Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
1.
Am J Obstet Gynecol ; 223(4): 578.e1-578.e11, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32343954

RÉSUMÉ

BACKGROUND: Ureaplasma parvum infection is a prevalent cause of intrauterine infection associated with preterm birth, preterm premature rupture of membranes, fetal inflammatory response syndrome, and adverse postnatal sequelae. Elucidation of diagnostic and treatment strategies for infection-associated preterm labor may improve perinatal and long-term outcomes for these cases. OBJECTIVE: This study assessed the effect of intraamniotic Ureaplasma infection on fetal hemodynamic and cardiac function and the effect of maternal antibiotic treatment on these outcomes. STUDY DESIGN: Chronically catheterized pregnant rhesus monkeys were assigned to control (n=6), intraamniotic inoculation with Ureaplasma parvum (107 colony-forming units/mL, n=15), and intraamniotic infection plus azithromycin treatment (12.5 mg/kg twice a day intravenously, n=8) groups. At approximately 135 days' gestation (term=165 days), pulsed and color Doppler ultrasonography was used to obtain measurements of fetal hemodynamics (pulsatility index of umbilical artery, ductus venosus, descending aorta, ductus arteriosus, aortic isthmus, right pulmonary artery, middle cerebral artery and cerebroplacental ratio, and left and right ventricular cardiac outputs) and cardiac function (ratio of peak early vs late transmitral flow velocity [marker of ventricular function], Tei index [myocardial performance index]). These indices were stratified by amniotic fluid proinflammatory mediator levels and cardiac histology. RESULTS: Umbilical and fetal pulmonary artery vascular impedances were significantly increased in animals from the intraamniotic inoculation with Ureaplasma parvum group (P<.05). Azithromycin treatment restored values to control levels. Amniotic fluid prostaglandin F2 alpha levels were significantly higher in animals with abnormal umbilical artery pulsatility index (>1.1) than in those with normal blood flow (P<.05; Spearman ρ=0.6, P<.05). In the intraamniotic inoculation with Ureaplasma parvum group, left ventricular cardiac output was significantly decreased (P<.001), and more animals had abnormal right-to-left ventricular cardiac output ratios (defined as >1.6, P<.05). Amniotic fluid interleukin-6 concentrations were elevated in cases of abnormal right-to-left ventricular cardiac output ratios compared with those in normal cases (P<.05). CONCLUSION: Fetal hemodynamic alterations were associated with intraamniotic Ureaplasma infection and ameliorated after maternal antibiotic treatment. Doppler ultrasonographic measurements merit continuing investigation as a diagnostic method to identify fetal cardiovascular and hemodynamic compromise associated with intrauterine infection or inflammation and in the evaluation of therapeutic interventions or clinical management of preterm labor.


Sujet(s)
Antibactériens/usage thérapeutique , Azithromycine/usage thérapeutique , Chorioamnionite/traitement médicamenteux , Coeur foetal/physiopathologie , Hémodynamique/physiologie , Complications infectieuses de la grossesse/traitement médicamenteux , Infections à Ureaplasma/traitement médicamenteux , Administration par voie intraveineuse , Amnios , Liquide amniotique/immunologie , Animaux , Aorte/imagerie diagnostique , Vitesse du flux sanguin , Débit cardiaque/physiologie , Chorioamnionite/immunologie , Chorioamnionite/physiopathologie , Modèles animaux de maladie humaine , Ligament artériel/imagerie diagnostique , Échocardiographie-doppler , Femelle , Injections , Interleukine-6/immunologie , Macaca mulatta , Artère cérébrale moyenne/imagerie diagnostique , Grossesse , Complications infectieuses de la grossesse/immunologie , Complications infectieuses de la grossesse/physiopathologie , Artère pulmonaire/imagerie diagnostique , Écoulement pulsatoire , Échographie-doppler , Échographie prénatale , Artères ombilicales/imagerie diagnostique , Ureaplasma , Infections à Ureaplasma/immunologie , Infections à Ureaplasma/physiopathologie
2.
J Infect Dis ; 209(6): 898-904, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24179112

RÉSUMÉ

BACKGROUND: Our goals were to describe azithromycin (AZI) pharmacokinetics in maternal plasma (MP), fetal plasma (FP), and amniotic fluid (AF) following intra-amniotic infection (IAI) with Ureaplasma in pregnant rhesus monkeys and to explore concentration-response relationships. METHODS: Following intra-amniotic inoculation of Ureaplasma parvum, rhesus monkeys received AZI (12.5 mg/kg every 12 hours intravenously for 10 days; n = 10). Intensive pharmacokinetic sampling of MP, FP, and AF was scheduled following the first (ie, single) dose and the last (ie, multiple) dose. Noncompartmental and pharmacokinetic modeling methods were used. RESULTS: The AF area under the concentration-time curve at 12 hours was 0.22 µg×h/mL following a single dose and 6.3 µg×h/mL at day 10. MP and AF accumulation indices were 8.4 and 19, respectively. AZI AF half-life following the single dose and multiple dose were 156 and 129 hours, respectively. The median MP:FP ratio in concomitantly drawn samples was 3.2 (range, 1.3-9.6; n = 9). Eradication of U. parvum occurred at 6.6 days, with a 95% effective concentration (EC95) of 39 ng/mL for the maximum AZI AF concentration. CONCLUSIONS: Our study demonstrates that a maternal multiple-dose AZI regimen is effective in eradicating U. parvum IAI by virtue of intra-amniotic accumulation and suggests that antenatal therapy has the potential to mitigate complications associated with U. parvum infection in pregnancy, such as preterm labor and fetal sequelae.


Sujet(s)
Antibactériens/pharmacocinétique , Azithromycine/pharmacocinétique , Chorioamnionite/traitement médicamenteux , Complications infectieuses de la grossesse/traitement médicamenteux , Infections à Ureaplasma/traitement médicamenteux , Administration par voie intraveineuse , Liquide amniotique/métabolisme , Liquide amniotique/microbiologie , Animaux , Antibactériens/sang , Antibactériens/usage thérapeutique , Azithromycine/administration et posologie , Azithromycine/sang , Azithromycine/usage thérapeutique , Chorioamnionite/métabolisme , Modèles animaux de maladie humaine , Femelle , Sang foetal/métabolisme , Sang foetal/microbiologie , Macaca mulatta , Grossesse , Complications infectieuses de la grossesse/métabolisme , Infections à Ureaplasma/métabolisme
3.
Am J Obstet Gynecol ; 207(6): 475.e1-475.e14, 2012 Dec.
Article de Anglais | MEDLINE | ID: mdl-23111115

RÉSUMÉ

OBJECTIVE: We assessed the efficacy of a maternal multidose azithromycin (AZI) regimen, with and without antiinflammatory agents to delay preterm birth and to mitigate fetal lung injury associated with Ureaplasma parvum intraamniotic infection. STUDY DESIGN: Long-term catheterized rhesus monkeys (n = 16) received intraamniotic inoculation of U parvum (10(7) colony-forming U/mL, serovar 1). After contraction onset, rhesus monkeys received no treatment (n = 6); AZI (12.5 mg/kg, every 12 h, intravenous for 10 days; n = 5); or AZI plus dexamethasone and indomethacin (n = 5). Outcomes included amniotic fluid proinflammatory mediators, U parvum cultures and polymerase chain reaction, AZI pharmacokinetics, and the extent of fetal lung inflammation. RESULTS: Maternal AZI therapy eradicated U parvum intraamniotic infection from the amniotic fluid within 4 days. Placenta and fetal tissues were 90% culture negative at delivery. AZI therapy significantly delayed preterm delivery and prevented advanced fetal lung injury, although residual acute chorioamnionitis persisted. CONCLUSION: Specific maternal antibiotic therapy can eradicate U parvum from the amniotic fluid and key fetal organs, with subsequent prolongation of pregnancy, which provides a therapeutic window of opportunity to effectively reduce the severity of fetal lung injury.


Sujet(s)
Antibactériens/administration et posologie , Azithromycine/administration et posologie , Chorioamnionite/traitement médicamenteux , Lésion pulmonaire/prévention et contrôle , Naissance prématurée/prévention et contrôle , Infections à Ureaplasma/traitement médicamenteux , Ureaplasma/isolement et purification , Animaux , Anti-inflammatoires/administration et posologie , Anti-inflammatoires non stéroïdiens/administration et posologie , Chorioamnionite/microbiologie , Dexaméthasone/administration et posologie , Association de médicaments , Femelle , Maladies foetales/prévention et contrôle , Indométacine/administration et posologie , Macaca mulatta , Réaction de polymérisation en chaîne , Grossesse , Résultat thérapeutique , Ureaplasma/effets des médicaments et des substances chimiques , Infections à Ureaplasma/microbiologie
4.
Reprod Sci ; 17(1): 85-94, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-19843879

RÉSUMÉ

Causal, cellular, and inflammatory links between choriodecidual infection with group B streptococcus (GBS) and preterm labor were assessed in a nonhuman primate model. Rhesus monkeys received varying doses of a clinical isolate of GBS, type III or saline, via an indwelling catheter placed between the chorion/decidua and myometrium in the lower pole of the uterus. Choriodecidual inoculation of GBS was followed by a graded response in amniotic fluid (AF) leukocytes, proinflammatory cytokines, prostaglandin E(2) and F(2alpha), and uterine activity (P < .05). The magnitude of the inflammatory response in AF was related, in part, to the initial inoculum size and whether AF cultures remained negative or became positive for GBS. Microbial invasion of AF was associated with advanced inflammation and preterm labor. We provide experimental evidence that choriodeciduitis is a transitional stage of intrauterine infection, which may be self-limited, remain dormant, or progress to intraamniotic infection. These data, coupled with clinical observations, suggest that choriodecidual inflammation is an antecedent event in the pathogenesis of premature cervical ripening (functional cervical insufficiency), premature rupture of the fetal membranes, or preterm labor.


Sujet(s)
Chorion/anatomopathologie , Caduques/anatomopathologie , Inflammation/anatomopathologie , Myomètre/anatomopathologie , Travail obstétrical prématuré/anatomopathologie , Liquide amniotique/métabolisme , Analyse de variance , Animaux , Cellules cultivées , Chorion/métabolisme , Cytokines/métabolisme , Dinoprostone/métabolisme , Test ELISA , Femelle , Leucocytes/métabolisme , Leucocytes/anatomopathologie , Macaca mulatta , Myomètre/métabolisme , Travail obstétrical prématuré/métabolisme , Grossesse , Infections à streptocoques/métabolisme , Infections à streptocoques/anatomopathologie , Streptococcus agalactiae/métabolisme
5.
Reprod Sci ; 16(1): 56-70, 2009 Jan.
Article de Anglais | MEDLINE | ID: mdl-19122105

RÉSUMÉ

The authors assess causal, cellular and inflammatory links between intraamniotic infection with Ureaplasma parvum or Mycoplasma hominis and preterm labor in a nonhuman primate model. Long-term catheterized rhesus monkeys received intraamniotic inoculations of clinical isolates of Ureaplasma parvum serovar 1, M hominis, media control or physiological saline. Genital mycoplasmas were quantified in amniotic fluid (AF) and documented in fetal tissues by culture and PCR. In association with elevated AF colony counts for U parvum or M hominis, there was a sequential upregulation of AF leukocytes, proinflammatory cytokines, prostaglandin E2 and F2a, metalloproteinase-9 and uterine activity ( P< .05). Fetal membranes and lung were uniformly positive for both microorganisms; fetal blood and cerebrospinal fluid cultures and PCR were more often positive for M hominis than U parvum. Histopathologic findings of chorioamnionitis, a systemic fetal inflammatory response and pneumonitis worsen with duration of in utero infection. U parvum or M hominis, as sole pathogens, elicit a robust proinflammatory response which contributes to preterm labor and fetal lung injury.


Sujet(s)
Chorioamnionite/microbiologie , Infections à Mycoplasma/microbiologie , Mycoplasma hominis/croissance et développement , Travail obstétrical prématuré/microbiologie , Pneumopathie infectieuse/microbiologie , Infections à Ureaplasma/microbiologie , Ureaplasma/croissance et développement , Liquide amniotique/microbiologie , Animaux , Femelle , Macaca mulatta , Grossesse
6.
Semin Fetal Neonatal Med ; 14(4): 190-9, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19109084

RÉSUMÉ

There is strong evidence from clinical and experimental animal studies that ureaplasmas can invade the amnionic sac and induce an inflammatory response resulting in chorioamnionitis, preterm labor and neonatal lung injury. The ability of Ureaplasma spp. and Mycoplasma hominis to cause pneumonia, bacteremia, and meningitis in newborns can no longer be questioned. The association of Ureaplasma spp. with bronchopulmonary dysplasia has been supported by the majority of observational studies, but proof of causality is still lacking. The availability of molecular diagnostic technologies has enabled the designation of the two Ureaplasma biovars as individual species, but additional work must be done to establish whether there is differential pathogenicity between the Ureaplasma spp. or among their respective serovars. Future investigations to prevent prematurity should be directed toward identification and localization of specific micro-organisms combined with targeted antibiotic trials to determine whether such interventions can improve long-term infant outcomes.


Sujet(s)
Maladies néonatales/microbiologie , Infections à Mycoplasma/diagnostic , Infections opportunistes/diagnostic , Complications infectieuses de la grossesse/microbiologie , Infections à Ureaplasma/diagnostic , Antibactériens/usage thérapeutique , Femelle , Humains , Nouveau-né , Maladies néonatales/traitement médicamenteux , Infections à Mycoplasma/traitement médicamenteux , Mycoplasma hominis/isolement et purification , Infections opportunistes/traitement médicamenteux , Infections opportunistes/microbiologie , Grossesse , Complications infectieuses de la grossesse/diagnostic , Ureaplasma/isolement et purification , Infections à Ureaplasma/traitement médicamenteux
7.
Reprod Sci ; 15(2): 121-7, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18187405

RÉSUMÉ

Intrauterine infection, which occurs in most early preterm births, triggers an immune response culminating in preterm labor. The authors hypothesize that blockade of lipopolysaccharide (LPS)-induced immune responses by a toll-like receptor 4 antagonist (TLR4A) would prevent elevations in amniotic fluid (AF) cytokines, prostaglandins, and uterine contractility. Chronically catheterized rhesus monkeys at 128 to 147 days' gestation received intra-amniotic infusions of either (1) saline (n = 6), (2) LPS (0.15-10 microg; n = 4), or (3) TLR4A pretreatment with LPS (10 microg) 1 hour later (n = 4). AF cytokines, prostaglandins, and uterine contractility were compared using 1-way ANOVA with Bonferroni-adjusted pairwise comparisons. Compared with saline controls, LPS induced significant elevations in AF interleukin-8 (IL-8), tumor necrosis factor (TNF)- alpha, PGE(2), PGF(2)(alpha), and uterine contractility (P < .05). In contrast, TLR4A pretreatment inhibited LPS-induced uterine activity and was associated with significantly lower AF IL-8, TNF-alpha, PGE(2), and PGF(2)( alpha) versus LPS alone (P < .05). Toll-like receptor antagonists, together with antibiotics, may delay or prevent infection-associated preterm birth.


Sujet(s)
Lipopolysaccharides/immunologie , Travail obstétrical prématuré/immunologie , Complications infectieuses de la grossesse/immunologie , Récepteur de type Toll-4/antagonistes et inhibiteurs , Contraction utérine/effets des médicaments et des substances chimiques , Liquide amniotique/microbiologie , Animaux , Chorioamnionite , Cytokines/immunologie , Modèles animaux de maladie humaine , Escherichia coli , Femelle , Lipopolysaccharides/administration et posologie , Macaca mulatta , Travail obstétrical prématuré/prévention et contrôle , Grossesse , Prostaglandines/immunologie , Contraction utérine/immunologie , Maladies de l'utérus/microbiologie
8.
Am J Obstet Gynecol ; 197(5): 518.e1-8, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17980193

RÉSUMÉ

OBJECTIVE: The purpose of this study was to determine whether treatment with ampicillin together with dexamethasone and indomethacin delays preterm birth that is induced by intraamniotic group B Streptococcus in a nonhuman primate model. STUDY DESIGN: After contraction onset that was induced by group B Streptococcus (10(6) colony-forming units/mL), chronically instrumented rhesus macaques received either no treatment (controls; n = 6); ampicillin (n = 4); or ampicillin + dexamethasone + indomethacin (n = 5). Outcomes included the interval from contraction onset until delivery and concentrations of amniotic fluid inflammatory mediators. RESULTS: Mean interval from contraction onset until delivery was 33 +/- 8.7 hours in controls, 82 +/- 28.0 hours with ampicillin (P = .18, vs controls), and 213 +/- 50.8 hours with ampicillin + dexamethasone + indomethacin (P = .004, vs controls). Ampicillin eradicated group B Streptococcus; however, uterine activity, amniotic fluid cytokines, prostaglandins, and matrix metalloproteinase-9 remained elevated. Ampicillin + dexamethasone + indomethacin suppressed interleukin-1beta, tumor necrosis factor-alpha, and prostaglandins E2 and F2alpha but did not alter matrix metalloproteinase expression or chorioamnionitis. CONCLUSION: The combination of ampicillin + dexamethasone + indomethacin suppressed inflammation and significantly prolonged gestation.


Sujet(s)
Ampicilline/usage thérapeutique , Antibactériens/usage thérapeutique , Chorioamnionite , Dexaméthasone/usage thérapeutique , Glucocorticoïdes/usage thérapeutique , Facteurs immunologiques/usage thérapeutique , Indométacine/usage thérapeutique , Travail obstétrical prématuré/prévention et contrôle , Amnios/microbiologie , Liquide amniotique/composition chimique , Liquide amniotique/microbiologie , Animaux , Césarienne , Chorion/microbiologie , Cytokines/analyse , Modèles animaux de maladie humaine , Association de médicaments , Femelle , Macaca mulatta , Matrix metalloproteinase 9/composition chimique , Grossesse
9.
J Proteome Res ; 6(1): 89-96, 2007 Jan.
Article de Anglais | MEDLINE | ID: mdl-17203952

RÉSUMÉ

Intra-amniotic infection (IAI) is associated with preterm birth and perinatal mortality. To identify potential biomarkers, we performed a comprehensive survey of the cervical-vaginal fluid (CVF) proteome from a primate IAI model utilizing multidimensional protein identification technology (LC/LC-MS/MS) and MALDI-TOF-MS analyses. Analyses of CVF proteome identified 205 unique proteins and differential expression of 27 proteins in controls and IAI samples. Protein expression signatures and immunodetection of specific biomarkers identified can be employed for noninvasive detection of IAI.


Sujet(s)
Liquide amniotique/microbiologie , Marqueurs biologiques/composition chimique , Liquides biologiques/métabolisme , Col de l'utérus/métabolisme , Infections/diagnostic , Protéomique/méthodes , Vagin/métabolisme , Animaux , Liquides biologiques/microbiologie , Col de l'utérus/microbiologie , Femelle , Régulation de l'expression des gènes , Macaca mulatta , Grossesse , Gestation animale , Naissance prématurée/prévention et contrôle , Ureaplasma/métabolisme , Vagin/microbiologie
10.
Reprod Sci ; 14(8 Suppl): 42-5, 2007 Dec.
Article de Anglais | MEDLINE | ID: mdl-18089609

RÉSUMÉ

The human fetal membranes are complex tissues that perform many important functions during gestation. The extracellular matrix provides their strength to withstand the forces directed from the fetus and myometrium. Relaxin is a collagenolytic hormone that causes increased production of the matrix metalloproteinases. Its expression from the decidua is increased in patients with preterm premature rupture of the membranes, and its leucine-rich G receptor 7 is upregulated at preterm. The authors previously showed that relaxin is not involved in the infection-mediated cytokine response, but in the absence of infection, it causes increased secretion of both interleukin -6 and interleukin-8 from the membranes. In this article, the authors propose that relaxin is one of a number of sterile stimuli capable of causing increased proinflammatory cytokines, similar to but less robust than the effects of infection. These probably represent distinct inflammatory pathways involving different intracellular signaling events, which can result in either preterm premature rupture of the membranes or preterm labor. The current challenge is to fully understand these pathways and to clarify their similarities and differences.


Sujet(s)
Membranes extraembryonnaires/métabolisme , Relaxine/métabolisme , Transduction du signal , Cytokines/métabolisme , Caduques/métabolisme , Matrice extracellulaire/métabolisme , Femelle , Rupture prématurée des membranes foetales/métabolisme , Humains , Inflammation/métabolisme , Protéines membranaires/métabolisme , Grossesse , Naissance prématurée/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs peptidiques , Résistance à la traction
11.
Am J Obstet Gynecol ; 195(6): 1578-89, 2006 Dec.
Article de Anglais | MEDLINE | ID: mdl-17132473

RÉSUMÉ

OBJECTIVES: The purpose of this study was to determine the relative contributions of individual proinflammatory cytokines and chemokines to the triggering of preterm labor. STUDY DESIGN: Eighteen chronically instrumented pregnant rhesus monkeys at 135 +/- 3 days gestation (term = 167 days) received 1 of 5 intraamniotic infusions: (1) interleukin-1beta (IL-1beta) (10 microg; n = 5), (2) tumor necrosis factor-alpha (TNF-alpha) (10-100 microg; n = 5), (3) IL-6 (20 microg twice a day; n = 2), (4) IL-8 (20 microg twice a day; n = 2), and (5) saline control (n = 4). Primary study outcomes were the mean uterine hourly contraction area (mm Hg x s/h) in 24 hours during peak response to cytokine infusion (all groups) and the interval from cytokine infusion until labor onset (IL-1beta, IL-6, and IL-8 groups). Secondary outcomes were quantities of amniotic fluid cytokines and chemokines (IL-1beta, TNF-alpha, IL-6, and IL-8), prostaglandins E2 and F2alpha, leukocytes, and matrix metalloproteinase-9 (MMP-9). Histopathology of fetal lungs and placental membranes was assessed. RESULTS: IL-1beta stimulated the most intense contraction patterns, resulting in preterm labor in all cases. TNF-alpha induced a variable degree of uterine activity among individual animals stimulating either preterm labor (n = 2) or a uterine contraction pattern of moderate intensity (n = 3). Despite prolonged elevations in amniotic fluid levels, neither IL-6 nor IL-8 induced preterm labor or an increase in uterine activity until near term. The mean interval from the initiation of IL-6 and IL-8 infusion to the onset of labor was significantly longer than after IL-1beta (21.9 vs 1.1 days; P < .01), and did not differ from the saline control group (27.6 days; P = NS). Intraamniotic infusion of IL-1beta or TNF-alpha was associated with significant elevations in all tested amniotic fluid cytokines, IL-8, prostaglandins, MMP-9 and leukocytes compared with gestational age-matched saline controls. IL-6 and IL-8 infusions were not associated with increases in IL-1beta or TNF-alpha and only produced a moderate increase in amniotic fluid prostaglandins. All cytokine infusions induced histologic chorioamnionitis and an accumulation of neutrophils in fetal lungs. CONCLUSION: Preterm labor was induced by intraamniotic infusions of IL-1beta and TNF-alpha, but not by IL-6 or IL-8 although inflammatory changes in fetal membranes and lungs were uniformly present. Our results indicate a primary role for IL-1beta and TNF-alpha in the triggering of preterm labor associated with inflammation or infection.


Sujet(s)
Liquide amniotique , Interleukine-1 bêta/administration et posologie , Travail obstétrical prématuré/induit chimiquement , Facteur de nécrose tumorale alpha/administration et posologie , Liquide amniotique/cytologie , Liquide amniotique/métabolisme , Animaux , Chorioamnionite/induit chimiquement , Cytokines/métabolisme , Membranes extraembryonnaires/effets des médicaments et des substances chimiques , Membranes extraembryonnaires/anatomopathologie , Femelle , Âge gestationnel , Inflammation/induit chimiquement , Inflammation/anatomopathologie , Injections , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Interleukine-6/administration et posologie , Interleukine-6/pharmacologie , Interleukine-8/administration et posologie , Interleukine-8/métabolisme , Interleukine-8/pharmacologie , Poumon/effets des médicaments et des substances chimiques , Poumon/embryologie , Poumon/anatomopathologie , Macaca mulatta , Matrix metalloproteinase 9/métabolisme , Infiltration par les neutrophiles , Grossesse , Prostaglandines/métabolisme , Protéines recombinantes/administration et posologie , Protéines recombinantes/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Contraction utérine , Utérus/effets des médicaments et des substances chimiques
12.
JAMA ; 292(4): 462-9, 2004 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-15280344

RÉSUMÉ

CONTEXT: Intra-amniotic infection (IAI) is commonly associated with preterm birth and adverse neonatal sequelae. Early diagnosis of IAI, however, has been hindered by insensitive or nonspecific tests. OBJECTIVE: To identify unique protein signatures in rhesus monkeys with experimental IAI, a proteomics-based analysis of amniotic fluid was used to develop diagnostic biomarkers for subclinical IAI in amniotic fluid and blood of women with preterm labor. DESIGN, SETTING, AND PARTICIPANTS: Surface-enhanced laser desorption-ionization/time-of-flight mass spectrometry, gel electrophoresis, and tandem mass spectrometry were used to characterize amniotic fluid peptides in 19 chronically instrumented pregnant rhesus monkeys before and after experimental IAI. Candidate biomarkers were determined by liquid chromatography-tandem mass spectrometry. Polyclonal antibodies were generated from synthetic peptides for validation of biomarkers of IAI. Amniotic fluid peptide profiles identified in experimental IAI were subsequently tested in a cohort of 33 women admitted to Seattle, Wash, hospitals between June 25, 1991, and June 30, 1997, with preterm delivery at 35 weeks or earlier associated with subclinical IAI (n = 11), preterm delivery at 35 weeks or earlier without IAI (n = 11), and preterm contractions with subsequent term delivery at later than 35 weeks (n = 11). MAIN OUTCOME MEASURES: Identification of peptide biomarkers for occult IAI. RESULTS: Protein expression profiles in amniotic fluid showed unique signatures of overexpression of polypeptides in the 3- to 5-kDa and 10- to 12-kDa molecular weight ranges in all animals after infection and in no animal prior to infection. In women, the 10- to 12-kDa signature was identified in all 11 patients with subclinical IAI, in 2 of 11 with preterm delivery without IAI, and in 0 of 11 with preterm labor and term delivery without infection (P<.001). Peptide fragment analysis of the diagnostic peak in amniotic fluid identified calgranulin B and a unique fragment of insulinlike growth factor binding protein 1, which were also expressed in maternal serum. Mapping of other amniotic fluid proteins differentially expressed in IAI identified several immunoregulators not previously described in amniotic fluid. CONCLUSIONS: This proteomics-based characterization of the differential expression of amniotic fluid proteins in IAI identified a distinct proteomic profile in an experimental primate chorioamnionitis model that detected subclinical IAI in a human cohort with preterm labor. These diagnostic protein expression signatures, complemented by immunodetection of specific biomarkers in amniotic fluid and in maternal serum, might have application in the early detection of IAI.


Sujet(s)
Liquide amniotique/composition chimique , Marqueurs biologiques/analyse , Chorioamnionite/métabolisme , Travail obstétrical prématuré/métabolisme , Adulte , Liquide amniotique/microbiologie , Animaux , Technique de Western , Calgranuline B/métabolisme , Chorioamnionite/microbiologie , Femelle , Chromatographie gazeuse-spectrométrie de masse , Humains , Infections/métabolisme , Protéine-1 de liaison aux IGF , Protéines de liaison aux IGF/métabolisme , Macaca mulatta , Grossesse , Protéines de la grossesse/métabolisme , Protéome/analyse , Spectrométrie de masse MALDI
13.
Am J Obstet Gynecol ; 190(6): 1669-75; discussion 1675-8, 2004 Jun.
Article de Anglais | MEDLINE | ID: mdl-15284765

RÉSUMÉ

OBJECTIVE: This study was undertaken to evaluate the diagnostic management and the reproductive outcome after surgical repair of a rare reproductive malformation. STUDY DESIGN: Sixteen women with a complete septate uterus, double cervix, and a longitudinal vaginal septum were referred for evaluation. Presenting complaints were chiefly pregnancy loss in parous women (n=9) and dyspareunia in nulligravid women (n=7). The combination of hysterosalpingography, ultrasonography, and/or magnetic resonance imaging was used to correctly identify the anomaly in 15 of the 16 cases. Both hysteroscopic (n=11) and transabdominal (n=5) surgical techniques were used to repair the uterine septum. RESULTS: In no case was the correct diagnosis made before referral; the uterus didelphys was the most common misdiagnosis. The preoperative pregnancy loss was 81%. Postoperatively, 12 women conceived for a total of 17 pregnancies; there were 14 term live births or ongoing pregnancies in the third trimester (82%), with a first trimester spontaneous abortion rate of 18%. In 9 women who conceived after hysteroscopic surgery, term live births occurred in 9 of 12 (75%) conceptions. A modified Tompkins metroplasty was performed in 5 women with subsequent term live births or ongoing third trimester pregnancies in 5 of 5 (100%) patients. CONCLUSION: The identification of a duplicated cervix and a vaginal septum is consistent with several uterine malformations, which leads to frequent misdiagnosis and errors in management. Significant pregnancy wastage, obstetric complications, and dyspareunia are common, and surgical treatment is therefore advisable. Making the best choice between hysteroscopic or transabdominal metroplasty depends on the anatomic features of the cervix and the uterine cavity, but optimal patient management requires familiarity with both techniques.


Sujet(s)
Col de l'utérus/malformations , Malformations/chirurgie , Hystéroscopie/méthodes , Grossesse/statistiques et données numériques , Utérus/malformations , Vagin/malformations , Malformations multiples/diagnostic , Malformations multiples/chirurgie , Adolescent , Adulte , Col de l'utérus/chirurgie , Études de cohortes , Malformations/diagnostic , Femelle , Études de suivi , Procédures de chirurgie gynécologique/méthodes , Humains , Hystérosalpingographie/méthodes , Infertilité féminine/prévention et contrôle , Imagerie par résonance magnétique , Antécédents gynécologiques et obstétricaux , Appréciation des risques , Utérus/chirurgie , Vagin/chirurgie
14.
Am J Obstet Gynecol ; 188(1): 252-63, 2003 Jan.
Article de Anglais | MEDLINE | ID: mdl-12548226

RÉSUMÉ

OBJECTIVE: The purpose of this study was to determine whether treatment with the immune modulators dexamethasone or interleukin-10 prevents interleukin-1beta-induced uterine contractions in a nonhuman primate model. STUDY DESIGN: Thirteen chronically instrumented rhesus monkeys at 135 +/- 1 days of gestation (term, 167 days) received one of three interventions: (1) intra-amniotic interleukin-1beta (10 microg) infusion with maternal dexamethasone (1 mg/kg) intravenously every 6 hours for 1 day before interleukin-1beta and for 2 days thereafter (n = 4), (2) intra-amniotic interleukin-1beta infusion with maternal interleukin-10 (25 microg/kg) given intravenously and 100 microg interleukin-10 given intra-amniotically before the interleukin-1beta and continued every 8 hours for 3 days (n = 5), and (3) intra-amniotic interleukin-1beta administered alone (n = 5). Uterine activity was monitored continuously and quantified as the hourly contraction area (millimeters of mercury times seconds per hour) in all groups until delivery. Amniotic fluid was sampled for leukocyte counts and assayed for prostaglandins E(2) and F(2)alpha, cytokines interleukin-1beta, interleukin-6, interleukin-8, tumor necrosis factor-alpha, interleukin-10, and interleukin-1 receptor antagonist by specific assays. Maternal and fetal blood were assayed for cortisol, dehydroepiandrosterone sulfate, and estradiol. RESULTS: Interleukin-1beta infusion in the absence of immune modulators resulted in an increase in uterine activity and amniotic fluid proinflammatory cytokines, prostaglandins, and leukocytes. Dexamethasone and interleukin-10 treatment significantly reduced interleukin-1beta-induced uterine contractility (P <.05) and amniotic fluid prostaglandins (P <.05) but not interleukin-8 or interleukin-1 receptor antagonist. Amniotic fluid interleukin-6 and maternal and fetal cortisol, dehydroepiandrosterone sulfate, and estradiol concentrations were reduced by dexamethasone (P <.05), whereas tumor necrosis factor-alpha levels and leukocyte counts were attenuated by interleukin-10 treatment (P <.05). An inverse relationship was noted between amniotic fluid interleukin-10 concentrations and interleukin-1beta-induced uterine activity (r = -0.74, P <.05). CONCLUSION: Dexamethasone and interleukin-10 exert similar inhibitory effects on interleukin-1beta-induced uterine activity, which appears to be mediated by a decrease in prostaglandin production. Reduced estrogen biosynthesis or suppression of tumor necrosis factor-alpha and leukocyte migration may contribute to the tocolytic actions of dexamethasone and interleukin-10, respectively. Dexamethasone and interleukin-10 are likely to be useful adjuncts in the treatment of preterm labor that is associated with inflammation or infection.


Sujet(s)
Dexaméthasone/pharmacologie , Glucocorticoïdes/pharmacologie , Interleukine-10/pharmacologie , Interleukine-1/pharmacologie , Contraction utérine/effets des médicaments et des substances chimiques , Liquide amniotique/composition chimique , Liquide amniotique/cytologie , Animaux , Cytokines/analyse , Dinoprost/analyse , Dinoprostone/analyse , Interactions médicamenteuses , Femelle , Âge gestationnel , Rythme cardiaque foetal , Antagoniste du récepteur à l'interleukine-1 , Interleukine-6/analyse , Interleukine-8/analyse , Numération des leucocytes , Macaca mulatta , Grossesse , Prostaglandines/analyse , Sialoglycoprotéines/analyse , Facteur de nécrose tumorale alpha/analyse
15.
Am J Obstet Gynecol ; 187(5): 1426-7; author reply 1427-8, 2002 Nov.
Article de Anglais | MEDLINE | ID: mdl-12439545
16.
Am J Obstet Gynecol ; 186(1): 128-38, 2002 Jan.
Article de Anglais | MEDLINE | ID: mdl-11810098

RÉSUMÉ

OBJECTIVE: The purpose of this study was to examine the roles of intrauterine infection, inflammation, and spontaneous labor on the expression of matrix metalloproteinase-9 in fetal membranes and amniotic fluid of late pregnant rhesus monkeys. STUDY DESIGN: Pregnant rhesus monkeys with timed gestations were chronically catheterized to allow serial sampling of amniotic fluid before and during experimentally induced intrauterine inflammation. Six animals received group B streptococci into the chorionic-decidual space, and 4 animals received intra-amniotic interleukin-1 beta infusions (10 microg). Three additional animals were serially sampled by amniocentesis through late pregnancy until spontaneous term labor. Amniotic fluid samples were examined by zymography for matrix metalloproteinase-9 and -2 and Western immunoblot for matrix metalloproteinase-9 and -2 and tissue inhibitors of metalloproteinase-1 and -2. Fetal membranes were obtained at cesarean delivery during labor (before rupture), formalin fixed, and embedded in paraffin for immunocytochemistry of matrix metalloproteinase-9 and in situ hybridization of matrix metalloproteinase-9 messenger RNA. Tissues from 2 additional animals were collected as age-matched non-labor controls. RESULTS: In amniotic fluid, the 92-kd latent matrix metalloproteinase-9 was detectable in late pregnancy and increased dramatically, followed by the appearance of the 83-kd active form before spontaneous term delivery. Amniotic fluid matrix metalloproteinase-2 levels (both latent and active forms) remained relatively constant throughout pregnancy and in labor. Both bacteria and interleukin-1 beta rapidly increased the signal of latent matrix metalloproteinase-9 without a consistent increase in the active form before the onset of labor. Chorionic-decidual inoculation of group B streptococci was followed by the expression of latent matrix metalloproteinase-9 before the appearance of group B streptococci in amniotic fluid or the onset of labor. Matrix metalloproteinase-2 increased to a new steady-state level or remained unchanged after group B streptococci inoculation or interleukin-1 beta infusion, respectively. Amniotic fluid tissue inhibitors of metalloproteinase-1 declined and tissue inhibitors of metalloproteinase-2 remained unchanged during early group B streptococci infection, after interleukin-1 beta infusion and on the day of spontaneous term labor. However, both tissue inhibitors of metalloproteinase-1 and -2 levels increased after preterm labor that was induced by group B streptococci. Immunocytochemistry localized matrix metalloproteinase-9 protein to amnion and chorion epithelial and mesenchymal cells and decidual stromal cells. Granular matrix metalloproteinase-9 staining was observed in the connective tissue layer of inflamed fetal membranes. In situ hybridization for messenger RNA confirmed the production of matrix metalloproteinase-9 by amnion and chorion. CONCLUSION: Bacterial- and interleukin-1 beta-induced preterm labor and spontaneous term labor are preceded and accompanied by progressive increases in amniotic fluid matrix metalloproteinase-9 (92 kd) in rhesus monkeys. Amniotic fluid matrix metalloproteinase-9 may serve as a clinical marker for the onset of both preterm and term labor.


Sujet(s)
Liquide amniotique/métabolisme , Membranes extraembryonnaires/métabolisme , Interleukine-1/pharmacologie , Travail obstétrical/métabolisme , Matrix metalloproteinase 9/métabolisme , Gestation animale/métabolisme , Infections à streptocoques/métabolisme , Maladies de l'utérus/métabolisme , Amnios/métabolisme , Amnios/microbiologie , Animaux , Chorion/métabolisme , Chorion/microbiologie , Accouchement (procédure) , Membranes extraembryonnaires/microbiologie , Femelle , Immunohistochimie , Hybridation in situ , Macaca mulatta , Grossesse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...