Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Pulm Ther ; 8(3): 297-310, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35969360

ABSTRACT

INTRODUCTION: Available therapies for acute cough, a condition frequently caused by a viral upper respiratory tract infection (URTI), have shown limited evidence of efficacy. Gefapixant, a P2X3-receptor antagonist, has demonstrated efficacy and safety in studies of the treatment of refractory or unexplained chronic cough, but its efficacy for treating acute cough has not been previously studied. METHODS: This was a phase 2a, randomized, double-blind, placebo-controlled, parallel-group, pilot study. Healthy volunteers were randomized 1:1 to receive twice-daily gefapixant 45 mg or placebo and inoculated with human rhinovirus 16 to induce URTI and cough. Participants were observed while quarantined for 7 days after the start of treatment. The primary endpoint was awake cough frequency on day 3, which was objectively measured with a cough-recording device. Secondary endpoints included change from baseline to day 3 in subjective cough severity measures (cough severity visual analog scale, Cough Severity Diary) and cough-specific quality of life (Leicester Cough Questionnaire-acute). RESULTS: Of the 46 participants who met inclusion criteria [mean (standard deviation, SD) age, 24.6 (6.5) years; females, n = 8], 40 completed the study (gefapixant, n = 21; placebo, n = 19). There was no significant difference in awake cough frequency on day 3 between the gefapixant and placebo groups [least squares means, 2.4 versus 2.7 coughs per hour, respectively; mean difference (95% confidence interval, CI), -0.3 (-2.3, 1.7); P = 0.75]. There were no significant between-group differences for any of the secondary endpoints. Peak cough frequency was low and occurred later in the study than expected (days 4-5). The safety profile was consistent with that of previous studies of gefapixant. CONCLUSION: Compared with placebo, gefapixant did not reduce the frequency or severity of acute cough secondary to induced URTI. Induced viral URTI produced mild symptoms, including lower cough frequency than observed in previous studies of patients selected for acute cough associated with naturally occurring URTI. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03569033; EudraCT, 2017-000472-28; protocol number, MK-7264-013.

2.
Nat Med ; 28(5): 1031-1041, 2022 05.
Article in English | MEDLINE | ID: mdl-35361992

ABSTRACT

Since its emergence in 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused hundreds of millions of cases and continues to circulate globally. To establish a novel SARS-CoV-2 human challenge model that enables controlled investigation of pathogenesis, correlates of protection and efficacy testing of forthcoming interventions, 36 volunteers aged 18-29 years without evidence of previous infection or vaccination were inoculated with 10 TCID50 of a wild-type virus (SARS-CoV-2/human/GBR/484861/2020) intranasally in an open-label, non-randomized study (ClinicalTrials.gov identifier NCT04865237 ; funder, UK Vaccine Taskforce). After inoculation, participants were housed in a high-containment quarantine unit, with 24-hour close medical monitoring and full access to higher-level clinical care. The study's primary objective was to identify an inoculum dose that induced well-tolerated infection in more than 50% of participants, with secondary objectives to assess virus and symptom kinetics during infection. All pre-specified primary and secondary objectives were met. Two participants were excluded from the per-protocol analysis owing to seroconversion between screening and inoculation, identified post hoc. Eighteen (~53%) participants became infected, with viral load (VL) rising steeply and peaking at ~5 days after inoculation. Virus was first detected in the throat but rose to significantly higher levels in the nose, peaking at ~8.87 log10 copies per milliliter (median, 95% confidence interval (8.41, 9.53)). Viable virus was recoverable from the nose up to ~10 days after inoculation, on average. There were no serious adverse events. Mild-to-moderate symptoms were reported by 16 (89%) infected participants, beginning 2-4 days after inoculation, whereas two (11%) participants remained asymptomatic (no reportable symptoms). Anosmia or dysosmia developed more slowly in 15 (83%) participants. No quantitative correlation was noted between VL and symptoms, with high VLs present even in asymptomatic infection. All infected individuals developed serum spike-specific IgG and neutralizing antibodies. Results from lateral flow tests were strongly associated with viable virus, and modeling showed that twice-weekly rapid antigen tests could diagnose infection before 70-80% of viable virus had been generated. Thus, with detailed characterization and safety analysis of this first SARS-CoV-2 human challenge study in young adults, viral kinetics over the course of primary infection with SARS-CoV-2 were established, with implications for public health recommendations and strategies to affect SARS-CoV-2 transmission. Future studies will identify the immune factors associated with protection in those participants who did not develop infection or symptoms and define the effect of prior immunity and viral variation on clinical outcome.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , Humans , Kinetics , Treatment Outcome , Viral Load , Young Adult
3.
N Engl J Med ; 386(7): 655-666, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35172056

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) infection causes substantial morbidity and mortality among infants, older adults, and immunocompromised adults. EDP-938, a nonfusion replication inhibitor of RSV, acts by modulating the viral nucleoprotein. METHODS: In a two-part, phase 2a, randomized, double-blind, placebo-controlled challenge trial, we assigned participants who had been inoculated with RSV-A Memphis 37b to receive EDP-938 or placebo. Different doses of EDP-938 were assessed. Nasal-wash samples were obtained from day 2 until day 12 for assessments. Clinical symptoms were assessed by the participants, and pharmacokinetic profiles were obtained. The primary end point was the area under the curve (AUC) for the RSV viral load, as measured by reverse-transcriptase-quantitative polymerase-chain-reaction assay. The key secondary end point was the AUC for the total symptom score. RESULTS: In part 1 of the trial, 115 participants were assigned to receive EDP-938 (600 mg once daily [600-mg once-daily group] or 300 mg twice daily after a 500-mg loading dose [300-mg twice-daily group]) or placebo. In part 2, a total of 63 participants were assigned to receive EDP-938 (300 mg once daily after a 600-mg loading dose [300-mg once-daily group] or 200 mg twice daily after a 400-mg loading dose [200-mg twice-daily group]) or placebo. In part 1, the AUC for the mean viral load (hours × log10 copies per milliliter) was 204.0 in the 600-mg once-daily group, 217.7 in the 300-mg twice-daily group, and 790.2 in the placebo group. The AUC for the mean total symptom score (hours × score, with higher values indicating greater severity) was 124.5 in the 600-mg once-daily group, 181.8 in the 300-mg twice-daily group, and 478.8 in the placebo group. The results in part 2 followed a pattern similar to that in part 1: the AUC for the mean viral load was 173.9 in the 300-mg once-daily group, 196.2 in the 200-mg twice-daily group, and 879.0 in the placebo group, and the AUC for the mean total symptom score was 99.3, 89.6, and 432.2, respectively. In both parts, mucus production was more than 70% lower in each EDP-938 group than in the placebo group. The four EDP-938 regimens had a safety profile similar to that of placebo. Across all dosing regimens, the EDP-938 median time to maximum concentration ranged from 4 to 5 hours, and the geometric mean half-life ranged from 13.7 to 14.5 hours. CONCLUSIONS: All EDP-938 regimens were superior to placebo with regard to lowering of the viral load, total symptom scores, and mucus weight without apparent safety concerns. (ClinicalTrials.gov number, NCT03691623.).


Subject(s)
Antiviral Agents , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Adult , Female , Humans , Male , Administration, Oral , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacology , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/isolation & purification , Viral Load/drug effects
4.
J Infect Dis ; 226(3): 396-406, 2022 08 26.
Article in English | MEDLINE | ID: mdl-33400792

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is a significant cause of severe lower respiratory tract disease in children and older adults, but has no approved vaccine. This study assessed the potential of Ad26.RSV.preF to protect against RSV infection and disease in an RSV human challenge model. METHODS: In this double-blind, placebo-controlled study, healthy adults aged 18-50 years were randomized 1:1 to receive 1 × 1011 vp Ad26.RSV.preF or placebo intramuscularly. Twenty-eight days postimmunization, volunteers were challenged intranasally with RSV-A (Memphis 37b). Assessments included viral load (VL), RSV infections, clinical symptom score (CSS), safety, and immunogenicity. RESULTS: Postchallenge, VL, RSV infections, and disease severity were lower in Ad26.RSV.preF (n = 27) vs placebo (n = 26) recipients: median VL area under the curve (AUC) quantitative real-time polymerase chain reaction: 0.0 vs 236.0 (P = .012; predefined primary endpoint); median VL-AUC quantitative culture: 0.0 vs 109; RSV infections 11 (40.7%) vs 17 (65.4%); median RSV AUC-CSS 35 vs 167, respectively. From baseline to 28 days postimmunization, geometric mean fold increases in RSV A2 neutralizing antibody titers of 5.8 and 0.9 were observed in Ad26.RSV.preF and placebo, respectively. Ad26.RSV.preF was well tolerated. CONCLUSIONS: Ad26.RSV.preF demonstrated protection from RSV infection through immunization in a human challenge model, and therefore could potentially protect against natural RSV infection and disease. CLINICAL TRIALS REGISTRATION: NCT03334695; CR108398, 2017-003194-33 (EudraCT); VAC18193RSV2002.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Aged , Antibodies, Neutralizing , Antibodies, Viral , Child , Humans , Immunization , Viral Fusion Proteins
5.
BMC Res Notes ; 11(1): 620, 2018 Aug 29.
Article in English | MEDLINE | ID: mdl-30157933

ABSTRACT

OBJECTIVE: This manuscript aims to provide an overview of the unique considerations and best practice principles associated with the manufacture of human viral challenge agents. RESULTS: Considerations are discussed on the entire process from strain and viral source selection through manufacturing, safety and efficacy testing. The human viral challenge (HVC) model is an important tool to help accelerate the drug development process but producing viruses suitable for use in the model presents a unique set of challenges. There are many case by case decisions and risk assessments to consider and no clear international standard to produce viruses for this purpose. The authors present challenge virus manufacturing considerations from the current literature, regulatory guidance and their own direct experience in producing challenge viruses. The use of these viral stocks in clinical studies, as published in peer-reviewed journals, is also briefly described.


Subject(s)
Drug Discovery , Viruses , Antibodies, Viral , Humans , Risk Assessment
6.
Respir Res ; 19(1): 123, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29929556

ABSTRACT

The Human Viral Challenge (HVC) model has, for many decades, helped in the understanding of respiratory viruses and their role in disease pathogenesis. In a controlled setting using small numbers of volunteers removed from community exposure to other infections, this experimental model enables proof of concept work to be undertaken on novel therapeutics, including vaccines, immunomodulators and antivirals, as well as new diagnostics.Crucially, unlike conventional phase 1 studies, challenge studies include evaluable efficacy endpoints that then guide decisions on how to optimise subsequent field studies, as recommended by the FDA and thus licensing studies that follow. Such a strategy optimises the benefit of the studies and identifies possible threats early on, minimising the risk to subsequent volunteers but also maximising the benefit of scarce resources available to the research group investing in the research. Inspired by the principles of the 3Rs (Replacement, Reduction and Refinement) now commonly applied in the preclinical phase, HVC studies allow refinement and reduction of the subsequent development phase, accelerating progress towards further statistically powered phase 2b studies. The breadth of data generated from challenge studies allows for exploration of a wide range of variables and endpoints that can then be taken through to pivotal phase 3 studies.We describe the disease burden for acute respiratory viral infections for which current conventional development strategies have failed to produce therapeutics that meet clinical need. The Authors describe the HVC model's utility in increasing scientific understanding and in progressing promising therapeutics through development.The contribution of the model to the elucidation of the virus-host interaction, both regarding viral pathogenicity and the body's immunological response is discussed, along with its utility to assist in the development of novel diagnostics.Future applications of the model are also explored.


Subject(s)
Antiviral Agents/therapeutic use , Respiratory Tract Infections/drug therapy , Viral Vaccines/therapeutic use , Antiviral Agents/pharmacology , Clinical Trials as Topic/methods , Drug Evaluation, Preclinical/methods , Humans , Influenza, Human/drug therapy , Influenza, Human/physiopathology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/physiology , Respiratory Tract Infections/physiopathology , Rhinovirus/drug effects , Rhinovirus/physiology , Viral Load/drug effects , Viral Load/physiology , Viral Vaccines/pharmacology
8.
PLoS One ; 11(1): e0145902, 2016.
Article in English | MEDLINE | ID: mdl-26761707

ABSTRACT

BACKGROUND: Influenza and its associated diseases are a major cause of morbidity and mortality. The United States Advisory Committee on Immunization Practices recommends influenza vaccination for everyone over 6 months of age. The failure of the flu vaccine in 2014-2015 demonstrates the need for a model that allows the rapid development of novel antivirals, universal/intra-seasonal vaccines, immunomodulators, monoclonal antibodies and other novel treatments. To this end we manufactured a new H3N2 influenza virus in compliance with Good Manufacturing Practice for use in the Human Viral Challenge Model. METHODS AND STRAIN SELECTION: We chose an H3N2 influenza subtype, rather than H1N1, given that this strain has the most substantial impact in terms of morbidity or mortality annually as described by the Centre for Disease Control. We first subjected the virus batch to rigorous adventitious agent testing, confirmed the virus to be wild-type by Sanger sequencing and determined the virus titres appropriate for human use via the established ferret model. We built on our previous experience with other H3N2 and H1N1 viruses to develop this unique model. HUMAN CHALLENGE AND CONCLUSIONS: We conducted an initial safety and characterisation study in healthy adult volunteers, utilising our unique clinical quarantine facility in London, UK. In this study we demonstrated this new influenza (H3N2) challenge virus to be both safe and pathogenic with an appropriate level of disease in volunteers. Furthermore, by inoculating volunteers with a range of different inoculum titres, we established the minimum infectious titre required to achieve reproducible disease whilst ensuring a sensitive model that can be translated to design of subsequent field based studies. TRIAL REGISTRATION: ClinicalTrials.gov NCT02525055.


Subject(s)
Antibodies, Monoclonal/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Adult , Animals , Antibodies, Viral/immunology , Antiviral Agents/chemistry , Double-Blind Method , Female , Ferrets , Healthy Volunteers , Humans , Influenza Vaccines/chemistry , Influenza, Human/virology , London , Male , Middle Aged , Polymerase Chain Reaction , Virus Shedding , Young Adult
9.
J Virol Methods ; 224: 83-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26335961

ABSTRACT

Retroscreen (hVIVO) have developed an RSV human viral challenge model (hVCM) for testing the efficacy of novel antiviral therapies by monitoring changes in viral load and symptoms. The integrated cycler technology and Simplexa™ kits (Focus Diagnostics) currently provide fast, qualitative and sensitive diagnostic testing in hospitals and other healthcare facilities for patients with well-established respiratory illness. We have developed a novel use of qualitative integrated cycler PCR (qicPCR) technology to identify onset of RSV infection enabling an informed dosing clinical protocol in the RSV hVCM. We have validated qicPCR detection of RSV in spiked nasal wash aspirates and demonstrate that the qicPCR assay is 94% concordant with RSV plaque assay data in nasal wash samples from 53 RSV inoculated human volunteers in the hVCM. The use of qicPCR for informed dosing was successfully implemented in a recent clinical trial demonstrating efficacy of the RSV entry inhibitor GS-5806 in the hVCM (NCT01756482). Comparison of qicPCR positivity in relation to nasal wash viral load measured by both RT-qPCR and plaque assay shows that the therapeutic exposure was correctly initiated prior to onset and peak of RSV viral shedding and symptoms in the majority of volunteers.


Subject(s)
Antiviral Agents/administration & dosage , Nasal Cavity/virology , Polymerase Chain Reaction/methods , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Viruses/isolation & purification , Viral Load/methods , Clinical Trials as Topic , Humans , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology
10.
PLoS One ; 9(5): e93761, 2014.
Article in English | MEDLINE | ID: mdl-24850536

ABSTRACT

We investigated the protective efficacy of two intranasal chitosan (CSN and TM-CSN) adjuvanted H5N1 Influenza vaccines against highly pathogenic avian Influenza (HPAI) intratracheal and intranasal challenge in a ferret model. Six groups of 6 ferrets were intranasally vaccinated twice, 21 days apart, with either placebo, antigen alone, CSN adjuvanted antigen, or TM-CSN adjuvanted antigen. Homologous and intra-subtypic antibody cross-reacting responses were assessed. Ferrets were inoculated intratracheally (all treatments) or intranasally (CSN adjuvanted and placebo treatments only) with clade 1 HPAI A/Vietnam/1194/2004 (H5N1) virus 28 days after the second vaccination and subsequently monitored for morbidity and mortality outcomes. Clinical signs were assessed and nasal as well as throat swabs were taken daily for virology. Samples of lung tissue, nasal turbinates, brain, and olfactory bulb were analysed for the presence of virus and examined for histolopathological findings. In contrast to animals vaccinated with antigen alone, the CSN and TM-CSN adjuvanted vaccines induced high levels of antibodies, protected ferrets from death, reduced viral replication and abrogated disease after intratracheal challenge, and in the case of CSN after intranasal challenge. In particular, the TM-CSN adjuvanted vaccine was highly effective at eliciting protective immunity from intratracheal challenge; serologically, protective titres were demonstrable after one vaccination. The 2-dose schedule with TM-CSN vaccine also induced cross-reactive antibodies to clade 2.1 and 2.2 H5N1 viruses. Furthermore ferrets immunised with TM-CSN had no detectable virus in the respiratory tract or brain, whereas there were signs of virus in the throat and lungs, albeit at significantly reduced levels, in CSN vaccinated animals. This study demonstrated for the first time that CSN and in particular TM-CSN adjuvanted intranasal vaccines have the potential to protect against significant mortality and morbidity arising from infection with HPAI H5N1 virus.


Subject(s)
Chitosan/analogs & derivatives , Chitosan/administration & dosage , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Vaccination , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Animals, Outbred Strains , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dogs , Ferrets , Humans , Influenza, Human/blood , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Male , Nose/immunology , Nose/virology , Trachea/immunology , Trachea/virology , Vaccine Potency , Viral Load
11.
Eur J Immunol ; 39(6): 1587-96, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19424969

ABSTRACT

Bacterial products (such as endotoxins and flagellin) trigger innate immune responses through TLRs. Flagellin-induced signalling involves TLR5 and MyD88 and, according to some reports, TLR4. Whereas epithelial and dendritic cells are stimulated by flagellin in vitro, the cell contribution to the in vivo response is still unclear. Here, we studied the respective roles of radioresistant and radiosensitive cells in flagellin-induced airway inflammation in mice. We found that i.n. delivery of flagellin elicits a transient change in respiratory function and an acute, pro-inflammatory response in the lungs, characterized by TLR5- and MyD88-dependent chemokine secretion and neutrophil recruitment. In contrast, TLR4, CD14 and TRIF were not essential for flagellin-mediated responses, indicating that TLR4 does not cooperate with TLR5 in the lungs. Respiratory function, chemokine secretion and airway infiltration by neutrophils were dependent on radioresistant, TLR5-expressing cells. Furthermore, lung haematopoietic cells also responded to flagellin by activating TNF-alpha production. We suggest that the radioresistant lung epithelial cells are essential for initiating early, TLR5-dependent signalling in response to flagellin and thus triggering the lung's innate immune responses.


Subject(s)
Epithelial Cells/immunology , Flagellin/immunology , Immunity, Innate/immunology , Radiation Tolerance , Respiratory Mucosa/immunology , Toll-Like Receptor 5/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Administration, Intranasal , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoconstriction/immunology , Cell Movement/genetics , Cell Movement/immunology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/metabolism , Gene Expression/immunology , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neutrophils/cytology , Peroxidase/metabolism , Plethysmography, Whole Body , Radiation Chimera/immunology , Radiation Chimera/metabolism , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 5/genetics
12.
J Immunol ; 180(2): 1169-78, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18178857

ABSTRACT

Acute cigarette smoke exposure of the airways (two cigarettes twice daily for three days) induces acute inflammation in mice. In this study, we show that airway inflammation is dependent on Toll-like receptor 4 and IL-1R1 signaling. Cigarette smoke induced a significant recruitment of neutrophils in the bronchoalveolar space and pulmonary parenchyma, which was reduced in TLR4-, MyD88-, and IL-1R1-deficient mice. Diminished neutrophil influx was associated with reduced IL-1, IL-6, and keratinocyte-derived chemokine levels and matrix metalloproteinase-9 activity in the bronchoalveolar space. Further, cigarette smoke condensate (CSC) induced a macrophage proinflammatory response in vitro, which was dependent on MyD88, IL-1R1, and TLR4 signaling, but not attributable to LPS. Heat shock protein 70, a known TLR4 agonist, was induced in the airways upon smoke exposure, which probably activates the innate immune system via TLR4/MyD88, resulting in airway inflammation. CSC-activated macrophages released mature IL-1beta only in presence of ATP, whereas CSC alone promoted the TLR4/MyD88 signaling dependent production of IL-1alpha and pro-IL-1beta implicating cooperation between TLRs and the inflammasome. In conclusion, acute cigarette exposure results in LPS-independent TLR4 activation, leading to IL-1 production and IL-1R1 signaling, which is crucial for cigarette smoke induced inflammation leading to chronic obstructive pulmonary disease with emphysema.


Subject(s)
Myeloid Differentiation Factor 88/genetics , Nicotiana/toxicity , Pneumonia/chemically induced , Pneumonia/genetics , Receptors, Interleukin-1 Type I/genetics , Smoke , Toll-Like Receptor 4/genetics , Animals , HSP70 Heat-Shock Proteins/metabolism , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Mutant Strains , Neutrophils/drug effects , Neutrophils/immunology , Pneumonia/immunology , Signal Transduction/genetics
13.
Int J Exp Pathol ; 88(6): 387-91, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18039275

ABSTRACT

Recent studies on endotoxin/lipopolysaccharide (LPS)-induced acute inflammatory response in the lung are reviewed. The acute airway inflammatory response to inhaled endotoxin is mediated through Toll-like receptor 4 (TLR4) and CD14 signalling as mice deficient for TLR4 or CD14 are unresponsive to endotoxin. Acute bronchoconstriction, tumour necrosis factor (TNF), interleukin (IL)-12 and keratinocyte-derived chemokine (KC) production, protein leak and neutrophil recruitment in the lung are abrogated in mice deficient for the adaptor molecules myeloid differentiation factor 88 (MyD88) and Toll/Interleukin-1 receptor (TIR)-domain-containing adaptor protein (TIRAP), but independent of TIR-domain-containing adaptor-inducing interferon-beta (TRIF). In particular, LPS-induced TNF is required for bronchoconstriction, but dispensable for inflammatory cell recruitment. Lipopolysaccharide induces activation of the p38 mitogen-activated protein kinase (MAPK). Inhibition of pulmonary MAPK activity abrogates LPS-induced TNF production, bronchoconstriction, neutrophil recruitment into the lungs and broncho-alveolar space. In conclusion, TLR4-mediated, bronchoconstriction and acute inflammatory lung pathology to inhaled endotoxin are dependent on TLR4/CD14/MD2 expression using the adapter proteins TIRAP and MyD88, while TRIF, IL-1R1 or IL-18R signalling pathways are dispensable. Further downstream in this axis of signalling, TNF blockade reduces only acute bronchoconstriction, while MAPK inhibition abrogates completely endotoxin-induced inflammation.


Subject(s)
Lung/immunology , MAP Kinase Signaling System , Pneumonia/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Bronchoconstriction , Cytokines/immunology , Enzyme Activation , Humans , Lipopolysaccharides , Mice , Mice, Transgenic , Pneumonia/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Clin Invest ; 117(12): 3786-99, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17992263

ABSTRACT

The molecular mechanisms of acute lung injury resulting in inflammation and fibrosis are not well established. Here we investigate the roles of the IL-1 receptor 1 (IL-1R1) and the common adaptor for Toll/IL-1R signal transduction, MyD88, in this process using a murine model of acute pulmonary injury. Bleomycin insult results in expression of neutrophil and lymphocyte chemotactic factors, chronic inflammation, remodeling, and fibrosis. We demonstrate that these end points were attenuated in the lungs of IL-1R1- and MyD88-deficient mice. Further, in bone marrow chimera experiments, bleomycin-induced inflammation required primarily MyD88 signaling from radioresistant resident cells. Exogenous rIL-1beta recapitulated a high degree of bleomycin-induced lung pathology, and specific blockade of IL-1R1 by IL-1 receptor antagonist dramatically reduced bleomycin-induced inflammation. Finally, we found that lung IL-1beta production and inflammation in response to bleomycin required ASC, an inflammasome adaptor molecule. In conclusion, bleomycin-induced lung pathology required the inflammasome and IL-1R1/MyD88 signaling, and IL-1 represented a critical effector of pathology and therapeutic target of chronic lung inflammation and fibrosis.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Interleukin-1 Type I/metabolism , Respiratory Distress Syndrome/metabolism , Signal Transduction , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Bone Marrow Transplantation , Chronic Disease , Disease Models, Animal , Humans , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/pharmacology , Interleukin-8/genetics , Interleukin-8/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Receptors, Interleukin-1 Type I/agonists , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Receptors, Interleukin-1 Type I/genetics , Recombinant Proteins/pharmacology , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transplantation Chimera/genetics , Transplantation Chimera/metabolism
15.
Eur J Immunol ; 37(3): 768-79, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17301950

ABSTRACT

Acute and chronic airway inflammations caused by environmental agents including endotoxin represent an increasing health problem. Local TNF production may contribute to lung dysfunction and inflammation, although pulmonary neutrophil recruitment occurs in the absence of TNF. First, we demonstrate that membrane-bound TNF is sufficient to mediate the inflammatory responses to lipopolysaccharide (LPS). Secondly, using cell type-specific TNF-deficient mice we show that TNF derived from either macrophage/neutrophil (M/N) or T lymphocytes have differential effects on LPS-induced respiratory dysfunction (enhanced respiratory pause, Penh) and pulmonary neutrophil recruitment. While Penh, vascular leak, neutrophil recruitment, TNF, and thymus- and activation-regulated chemokine/CCL17 (TARC) expression in the lung were reduced in M/N-deficient mice, T cell-specific TNF-deficient mice displayed augmented Penh, vascular leak, neutrophil influx, increased CD11c+ cells and expression of TNF, TARC and murine CXC chemokines KC/CXCL1 in the lung. In conclusion, inactivation of TNF in either M/N or T cells has differential effects on LPS-induced lung disease, suggesting that selective deletion of TNF in T cells may aggravate airway pathology.


Subject(s)
Bronchi/immunology , Down-Regulation/immunology , Endotoxins/toxicity , Lung/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factors/physiology , Administration, Inhalation , Animals , Bronchi/pathology , Cells, Cultured , Endotoxins/antagonists & inhibitors , Lung/pathology , Mice , Mice, Inbred C57BL , Plethysmography, Whole Body , T-Lymphocytes/immunology
16.
Lab Invest ; 86(11): 1126-35, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16983331

ABSTRACT

Inhaled endotoxins induce an acute inflammatory response in the airways mediated through Toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88). However, the relative roles of the TLR4 adaptor proteins TIRAP and TRIF and of the MyD88-dependent IL-1 and IL-18 receptor pathways in this response are unclear. Here, we demonstrate that endotoxin-induced acute bronchoconstriction, vascular damage resulting in protein leak, Th1 cytokine and chemokine secretion and neutrophil recruitment in the airways are abrogated in mice deficient for either TIRAP or MyD88, but not in TRIF deficient mice. The contribution of other TLR-independent, MyD88-dependent signaling pathways was investigated in IL-1R1, IL-18R and caspase-1 (ICE)-deficient mice, which displayed normal airway responses to endotoxin. In conclusion, the TLR4-mediated, bronchoconstriction and acute inflammatory lung pathology to inhaled endotoxin critically depend on the expression of both adaptor proteins, TIRAP and MyD88, suggesting cooperative roles, while TRIF, IL-1R1, IL-18R signaling pathways are dispensable.


Subject(s)
Bronchi/drug effects , Bronchoconstriction/drug effects , Endotoxins/toxicity , Lipopolysaccharides/toxicity , Membrane Glycoproteins/physiology , Myeloid Differentiation Factor 88/physiology , Receptors, Interleukin-1/physiology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Bronchi/metabolism , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Cells, Cultured , Interleukin-1/metabolism , Interleukin-18/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism
17.
J Immunol ; 175(10): 6861-9, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16272344

ABSTRACT

Inhaled endotoxin induces an inflammatory response that contributes to the development and severity of asthma and other forms of airway disease. Here, we show that inhaled endotoxin-induced acute bronchoconstriction, TNF, IL-12p40, and KC production, protein leak, and neutrophil recruitment in the lung are abrogated in mice deficient for the adaptor molecule MyD88. Bronchoconstriction, inflammation, and protein leak are normal in Toll/IL-1R domain-containing adaptor inducing IFN-beta-deficient mice. MyD88 is involved in TLR, but also in IL-1R-associated kinase 1-mediated IL-1R and -18R signaling. We exclude a role for IL-1 and IL-18 pathways in this response, as IL-1R1 and caspase-1 (ICE)-deficient mice develop lung inflammation while TLR4-deficient mice are unresponsive to inhaled LPS. Significantly, using bone marrow chimera, we demonstrate that both hemopoietic and resident cells are necessary for a full MyD88-dependent response to inhaled endotoxin; bronchoconstriction depends on resident cells while cytokine secretion is mediated by hemopoietic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, Differentiation/metabolism , Bone Marrow Cells/drug effects , Bronchoconstriction/drug effects , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/immunology , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Administration, Inhalation , Animals , Antigens, Differentiation/genetics , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Chimera , Cytokines/biosynthesis , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Lipopolysaccharides/administration & dosage , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88 , Neutrophils/drug effects , Pneumonia/etiology , Pneumonia/immunology , Pneumonia/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics
18.
J Immunol ; 175(1): 262-9, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972657

ABSTRACT

The administration of endotoxins from Gram-negative bacteria induces manifestations reminding of acute respiratory distress syndrome. p38 MAPKs have been implicated in this pathology. In this study, we show that the specific p38 alpha,beta MAPK inhibitor, compound 37, prevents LPS-induced bronchoconstriction and neutrophil recruitment into the lungs and bronchoalveolar space in a dose-dependent manner in C57BL/6 mice. Furthermore, TNF induction and TNF signals were blocked. In TNF-deficient mice, bronchoconstriction, but not neutrophil sequestration, in the lung was abrogated after LPS administration. Therefore, TNF inhibition does not explain all of the effects of the p38 MAPK inhibitor. The p38 alpha,beta MAPK inhibitor also prevented LPS-induced neutrophilia in TNF-deficient mice. In conclusion, LPS provokes acute bronchoconstriction that is TNF dependent and p38 MAPK mediated, whereas the neutrophil recruitment is independent of TNF but depends on LPS/TLR4-induced signals mediated by p38 MAPK.


Subject(s)
Bronchoconstriction/drug effects , Bronchoconstriction/physiology , Neutrophils/drug effects , Neutrophils/physiology , Respiratory Distress Syndrome/physiopathology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Interleukin-6/biosynthesis , Leukocytosis/chemically induced , Leukocytosis/pathology , Leukocytosis/physiopathology , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/pathology , Lung/physiopathology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/pathology , Respiratory Distress Syndrome/chemically induced , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...