Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
9.
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
10.
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
11.
Front Immunol ; 11: 574862, 2020.
Article in English | MEDLINE | ID: mdl-33042157

ABSTRACT

It is currently believed that innate immunity is unable to prevent the spread of SARS-CoV-2 from the upper airways to the alveoli of high-risk groups of patients. SARS-CoV-2 replication in ACE-2-expressing pneumocytes can drive the diffuse alveolar injury through the cytokine storm and immunothrombosis by upregulating the transcription of chemokine/cytokines, unlike several other respiratory viruses. Here we report histopathology data obtained in post-mortem lung biopsies of COVID-19, showing the increased density of perivascular and septal mast cells (MCs) and IL-4-expressing cells (n = 6), in contrast to the numbers found in pandemic H1N1-induced pneumonia (n = 10) or Control specimens (n = 10). Noteworthy, COVID-19 lung biopsies showed a higher density of CD117+ cells, suggesting that c-kit positive MCs progenitors were recruited earlier to the alveolar septa. These findings suggest that MC proliferation/differentiation in the alveolar septa might be harnessed by the shift toward IL-4 expression in the inflamed alveolar septa. Future studies may clarify whether the fibrin-dependent generation of the hyaline membrane, processes that require the diffusion of procoagulative plasma factors into the alveolar lumen and the endothelial dysfunction, are preceded by MC-driven formation of interstitial edema in the alveolar septa.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Mast Cells/immunology , Pneumonia, Viral/immunology , Pulmonary Alveoli/immunology , Pulmonary Edema/immunology , Thrombosis/immunology , Adult , Aged , Aged, 80 and over , COVID-19 , Coronavirus Infections/pathology , Coronavirus Infections/virology , Female , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/pathology , Influenza, Human/virology , Interleukin-4/immunology , Male , Mast Cells/pathology , Middle Aged , Pandemics , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Proto-Oncogene Proteins c-kit/immunology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Pulmonary Edema/pathology , Pulmonary Edema/virology , SARS-CoV-2 , Thrombosis/pathology , Thrombosis/virology
12.
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
13.
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
14.
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
15.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333319

ABSTRACT

Severe influenza (IAV) infection can develop into bronchopneumonia and edema, leading to acquired respiratory distress syndrome (ARDS) and pathophysiology. Underlying causes for pulmonary edema and aberrant fluid regulation largely remain unknown, particularly regarding the role of viral-mediated mechanisms. Herein, we show that distinct IAV strains reduced the functions of the epithelial sodium channel (ENaC) and the cystic fibrosis transmembrane regulator (CFTR) in murine respiratory and alveolar epithelia in vivo, as assessed by measurements of nasal potential differences and single-cell electrophysiology. Reduced ion channel activity was distinctly limited to virally infected cells in vivo and not bystander uninfected lung epithelium. Multiple lines of evidence indicated ENaC and CFTR dysfunction during the acute infection period; however, only CFTR dysfunction persisted beyond the infection period. ENaC, CFTR, and Na,K-ATPase activities and protein levels were also reduced in virally infected human airway epithelial cells. Reduced ENaC and CFTR led to changes in airway surface liquid morphology of human tracheobronchial cultures and airways of IAV-infected mice. Pharmacologic correction of CFTR function ameliorated IAV-induced physiologic changes. These changes are consistent with mucous stasis and pulmonary edema; furthermore, they indicate that repurposing therapeutic interventions correcting CFTR dysfunction may be efficacious for treatment of IAV lung pathophysiology.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Sodium Channels/metabolism , Influenza A virus/pathogenicity , Influenza, Human/pathology , Respiratory Mucosa/pathology , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Bronchi/metabolism , Bronchi/pathology , Bronchi/virology , Cells, Cultured , Disease Models, Animal , Dogs , Epithelial Cells , Female , Humans , Influenza, Human/complications , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Male , Mice , Primary Cell Culture , Pulmonary Edema/pathology , Pulmonary Edema/virology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Sodium-Potassium-Exchanging ATPase/metabolism , Water-Electrolyte Balance/drug effects
16.
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
17.
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
18.
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
19.
Sci Rep ; 6: 28876, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27357918

ABSTRACT

Enterovirus 71 (EV-A71) is a neurotropic virus that sporadically causes fatal neurologic illness among infected children. Animal models of EV-A71 infection exist, but they do not recapitulate in animals the spectrum of disease and pathology observed in fatal human cases. Specifically, neurogenic pulmonary oedema (NPE)-the main cause of EV-A71 infection-related mortality-is not observed in any of these models. This limits their utility in understanding viral pathogenesis of neurologic infections. We report the development of a mouse model of EV-A71 infection displaying NPE in severely affected animals. We inoculated one-week-old BALB/c mice with an adapted EV-A71 strain and identified clinical signs consistent with observations in human cases and other animal models. We also observed respiratory distress in some mice. At necropsy, we found their lungs to be heavier and incompletely collapsed compared to other mice. Serum levels of catecholamines and histopathology of lung and brain tissues of these mice strongly indicated onset of NPE. The localization of virally-induced brain lesions also suggested a potential pathogenic mechanism for EV-A71-induced NPE. This novel mouse model of virally-induced NPE represents a valuable resource for studying viral mechanisms of neuro-pathogenesis and pre-clinical testing of potential therapeutics and prophylactics against EV-A71-related neurologic complications.


Subject(s)
Enterovirus A, Human/physiology , Enterovirus Infections/pathology , Pulmonary Edema/pathology , Animals , Antibodies, Neutralizing/blood , Brain/metabolism , Brain/pathology , Catecholamines/metabolism , Disease Models, Animal , Enterovirus A, Human/immunology , Enterovirus Infections/metabolism , Enterovirus Infections/mortality , Enterovirus Infections/virology , Humans , Kaplan-Meier Estimate , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Pulmonary Edema/metabolism , Pulmonary Edema/mortality , Pulmonary Edema/virology , Severity of Illness Index
20.
J Ethnopharmacol ; 192: 217-224, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27401293

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Mahuang-Xixin-Fuzi Decoction (MXF) as a famous formula for the treatment of colds, fever, nasal congestion and headache with elder people, has always been widely used in traditional Chinese medicine. The present study is aimed at investigating the treatment effect of MXF on Kidney-Yang deficiency syndrome in mice simultaneously infected with H1N1 virus. MATERIALS AND METHODS: We employed the Kidney-Yang deficiency mouse model to investigate the effect of MXF against influenza A virus (A/FM/1/47, H1N1). Mice were infected with the virus after fifteen days Kidney-Yang deficiency syndrome was established (intraperitoneal injection of estradiol benzoate), while MXF was orally administrated with 1.2-4.7g/kg/d for 6 consecutive days after inoculation. Body weight, rectal temperature, morbidity, and mortality were recorded daily. Histopathologic changes, antioxidant activity of SOD and MDA were detected. Moreover, levels of inflammatory cytokines including IL-6, IL-10, MCP-1, TNF-α were measured in the sera of mice. RESULTS: We found that the extract of MXF at dosages of 2.3-4.7g/kg could effectively diminish mortality rate, ameliorate lung edema and inflammation. Administration of MXF decoction significantly depressed the expression of IL-6, MCP-1 and TNF-α, and markedly increased expression of IL-10 in serum. Simultaneously, the extract was also found to reduce MDA and increase SOD in the lung tissue of mice. CONCLUSION: These data support the notion that the extract of MXF could treat Kidney-Yang deficiency syndrome in mice simultaneously infected with influenza A virus by reducing inflammation and increasing antioxidant activities.


Subject(s)
Antiviral Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Kidney Diseases/drug therapy , Orthomyxoviridae Infections/drug therapy , Yang Deficiency/drug therapy , Administration, Oral , Animals , Antioxidants/pharmacology , Antiviral Agents/administration & dosage , Chemokine CCL2/blood , Disease Models, Animal , Drugs, Chinese Herbal/administration & dosage , Estradiol/analogs & derivatives , Inflammation Mediators/blood , Influenza A Virus, H1N1 Subtype/pathogenicity , Interleukin-6/blood , Kidney Diseases/blood , Kidney Diseases/chemically induced , Lung/drug effects , Lung/metabolism , Malondialdehyde/metabolism , Mice , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , Pulmonary Edema/blood , Pulmonary Edema/prevention & control , Pulmonary Edema/virology , Ribavirin/pharmacology , Superoxide Dismutase/metabolism , Time Factors , Tumor Necrosis Factor-alpha/blood , Yang Deficiency/blood , Yang Deficiency/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...