Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
Malays J Pathol ; 46(1): 51-62, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38682844

ABSTRACT

Small animal models play an important role in investigating and revealing the molecular determinants and mechanisms underlying neuro-virulence of enterovirus A71 (EV-A71). In our previous study, we successfully developed two mouse cell-line replication competent EV-A71 strains (EV71:TLLm and EV71:TLLmv) which were capable of inducing neuro-invasion in BALB/c mice. The more virulent EV71:TLLmv exhibited ability to induce acute encephalomyelitis accompanied by neurogenic pulmonary oedema. EV71:TLLcho virus strain was generated from EV71:TLLm by a series of passages in CHO-K1 cells. EV71:TLLcho demonstrated a broader range of infectivity across various mammalian cell lines and exhibited complete cytopathic effects (CPE) within 48 hours post-inoculation in comparison to EV71:TLLm or EV71:TLLmv. EV71:TLLcho consistently yielded higher levels of viral replication at all time points examined. In comparison to EV71:TLLm, EV71:TLLcho consistently induced more severe disease and increased mortality in one-week old BALB/c mice. However, unlike mice challenged with EV71:TLLmv, none of the mice challenged with EV71:TLLcho progressed to severe acute encephalomyelitis and developed neurogenic pulmonary oedema.


Subject(s)
Disease Models, Animal , Enterovirus A, Human , Enterovirus Infections , Mice, Inbred BALB C , Pulmonary Edema , Animals , Pulmonary Edema/virology , Pulmonary Edema/pathology , Enterovirus Infections/complications , Enterovirus Infections/virology , Mice , Virus Replication , Humans
2.
Trends Immunol ; 41(10): 856-859, 2020 10.
Article in English | MEDLINE | ID: mdl-32863134

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mainly affects the lungs. Sarcoidosis is an autoinflammatory disease characterized by the diffusion of granulomas in the lungs and other organs. Here, we discuss how the two diseases might involve some common mechanistic cellular pathways around the regulation of autophagy.


Subject(s)
Autophagy/drug effects , Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Pulmonary Edema/drug therapy , Sarcoidosis/drug therapy , Severe Acute Respiratory Syndrome/drug therapy , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Autophagy/genetics , Azithromycin/therapeutic use , Betacoronavirus/growth & development , COVID-19 , Chloroquine/therapeutic use , Coronavirus Infections/epidemiology , Coronavirus Infections/genetics , Coronavirus Infections/virology , Host-Pathogen Interactions/drug effects , Humans , Isoniazid/therapeutic use , Lung/drug effects , Lung/pathology , Lung/virology , Pandemics , Pneumonia, Viral/epidemiology , Pneumonia, Viral/genetics , Pneumonia, Viral/virology , Pulmonary Edema/epidemiology , Pulmonary Edema/genetics , Pulmonary Edema/virology , Rifampin/therapeutic use , SARS-CoV-2 , Sarcoidosis/epidemiology , Sarcoidosis/genetics , Sarcoidosis/virology , Severe Acute Respiratory Syndrome/epidemiology , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/virology , Severity of Illness Index
3.
Arch Virol ; 167(12): 2483-2501, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36171507

ABSTRACT

Enterovirus A71 (EV-A71) is one of the major pathogens responsible for hand, foot, and mouth disease (HFMD). Many HFMD outbreaks have been reported throughout the world in the past decades. Compared with other viruses, EV-A71 infection is more frequently associated with severe neurological complications and even death in children. EV-A71 can also infect adults and cause severe complications and death, although such cases are very uncommon. Although fatal cases of EV-A71 infection have been reported, the underlying mechanisms of EV-A71 infection, especially the mode of viral spread into the central nervous system (CNS) and mechanisms of pulmonary edema, which is considered to be the direct cause of death, have not yet been fully clarified, and more studies are needed. Here, we first summarize the pathological findings in various systems of patients with fatal EV-A71 infections, focussing in detail on gross changes, histopathological examination, tissue distribution of viral antigens and nucleic acids, systemic inflammatory cell infiltration, and tissue distribution of viral receptors and their co-localization with viral antigens. We then present our conclusions about viral dissemination, neuropathogenesis, and the mechanism of pulmonary edema in EV-A71 infection, based on pathological findings.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Child , Humans , Antigens, Viral/metabolism , Enterovirus/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/complications , Enterovirus Infections/pathology , Pulmonary Edema/virology
4.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34576058

ABSTRACT

Sporadic occurrences and outbreaks of hand, foot, and mouth disease (HFMD) caused by Coxsackievirus A2 (CVA2) have frequently reported worldwide recently, which pose a great challenge to public health. Epidemiological studies have suggested that the main cause of death in critical patients is pulmonary edema. However, the pathogenesis of this underlying comorbidity remains unclear. In this study, we utilized the 5-day-old BALB/c mouse model of lethal CVA2 infection to evaluate lung damage. We found that the permeability of lung microvascular was significantly increased after CVA2 infection. We also observed the direct infection and apoptosis of lung endothelial cells as well as the destruction of tight junctions between endothelial cells. CVA2 infection led to the degradation of tight junction proteins (e.g., ZO-1, claudin-5, and occludin). The gene transcription levels of von Willebrand factor (vWF), endothelin (ET), thrombomodulin (THBD), granular membrane protein 140 (GMP140), and intercellular cell adhesion molecule-1 (ICAM-1) related to endothelial dysfunction were all significantly increased. Additionally, CVA2 infection induced the increased expression of inflammatory cytokines (IL-6, IL-1ß, and MCP-1) and the activation of p38 mitogen-activated protein kinase (MAPK). In conclusion, the disruption of the endothelial barrier contributes to acute lung injury induced by CVA2 infection; targeting p38-MAPK signaling may provide a therapeutic approach for pulmonary edema in critical infections of HFMD.


Subject(s)
Acute Lung Injury/genetics , Coxsackievirus Infections/genetics , Hand, Foot and Mouth Disease/genetics , Pulmonary Edema/genetics , Acute Lung Injury/complications , Acute Lung Injury/pathology , Acute Lung Injury/virology , Animals , Apoptosis/genetics , Claudin-5/genetics , Coxsackievirus Infections/complications , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Cytokines/genetics , Disease Models, Animal , Endothelial Cells/pathology , Endothelial Cells/virology , Hand, Foot and Mouth Disease/complications , Hand, Foot and Mouth Disease/pathology , Hand, Foot and Mouth Disease/virology , Humans , Mice , Occludin/genetics , Pulmonary Edema/complications , Pulmonary Edema/pathology , Pulmonary Edema/virology , Tight Junctions/genetics , Tight Junctions/pathology , Zonula Occludens-1 Protein/genetics , p38 Mitogen-Activated Protein Kinases/genetics
5.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1239-L1243, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32401673

ABSTRACT

Lethality of coronavirus disease (COVID-19) during the 2020 pandemic, currently still in the exponentially accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We argue for inhibition of the transient receptor potential vanilloid 4 (TRPV4) calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in COVID-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in COVID-19 patients with respiratory malfunction and at risk for lung edema. Perplexingly, among the currently pursued therapeutic strategies against COVID-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce COVID-19 lethality but will also preempt a distressing healthcare scenario with insufficient capacity to provide ventilator-assisted respiration.


Subject(s)
Betacoronavirus , Coronavirus Infections , Lung/virology , Pandemics , Pneumonia, Viral , Pulmonary Edema/prevention & control , TRPV Cation Channels/antagonists & inhibitors , COVID-19 , Calcium/metabolism , Coronavirus Infections/virology , Humans , Lung/metabolism , Pneumonia, Viral/virology , Pulmonary Edema/virology , Respiration, Artificial , SARS-CoV-2
6.
Am J Forensic Med Pathol ; 41(1): 48-51, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31977345

ABSTRACT

Chikungunya is a mosquito-transmitted viral illness with clinical hallmarks of rash, fever, arthralgia, and myalgia. It is rarely fatal, although vulnerable populations, to include elderly, children, and those with multiple comorbid illnesses, are more susceptible to severe infection and death. There have been multiple areas of the world with periodic chikungunya epidemics. With increased immigration, foreign travel, epidemics, and global spread of the virus, it is prudent to consider chikungunya as a diagnosis both clinically and postmortem when a patient presents with rash, fevers, and arthralgia. We present a case of a patient with recent foreign travel, a rash, fever, and arthralgia with mosquito bites who succumbed to chikungunya viral infection with pneumonia. His diagnosis was established postmortem. A review of the literature is included in this report. This case stresses the delayed time to diagnose chikungunya with serologic testing and the importance of using reverse transcriptase-polymerase chain reaction to aid in rapid and accurate diagnosis and management.


Subject(s)
Chikungunya Fever/diagnosis , Travel-Related Illness , Arthralgia/virology , Chikungunya virus/genetics , El Salvador , Endemic Diseases , Exanthema/pathology , Exanthema/virology , Forensic Pathology , Humans , Los Angeles , Lung/pathology , Male , Middle Aged , Pneumonia, Viral/etiology , Polymerase Chain Reaction , Pulmonary Edema/pathology , Pulmonary Edema/virology
7.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29084896

ABSTRACT

Staphylococcus aureus (S. aureus) infections are among the most common and severe infections, garnering notoriety in an era of increasing resistance to antibiotics. It is therefore important to define molecular mechanisms by which this pathogen attacks host cells. Here, we demonstrate that alpha-toxin, one of the major toxins of S. aureus, induces activation of acid sphingomyelinase and concomitant release of ceramide in endothelial cells treated with the toxin. Activation of acid sphingomyelinase by alpha-toxin is mediated via ADAM10. Infection experiments employing alpha-toxin-deficient S. aureus and the corresponding wild-type strain reveal that activation of acid sphingomyelinase in endothelial cells requires alpha-toxin expression by the pathogen. Activation of acid sphingomyelinase is linked to degradation of tight junctions in endothelial cells in vitro, which is blocked by pharmacological inhibition of acid sphingomyelinase. Most importantly, alpha-toxin induces severe degradation of tight junctions in the lung and causes lung edema in vivo, which is prevented by genetic deficiency of acid sphingomyelinase. These data indicate a novel and important role of the acid sphingomyelinase/ceramide system for the endothelial response to toxins and provide a molecular link between alpha-toxin and the degradation of tight junctions. The data also suggest that inhibition of acid sphingomyelinase may provide a novel treatment option to prevent lung edema caused by S. aureus alpha-toxin.


Subject(s)
Bacterial Toxins/metabolism , Ceramides/metabolism , Endothelial Cells/metabolism , Hemolysin Proteins/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Staphylococcus aureus/metabolism , Tight Junctions/metabolism , ADAM10 Protein/metabolism , Animals , Cells, Cultured , Endothelial Cells/virology , Lung/metabolism , Lung/virology , Mice , Mice, Inbred C57BL , Pulmonary Edema/metabolism , Pulmonary Edema/virology , Staphylococcal Infections/metabolism , Staphylococcal Infections/virology , Tight Junctions/virology
8.
Lab Invest ; 98(8): 1039-1051, 2018 08.
Article in English | MEDLINE | ID: mdl-29765110

ABSTRACT

Enterovirus (EV) 71 infection has been widely acknowledged as the leading cause of severe hand, foot and mouth disease (HFMD), which may rapidly lead to fatal pulmonary edema. In this study, we established a mouse model for EV71 infection exhibiting high incidence of severe symptoms with pulmonary edema. Mast cells (MCs) accumulation, activation and allergic inflammation were found in the brains, lungs and skeletal muscle of mice after EV71 infection, especially in the lungs of mice. Levels of histamine, platelet-activating factor (PAF), interleukin (IL)-4, IL-5, IL-13, tumor necrosis factor-α (TNF-α), nitric oxide (NO), endocrine gland-derived vascular endothelial growth factor (EG-VEGF) and noradrenaline (NA) were increased in EV71-infected lungs. In addition, EV71 infection reduced the number of pulmonary T cells, dendritic cells (DCs) and monocytes, and increased the number of lung eosinophils, Tregs and MCs. MCs number and tryptase expression in target organs or tissues posed a trend towards an increase from control to severe mice. There were positive correlations between MCs number in the brains (r = 0.701, P = 0.003), lungs (r = 0.802, P < 0.0001), skeletal muscles (r = 0.737, P = 0.001) and mean clinical score. Thus, our results suggested that MCs contributed to the pulmonary edema during EV71 infection.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Mast Cells/immunology , Pulmonary Edema/immunology , Animals , Animals, Newborn , Brain/immunology , Brain/metabolism , Brain/virology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Histamine/immunology , Histamine/metabolism , Humans , Lung/immunology , Lung/metabolism , Lung/virology , Mast Cells/metabolism , Mast Cells/virology , Mice, Inbred BALB C , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Pulmonary Edema/metabolism , Pulmonary Edema/virology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
9.
Virol J ; 14(1): 243, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29282065

ABSTRACT

BACKGROUND: Enterovirus (EV) infection has been a serious health issue in Asia-Pacific region. It has been indicated that the occurrence of fatal hand foot and mouth disease (HFMD) cases following EV71 infection is mainly attributed to pulmonary edema. However, the development of pulmonary disorders after EV71 infection remains largely unknown. To establish an EV71-infected animal model and further explore the underlying association of central nervous system (CNS) invasion with pulmonary edema, we isolated a clinical source EV71 strain (ZZ1350) from a severe case in Henan Province. METHODS: We evaluated the cytotoxicity of ZZ1350 strain and the susceptibility in 3-day-old BALB/c mice with intraperitoneal, intracerebral and intramuscular inoculation. Various histopathological and immunohistochemical techniques were applied to determine the target organs or tissue damage after infection. Correlation analysis was used to identify the relationship between CNS injury and pulmonary disorders. RESULTS: Our experimental results suggested that ZZ1350 (C4 subtype) had high cytotoxicity against African green monkey kidney (Vero) cells and human rhabdomyosarcoma (RD) cells and neonatal BALB/c mice were highly susceptible to the infection with ZZ1350 through three different inoculation routes (2 × 106 pfu/mouse) exhibiting severe neurological and respiratory symptoms that were similar to clinical observation. Viral replication was found in brain, spinal cord, skeletal muscle, lung, spleen, liver, heart of infected mice and these sections also showed histopathological changes. We found that brain histology score was positive correlated with lung histology score in total experimental mice and mice under the three inoculation routes (P < 0.05). At the same time, there were positive correlations between spinal cord score and lung score in total experimental mice and mice with intracerebral inoculation (P < 0.05). CONCLUSIONS: ZZ1350 strain is effective to establish animal model of EV71 infection with severe neurological and respiratory symptoms. The development of pulmonary disorders after EV71 infection is associated with severity of CNS damage.


Subject(s)
Brain Injuries/virology , Enterovirus A, Human/pathogenicity , Enterovirus Infections/complications , Lung/virology , Pulmonary Edema/virology , Spinal Cord Injuries/virology , Animals , Brain Injuries/pathology , Cell Line, Tumor , Cell Survival , China , Chlorocebus aethiops , Disease Models, Animal , Disease Susceptibility , Enterovirus A, Human/isolation & purification , Enterovirus Infections/pathology , Humans , Lung/pathology , Mice , Spinal Cord Injuries/pathology , Vero Cells
10.
Article in English | MEDLINE | ID: mdl-26521518

ABSTRACT

We studied risk factors associated with severe hand, foot and mouth disease (HFMD) caused by enteroviruses among patients aged less than 15 years admitted to King Narai Hospital, Lopburi, Thailand during 2011-2013. Cases were divided into either mild or severe. Severe cases were those with encephalitis, meningitis, myocarditis, pneumonia, pulmonary edema or respiratory failure. Risk factors for severe infection were evaluated using univariate and multivariate logistic regression analysis. One hundred eighteen patients met the case definition of HFMD. Of these, 95 (80.5%) were classified as mild cases, and 23 (19.5%) as severe cases; there were 5 deaths (4.2%). Of the 23 severe cases, 9 were infected with coxsackievirus A16 (CA16), 8 with enterovirus 71 (EV71) and 4 with both EV71 and CA16. The most common presentations among the severe caseswere: seizures (74%), pneumonia (39%), encephalitis (39%), and meningitis (13%). The clinical manifestations significantly related to severe HFMD on univariate analysis were highest body temperature 39.00C, duration of fever 23 days, absence of skin lesions, diarrhea, dyspnea, seizures and hyperglycemia. The clinical manifestations significantly related to severe HFMD on both univariate and multivariate analyses were age less than 1 year, absence of oral lesions and drowsiness/lethargy. Clinicians should be aware of these factors. Early recognition of severe cases is important to increase the rates of successful outcomes and reduce mortality.


Subject(s)
Enterovirus A, Human , Hand, Foot and Mouth Disease/complications , Adolescent , Aged , Diarrhea/etiology , Diarrhea/virology , Encephalitis, Viral/etiology , Encephalitis, Viral/virology , Enterovirus , Female , Fever/epidemiology , Fever/virology , Hand, Foot and Mouth Disease/virology , Humans , Logistic Models , Male , Meningitis, Viral/etiology , Meningitis, Viral/virology , Multivariate Analysis , Myocarditis/etiology , Myocarditis/virology , Pneumonia, Viral/etiology , Pneumonia, Viral/virology , Pulmonary Edema/etiology , Pulmonary Edema/virology , Respiratory Insufficiency/etiology , Respiratory Insufficiency/virology , Retrospective Studies , Risk Factors , Seizures/etiology , Seizures/virology , Severity of Illness Index , Thailand
11.
Crit Care Med ; 41(7): 1754-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685637

ABSTRACT

OBJECTIVE: Enterovirus 71-induced brainstem encephalitis with pulmonary edema and/or neurogenic shock (stage 3B) is associated with rapid mortality in children. In a small pilot study, we found that milrinone reduced early mortality compared with historical controls. This prospective, randomized control trial was designed to provide more definitive evidence of the ability of milrinone to reduce the 1-week mortality of stage 3B enterovirus 71 infections. DESIGN: Prospective, unicenter, open-label, randomized, controlled study. SETTING: Inpatient ward of a large tertiary teaching hospital in Ho Chi Minh City, Vietnam. PATIENTS: Children (≤ 18 yr old) admitted with proven enterovirus 71-induced pulmonary edema and/or neurogenic shock. INTERVENTIONS: Patients were randomly assigned to receive intravenous milrinone (0.5 µg/kg/min) (n = 22) or conventional management (n = 19). Both groups received dopamine or dobutamine and intravenous immunoglobulin. MEASUREMENTS AND MAIN RESULTS: The primary endpoint was 1-week mortality. The secondary endpoints included length of ventilator dependence and hospital stay and adverse events. The median age was 2 years with a predominance of boys in both groups. The 1-week mortality was significantly lower, 18.2% (4/22) in the milrinone compared with 57.9% (11/19) in the conventional management group (relative risk = 0.314 [95% CI, 0.12-0.83], p = 0.01). The median duration of ventilator-free days was longer in the milrinone treatment group (p = 0.01). There was no apparent neurologic sequela in the survivors in either group, and no drug-related adverse events were documented. CONCLUSIONS: Milrinone significantly reduced the 1-week mortality of enterovirus 71-induced pulmonary edema and/or neurogenic shock without adverse effects. Further studies are needed to determine whether milrinone might be useful to prevent progression of earlier stages of brainstem encephalitis.


Subject(s)
Cardiotonic Agents/therapeutic use , Enterovirus Infections/virology , Milrinone/therapeutic use , Pulmonary Edema/drug therapy , Shock/drug therapy , Cardiotonic Agents/administration & dosage , Child, Preschool , Dobutamine/therapeutic use , Dopamine/therapeutic use , Enterovirus Infections/mortality , Female , Humans , Immunoglobulins/therapeutic use , Infant , Infusions, Intravenous , Length of Stay , Male , Milrinone/administration & dosage , Prospective Studies , Pulmonary Edema/mortality , Pulmonary Edema/virology , Respiration, Artificial , Shock/mortality , Shock/virology , Vietnam
12.
J Infect Dis ; 205(2): 252-61, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22147799

ABSTRACT

For the first time, obesity appeared as a risk factor for developing severe 2009 pandemic influenza infection. Given the increase in obesity, there is a need to understand the mechanisms underlying poor outcomes in this population. In these studies, we examined the severity of pandemic influenza virus in obese mice and evaluated antiviral effectiveness. We found that genetically and diet-induced obese mice challenged with either 2009 influenza A virus subtype H1N1 or 1968 subtype H3N2 strains were more likely to have increased mortality and lung pathology associated with impaired wound repair and subsequent pulmonary edema. Antiviral treatment with oseltamivir enhanced survival of obese mice. Overall, these studies demonstrate that impaired wound lung repair in the lungs of obese animals may result in severe influenza virus infection. Alternative approaches to prevention and control of influenza may be needed in the setting of obesity.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Lung/pathology , Obesity/complications , Orthomyxoviridae Infections/drug therapy , Oseltamivir/therapeutic use , Albumins/analysis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Kaplan-Meier Estimate , Ki-67 Antigen/metabolism , Leukocyte Count , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Monocytes , Neutrophil Infiltration , Obesity/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Pulmonary Edema/virology , Severity of Illness Index , Viral Load , Wound Healing
13.
Am J Respir Cell Mol Biol ; 47(4): 543-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22679275

ABSTRACT

Acute respiratory disease is associated with significant morbidity and mortality in influenza. Because antiviral drugs are only effective early in infection, new agents are needed to treat nonvaccinated patients presenting with late-stage disease, particularly those who develop acute respiratory distress syndrome. We found previously that the de novo pyrimidine synthesis inhibitor A77-1726 reversed the influenza-induced impairment of alveolar fluid clearance. Patients with acute respiratory distress syndrome and intact alveolar fluid clearance demonstrate lower mortality than those with compromised fluid clearance. We therefore investigated the effects of treatment with nebulized A77-1726 (67.5 mg/kg) on indices of cardiopulmonary function relevant to the diagnosis of acute respiratory distress syndrome. BALB/cAnNCr mice (8-12 wk old) were inoculated intranasally with 10,000 plaque-forming units/mouse influenza A/WSN/33 (H1N1). Pulse oximetry was performed daily. Alveolar fluid clearance, lung water, and lung mechanics were measured at 2 and 6 days after inoculation in live, ventilated mice by BSA instillation, magnetic resonance imaging, and forced-oscillation techniques, respectively. A77-1726 treatment at 1 day after inoculation delayed mortality. Treatment on Days 1 or 5 reduced viral replication on Day 6, and improved alveolar fluid clearance, peripheral oxygenation, and cardiac function. Nebulized A77-1726 also reversed influenza-induced increases in lung water content and volume, improved pulmonary mechanics, reduced bronchoalveolar lavage fluid ATP and neutrophil content, and increased IL-6 concentrations. The ability of A77-1726 to improve cardiopulmonary function in influenza-infected mice and to reduce the severity of ongoing acute respiratory distress syndrome late in infection suggests that pyrimidine synthesis inhibitors are promising therapeutic candidates for the management of severe influenza.


Subject(s)
Aniline Compounds/administration & dosage , Antiviral Agents/administration & dosage , Hydroxybutyrates/administration & dosage , Influenza A Virus, H1N1 Subtype/physiology , Orthomyxoviridae Infections/drug therapy , Respiratory Distress Syndrome/prevention & control , Administration, Inhalation , Airway Resistance/drug effects , Aniline Compounds/pharmacology , Animals , Antiviral Agents/pharmacology , Bronchoalveolar Lavage Fluid , Carotid Arteries/physiopathology , Crotonates , Cytokines/metabolism , Drug Evaluation, Preclinical , Heart Rate/drug effects , Hydroxybutyrates/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Lung/drug effects , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Nitriles , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Oxygen/blood , Pulmonary Edema/immunology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Pulmonary Edema/virology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/virology , Respiratory Rate/drug effects , Toluidines , Virus Replication/drug effects
14.
Vet Pathol ; 49(2): 386-92, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21653204

ABSTRACT

Six cases of fatal myocarditis associated with encephalomyocarditis virus occurred over a 14-month period in a group of outdoor-housed juvenile rhesus macaques. All animals were younger than 3 years of age and died or were euthanized following acute onset of dyspnea or pulmonary effusion (3 of 6) or were found dead without premonitory signs (3 of 6). Gross findings included pulmonary congestion (6 of 6), variable degrees of pleural effusion (4 of 6), multifocal pale tan foci throughout the myocardium (3 of 6), hepatomegaly and hepatic congestion (3 of 6), and pericardial effusion (1 of 6). Histologically, affected myocardium was infiltrated multifocally by lymphoplasmacytic and histiocytic inflammation admixed with necrotic and degenerate myofibers and infrequent mineralization (6 of 6). Pulmonary edema was present in all animals. Encephalomyocarditis virus was confirmed in 6 of 6 hearts by immunohistochemistry, and virus was isolated from one case by polymerase chain reaction. Sequencing of virus isolated from 1 affected animal indicated infection with a novel encephalomyocarditis virus. Encephalomyocarditis virus should be considered as a differential etiology in outbreaks of myocarditis and pulmonary edema in juvenile primates.


Subject(s)
Cardiovirus Infections/veterinary , Encephalomyocarditis virus/isolation & purification , Macaca mulatta , Monkey Diseases/virology , Myocarditis/veterinary , Animals , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , Chlorocebus aethiops , DNA, Complementary/chemistry , DNA, Complementary/genetics , Disease Outbreaks/veterinary , Encephalomyocarditis virus/genetics , Female , Humans , Male , Microscopy, Electron, Transmission/veterinary , Monkey Diseases/pathology , Myocarditis/pathology , Myocarditis/virology , Myocardium/pathology , Myocardium/ultrastructure , Pulmonary Edema/pathology , Pulmonary Edema/veterinary , Pulmonary Edema/virology , RNA, Viral/genetics , Sequence Analysis, DNA , Vero Cells
15.
Cells ; 10(11)2021 11 07.
Article in English | MEDLINE | ID: mdl-34831290

ABSTRACT

The bronchial vascular endothelial network plays important roles in pulmonary pathology during respiratory viral infections, including respiratory syncytial virus (RSV), influenza A(H1N1) and importantly SARS-Cov-2. All of these infections can be severe and even lethal in patients with underlying risk factors.A major obstacle in disease prevention is the lack of appropriate efficacious vaccine(s) due to continuous changes in the encoding capacity of the viral genome, exuberant responsiveness of the host immune system and lack of effective antiviral drugs. Current management of these severe respiratory viral infections is limited to supportive clinical care. The primary cause of morbidity and mortality is respiratory failure, partially due to endothelial pulmonary complications, including edema. The latter is induced by the loss of alveolar epithelium integrity and by pathological changes in the endothelial vascular network that regulates blood flow, blood fluidity, exchange of fluids, electrolytes, various macromolecules and responses to signals triggered by oxygenation, and controls trafficking of leukocyte immune cells. This overview outlines the latest understanding of the implications of pulmonary vascular endothelium involvement in respiratory distress syndrome secondary to viral infections. In addition, the roles of infection-induced cytokines, growth factors, and epigenetic reprogramming in endothelial permeability, as well as emerging treatment options to decrease disease burden, are discussed.


Subject(s)
Endothelial Cells/pathology , Oxidative Stress , Respiratory Distress Syndrome/pathology , Virus Diseases/pathology , Epigenesis, Genetic , Humans , Influenza A Virus, H1N1 Subtype/physiology , Pulmonary Edema/genetics , Pulmonary Edema/pathology , Pulmonary Edema/virology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/virology , Respiratory Syncytial Viruses/pathogenicity , SARS-CoV-2/pathogenicity , Virus Diseases/genetics , Virus Diseases/virology
16.
Nephrol Ther ; 17(4): 226-232, 2021 Aug.
Article in French | MEDLINE | ID: mdl-33563573

ABSTRACT

BACKGROUND: The effect of COVID-19 pandemic on end stage renal disease patient who should initiated dialysis are limited in Sub-Saharan Africa is unknown. We sought to describe the epidemiologic and clinical profile of newly admitted patient in chronic haemodialysis during the COVID-19 pandemic in Cameroon and evaluate their survival between 90days of dialysis initiation. MATERIAL AND METHOD: We conducted a cohort study of 6months from April to October 2020. End stage renal disease patients newly admitted in the haemodialysis facility of the General Hospital of Douala were included. Patients with confirmed or suspected COVID-19 were identified. Socio-demographic, clinical and biological data at dialysis initiation as well as mortality between the 90days of dialysis initiation were registered. RESULTS: A total of 57 incident patients were recorded from April to October 2020 with a monthly mean of 9.5 patients. The mean age was 46.95±13.12years. Twenty-four COVID-19 were identified with a frequency of 49% among emergency admission. Pulmonary œdema (79.2% vs. 42.4%; P=0.006) and uremic encephalopathy (83.4% vs. 53.6%; P=0.022) were more common in COVID-19. The overall survival at 90days was 48% with a tendency to poor survival among COVID-19 and patients with low socioeconomic level. In Cox regression, low socioeconomic level increase the risk of instant death by 3.08. CONCLUSION: SARS-CoV2 seem to increase nephrology emergency and poor survival in haemodialysis at 90days.


Subject(s)
COVID-19/mortality , Hospitalization , Kidney Failure, Chronic/mortality , Renal Dialysis , Brain Diseases/epidemiology , Brain Diseases/etiology , Cameroon/epidemiology , Female , Hospitals, General , Humans , Incidence , Kidney Failure, Chronic/therapy , Male , Middle Aged , Pandemics , Prospective Studies , Pulmonary Edema/epidemiology , Pulmonary Edema/virology , Social Class , Uremia/epidemiology , Uremia/virology
17.
Nat Struct Mol Biol ; 28(9): 755-761, 2021 09.
Article in English | MEDLINE | ID: mdl-34518695

ABSTRACT

Bradykinin and kallidin are endogenous kinin peptide hormones that belong to the kallikrein-kinin system and are essential to the regulation of blood pressure, inflammation, coagulation and pain control. Des-Arg10-kallidin, the carboxy-terminal des-Arg metabolite of kallidin, and bradykinin selectively activate two G protein-coupled receptors, type 1 and type 2 bradykinin receptors (B1R and B2R), respectively. The hyperactivation of bradykinin receptors, termed 'bradykinin storm', is associated with pulmonary edema in COVID-19 patients, suggesting that bradykinin receptors are important targets for COVID-19 intervention. Here we report two G protein-coupled complex structures of human B1R and B2R bound to des-Arg10-kallidin and bradykinin, respectively. Combined with functional analysis, our structures reveal the mechanism of ligand selectivity and specific activation of the bradykinin receptor. These findings also provide a framework for guiding drug design targeting bradykinin receptors for the treatment of inflammation, cardiovascular disorders and COVID-19.


Subject(s)
Bradykinin/metabolism , COVID-19/pathology , Kallidin/metabolism , Receptors, Bradykinin/metabolism , Cryoelectron Microscopy , Enzyme Activation/physiology , Humans , Protein Structure, Tertiary , Pulmonary Edema/pathology , Pulmonary Edema/virology , SARS-CoV-2
18.
Sci Rep ; 11(1): 11524, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075155

ABSTRACT

Nearly 5% of patients suffering from COVID-19 develop acute respiratory distress syndrome (ARDS). Extravascular lung water index (EVLWI) is a marker of pulmonary oedema which is associated with mortality in ARDS. In this study, we evaluate whether EVLWI is higher in patients with COVID-19 associated ARDS as compared to COVID-19 negative, ventilated patients with ARDS and whether EVLWI has the potential to monitor disease progression. EVLWI and cardiac function were monitored by transpulmonary thermodilution in 25 patients with COVID-19 ARDS subsequent to intubation and compared to a control group of 49 non-COVID-19 ARDS patients. At intubation, EVLWI was noticeably elevated and significantly higher in COVID-19 patients than in the control group (17 (11-38) vs. 11 (6-26) mL/kg; p < 0.001). High pulmonary vascular permeability index values (2.9 (1.0-5.2) versus 1.9 (1.0-5.2); p = 0.003) suggested a non-cardiogenic pulmonary oedema. By contrast, the cardiac parameters SVI, GEF and GEDVI were comparable in both cohorts. High EVLWI values were associated with viral persistence, prolonged intensive care treatment and in-hospital mortality (23.2 ± 6.7% vs. 30.3 ± 6.0%, p = 0.025). Also, EVLWI showed a significant between-subjects (r = - 0.60; p = 0.001) and within-subjects correlation (r = - 0.27; p = 0.028) to Horowitz index. Compared to non COVID-19 ARDS, COVID-19 results in markedly elevated EVLWI-values in patients with ARDS. High EVLWI reflects a non-cardiogenic pulmonary oedema in COVID-19 ARDS and could serve as parameter to monitor ARDS progression on ICU.


Subject(s)
COVID-19/complications , Extravascular Lung Water/immunology , Pulmonary Edema/mortality , Respiratory Distress Syndrome/mortality , Adult , Aged , Aged, 80 and over , COVID-19/diagnosis , COVID-19/immunology , COVID-19/mortality , Capillary Permeability , Disease Progression , Extravascular Lung Water/virology , Female , Hospital Mortality , Humans , Intensive Care Units/statistics & numerical data , Length of Stay/statistics & numerical data , Lung/blood supply , Lung/physiopathology , Male , Middle Aged , Monitoring, Physiologic/methods , Monitoring, Physiologic/statistics & numerical data , Prognosis , Pulmonary Edema/diagnosis , Pulmonary Edema/immunology , Pulmonary Edema/virology , Respiration, Artificial , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/therapy , Risk Assessment/methods , SARS-CoV-2/isolation & purification , Severity of Illness Index , Thermodilution/methods , Thermodilution/statistics & numerical data , Young Adult
19.
SLAS Discov ; 26(9): 1079-1090, 2021 10.
Article in English | MEDLINE | ID: mdl-34269109

ABSTRACT

The recent renascence of phenotypic drug discovery (PDD) is catalyzed by its ability to identify first-in-class drugs and deliver results when the exact molecular mechanism is partially obscure. Acute respiratory distress syndrome (ARDS) is a severe, life-threatening condition with a high mortality rate that has increased in frequency due to the COVID-19 pandemic. Despite decades of laboratory and clinical study, no efficient pharmacological therapy for ARDS has been found. An increase in endothelial permeability is the primary event in ARDS onset, causing the development of pulmonary edema that leads to respiratory failure. Currently, the detailed molecular mechanisms regulating endothelial permeability are poorly understood. Therefore, the use of the PDD approach in the search for efficient ARDS treatment can be more productive than classic target-based drug discovery (TDD), but its use requires a new cell-based assay compatible with high-throughput (HTS) and high-content (HCS) screening. Here we report the development of a new plate-based image cytometry method to measure endothelial barrier function. The incorporation of image cytometry in combination with digital image analysis substantially decreases assay variability and increases the signal window. This new method simultaneously allows for rapid measurement of cell monolayer permeability and cytological analysis. The time-course of permeability increase in human pulmonary artery endothelial cells (HPAECs) in response to the thrombin and tumor necrosis factor α treatment correlates with previously published data obtained by transendothelial resistance (TER) measurements. Furthermore, the proposed image cytometry method can be easily adapted for HTS/HCS applications.


Subject(s)
COVID-19/diagnostic imaging , High-Throughput Screening Assays/methods , Image Cytometry/methods , Respiratory Distress Syndrome/diagnostic imaging , COVID-19/diagnosis , COVID-19/virology , Cell Membrane Permeability/genetics , Drug Discovery , Endothelial Cells/ultrastructure , Endothelial Cells/virology , Humans , Image Processing, Computer-Assisted , Pandemics/prevention & control , Phenotype , Pulmonary Artery/diagnostic imaging , Pulmonary Artery/pathology , Pulmonary Artery/virology , Pulmonary Edema/diagnosis , Pulmonary Edema/diagnostic imaging , Pulmonary Edema/virology , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/virology , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/diagnostic imaging , Respiratory Insufficiency/virology , SARS-CoV-2/pathogenicity , Thrombin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
20.
Vascul Pharmacol ; 137: 106829, 2021 04.
Article in English | MEDLINE | ID: mdl-33422689

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is an important player of the renin-angiotensin-aldosterone system (RAAS) in regulating the conversion of angiotensin II into angiotensin (1-7). While expressed on the surface of human cells, such as lung, heart, kidney, neurons, and endothelial cells (EC), ACE2 is the entry receptor for SARS-CoV-2. Here, we would like to highlight that ACE2 is predominant on the EC membrane. Many of coronavirus disease 2019 (COVID-19) symptoms have been associated with the large recruitment of immune cells, directly affecting EC. Additionally, cytokines, hypoxia, and complement activation can trigger the activation of EC leading to the coagulation cascade. The EC dysfunction plus the inflammation due to SARS-CoV-2 infection may lead to abnormal coagulation, actively participating in thrombo-inflammatory processes resulting in vasculopathy and indicating poor prognosis in patients with COVID-19. Considering the intrinsic relationship between EC and the pathophysiology of SARS-CoV-2, EC-associated therapies such as anticoagulants, fibrinolytic drugs, immunomodulators, and molecular therapies have been proposed. In this review, we will discuss the role of EC in the lung inflammation and edema, in the disseminate coagulation process, ACE2 positive cancer patients, and current and future EC-associated therapies to treat COVID-19.


Subject(s)
COVID-19/virology , Cardiovascular Diseases/virology , Endothelium, Vascular/virology , Inflammation/virology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/metabolism , Animals , Blood Coagulation , COVID-19/complications , COVID-19/therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Host-Pathogen Interactions , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Inflammation Mediators/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/virology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL