Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 648
Filter
1.
Cells ; 13(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38786077

ABSTRACT

Patients with COVID-19 have coagulation and platelet disorders, with platelet alterations and thrombocytopenia representing negative prognostic parameters associated with severe forms of the disease and increased lethality. METHODS: The aim of this study was to study the expression of platelet glycoprotein IIIa (CD61), playing a critical role in platelet aggregation, together with TRL-2 as a marker of innate immune activation. RESULTS: A total of 25 patients were investigated, with the majority (24/25, 96%) having co-morbidities and dying from a fatal form of SARS-CoV-2(+) infection (COVID-19+), with 13 men and 12 females ranging in age from 45 to 80 years. When compared to a control group of SARS-CoV-2 (-) negative lungs (COVID-19-), TLR-2 expression was up-regulated in a subset of patients with deadly COVID-19 fatal lung illness. The proportion of Spike-1 (+) patients found by PCR and ISH correlates to the proportion of Spike-S1-positive cases as detected by digital pathology examination. Furthermore, CD61 expression was considerably higher in the lungs of deceased patients. In conclusion, we demonstrate that innate immune prolonged hyperactivation is related to platelet/megakaryocyte over-expression in the lung. CONCLUSIONS: Microthrombosis in deadly COVID-19+ lung disease is associated with an increase in the number of CD61+ platelets and megakaryocytes in the pulmonary interstitium, as well as their functional activation; this phenomenon is associated with increased expression of innate immunity TLR2+ cells, which binds the SARS-CoV-2 E protein, and significantly with the persistence of the Spike-S1 viral sequence.


Subject(s)
COVID-19 , Lung , Megakaryocytes , SARS-CoV-2 , Thrombosis , Toll-Like Receptor 2 , Up-Regulation , Humans , COVID-19/pathology , COVID-19/immunology , COVID-19/metabolism , Male , Female , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Megakaryocytes/metabolism , Megakaryocytes/pathology , Megakaryocytes/virology , Aged , Middle Aged , Aged, 80 and over , Lung/pathology , Lung/virology , Lung/metabolism , Up-Regulation/genetics , Thrombosis/pathology , Integrin beta3/metabolism , Integrin beta3/genetics , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Pneumonia, Viral/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , Pneumonia, Viral/metabolism , Immunity, Innate , Pandemics
2.
J Sep Sci ; 47(11): e2400090, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38819782

ABSTRACT

Ephedra herb (EH), an important medicine prescribed in herbal formulas by Traditional Chinese Medicine practitioners, has been widely used in the treatment of viral pneumonia in China. However, the molecular basis of EH in viral pneumonia remains unclear. In this study, a ternary correlation multi-symptom network strategy was established based on in vivo chemical profile identification and metabolomics to explore the molecular basis of EH against viral pneumonia. Results showed that 143 compounds of EH and 70 prototype components were identified in vivo. EH could reduce alveolar-capillary barrier disruption in rats with viral pneumonia and significantly downregulate the expression of inflammatory factors and bronchoalveolar lavage fluid. Plasma metabolomics revealed that EH may be involved in the regulation of arachidonic acid, tryptophan, tyrosine, nicotinate, and nicotinamide metabolism. The multi-symptom network showed that 12 compounds have an integral function in the treatment of viral pneumonia by intervening in many pathways related to viruses, immunity and inflammation, and lung injury. Further verification demonstrated that sinapic acid and frambinone can regulate the expression of related genes. It has been shown to be a promising representative of the pharmacological constituents of ephedra.


Subject(s)
Drugs, Chinese Herbal , Ephedra , Metabolomics , Rats, Sprague-Dawley , Animals , Rats , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Ephedra/chemistry , Male , Pneumonia, Viral/drug therapy , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology
3.
Nat Commun ; 15(1): 4235, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762489

ABSTRACT

Inflammation induced by lung infection is a double-edged sword, moderating both anti-viral and immune pathogenesis effects; the mechanism of the latter is not fully understood. Previous studies suggest the vasculature is involved in tissue injury. Here, we report that expression of Sparcl1, a secreted matricellular protein, is upregulated in pulmonary capillary endothelial cells (EC) during influenza-induced lung injury. Endothelial overexpression of SPARCL1 promotes detrimental lung inflammation, with SPARCL1 inducing 'M1-like' macrophages and related pro-inflammatory cytokines, while SPARCL1 deletion alleviates these effects. Mechanistically, SPARCL1 functions through TLR4 on macrophages in vitro, while TLR4 inhibition in vivo ameliorates excessive inflammation caused by endothelial Sparcl1 overexpression. Finally, SPARCL1 expression is increased in lung ECs from COVID-19 patients when compared with healthy donors, while fatal COVID-19 correlates with higher circulating SPARCL1 protein levels in the plasma. Our results thus implicate SPARCL1 as a potential prognosis biomarker for deadly COVID-19 pneumonia and as a therapeutic target for taming hyperinflammation in pneumonia.


Subject(s)
COVID-19 , Endothelial Cells , Lung , Macrophage Activation , SARS-CoV-2 , Animals , Humans , COVID-19/immunology , COVID-19/virology , COVID-19/metabolism , COVID-19/pathology , Mice , Endothelial Cells/metabolism , Endothelial Cells/virology , Endothelial Cells/immunology , SARS-CoV-2/physiology , Lung/virology , Lung/pathology , Lung/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Mice, Inbred C57BL , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Pneumonia, Viral/metabolism , Male , Macrophages/metabolism , Macrophages/immunology , Female , Mice, Knockout , Extracellular Matrix Proteins
4.
EBioMedicine ; 103: 105132, 2024 May.
Article in English | MEDLINE | ID: mdl-38677182

ABSTRACT

BACKGROUND: SARS-CoV-2 infection is considered as a relapsing inflammatory process with a dysregulation of IL-6 signalling. Classic IL-6 signalling is thought to represent a defence mechanism against pathogens. In contrast, IL-6 trans-signalling has pro-inflammatory effects. In severe COVID-19, therapeutic strategies have focused on global inhibition of IL-6, with controversial results. We hypothesized that specific blockade of IL-6 trans-signalling could inhibit inflammatory response preserving the host defence activity inherent to IL-6 classic signalling. METHODS: To test the role of the specific IL-6 trans-signalling inhibition by sgp130Fc in short- and long-term consequences of COVID-19, we used the established K18-hACE2 transgenic mouse model. Histological as well as immunohistochemical analysis, and pro-inflammatory marker profiling were performed. To investigate IL-6 trans-signalling in human cells we used primary lung microvascular endothelial cells and fibroblasts in the presence/absence of sgp130Fc. FINDINGS: We report that targeting IL-6 trans-signalling by sgp130Fc attenuated SARS-CoV-2-related clinical symptoms and mortality. In surviving mice, the treatment caused a significant decrease in lung damage. In vitro, IL-6 trans-signalling induced strong and persisting JAK1/STAT3 activation in endothelial cells and lung fibroblasts with proinflammatory effects, which were attenuated by sgp130Fc. Our data also suggest that in those cells with scant amounts of IL-6R, the induction of gp130 and IL-6 by IL-6:sIL-6R complex sustains IL-6 trans-signalling. INTERPRETATION: IL-6 trans-signalling fosters progression of COVID-19, and suggests that specific blockade of this signalling mode could offer a promising alternative to mitigate both short- and long-term consequences without affecting the beneficial effects of IL-6 classic signalling. These results have implications for the development of new therapies of lung injury and endotheliopathy in COVID-19. FUNDING: The project was supported by ISCIII, Spain (COV-20/00792 to MB, PI23/01351 to MARH) and the European Commission-Next generation EU (European Union) (Regulation EU 2020/2094), through CSIC's Global Health Platform (PTI Salud Global, SGL2103029 to MB). PID2019-110587RB-I00 (MB) supported by MICIN/AEI/10.13039/501100011033/and PID2022-143034OB-I00 (MB) by MICIN/AEI/10.13039/501100011033/FEDER. MAR-H acknowledges support from ISCIII, Spain and the European Commission-Next generation EU (European Union), through CSIC's Global Health PTI.


Subject(s)
COVID-19 , Cytokine Receptor gp130 , Disease Models, Animal , Interleukin-6 , Mice, Transgenic , SARS-CoV-2 , Signal Transduction , Animals , Interleukin-6/metabolism , COVID-19/metabolism , Humans , Mice , Signal Transduction/drug effects , Cytokine Receptor gp130/metabolism , Cytokine Receptor gp130/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Lung/pathology , Lung/virology , Lung/metabolism , Endothelial Cells/metabolism , COVID-19 Drug Treatment , Betacoronavirus , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Coronavirus Infections/pathology , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/antagonists & inhibitors , Severity of Illness Index
5.
Mol Ther ; 32(5): 1510-1525, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38454605

ABSTRACT

The acute respiratory virus infection can induce uncontrolled inflammatory responses, such as cytokine storm and viral pneumonia, which are the major causes of death in clinical cases. Cyclophilin A (CypA) is mainly distributed in the cytoplasm of resting cells and released into the extracellular space in response to inflammatory stimuli. Extracellular CypA (eCypA) is upregulated and promotes inflammatory response in severe COVID-19 patients. However, how eCypA promotes virus-induced inflammatory response remains elusive. Here, we observe that eCypA is induced by influenza A and B viruses and SARS-CoV-2 in cells, mice, or patients. Anti-CypA mAb reduces pro-inflammatory cytokines production, leukocytes infiltration, and lung injury in virus-infected mice. Mechanistically, eCypA binding to integrin ß2 triggers integrin activation, thereby facilitating leukocyte trafficking and cytokines production via the focal adhesion kinase (FAK)/GTPase and FAK/ERK/P65 pathways, respectively. These functions are suppressed by the anti-CypA mAb that specifically blocks eCypA-integrin ß2 interaction. Overall, our findings reveal that eCypA-integrin ß2 signaling mediates virus-induced inflammatory response, indicating that eCypA is a potential target for antibody therapy against viral pneumonia.


Subject(s)
COVID-19 , Cyclophilin A , Cyclophilin A/metabolism , Animals , Humans , Mice , COVID-19/metabolism , COVID-19/virology , COVID-19/immunology , CD18 Antigens/metabolism , SARS-CoV-2 , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Pneumonia, Viral/metabolism , Pneumonia, Viral/immunology , Cytokines/metabolism , Antibodies, Monoclonal/pharmacology , Signal Transduction , Influenza A virus , Disease Models, Animal
6.
Proc Natl Acad Sci U S A ; 121(7): e2314747121, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38315853

ABSTRACT

Macrophages are integral components of the innate immune system, playing a dual role in host defense during infection and pathophysiological states. Macrophages contribute to immune responses and aid in combatting various infections, yet their production of abundant proinflammatory cytokines can lead to uncontrolled inflammation and worsened tissue damage. Therefore, reducing macrophage-derived proinflammatory cytokine release represents a promising approach for treating various acute and chronic inflammatory disorders. However, limited macrophage-specific delivery vehicles have hindered the development of macrophage-targeted therapies. In this study, we screened a pool of 112 lipid nanoparticles (LNPs) to identify an optimal LNP formulation for efficient siRNA delivery. Subsequently, by conjugating the macrophage-specific antibody F4/80 to the LNP surface, we constructed MacLNP, an enhanced LNP formulation designed for targeted macrophage delivery. In both in vitro and in vivo experiments, MacLNP demonstrated a significant enhancement in targeting macrophages. Specifically, delivery of siRNA targeting TAK1, a critical kinase upstream of multiple inflammatory pathways, effectively suppressed the phosphorylation/activation of NF-kB. LNP-mediated inhibition of NF-kB, a key upstream regulator in the classic inflammatory signaling pathway, in the murine macrophage cell line RAW264.7 significantly reduced the release of proinflammatory cytokines after stimulation with the viral RNA mimic Poly(I:C). Finally, intranasal administration of MacLNP-encapsulated TAK1 siRNA markedly ameliorated lung injury induced by influenza infection. In conclusion, our findings validate the potential of targeted macrophage interventions in attenuating inflammatory responses, reinforcing the potential of LNP-mediated macrophage targeting to treat pulmonary inflammatory disorders.


Subject(s)
Liposomes , Nanoparticles , Pneumonia, Viral , Mice , Humans , Animals , NF-kappa B/metabolism , Lipids/pharmacology , Macrophages/metabolism , RNA, Small Interfering/metabolism , Cytokines/metabolism , Pneumonia, Viral/metabolism
7.
Front Immunol ; 14: 1169968, 2023.
Article in English | MEDLINE | ID: mdl-37180156

ABSTRACT

Introduction: Human adenovirus (HAdV) is a common respiratory virus, which can lead to severe pneumonia in children and immunocompromised persons, and canonical inflammasomes are reported to be involved in anti-HAdV defense. However, whether HAdV induced noncanonical inflammasome activation has not been explored. This study aims to explore the broad roles of noncanonical inflammasomes during HAdV infection to investigate the regulatory mechanism of HAdV-induced pulmonary inflammatory damage. Methods: We mined available data on GEO database and collected clinical samples from adenovirus pneumonia pediatric patients to investigate the expression of noncanonical inflammasome and its clinical relevance. An in vitro cell model was employed to investigate the roles of noncanonical inflammasomes in macrophages in response to HAdV infection. Results: Bioinformatics analysis showed that inflammasome-related genes, including caspase-4 and caspase-5, were enriched in adenovirus pneumonia. Moreover, caspase-4 and caspase-5 expression levels were significantly increased in the cells isolated from peripheral blood and broncho-alveolar lavage fluid (BALF) of pediatric patients with adenovirus pneumonia, and positively correlated with clinical parameters of inflammatory damage. In vitro experiments revealed that HAdV infection promoted caspase-4/5 expression, activation and pyroptosis in differentiated THP-1 (dTHP-1) human macrophages via NF-κB, rather than STING signaling pathway. Interestingly, silencing of caspase-4 and caspase-5 in dTHP-1 cells suppressed HAdV-induced noncanonical inflammasome activation and macrophage pyroptosis, and dramatically decreased the HAdV titer in cell supernatants, by influencing virus release rather than other stages of virus life cycle. Discussion: In conclusion, our study demonstrated that HAdV infection induced macrophage pyroptosis by triggering noncanonical inflammasome activation via a NF-kB-dependent manner, which may explore new perspectives on the pathogenesis of HAdV-induced inflammatory damage. And high expression levels of caspase-4 and caspase-5 may be a biomarker for predicting the severity of adenovirus pneumonia.


Subject(s)
Adenoviridae Infections , Adenovirus Infections, Human , Pneumonia, Viral , Humans , Child , Inflammasomes/metabolism , Pyroptosis , Adenovirus Infections, Human/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Caspases/metabolism , Pneumonia, Viral/metabolism , Adenoviridae Infections/complications
8.
Cell Death Dis ; 14(1): 66, 2023 01 28.
Article in English | MEDLINE | ID: mdl-36707501

ABSTRACT

Coronavirus disease 2019 (COVID-19) treatments are still urgently needed for critically and severely ill patients. Human umbilical cord-mesenchymal stem cells (hUC-MSCs) infusion has therapeutic benefits in COVID-19 patients; however, uncertain therapeutic efficacy has been reported in severe patients. In this study, we selected an appropriate cytokine, IL-18, based on the special cytokine expression profile in severe pneumonia of mice induced by H1N1virus to prime hUC-MSCs in vitro and improve the therapeutic effect of hUC-MSCs in vivo. In vitro, we demonstrated that IL-18-primed hUC-MSCs (IL18-hUCMSC) have higher proliferative ability than non-primed hUC-MSCs (hUCMSCcon). In addition, VCAM-1, MMP-1, TGF-ß1, and some chemokines (CCL2 and CXCL12 cytokines) are more highly expressed in IL18-hUCMSCs. We found that IL18-hUCMSC significantly enhanced the immunosuppressive effect on CD3+ T-cells. In vivo, we demonstrated that IL18-hUCMSC infusion could reduce the body weight loss caused by a viral infection and significantly improve the survival rate. Of note, IL18-hUCMSC can also significantly attenuate certain clinical symptoms, including reduced activity, ruffled fur, hunched backs, and lung injuries. Pathologically, IL18-hUCMSC transplantation significantly enhanced the inhibition of inflammation, viral load, fibrosis, and cell apoptosis in acute lung injuries. Notably, IL18-hUCMSC treatment has a superior inhibitory effect on T-cell exudation and proinflammatory cytokine secretion in bronchoalveolar lavage fluid (BALF). Altogether, IL-18 is a promising cytokine that can prime hUC-MSCs to improve the efficacy of precision therapy against viral-induced pneumonia, such as COVID-19.


Subject(s)
COVID-19 , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Pneumonia, Viral , Humans , Mice , Animals , Interleukin-18/metabolism , Umbilical Cord/metabolism , T-Lymphocytes/metabolism , COVID-19/metabolism , Cytokines/metabolism , Pneumonia, Viral/therapy , Pneumonia, Viral/metabolism , Immunosuppression Therapy , Mesenchymal Stem Cells/metabolism
9.
Pathol Int ; 72(10): 506-518, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36066006

ABSTRACT

Pandemic influenza virus A(H1N1)pdm09 infection occurred in healthy children and young adults, but asthmatic patients presented more rapid progression of respiratory distress and plastic bronchitis. To investigate the pathogenesis of worsening respiratory symptoms after A(H1N1)pdm09 infection, we focused on matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinases-1 (TIMP-1). MMP-9 and TIMP-1 levels in bronchoalveolar lavage fluid and serum from mice with and without asthma were evaluated after A(H1N1)pdm09 or seasonal A(H1N1) infection. MMP-9 levels were more elevated in Asthma/A(H1N1)pdm09-infected mice than in non-Asthma/A(H1N1)pdm09-infected mice on both 3 and 7 days post-infection. Immunohistochemical findings in this pneumonia model showed that MMP-9 and TIMP-1 positive cells were observed in blood vessels and bronchus of lung tissue in severe pathological findings of pneumonia with asthma. Microscopically, shedding cells and secretions were conspicuous in the trachea on days 3 and 7 post-infection, in the A(H1N1)pdm09-infected mice with asthma. Our results suggest that MMP-9 and TIMP-1 expressions are related to severe pneumonia in the A(H1N1)pdm09 infection with asthma, leading to cause epithelial cell shedding.


Subject(s)
Asthma , Matrix Metalloproteinase 9 , Orthomyxoviridae Infections , Pneumonia, Viral , Tissue Inhibitor of Metalloproteinase-1 , Animals , Asthma/metabolism , Disease Models, Animal , Influenza A Virus, H1N1 Subtype , Matrix Metalloproteinase 9/metabolism , Mice , Orthomyxoviridae Infections/metabolism , Plastics , Pneumonia, Viral/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism
10.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35177474

ABSTRACT

Viral causes of pneumonia pose constant threats to global public health, but there are no specific treatments currently available for the condition. Antivirals are ineffective when administered late after the onset of symptoms. Pneumonia is caused by an exaggerated inflammatory cytokine response to infection, but tissue necrosis and damage caused by virus also contribute to lung pathology. We hypothesized that viral pneumonia can be treated effectively if both virus and inflammation are simultaneously targeted. Combined treatment with the antiviral drug cidofovir and etanercept, which targets tumor necrosis factor (TNF), down-regulated nuclear factor kappa B-signaling and effectively reduced morbidity and mortality during respiratory ectromelia virus (ECTV) infection in mice even when treatment was initiated after onset of clinical signs. Treatment with cidofovir alone reduced viral load, but animals died from severe lung pathology. Treatment with etanercept had no effect on viral load but diminished levels of inflammatory cytokines and chemokines including TNF, IL-6, IL-1ß, IL-12p40, TGF-ß, and CCL5 and dampened activation of the STAT3 cytokine-signaling pathway, which transduces signals from multiple cytokines implicated in lung pathology. Consequently, combined treatment with a STAT3 inhibitor and cidofovir was effective in improving clinical disease and lung pathology in ECTV-infected mice. Thus, the simultaneous targeting of virus and a specific inflammatory cytokine or cytokine-signaling pathway is effective in the treatment of pneumonia. This approach might be applicable to pneumonia caused by emerging and re-emerging viruses, like seasonal and pandemic influenza A virus strains and severe acute respiratory syndrome coronavirus 2.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Antiviral Agents/therapeutic use , Cidofovir/therapeutic use , Etanercept/administration & dosage , Pneumonia, Viral/drug therapy , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Cidofovir/pharmacology , Cytokines/metabolism , Drug Evaluation, Preclinical , Drug Therapy, Combination , Ectromelia virus/drug effects , Female , Lung/drug effects , Lung/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Pneumonia, Viral/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Viral Load/drug effects
11.
Front Immunol ; 12: 785457, 2021.
Article in English | MEDLINE | ID: mdl-34868067

ABSTRACT

Respirovirus such as influenza virus infection induces pulmonary anti-viral immune response, orchestration of innate and adaptive immunity restrain viral infection, otherwise causes severe diseases such as pneumonia. Chemokines regulate leukocyte recruitment to the inflammation site. One chemokine CXCL5, plays a scavenging role to regulate pulmonary host defense against bacterial infection, but its role in pulmonary influenza virus infection is underdetermined. Here, using an influenza (H1N1) infected CXCL5-/- mouse model, we found that CXCL5 not only responds to neutrophil infiltration into infected lungs at the innate immunity stage, but also affects B lymphocyte accumulation in the lungs by regulating the expression of the B cell chemokine CXCL13. Inhibition of CXCL5-CXCR2 axis markedly induces CXCL13 expression in CD64+CD44hiCD274hi macrophages/monocytes in infected lungs, and in vitro administration of CXCL5 to CD64+ alveolar macrophages suppresses CXCL13 expression via the CXCL5-CXCR2 axis upon influenza challenge. CXCL5 deficiency leads to increased B lymphocyte accumulation in infected lungs, contributing to an enhanced B cell immune response and facilitating induced bronchus-associated lymphoid tissue formation in the infected lungs during the late infection and recovery stages. These data highlight multiple regulatory roles of CXCL5 in leukocyte chemotaxis during pulmonary influenza infection.


Subject(s)
Adaptive Immunity , Chemokine CXCL5/metabolism , Chemotaxis/immunology , Immunity, Innate , Influenza, Human/complications , Pneumonia, Viral/etiology , Pneumonia, Viral/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Chemokine CXCL5/genetics , Chemotaxis/genetics , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions , Humans , Immunophenotyping , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/pathology , Influenza, Human/virology , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Pneumonia, Viral/pathology , Signal Transduction
12.
Physiol Rep ; 9(21): e15081, 2021 11.
Article in English | MEDLINE | ID: mdl-34755490

ABSTRACT

Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Orthomyxoviridae/pathogenicity , Pneumonia, Viral/drug therapy , Angiopoietin-2/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Cells, Cultured , Cytokines/metabolism , Humans , Lung/metabolism , Lung/virology , Mice , Mice, Inbred C57BL , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Receptor, TIE-2/metabolism , Viral Load
13.
Sci Rep ; 11(1): 20621, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663864

ABSTRACT

Pneumonia is the main reason for mortality among children under five years, causing 1.6 million deaths every year; late research has exhibited that mortality is increasing in the elderly. A few biomarkers used for its diagnosis need specificity and precision, as they are related to different infections, for example, pulmonary tuberculosis and Human Immunodeficiency Virus. There is a quest for new biomarkers worldwide to diagnose the disease to defeat these previously mentioned constraints. Antimicrobial peptides (AMPs) are promising indicative specialists against infection. This research work used AMPs as biomarkers to detect viral pneumonia pathogens, for example, Respiratory syncytial virus, Influenza A and B viruses utilizing in silico technologies, such as Hidden Markov Model (HMMER). HMMER was used to distinguish putative anti-viral pneumonia AMPs against the recognized receptor proteins of Respiratory syncytial virus, Influenza A, and B viruses. The physicochemical parameters of these putative AMPs were analyzed, and their 3-D structures were determined utilizing I-TASSER. Molecular docking interaction of these AMPs against the recognized viral pneumonia proteins was carried out using the PATCHDOCK and HDock servers. The results demonstrated 27 anti-viral AMPs ranked based on their E values with significant physicochemical parameters in similarity with known experimentally approved AMPs. The AMPs additionally had a high anticipated binding potential to the pneumonia receptors of these microorganisms sensitively. The tendency of the putative anti-viral AMPs to bind pneumonia proteins showed that they would be promising applicant biomarkers to identify these viral microorganisms in the point-of-care (POC) pneumonia diagnostics. The high precision observed for the AMPs legitimizes HMM's utilization in the disease diagnostics' discovery process.


Subject(s)
Antimicrobial Peptides/analysis , Antimicrobial Peptides/chemistry , Pneumonia, Viral/drug therapy , Antimicrobial Peptides/genetics , Antiviral Agents/therapeutic use , Biomarkers/analysis , Computational Biology/methods , Computer Simulation , Databases, Factual , Humans , Influenza A virus , Influenza B virus , Markov Chains , Molecular Docking Simulation , Pneumonia, Viral/metabolism , Protein Binding , Respiratory Syncytial Viruses
14.
Med Sci Monit ; 27: e930776, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34635632

ABSTRACT

During the coronavirus disease 2019 (COVID-19) pandemic due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, patients presented with COVID-19 pneumonia of varying severity. The phenomenon of severe hypoxemia without signs of respiratory distress is also known as silent or hidden hypoxemia. Although silent hypoxemia is not unique to pneumonia due to SARS-CoV-2 infection, this phenomenon is now recognized to be associated with severe COVID-19 pneumonia. Proper management of critically ill patients is the key to reducing mortality. Herein, we summarize the possible and rare factors contributing to silent hypoxemia in patients with COVID-19. Microvascular thrombosis causes dead space ventilation in the lungs, and the flow of pulmonary capillaries is reduced, which leads to an imbalance in the V/Q ratio. The dissociation curve of oxyhemoglobin shifts to the left and limits the release of oxygen to the tissue. SARS-CoV-2 interferes with the synthesis of hemoglobin and reduces the ability to carry oxygen. The accumulation of endogenous carbon monoxide and carboxyhemoglobin will reduce the total oxygen carrying capacity and interfere with pulse oxygen saturation readings. There are also some non-specific factors that cause the difference between pulse oximetry and oxygen partial pressure. We propose some potentially more effective clinical alternatives and recommendations for optimizing the clinical management processes of patients with COVID-19. This review aims to describe the prevalence of silent hypoxemia in COVID-19 pneumonia, to provide an update on what is known of the pathophysiology, and to highlight the importance of diagnosing silent hypoxemia in patients with COVID-19 pneumonia.


Subject(s)
COVID-19/metabolism , Hypoxia/virology , Pneumonia, Viral/virology , Asymptomatic Diseases/epidemiology , COVID-19/epidemiology , COVID-19/virology , Humans , Hypoxia/epidemiology , Hypoxia/metabolism , Lung/cytology , Lung/metabolism , Lung/virology , Microvessels/metabolism , Oximetry , Oxygen/metabolism , Pneumonia, Viral/metabolism , Prevalence , SARS-CoV-2/isolation & purification , Thrombosis/metabolism , Thrombosis/virology
15.
Cell Rep ; 37(1): 109798, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34587481

ABSTRACT

Despite the worldwide effect of the coronavirus disease 2019 (COVID-19) pandemic, the underlying mechanisms of fatal viral pneumonia remain elusive. Here, we show that critical COVID-19 is associated with enhanced eosinophil-mediated inflammation when compared to non-critical cases. In addition, we confirm increased T helper (Th)2-biased adaptive immune responses, accompanying overt complement activation, in the critical group. Moreover, enhanced antibody responses and complement activation are associated with disease pathogenesis as evidenced by formation of immune complexes and membrane attack complexes in airways and vasculature of lung biopsies from six fatal cases, as well as by enhanced hallmark gene set signatures of Fcγ receptor (FcγR) signaling and complement activation in myeloid cells of respiratory specimens from critical COVID-19 patients. These results suggest that SARS-CoV-2 infection may drive specific innate immune responses, including eosinophil-mediated inflammation, and subsequent pulmonary pathogenesis via enhanced Th2-biased immune responses, which might be crucial drivers of critical disease in COVID-19 patients.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Complement System Proteins/immunology , Eosinophils/immunology , Inflammation/immunology , Pneumonia, Viral/immunology , SARS-CoV-2/immunology , Adaptive Immunity , Adult , Aged , Aged, 80 and over , Antigen-Antibody Complex/metabolism , COVID-19/metabolism , COVID-19/virology , Complement Activation , Complement Membrane Attack Complex/metabolism , Eosinophils/virology , Female , Humans , Inflammation/metabolism , Inflammation/virology , Lung Injury/immunology , Lung Injury/pathology , Lung Injury/virology , Male , Middle Aged , Pneumonia, Viral/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Severity of Illness Index , Signal Transduction , Th2 Cells/immunology , Viral Load , Young Adult
16.
Toxicol Appl Pharmacol ; 426: 115645, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34271066

ABSTRACT

Elevated ambient temperatures and extreme weather events have increased the incidence of wildfires world-wide resulting in increased wood smoke particle (WSP). Epidemiologic data suggests that WSP exposure associates with exacerbations of respiratory diseases, and with increased respiratory viral infections. To assess the impact of WSP exposure on host response to viral pneumonia, we performed WSP exposures in rodents followed by infection with mouse adapted influenza (HINI-PR8). C57BL/6 male mice aged 6-8 weeks were challenged with WSP or PBS by oropharyngeal aspiration in acute (single dose) or sub-acute exposures (day 1, 3, 5, 7 and 10). Additional groups underwent sub-acute exposure followed by infection by influenza or heat-inactivated (HI) virus. Following exposures/infection, bronchoalveolar lavage (BAL) was performed to assess for total cell counts/differentials, total protein, protein carbonyls and hyaluronan. Lung tissue was assessed for viral counts by real time PCR. When compared to PBS, acute WSP exposure associated with an increase in airspace macrophages. Alternatively, sub-acute exposure resulted in a dose dependent increase in airspace neutrophils. Sub-acute WSP exposure followed by influenza infection was associated with improved respiratory viral outcomes including reduced weight loss and increased blood oxygen saturation, and decreased protein carbonyls and viral titers. Flow cytometry demonstrated dynamic changes in pulmonary macrophage and T cell subsets based on challenge with WSP and influenza. This data suggests that sub-acute WSP exposure can improve host response to acute influenza infection.


Subject(s)
Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections , Pneumonia, Viral , Smoke , Wildfires , Administration, Inhalation , Animals , Influenza A Virus, H1N1 Subtype/physiology , Lung/immunology , Lung/metabolism , Lung/virology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Severity of Illness Index , Transcriptome , Virus Replication , Wood
17.
Crit Care ; 25(1): 234, 2021 07 03.
Article in English | MEDLINE | ID: mdl-34217339

ABSTRACT

BACKGROUND: The coronavirus disease 2019 (COVID-19) has induced a worldwide epidemiological event with a high infectivity and mortality. However, the predicting biomarkers and their potential mechanism in the progression of COVID-19 are not well known. OBJECTIVE: The aim of this study is to identify the candidate predictors of COVID-19 and investigate their underlying mechanism. METHODS: The retrospective study was conducted to identify the potential laboratory indicators with prognostic values of COVID-19 disease. Then, the prognostic nomogram was constructed to predict the overall survival of COVID-19 patients. Additionally, the scRNA-seq data of BALF and PBMCs from COVID-19 patients were downloaded to investigate the underlying mechanism of the most important prognostic indicators in lungs and peripherals, respectively. RESULTS: In total, 304 hospitalized adult COVID-19 patients in Wuhan Jinyintan Hospital were included in the retrospective study. CEA was the only laboratory indicator with significant difference in the univariate (P < 0.001) and multivariate analysis (P = 0.020). The scRNA-seq data of BALF and PBMCs from COVID-19 patients were downloaded to investigate the underlying mechanism of CEA in lungs and peripherals, respectively. The results revealed the potential roles of CEA were significantly distributed in type II pneumocytes of BALF and developing neutrophils of PBMCs, participating in the progression of COVID-19 by regulating the cell-cell communication. CONCLUSION: This study identifies the prognostic roles of CEA in COVID-19 patients and implies the potential roles of CEACAM8-CEACAM6 in the progression of COVID-19 by regulating the cell-cell communication of developing neutrophils and type II pneumocyte.


Subject(s)
COVID-19/metabolism , Carcinoembryonic Antigen/metabolism , Pneumonia, Viral/metabolism , Adult , Aged , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/chemistry , COVID-19/mortality , Cell Communication , China/epidemiology , Disease Progression , Hospitalization , Humans , Male , Middle Aged , Neutrophils/metabolism , Nomograms , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , Predictive Value of Tests , Prognosis , Retrospective Studies , SARS-CoV-2 , Survival Analysis
18.
Infect Dis Now ; 51(5): 429-434, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34146758

ABSTRACT

INTRODUCTION: The SARS-CoV-2 virus affects many organs, especially the lungs, with widespread inflammation. We aimed to compare the endogenous oxidative damage markers of coenzyme Q10, nicotinamide dinucleotide oxidase 4, malondialdehyde, and ischemia-modified albumin levels in patients with pneumonia caused by SARS-CoV-2 and in an healthy control group. We also aimed to compare these parameters between patients with severe and non-severe pulmonary involvement. METHODS: The study included 58 adult patients with SARS-CoV-2 pneumonia and 30 healthy volunteers. CoQ10 and MDA levels were determined by high-pressure liquid chromatography. NOX4 and IMA levels were determined by ELISA assay and colorimetric method. RESULTS: Higher levels of CoQ10, MDA, NOX4, and IMA and lower levels of COQ10H were observed in patients with SARS-CoV-2 pneumonia than in the control group. MDA, IMA, NOX4, and CoQ10 levels were significantly higher in patients with severe pulmonary involvement than in patients with non-severe pulmonary involvement, but no significant difference was observed in CoQ10H levels. CoQ10 levels were significantly and positively correlated with both ferritin and CRP levels. CONCLUSION: SARS-CoV-2 pneumonia is significantly associated with increased endogenous oxidative damage. Oxidative damage seems to be associated with pulmonary involvement severity.


Subject(s)
COVID-19/blood , COVID-19/metabolism , Oxidative Stress , Pneumonia, Viral/blood , Pneumonia, Viral/metabolism , Adult , Aged , Biomarkers/blood , Female , Humans , Male , Middle Aged , Severity of Illness Index
19.
Am J Epidemiol ; 190(10): 2094-2106, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33984860

ABSTRACT

Longitudinal trajectories of vital signs and biomarkers during hospital admission of patients with COVID-19 remain poorly characterized despite their potential to provide critical insights about disease progression. We studied 1884 patients with severe acute respiratory syndrome coronavirus 2 infection from April 3, 2020, to June 25, 2020, within 1 Maryland hospital system and used a retrospective longitudinal framework with linear mixed-effects models to investigate relevant biomarker trajectories leading up to 3 critical outcomes: mechanical ventilation, discharge, and death. Trajectories of 4 vital signs (respiratory rate, ratio of oxygen saturation (Spo2) to fraction of inspired oxygen (Fio2), pulse, and temperature) and 4 laboratory values (C-reactive protein (CRP), absolute lymphocyte count (ALC), estimated glomerular filtration rate, and D-dimer) clearly distinguished the trajectories of patients with COVID-19. Before any ventilation, log(CRP), log(ALC), respiratory rate, and Spo2-to-Fio2 ratio trajectories diverge approximately 8-10 days before discharge or death. After ventilation, log(CRP), log(ALC), respiratory rate, Spo2-to-Fio2 ratio, and estimated glomerular filtration rate trajectories again diverge 10-20 days before death or discharge. Trajectories improved until discharge and remained unchanged or worsened until death. Our approach characterizes the distribution of biomarker trajectories leading up to competing outcomes of discharge versus death. Moving forward, this model can contribute to quantifying the joint probability of biomarkers and outcomes when provided clinical data up to a given moment.


Subject(s)
Biomarkers/metabolism , COVID-19/metabolism , Outcome Assessment, Health Care , Pneumonia, Viral/metabolism , COVID-19/diagnosis , COVID-19/epidemiology , Case-Control Studies , Disease Progression , Female , Humans , Longitudinal Studies , Male , Maryland/epidemiology , Pandemics , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Predictive Value of Tests , Retrospective Studies , SARS-CoV-2 , Vital Signs
20.
Cells ; 10(5)2021 04 21.
Article in English | MEDLINE | ID: mdl-33919158

ABSTRACT

Extracellular vesicles (EVs) refer to a heterogenous population of membrane-bound vesicles that are released by cells under physiological and pathological conditions. The detection of EVs in the majority of the bodily fluids, coupled with their diverse cargo comprising of DNA, RNA, lipids, and proteins, have led to the accumulated interests in leveraging these nanoparticles for diagnostic and therapeutic purposes. In particular, emerging studies have identified enhanced levels of a wide range of specific subclasses of non-coding RNAs (ncRNAs) in EVs, thereby suggesting the existence of highly selective and regulated molecular processes governing the sorting of these RNAs into EVs. Recent studies have also illustrated the functional relevance of these enriched ncRNAs in a variety of human diseases. This review summarizes the current state of knowledge on EV-ncRNAs, as well as their functions and significance in lung infection and injury. As a majority of the studies on EV-ncRNAs in lung diseases have focused on EV-microRNAs, we will particularly highlight the relevance of these molecules in the pathophysiology of these conditions, as well as their potential as novel biomarkers therein. We also outline the current challenges in the EV field amidst the tremendous efforts to propel the clinical utility of EVs for human diseases. The lack of published literature on the functional roles of other EV-ncRNA subtypes may in turn provide new avenues for future research to exploit their feasibility as novel diagnostic and therapeutic targets in human diseases.


Subject(s)
Extracellular Vesicles/physiology , Lung Injury/metabolism , Pneumonia, Bacterial/metabolism , Pneumonia, Viral/metabolism , RNA, Untranslated/physiology , Animals , Biomarkers/metabolism , Humans , Lung/metabolism , Lung/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...