disease 2019 (COVID-19). The real-world effectiveness of nirmatrelvir-ritonavir urgently needs to be determined using representative in-hospital patients with COVID-19 during the Omicron wave of the pandemic. METHODS: We performed a multicentre, retrospective study in five Chinese PLA General Hospital medical centers in Beijing, China. Patients hospitalized with COVID-19 from 10 December 2022 to 20 February 2023 were eligible for inclusion. A 1:1 propensity score matching was performed between the nirmatrelvir-ritonavir group and the control group. RESULTS: 1010 recipients of nirmatrelvir-ritonavir and 1010 matched controls were finally analyzed after matching. Compared with matched controls, the nirmatrelvir-ritonavir group had a lower incidence rate of all-cause death (4.6/1000 vs. 6.3/1000 person-days, p = 0.013) and a higher incidence rate of clinical improvement (47.6/1000 vs. 45.8/1000 person-days, p = 0.012). Nirmatrelvir-ritonavir was associated with a 22% lower all-cause mortality and a 14% higher incidence of clinical improvement. Initiation of nirmatrelvir-ritonavir within 5 days after symptom onset was associated with a 50% lower mortality and a 26% higher clinical improvement rate. By contrast, no significant associations were identified among patients receiving nirmatrelvir-ritonavir treatment more than 5 days after symptom onset. Nirmatrelvir-ritonavir was also associated with a 50% increase in survival days and a 12% decrease in days to clinical improvement. CONCLUSION: Among hospitalized patients with COVID-19 during the Omicron wave in Beijing, China, the early initiation of nirmatrelvir-ritonavir was associated with clinical benefits of lowering mortality and improving clinical recovery.


Subject(s)
COVID-19 , Lactams , Leucine , Nitriles , Proline , Ritonavir , Humans , Retrospective Studies , Beijing , Ritonavir/therapeutic use , COVID-19 Drug Treatment , China/epidemiology , Antiviral Agents/therapeutic use
2.
Int Immunopharmacol ; 126: 111097, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37988909

ABSTRACT

OBJECTIVE: We investigated the effects of the inducible NO synthase (iNOS) inhibitor, S-methylisothiourea (SMT), in a mouse model of smoke inhalation-induced acute lung injury (ALI) and explored the underlying molecular mechanism. METHODS AND ANALYSIS: A mouse model of smoke inhalation-induced ALI was established. RNA-sequencing (seq) analysis was conducted to identify the differentially expressed genes (DEGs). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed for functional annotation of DEGs. Moreover, an immunofluorescence assay using macrophage marker F4/80 was performed to assess macrophage infiltration. A hypoxia-induced HUVEC model was used to mimic smoke inhalation-induced injury in endothelial cells. Finally, a transwell assay was used to analyze the chemoattractive effects of endothelial cells on macrophages. RESULTS: SMT markedly alleviated the pulmonary pathological symptoms, edema, and inflammatory response in the mouse smoke inhalation-induced ALI model. RNA-seq analysis revealed that SMT may diminish lung injury by regulating the levels of genes associated with inflammatory responses, cell chemokines, and adhesion. In vivo data revealed that the protective effects of SMT against smoke inhalation-induced ALI were partly achieved by inhibiting the production of adhesion molecules and infiltration of macrophages. Furthermore, in vitro data from the hypoxia-induced HUVEC model revealed that SMT reduced macrophage chemotaxis by inhibiting the production of chemokines and adhesion molecules in endothelial cells. CONCLUSION: iNOS inhibitor SMT protects the lungs from smoke inhalation-induced ALI by reducing the production of pro-inflammatory cytokines, adhesion molecules, and chemokines in endothelial cells, thereby inhibiting inflammation and macrophage infiltration.


Subject(s)
Acute Lung Injury , Smoke Inhalation Injury , Rats , Mice , Animals , Endothelial Cells/metabolism , Rats, Sprague-Dawley , Acute Lung Injury/chemically induced , Lung/pathology , Inflammation/metabolism , Enzyme Inhibitors/pharmacology , Macrophages , Chemokines/metabolism , Smoke/adverse effects , Hypoxia/metabolism , Lipopolysaccharides/pharmacology
3.
Front Med (Lausanne) ; 10: 1236484, 2023.
Article in English | MEDLINE | ID: mdl-37608823

ABSTRACT

Background: This study explored novel biomarkers for diagnosing sepsis, a severe disease prevalent in clinical settings, particularly threatening to elderly patients. Methods: Using microarray gene expression datasets and fatty acid metabolism signatures, we identified differentially expressed genes between sepsis and healthy control groups. Correlations between candidate genes, immune cells, and immune function were assessed. Logistic regression analysis and single-gene GSEA analysis were performed to identify potential biomarkers. The biomarkers' association with different types of tumors was investigated. Results: Twelve genes related to fatty acid metabolism were excluded. CA4, OLAN, and VNN1 were found relevant to immune cells and function. Among these, only VNN1 showed statistical significance (p < 0.05), with a strong area under the ROC curve (0.995). High VNN1 expression indicated activation of certain metabolic pathways, while low expression suggested potential autoimmune responses. VNN1 was up-regulated in eight tumors and down-regulated in eight others. High VNN1 expression was linked to poor prognosis in six types of tumors, and low expression was linked to poor prognosis in four types of tumors. VNN1 expression showed correlations with stromal scores, immune scores, and cancer purity in different types of tumors. Conclusion: VNN1 holds promise as a potential biomarker for sepsis diagnosis and is significant in identifying immune infiltration in tumor tissue and predicting tumor prognosis.

4.
Trials ; 24(1): 552, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37612723

ABSTRACT

INTRODUCTION: The mortality rate of hospitalized patients with severe hospital-acquired pneumonia (SHAP) remains high. Empirical broad-spectrum antibiotic coverage and the misuse of high-grade antibiotics could lead to the emergence of multi-drug and even pandrug-resistant bacteria. In addition to metagenomic next-generation sequencing (mNGS), microbiological rapid on-site evaluation (M-ROSE) might be a useful technique to identify the pathogens in the early stage; however, the effect of M-ROSE guiding anti-infection treatment on prognostic outcomes of SHAP patients is still unclear. METHODS/DESIGN: This is a multicenter, single-blind, prospective, randomized controlled trial to evaluate the effect of M-ROSE guiding anti-infection treatment in SHAP patients, which will provide new strategies for the prevention and control of clinical multi-drug resistance bacteria. A total of 166 patients with SHAP, aged 18 years and over, will be recruited from seven centers in Beijing and randomly assigned to the intervention group (M-ROSE combined with mNGS) or the control group (mNGS only) in a 1:1 ratio using the central randomization system. Patients in the intervention group will accept M-ROSE and mNGS analysis, and the control group will accept mNGS analysis. Individualized anti-infective treatment and routine treatment will be selected according to the analysis results. The primary outcome is the ICU outcome (mortality). The safety of the intervention measures will be evaluated during the entire trial period. This trial will be the first randomized controlled trial to evaluate the effect of M-ROSE guiding treatment on mortality in patients with SHAP and may change the prevalence of multi-drug resistant bacteria. ETHICS AND DISSEMINATION: This trial adheres to the Declaration of Helsinki and guidelines of Good Clinical Practice. Signed informed consent will be obtained from all participants. The trial has been approved by the Chinese PLA General Hospital (Approval Number: 20220322001). TRIAL REGISTRATION: ClinicalTrials.gov NCT05300776. Registered on 25 March 2022.


Subject(s)
Anti-Infective Agents , Pneumonia , Humans , Adolescent , Adult , Prospective Studies , Rapid On-site Evaluation , Single-Blind Method , Hospitals, General , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
5.
Discov Med ; 35(177): 539-552, 2023 08.
Article in English | MEDLINE | ID: mdl-37553308

ABSTRACT

BACKGROUND: Influenza is an important respiratory tract pathogen that causes substantial seasonal and pandemic morbidity and mortality. The aim of this study was to systematically analyze the transcriptome characteristics of peripheral blood mononuclear cells (PBMCs) after influenza A virus infection by constructing a human lung microarray model composed of PBMCs to simulate the influenza A virus infection process. METHODS: A human lung microarray model was constructed using alveolar epithelial cells, vascular endothelial cells, alveolar macrophages and PBMCs, for simulation of the process of influenza A virus infection. The transcriptome characteristics of PBMCs after influenza A virus infection were analyzed by a single-cell RNA sequencing system. RESULTS: The study could realistically mimic the structure and physiological functions of the alveoli in vitro using immunofluorescence staining and expression of the specific marker. After the influenza A virus infected the upper lung chip channels, the epithelial cells underwent a high inflammatory response and spread to endothelial cells. Under experimental conditions, the Influenza A virus infection did not compromise the integrity of epithelial cells, but caused damage to endothelial cells and barrier dysfunction. Single-cell RNA sequencing of PBMCs showed that B and cluster of differentiation 4 (CD4) T cells played important immunomodulatory roles in response to influenza A virus infection, including significantly activating type I interferon signaling pathway, regulating cytokine and chemokine signaling pathway. Especially genes involved in cellular communication were significantly highly expressed post-infection. CONCLUSIONS: All these results suggested that the interactions among immune cells played a crucial role in endothelial cell injury and immune cell recruitment after influenza virus infection. This lung-on-chip infection model combined with single-cell RNA sequencing provided a unique platform that can closely investigate the lung immune response to influenza A virus infection and new therapeutic strategies for influenza.


Subject(s)
Influenza A virus , Influenza, Human , Lung Injury , Influenza, Human/complications , Influenza, Human/immunology , Lung Injury/etiology , Lung Injury/immunology , Biosensing Techniques , Humans , Endothelial Cells , Cytokines/immunology , Single-Cell Analysis , Leukocytes, Mononuclear/immunology
6.
Biomed Pharmacother ; 165: 115124, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37454589

ABSTRACT

The therapeutic efficacy of umbilical cord mesenchymal stem cells (UCMSCs) in acute respiratory distress syndrome (ARDS) is mainly limited by the efficiency of homing of UCMSCs toward tissue damage. C-X-C chemokine receptor type 7 (CXCR7), which is involved in the mobilization of UCMSCs, is only expressed on the surface of a small proportion of UCMSCs. This study examined whether overexpression of CXCR7 in UCMSCs (UCMSCsOE-CXCR7) could improve their homing efficiency, and therefore, improve their effectiveness in fibrosis repair at the site of lung injury caused by ARDS. A lentiviral vector expressing CXCR7 was built and then transfect into UCMSCs. The impacts of CXCR7 expression of the proliferationand homing of UCMSCs were examined in a lipopolysaccharide-induced ARDS mouse model. The potential role and underlying mechanism of CXCR7 were examined by performing scratch assays, transwell assays, and immunoassays. The therapeutic dose and treatment time of UCMSCsOE-CXCR7 were directly proportional to their therapeutic effect on lung injury. In addition, overexpression of CXCR7 increased SDF-1-induced proliferation and migration of lung epithelial cells (Base-2b cells), and upregulation of CXCR7 inhibited α-SMA expression, suggesting that CXCR7 may have a role in alleviating pulmonary fibrosis caused by ARDS. Overexpression of CXCR7 in UCMSCs may improve their therapeutic effect of acute lung injury mouse, The mechanism of fibrosis repair by CXCR7 is inhibition of Jag1 via suppression of the Wnt/ß-catenin pathway under the chemotaxis of SDF-1.


Subject(s)
Acute Lung Injury , Mesenchymal Stem Cells , Pulmonary Fibrosis , Respiratory Distress Syndrome , Animals , Mice , Acute Lung Injury/metabolism , beta Catenin/metabolism , Fibrosis , Mesenchymal Stem Cells/metabolism , Pulmonary Fibrosis/therapy , Pulmonary Fibrosis/metabolism , Respiratory Distress Syndrome/metabolism , Umbilical Cord/metabolism , Wnt Signaling Pathway
7.
Int J Biol Macromol ; 142: 846-854, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31622700

ABSTRACT

The green composite cassava starch films were prepared using stearic acid modified microcrystalline cellulose (M-MCC)/nanocellulose (M-NCC) as strength agent, which shows good mechanical and hydrophobic properties and is a candidate for food package in this work. Microcrystalline cellulose (MCC) was prepared with 98% phosphoric acid hydrolysis and mechanical stirring at 600 r/min and nanocellulose (NCC) was prepared with 65% sulfuric acid hydrolysis. After using stearic acid to modify MCC and NCC, the casting method was used to prepare the M-MCC/cassava starch composite films and M-NCC/cassava starch films. Physical, mechanical, hydrophobic and thermal properties were characterized using SEM, electronic universal testing machine, contact angle meter and TGA. The results showed that: M-MCC and M-NCC can lead to the enhancement of mechanical and hydrophobic properties of composite films; 0.5% M-MCC and 1.5% M-NCC had the highest enhancement effect on mechanical properties, leading the tensile strength of cassava starch film increased by 484.5% and 327.7% respectively; As to hydrophobic property, when 2% M-MCC and 0.5% M-NCC added, the hydrophobicity of the film increased by 65.0% and 30.3% respectively. Overall, the enhancement effect of M-MCC was better than M-NCC. But when M-MCC/M-NCC was added, the thermal stability of films reduced.


Subject(s)
Cellulose/chemistry , Manihot/chemistry , Nanostructures/chemistry , Starch/chemistry , Stearic Acids/chemistry , Elastic Modulus , Food Packaging/methods , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Membranes, Artificial , Permeability , Phosphoric Acids/chemistry , Sulfuric Acids/chemistry , Temperature , Tensile Strength
SELECTION OF CITATIONS
SEARCH DETAIL
...