Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Respir Cell Mol Biol ; 58(2): 253-260, 2018 02.
Article in English | MEDLINE | ID: mdl-28915064

ABSTRACT

Mycoplasma pneumoniae infection has been linked to poor asthma outcomes. M. pneumoniae produces an ADP-ribosylating and vacuolating toxin called community-acquired respiratory distress syndrome (CARDS) toxin that has a major role in inflammation and airway dysfunction. The objective was to evaluate the immunopathological effects in primates exposed to M. pneumoniae or CARDS toxin. A total of 13 baboons were exposed to M. pneumoniae or CARDS toxin. At Days 7 and 14, BAL fluid was collected and analyzed for cell count, percent of each type of cell, CARDS toxin by PCR, CARDS toxin by antigen capture, eosinophilic cationic protein, and cytokine profiles. Serum IgM, IgG, and IgE responses to CARDS toxin were measured. All animals had a necropsy for analysis of the histopathological changes on lungs. No animal developed signs of infection. The serological responses to CARDS toxin were variable. At Day 14, four of seven animals exposed to M. pneumoniae and all four animals exposed to CARDS toxin developed histological "asthma-like" changes. T cell intracellular cytokine analysis revealed an increasing ratio of IL-4/IFN-γ over time. Both M. pneumoniae and CARDS toxin exposure resulted in similar histopathological pulmonary changes, suggesting that CARDS toxin plays a major role in the inflammatory response.


Subject(s)
Asthma/immunology , Asthma/pathology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Lung/immunology , Lung/pathology , Mycoplasma pneumoniae/pathogenicity , Animals , CD4-Positive T-Lymphocytes/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Interleukin-13/immunology , Interleukin-4/immunology , Lung/microbiology , Mice , Mycoplasma pneumoniae/immunology , Papio
2.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29061706

ABSTRACT

Mycoplasma pneumoniae is an atypical bacterial respiratory pathogen known to cause a range of airway inflammation and lung and extrapulmonary pathologies. We recently reported that an M. pneumoniae-derived ADP-ribosylating and vacuolating toxin called community-acquired respiratory distress syndrome (CARDS) toxin is capable of triggering NLRP3 (NLR-family, leucine-rich repeat protein 3) inflammasome activation and interleukin-1ß (IL-1ß) secretion in macrophages. However, it is unclear whether the NLRP3 inflammasome is important for the immune response during M. pneumoniae acute infection. In the current study, we utilized in vitro and in vivo models of M. pneumoniae infection to characterize the role of the NLRP3 inflammasome during acute infection. M. pneumoniae-infected macrophages deficient for inflammasome components NLRP3, ASC (apoptosis speck-like protein containing a caspase activation and recruitment domain), or caspase-1 failed to process and secrete IL-1ß. The MyD88/NF-κB signaling pathway was found to be critical for proinflammatory gene expression in macrophages infected with M. pneumoniae C57BL/6 mice deficient for NLRP3 expression were unable to produce IL-1ß in the airways during acute infection, and lack of this inflammatory response led to deficient immune cell activation and delayed bacterial clearance. These findings are the first to report the importance of the NLRP3 inflammasome in regulating the inflammatory response and influencing the progression of M. pneumoniae during acute infection.


Subject(s)
Immunity, Innate/immunology , Inflammation/metabolism , Mycoplasma pneumoniae/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/metabolism , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/immunology , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/immunology , Caspase 1/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/microbiology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Pneumonia, Mycoplasma/microbiology , Signal Transduction/immunology
3.
Am J Respir Crit Care Med ; 196(5): 609-620, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28614669

ABSTRACT

RATIONALE: Up to one-third of patients hospitalized with pneumococcal pneumonia experience major adverse cardiac events (MACE) during or after pneumonia. In mice, Streptococcus pneumoniae can invade the myocardium, induce cardiomyocyte death, and disrupt cardiac function following bacteremia, but it is unknown whether the same occurs in humans with severe pneumonia. OBJECTIVES: We sought to determine whether S. pneumoniae can (1) translocate the heart, (2) induce cardiomyocyte death, (3) cause MACE, and (4) induce cardiac scar formation after antibiotic treatment during severe pneumonia using a nonhuman primate (NHP) model. METHODS: We examined cardiac tissue from six adult NHPs with severe pneumococcal pneumonia and three uninfected control animals. Three animals were rescued with antibiotics (convalescent animals). Electrocardiographic, echocardiographic, and serum biomarkers of cardiac damage were measured (troponin T, N-terminal pro-brain natriuretic peptide, and heart-type fatty acid binding protein). Histological examination included hematoxylin and eosin staining, immunofluorescence, immunohistochemistry, picrosirius red staining, and transmission electron microscopy. Immunoblots were used to assess the underlying mechanisms. MEASUREMENTS AND MAIN RESULTS: Nonspecific ischemic alterations were detected by electrocardiography and echocardiography. Serum levels of troponin T and heart-type fatty acid binding protein were increased (P < 0.05) after pneumococcal infection in both acutely ill and convalescent NHPs. S. pneumoniae was detected in the myocardium of all NHPs with acute severe pneumonia. Necroptosis and apoptosis were detected in the myocardium of both acutely ill and convalescent NHPs. Evidence of cardiac scar formation was observed only in convalescent animals by transmission electron microscopy and picrosirius red staining. CONCLUSIONS: S. pneumoniae invades the myocardium and induces cardiac injury with necroptosis and apoptosis, followed by cardiac scarring after antibiotic therapy, in an NHP model of severe pneumonia.


Subject(s)
Cardiotoxicity/etiology , Myocardium/pathology , Pneumonia, Pneumococcal/complications , Streptococcus pneumoniae/pathogenicity , Animals , Anti-Bacterial Agents/therapeutic use , Blotting, Western , Cardiotoxicity/blood , Disease Models, Animal , Echocardiography , Electrocardiography , Fatty Acid-Binding Proteins/blood , Female , Heart/microbiology , Male , Papio , Pneumonia, Pneumococcal/blood , Pneumonia, Pneumococcal/drug therapy , Troponin T/blood
4.
PLoS One ; 11(11): e0166092, 2016.
Article in English | MEDLINE | ID: mdl-27855182

ABSTRACT

RATIONALE: Streptococcus pneumoniae is the leading cause of community-acquired pneumonia and infectious death in adults worldwide. A non-human primate model is needed to study the molecular mechanisms that underlie the development of severe pneumonia, identify diagnostic tools, explore potential therapeutic targets, and test clinical interventions during pneumococcal pneumonia. OBJECTIVE: To develop a non-human primate model of pneumococcal pneumonia. METHODS: Seven adult baboons (Papio cynocephalus) were surgically tethered to a continuous monitoring system that recorded heart rate, temperature, and electrocardiography. Animals were inoculated with 109 colony-forming units of S. pneumoniae using bronchoscopy. Three baboons were rescued with intravenous ampicillin therapy. Pneumonia was diagnosed using lung ultrasonography and ex vivo confirmation by histopathology and immunodetection of pneumococcal capsule. Organ failure, using serum biomarkers and quantification of bacteremia, was assessed daily. RESULTS: Challenged animals developed signs and symptoms of pneumonia 4 days after infection. Infection was characterized by the presence of cough, tachypnea, dyspnea, tachycardia and fever. All animals developed leukocytosis and bacteremia 24 hours after infection. A severe inflammatory reaction was detected by elevation of serum cytokines, including Interleukin (IL)1Ra, IL-6, and IL-8, after infection. Lung ultrasonography precisely detected the lobes with pneumonia that were later confirmed by pathological analysis. Lung pathology positively correlated with disease severity. Antimicrobial therapy rapidly reversed symptomology and reduced serum cytokines. CONCLUSIONS: We have developed a novel animal model for severe pneumococcal pneumonia that mimics the clinical presentation, inflammatory response, and infection kinetics seen in humans. This is a novel model to test vaccines and treatments, measure biomarkers to diagnose pneumonia, and predict outcomes.


Subject(s)
Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae , Animals , Biomarkers , Biopsy , Cytokines/metabolism , Disease Models, Animal , Hemodynamics , Inflammation Mediators/metabolism , Lung/diagnostic imaging , Lung/microbiology , Lung/pathology , Papio , Phenotype , Pneumonia, Pneumococcal/diagnosis , Primates , Severity of Illness Index , Streptococcus pneumoniae/classification , Ultrasonography
5.
PLoS One ; 9(7): e102613, 2014.
Article in English | MEDLINE | ID: mdl-25058417

ABSTRACT

Mycoplasma pneumoniae causes a range of airway and extrapulmonary pathologies in humans. Clinically, M. pneumoniae is associated with acute exacerbations of human asthma and a worsening of experimentally induced asthma in mice. Recently, we demonstrated that Community Acquired Respiratory Distress Syndrome (CARDS) toxin, an ADP-ribosylating and vacuolating toxin synthesized by M. pneumoniae, is sufficient to induce an asthma-like disease in BALB/cJ mice. To test the potential of CARDS toxin to exacerbate preexisting asthma, we examined inflammatory responses to recombinant CARDS toxin in an ovalbumin (OVA) murine model of asthma. Differences in pulmonary inflammatory responses between treatment groups were analyzed by histology, cell differentials and changes in cytokine and chemokine concentrations. Additionally, assessments of airway hyperreactivity were evaluated through direct pulmonary function measurements. Analysis of histology revealed exaggerated cellular inflammation with a strong eosinophilic component in the CARDS toxin-treated group. Heightened T-helper type-2 inflammatory responses were evidenced by increased expression of IL-4, IL-13, CCL17 and CCL22 corresponding with increased airway hyperreactivity in the CARDS toxin-treated mice. These data demonstrate that CARDS toxin can be a causal factor in the worsening of experimental allergic asthma, highlighting the potential importance of CARDS toxin in the etiology and exacerbation of human asthma.


Subject(s)
Asthma/pathology , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Bronchial Hyperreactivity/pathology , Respiratory System/drug effects , Animals , Asthma/chemically induced , Asthma/immunology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CCL17/biosynthesis , Chemokine CCL17/immunology , Chemokine CCL22/biosynthesis , Chemokine CCL22/immunology , Eosinophils/immunology , Eosinophils/pathology , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interleukin-13/biosynthesis , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Recombinant Proteins/toxicity , Respiratory System/immunology , Respiratory System/pathology , Th2 Cells/immunology , Th2 Cells/pathology
6.
Am J Respir Cell Mol Biol ; 46(6): 815-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22281984

ABSTRACT

Mycoplasma pneumoniae causes acute and chronic lung infections in humans, leading to a variety of pulmonary and extrapulmonary sequelae. Of the airway complications of M. pneumoniae infection, M. pneumoniae-associated exacerbation of asthma and pediatric wheezing are emerging as significant sources of human morbidity. However, M. pneumoniae products capable of promoting allergic inflammation are unknown. Recently, we reported that M. pneumoniae produces an ADP-ribosylating and vacuolating toxin termed the community-acquired respiratory distress syndrome (CARDS) toxin. Here we report that naive mice exposed to a single dose of recombinant CARDS (rCARDS) toxin respond with a robust inflammatory response consistent with allergic disease. rCARDS toxin induced 30-fold increased expression of the Th-2 cytokines IL-4 and IL-13 and 70- to 80-fold increased expression of the Th-2 chemokines CCL17 and CCL22, corresponding to a mixed cellular inflammatory response comprised of a robust eosinophilia, accumulation of T cells and B cells, and mucus metaplasia. The inflammatory responses correlate temporally with toxin-dependent increases in airway hyperreactivity characterized by increases in airway restriction and decreases in lung compliance. Furthermore, CARDS toxin-mediated changes in lung function and histopathology are dependent on CD4(+) T cells. Altogether, the data suggest that rCARDS toxin is capable of inducing allergic-type inflammation in naive animals and may represent a causal factor in M. pneumoniae-associated asthma.


Subject(s)
Bacterial Toxins/toxicity , Eosinophils/cytology , Lung/drug effects , Lymphocytes/cytology , Mycoplasma pneumoniae/physiology , Animals , Bronchoalveolar Lavage Fluid , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lung/cytology , Mice , Real-Time Polymerase Chain Reaction
7.
Neonatology ; 100(2): 130-8, 2011.
Article in English | MEDLINE | ID: mdl-21372595

ABSTRACT

BACKGROUND: Interleukin (IL)-6, when complexed with soluble IL-6 receptor (sIL-6R), has emerged as an important modulator of chemokine expression and leukocyte recruitment during inflammation and in this state can be specifically antagonised by soluble gp130 (sgp130). The expression of these modifiers of IL-6 activity during ventilator-induced inflammation remains poorly understood. OBJECTIVES: To ascertain the expression pattern of IL-6, sIL-6R and sgp130 in response to mechanical ventilation in the preterm neonatal lung and define its relationship to associated markers of inflammation. METHODS: Inflammatory cell recruitment and expression of IL-6, sIL-6R, sgp130, IL-8 and monocyte chemotactic protein-1 (MCP-1) were quantified in tracheal aspirate fluid collected over a 14-day period from preterm (125 days) baboons undergoing mechanical ventilation. RESULTS: Over the period of ventilation, the ratio of agonistic IL-6/sIL-6R increased 4.3-fold between days 3 and 10-11 (p < 0.01) while the ratio of antagonistic sgp130/IL-6 decreased 2.6-fold over the same period (p < 0.05). Over the same period, the relative numbers of neutrophils compared to mononuclear cells shifted from an excess of 1.8 on day 1 to 0.6 on day 14 (p < 0.01). Both IL-8 and MCP-1 were elevated between days 1 and 10-11 of ventilation (p < 0.01). CONCLUSIONS: In the ventilated preterm baboon lung, expression of sIL-6R and dynamic modulation of sgp130 expression appear to modulate the activity and inflammatory potential of IL-6.


Subject(s)
Animals, Newborn/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Lung/metabolism , Papio cynocephalus/metabolism , Premature Birth , Respiration, Artificial/adverse effects , Animals , Biomarkers/metabolism , Chemokine CCL2/metabolism , Cytokine Receptor gp130/metabolism , Disease Models, Animal , Female , Inflammation/etiology , Inflammation/pathology , Interleukin-8/metabolism , Lung Diseases/metabolism , Lung Diseases/pathology , Neutrophils/pathology , Pregnancy , Receptors, Interleukin-6/metabolism
8.
Pediatrics ; 118(5): 2038-50, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17079577

ABSTRACT

OBJECTIVE: Using the 125-day baboon model of bronchopulmonary dysplasia treated with prenatal steroid and exogenous surfactant, we hypothesized that a delay of extubation from low tidal volume positive pressure ventilation to nasal continuous positive airway pressure at 5 days (delayed nasal continuous positive airway pressure group) would not induce more lung injury when compared with baboons aggressively weaned to nasal continuous positive airway pressure at 24 hours (early nasal continuous positive airway pressure group), because both received positive pressure ventilation. METHODS AND RESULTS: After delivery by cesarean section at 125 days (term: 185 days), infants received 2 doses of Curosurf (Chiesi Farmaceutica S.p.A., Parma, Italy) and daily caffeine citrate. The delay in extubation to 5 days resulted in baboons in the delayed nasal continuous positive airway pressure group having a lower arterial to alveolar oxygen ratio, high PaCO2, and worse respiratory function. The animals in the delayed nasal continuous positive airway pressure group exhibited a poor respiratory drive that contributed to more reintubations and time on mechanical ventilation. A few animals in both groups developed necrotizing enterocolitis and/or sepsis, but infectious pneumonias were not documented. Cellular bronchiolitis and peribronchiolar alveolar wall thickening were more frequently seen in the delayed nasal continuous positive airway pressure group. Bronchoalveolar lavage levels of interleukin-6, interleukin-8, monocyte chemotactic protein-1, macrophage inflammatory protein-1 alpha, and growth-regulated oncogene-alpha were significantly increased in the delayed nasal continuous positive airway pressure group. Standard and digital morphometric analyses showed no significant differences in internal surface area and nodal measurements between the groups. Platelet endothelial cell adhesion molecule vascular staining was not significantly different between the 2 nasal continuous positive airway pressure groups. CONCLUSIONS: Volutrauma and/or low-grade colonization of airways secondary to increased reintubations and ventilation times are speculated to play causative roles in the delayed nasal continuous positive airway pressure group findings.


Subject(s)
Bronchopulmonary Dysplasia/therapy , Continuous Positive Airway Pressure , Disease Models, Animal , Ventilator Weaning , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Female , Humans , Infant, Newborn , Male , Papio , Time Factors
9.
FASEB J ; 20(10): 1698-700, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16807366

ABSTRACT

Bronchopulmonary dysplasia (BPD), a chronic lung disease affecting preterm neonates, is associated with significant childhood and adult health problems. Histopathologic features of BPD include impaired vascular and distal airway development. We previously showed that activation of hypoxia-inducible factors (HIFs) by inhibition of prolyl hydroxylase domain-containing proteins (PHDs) is feasible and that it stimulates vascular endothelial growth factor (VEGF)-dependent angiogenesis in vitro. We tested the hypothesis that enhancement of angiogenesis by activation of HIFs improves lung growth and function in prematurely born neonates in vivo. Preterm baboons (125 day+14 day pro re nata O2 model, corresponding to 27 human gestational weeks) were treated for 14 days with intravenous (i.v.) FG-4095, a PHD inhibitor. Notably, 77% of diminished total alveolar surface area in untreated controls was recovered by FG-4095 treatment. Functional significance of the structural changes was indicated by improved oxygenation and lung compliance in FG-4095-treated newborns. Surfactant proteins B and C and saturated phosphatidylcholine were unchanged. Incidence of spontaneous ductus arteriosus closure was increased, likely contributing to lower ratio of pulmonary to systemic blood flow in FG-4095 group. These findings indicate that HIF stimulation by PHD inhibition ameliorates pathological and physiological consequences of BPD.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Lung Diseases/drug therapy , Lung/growth & development , Premature Birth , Animals , Animals, Newborn , Chronic Disease , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Hypoxia-Inducible Factor 1/physiology , Lung/pathology , Lung/physiopathology , Lung Diseases/etiology , Male , Neovascularization, Physiologic/drug effects , Papio , Respiratory Function Tests , Treatment Outcome
10.
Am J Physiol Lung Cell Mol Physiol ; 288(3): L450-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15591412

ABSTRACT

Nitric oxide (NO) serves multiple functions in the developing lung, and pulmonary NO production is decreased in a baboon model of chronic lung disease (CLD) after premature birth at 125 days (d) gestation (term = 185d). To determine whether postnatal NO administration alters the genesis of CLD, the effects of inhaled NO (iNO, 5 ppm) were assessed in the baboon model over 14d. iNO caused a decrease in pulmonary artery pressure in the first 2d and a greater rate of spontaneous closure of the ductus arteriosus, and lung compliance was greater and expiratory resistance was improved during the first week. With iNO, postmortem pressure-volume curves were shifted upward, lung DNA content and cell proliferation were increased, and lung growth was preserved to equal that which occurs during the same period in utero. In addition, the excessive elastin deposition characteristic of CLD was normalized by iNO, and there was evidence of stimulation of secondary crest development. Thus, in the baboon model of CLD, iNO improves early pulmonary function and alters lung growth and extracellular matrix deposition. As such, NO biosynthetic pathway dysfunction may contribute to the pathogenesis of CLD.


Subject(s)
Animals, Newborn , Bronchodilator Agents/administration & dosage , Elastin/metabolism , Lung Diseases/physiopathology , Lung/physiopathology , Nitric Oxide/administration & dosage , Administration, Inhalation , Animals , Bronchodilator Agents/pharmacology , Chronic Disease , Hemodynamics/drug effects , Image Processing, Computer-Assisted , Lung/growth & development , Lung/metabolism , Lung/pathology , Lung Diseases/metabolism , Nitric Oxide/pharmacology , Organ Size/drug effects , Papio , Pulmonary Alveoli/pathology , Pulmonary Artery/physiopathology , Pulmonary Circulation/drug effects
11.
Am J Respir Crit Care Med ; 169(9): 1054-62, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14962819

ABSTRACT

Using the 125-day baboon model of long-term bronchopulmonary dysplasia, we hypothesized that early use of nasal continuous positive airway pressure (nCPAP), a noninvasive ventilatory method, combined with prophylactic surfactant therapy would permit continuation of alveolar and vascular development in the lung. Retrospective human studies have shown that infants treated with nCPAP spend less time on mechanical ventilation and thereby sustain less volutrauma. After delivery by cesarean section at 125 days (term, 185 days), the infants received two doses of surfactant (Curosurf) and daily caffeine citrate. Weaning from low-volume positive pressure ventilation to nCPAP was attempted at 24 hours of age. Serial physiological parameters were recorded. Lung histopathology and morphometric measurements of nCPAP animals were done after necropsy at 28 days and data were compared with 125- and 156-day gestational controls. Documented episodes of clinical sepsis and pneumonia at postmortem examination were absent. nCPAP lungs showed enlarged thin-walled air spaces with minimal fibroproliferation and scattered secondary crests. Internal surface area and surface-to-volume ratio dimensions were similar to those of 156-day gestational control lungs, the intrauterine developmental control. nCPAP is an effective noninvasive ventilatory technique that minimizes lung injury in baboons at risk of developing bronchopulmonary dysplasia.


Subject(s)
Bronchopulmonary Dysplasia/therapy , Continuous Positive Airway Pressure/methods , Disease Models, Animal , Age Factors , Animals , Animals, Newborn , Biological Products/therapeutic use , Biopsy , Bronchoalveolar Lavage Fluid/chemistry , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/pathology , Caffeine/therapeutic use , Cesarean Section , Citrates/therapeutic use , Combined Modality Therapy , Continuous Positive Airway Pressure/instrumentation , Drug Combinations , Feasibility Studies , Humans , Infant, Newborn , Intensive Care, Neonatal/methods , Lung/growth & development , Papio , Parenteral Nutrition, Total/methods , Phospholipids/therapeutic use , Pulmonary Gas Exchange , Risk Factors , Time Factors , Ventilator Weaning/methods
12.
Pediatr Res ; 54(6): 797-807, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12930907

ABSTRACT

Current nonhuman models for bronchopulmonary dysplasia have not included perinatal infection. We studied the effects of antenatal Ureaplasma urealyticum (Uu) infection in the 125-d immature baboon. Ten 125-d gestation (term = 185 d) baboon dams were delivered after intra-amniotic inoculation with Uu. Serial blood and tracheal aspirate samples were analyzed for Uu colony-forming units, IL-6, IL-8, and cell counts. Physiologic parameters were serially recorded. Lung histology was examined after 14 d of ventilation and compared with unexposed controls. All Uu-exposed animals had >4 x 102 CFU in tracheal aspirate at 24 h. Four of nine Uu animals remained heavily colonized [(+) Uu] at necropsy (>6 x 103). Five animals had negative or low tracheal colony-forming units. All Uu animals had significant increases for white blood cells, IL-6, and IL-8 in amniotic and fetal lung fluid. Compared with controls, (+) Uu animals had significantly higher fraction of inspired oxygen, airway pressures, oxygenation index, and ventilation efficiency index between 48 and 240 h and had significantly elevated tracheal IL-6 and IL-8 concentrations between 72 and 240 h. Compared with controls (-) Uu animals had significantly better oxygenation index and ventilation efficiency index scores between 48 and 144 h. Lung histopathology in both Uu groups showed more severe bronchiolitis and interstitial pneumonitis compared with controls. Two patterns of disease were observed after Uu perinatal infection. Persistent colonization manifested a picture consistent with acute pneumonitis, worse lung function from 2 to 10 d, and prolonged elevated tracheal cytokines. Colonized animals that subsequently cleared Uu from the lung demonstrated early improved lung function compared with unexposed controls yet still manifested mixed bronchiolitis and interstitial pneumonitis at necropsy. Inherent immune system responses may determine outcome of perinatal Ureaplasma colonization.


Subject(s)
Bronchopulmonary Dysplasia/physiopathology , Ureaplasma Infections/physiopathology , Ureaplasma urealyticum , Age Factors , Amniotic Fluid/microbiology , Animals , Bronchopulmonary Dysplasia/microbiology , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Female , Humans , Infant, Newborn , Interleukin-6/blood , Interleukin-8/blood , Papio , Pregnancy , Respiratory Function Tests , Ureaplasma Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...