56 results on '"Wolfs TG"'
Search Results
2. Antenatal Ureaplasma infection induces ovine small intestinal goblet cell defects: a strong link with NEC pathology.
- Author
-
van Gorp C, de Lange IH, Hütten MC, López-Iglesias C, Massy KR, Kessels L, Kramer B, van de Wetering W, Spiller B, Birchenough GM, van Gemert WG, Zimmermann LJ, and Wolfs TG
- Subjects
- Humans, Pregnancy, Animals, Sheep, Female, Goblet Cells pathology, Intestinal Mucosa, Mucus, Chorioamnionitis pathology, Ureaplasma Infections complications, Ureaplasma Infections pathology
- Abstract
Disruption of the intestinal mucus barrier and intestinal epithelial endoplasmic reticulum (ER) stress contribute to necrotizing enterocolitis (NEC). Previously, we observed intestinal goblet cell loss and increased intestinal epithelial ER stress following chorioamnionitis. Here, we investigated how chorioamnionitis affects goblet cells by assessing their cellular characteristics. Importantly, goblet cell features are compared with those in clinical NEC biopsies. Mucus thickness was assessed as read-out of goblet cell function. Fetal lambs were intra-amniotically (IA) infected for 7d at 122 gestational age with Ureaplasma parvum serovar-3 , the main microorganism clinically associated with chorioamnionitis. After preterm delivery, mucus thickness, goblet cell numbers, gut inflammation, epithelial proliferation and apoptosis and intestinal epithelial ER stress were investigated in the terminal ileum. Next, goblet cell morphological alterations (TEM) were studied and compared to human NEC samples. Ileal mucus thickness and goblet cell numbers were elevated following IA UP exposure. Increased pro-apoptotic ER stress, detected by elevated CHOP-positive cell counts and disrupted organelle morphology of secretory cells in the intestinal epithelium, was observed in IA UP exposed animals. Importantly, comparable cellular morphological alterations were observed in the ileum from NEC patients. In conclusion, UP-driven chorioamnionitis leads to a thickened ileal mucus layer and mucus hypersecretion from goblet cells. Since this was associated with pro-apoptotic ER stress and organelle disruption, mucus barrier alterations seem to occur at the expense of goblet cell resilience and may therefore predispose to detrimental intestinal outcomes. The remarkable overlap of these in utero findings with observations in NEC patients underscores their clinical relevance.
- Published
- 2023
- Full Text
- View/download PDF
3. Polymer film-based microwell array platform for long-term culture and research of human bronchial organoids.
- Author
-
Baptista D, Tahmasebi Birgani Z, Widowski H, Passanha F, Stylianidis V, Knoops K, Gubbins E, Iriondo C, Skarp KP, Rottier RJ, Wolfs TG, van Blitterswijk C, LaPointe V, Habibović P, Reynaert NL, Giselbrecht S, and Truckenmüller R
- Abstract
The culture of lung organoids relies on drops of basement membrane matrices. This comes with limitations, for example, concerning the microscopic monitoring and imaging of the organoids in the drops. Also, the culture technique is not easily compatible with micromanipulations of the organoids. In this study, we investigated the feasibility of the culture of human bronchial organoids in defined x-, y- and z-positions in a polymer film-based microwell array platform. The circular microwells have thin round/U-bottoms. For this, single cells are first precultured in drops of basement membrane extract (BME). After they form cell clusters or premature organoids, the preformed structures are then transferred into the microwells in a solution of 50% BME in medium. There, the structures can be cultured toward differentiated and mature organoids for several weeks. The organoids were characterized by bright-field microscopy for size growth and luminal fusion over time, by scanning electron microscopy for overall morphology, by transmission electron microscopy for the existence of microvilli and cilia, by video microscopy for beating cilia and swirling fluid, by live-cell imaging, by fluorescence microscopy for the expression of cell-specific markers and for proliferating and apoptotic cells, and by ATP measurement for extended cell viability. Finally, we demonstrated the eased micromanipulation of the organoids in the microwells by the example of their microinjection., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:Roman Truckenmueller reports a relationship with 300MICRONS GmbH that includes: board membership and equity or stocks. Stefan Giselbrecht reports a relationship with 300MICRONS GmbH that includes: board membership and equity or stocks., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
4. Why -aVF can be used in STAN as a proxy for scalp electrode-derived signal; reply to comments by Kjellmer et al.
- Author
-
Delhaas T, Andriessen P, van Laar JO, Vullings R, Hermans BJ, Niemarkt HJ, Jellema RK, Ophelders DR, Wolfs TG, Kramer BW, and Zwanenburg A
- Subjects
- Animals, Electrodes, Female, Fetus, Humans, Hypoxia, Pregnancy, Sheep, Umbilical Cord, Electrocardiography, Scalp
- Abstract
The conclusion of our recent paper that performance of the STAN device in clinical practice is potentially limited by high false-negative and high false-positive STAN-event rates and loss of ST waveform assessment capacity during severe hypoxemia, evoked comments by Kjellmer, Lindecrantz and Rosén. These comments can be summarized as follows: 1) STAN analysis is based on a unipolar lead but the authors used a negative aVF lead, and they did not validate this methodology; 2) The fetuses used in the study were too young to display the signals that the authors were trying to detect. In response to these comments we now provide both a theoretical and an experimental underpinning of our approach. In an in vivo experiment in human we placed several electrodes over the head (simulating different places of a scalp electrode), simultaneously recorded Einthoven lead I and II, and constructed -aVF from these two frontal leads. Irrespective of scalp electrode placement, the correlation between any of unipolar scalp electrode-derived signals and constructed-aVF was excellent (≥ 0.92). In response to the second comment we refer to a study which demonstrated that umbilical cord occlusion resulted in rapid increase in T/QRS ratio that coincided with initial hypertension and bradycardia at all gestational ages which were tested from 0.6-0.8 gestation. The animals of our study were in this gestational range and, hence, our experimental setup can be used to assess STAN's quality to detect fetal hypoxia. In conclusion, we have clearly demonstrated the appropriateness of using-aVF as a proxy for a scalp electrode-derived signal in STAN in these preterm lambs. Investigation why STAN could not detect relevant ST-changes and instead produced erroneous alarms in our experimental setup is hampered by the fact that the exact STAN algorithm (signal processing and analysis) is not in the public domain., Competing Interests: The authors reconfirm and declare that no competing interests exist. Although Rik Vullings is scientific director and shareholder of Nemo Healthcare (Veldhoven, the Netherlands), a company that develops innovative technology for improved pregnancy monitoring, this does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2019
- Full Text
- View/download PDF
5. Plant-based sterols and stanols in health & disease: "Consequences of human development in a plant-based environment?"
- Author
-
Plat J, Baumgartner S, Vanmierlo T, Lütjohann D, Calkins KL, Burrin DG, Guthrie G, Thijs C, Te Velde AA, Vreugdenhil ACE, Sverdlov R, Garssen J, Wouters K, Trautwein EA, Wolfs TG, van Gorp C, Mulder MT, Riksen NP, Groen AK, and Mensink RP
- Subjects
- Asthma metabolism, Cardiovascular Diseases metabolism, Cholesterol metabolism, Cholesterol, LDL antagonists & inhibitors, Cholesterol, LDL metabolism, Humans, Inflammatory Bowel Diseases metabolism, Intestinal Absorption drug effects, Non-alcoholic Fatty Liver Disease metabolism, Phytosterols administration & dosage, Sitosterols administration & dosage, Asthma drug therapy, Cardiovascular Diseases drug therapy, Inflammatory Bowel Diseases drug therapy, Non-alcoholic Fatty Liver Disease drug therapy, Phytosterols pharmacology, Sitosterols pharmacology
- Abstract
Dietary plant sterols and stanols as present in our diet and in functional foods are well-known for their inhibitory effects on intestinal cholesterol absorption, which translates into lower low-density lipoprotein cholesterol concentrations. However, emerging evidence suggests that plant sterols and stanols have numerous additional health effects, which are largely unnoticed in the current scientific literature. Therefore, in this review we pose the intriguing question "What would have occurred if plant sterols and stanols had been discovered and embraced by disciplines such as immunology, hepatology, pulmonology or gastroenterology before being positioned as cholesterol-lowering molecules?" What would then have been the main benefits and fields of application of plant sterols and stanols today? We here discuss potential effects ranging from its presence and function intrauterine and in breast milk towards a potential role in the development of non-alcoholic steatohepatitis (NASH), cardiovascular disease (CVD), inflammatory bowel diseases (IBD) and allergic asthma. Interestingly, effects clearly depend on the route of entrance as observed in intestinal-failure associated liver disease (IFALD) during parenteral nutrition regimens. It is only until recently that effects beyond lowering of cholesterol concentrations are being explored systematically. Thus, there is a clear need to understand the full health effects of plant sterols and stanols., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
6. Prenatal Intra-Amniotic Endotoxin Induces Fetal Gut and Lung Immune Responses and Postnatal Systemic Inflammation in Preterm Pigs.
- Author
-
Nguyen DN, Thymann T, Goericke-Pesch SK, Ren S, Wei W, Skovgaard K, Damborg P, Brunse A, van Gorp C, Kramer BW, Wolfs TG, and Sangild PT
- Subjects
- Animals, Animals, Newborn, Chorioamnionitis chemically induced, Chorioamnionitis pathology, Female, Fetus drug effects, Fetus pathology, Gastrointestinal Tract drug effects, Gastrointestinal Tract pathology, Inflammation chemically induced, Inflammation pathology, Lung drug effects, Lung pathology, Pregnancy, Premature Birth, Swine, Chorioamnionitis immunology, Endotoxins toxicity, Fetus immunology, Gastrointestinal Tract immunology, Inflammation immunology, Lung immunology
- Abstract
Prenatal inflammation is a major risk for preterm birth and neonatal morbidity, but its effects on postnatal immunity and organ functions remain unclear. Using preterm pigs as a model for preterm infants, we investigated whether prenatal intra-amniotic (IA) inflammation modulates postnatal systemic immune status and organ functions. Preterm pigs exposed to IA lipopolysaccharide (LPS) for 3 days were compared with controls at birth and postnatal day 5 after formula feeding. IA LPS induced mild chorioamnionitis but extensive intra-amniotic inflammation. There were minor systemic effects at birth (increased blood neutrophil counts), but a few days later, prenatal LPS induced delayed neonatal arousal, systemic inflammation (increased blood leukocytes, plasma cytokines, and splenic bacterial counts), altered serum biochemistry (lower albumin and cholesterol and higher iron and glucose values), and increased urinary protein and sodium excretion. In the gut and lungs, IA LPS-induced inflammatory responses were observed mainly at birth (increased LPS, CXCL8, and IL-1β levels and myeloperoxidase-positive cell density, multiple increases in innate immune gene expressions, and reduced villus heights), but not on postnatal day 5 (except elevated lung CXCL8 and diarrhea symptoms). Finally, IA LPS did not affect postnatal gut brush-border enzymes, hexose absorption, permeability, or sensitivity to necrotizing enterocolitis on day 5. Short-term IA LPS exposure predisposes preterm pigs to postnatal systemic inflammation after acute fetal gut and lung inflammatory responses., (Copyright © 2018 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
7. Mesenchymal stem/stromal cells-a key mediator for regeneration after perinatal morbidity?
- Author
-
Mueller M, Wolfs TG, Schoeberlein A, Gavilanes AW, Surbek D, and Kramer BW
- Abstract
Perinatal complications in both term- and preterm-born infants are a leading cause of neonatal morbidities and mortality. Infants face different challenges in the neonatal intensive care unit with long-term morbidities such as perinatal brain injury and bronchopulmonary dysplasia being particularly devastating. While advances in perinatal medicine have improved our understanding of the pathogenesis, effective therapies to prevent and/or reduce the severity of these disorders are still lacking. The potential of mesenchymal stem/stromal cell (MSC) therapy has emerged during the last two decades, and an increasing effort is conducted to address brain- and lung-related morbidities in neonates at risk. Various studies support the notion that MSCs have protective effects. MSCs are an easy source and may be readily available after birth in a clinical setting. MSCs' mechanisms of action are diverse, including migration and homing, release of growth factors and immunomodulation, and the potential to replace injured cells. Here, we review the pathophysiology of perinatally acquired brain and lung injuries and focus on MSCs as potential candidates for therapeutic strategies summarizing preclinical and clinical evidence.
- Published
- 2016
- Full Text
- View/download PDF
8. Can the preterm lung recover from perinatal stress?
- Author
-
Hütten MC, Wolfs TG, and Kramer BW
- Abstract
After birth, adequate lung function is necessary for the successful adaptation of a preterm baby. Both prenatal and postnatal insults and therapeutic interventions have an immediate effect on lung function and gas exchange but also interfere with fetal and neonatal lung development. Prenatal insults like chorioamnionitis and prenatal interventions like maternal glucocorticosteroids interact but might also determine the preterm baby's lung response to postnatal interventions ("second hit") like supplementation of oxygen and drug therapy. We review current experimental and clinical findings on the influence of different perinatal factors on preterm lung development and discuss how well-established interventions in neonatal care might be adapted to attenuate postnatal lung injury.
- Published
- 2016
- Full Text
- View/download PDF
9. Intra-amniotic Candida albicans infection induces mucosal injury and inflammation in the ovine fetal intestine.
- Author
-
Nikiforou M, Jacobs EM, Kemp MW, Hornef MW, Payne MS, Saito M, Newnham JP, Janssen LE, Jobe AH, Kallapur SG, Kramer BW, and Wolfs TG
- Subjects
- Animals, Antifungal Agents pharmacology, Candida albicans drug effects, Candidiasis drug therapy, Candidiasis microbiology, Chorioamnionitis drug therapy, Chorioamnionitis microbiology, Cytokines genetics, Cytokines metabolism, Female, Fetus metabolism, Fetus microbiology, Fetus pathology, Fluconazole pharmacology, Host-Pathogen Interactions, Inflammation microbiology, Intestinal Mucosa embryology, Intestinal Mucosa pathology, Pregnancy, Sheep, Sheep Diseases pathology, Candida albicans physiology, Candidiasis veterinary, Chorioamnionitis veterinary, Inflammation veterinary, Intestinal Mucosa microbiology, Sheep Diseases microbiology
- Abstract
Chorioamnionitis is caused by intrauterine infection with microorganisms including Candida albicans (C.albicans). Chorioamnionitis is associated with postnatal intestinal pathologies including necrotizing enterocolitis. The underlying mechanisms by which intra-amniotic C.albicans infection adversely affects the fetal gut remain unknown. Therefore, we assessed whether intra-amniotic C.albicans infection would cause intestinal inflammation and mucosal injury in an ovine model. Additionally, we tested whether treatment with the fungistatic fluconazole ameliorated the adverse intestinal outcome of intra-amniotic C.albicans infection. Pregnant sheep received intra-amniotic injections with 10(7) colony-forming units C.albicans or saline at 3 or 5 days before preterm delivery at 122 days of gestation. Fetuses were given intra-amniotic and intra-peritoneal fluconazole treatments 2 days after intra-amniotic administration of C.albicans. Intra-amniotic C.albicans caused intestinal colonization and invasive growth within the fetal gut with mucosal injury and intestinal inflammation, characterized by increased CD3(+) lymphocytes, MPO(+) cells and elevated TNF-α and IL-17 mRNA levels. Fluconazole treatment in utero decreased intestinal C.albicans colonization, mucosal injury but failed to attenuate intestinal inflammation. Intra-amniotic C.albicans caused intestinal infection, injury and inflammation. Fluconazole treatment decreased mucosal injury but failed to ameliorate C.albicans-mediated mucosal inflammation emphasizing the need to optimize the applied antifungal therapeutic strategy.
- Published
- 2016
- Full Text
- View/download PDF
10. Comparison of ECG-based physiological markers for hypoxia in a preterm ovine model.
- Author
-
Zwanenburg A, Hermans BJ, Andriessen P, Niemarkt HJ, Jellema RK, Ophelders DR, Vullings R, Wolfs TG, Kramer BW, and Delhaas T
- Subjects
- Animals, Animals, Newborn, Disease Models, Animal, Female, Heart Rate, Hydrogen-Ion Concentration, Male, ROC Curve, Sensitivity and Specificity, Sheep, Time Factors, Electrocardiography, Hypoxia diagnosis, Ischemia diagnosis
- Abstract
Background: Current methods for assessing perinatal hypoxic conditions did not improve infant outcomes. Various waveform-based and interval-based ECG markers have been suggested, but not directly compared. We compare performance of ECG markers in a standardized ovine model for fetal hypoxia., Methods: Sixty-nine fetal sheep of 0.7 gestation had ECG recorded 4 h before, during, and 4 h after a 25-min period of umbilical cord occlusion (UCO), leading to severe hypoxia. Various ECG markers were calculated, among which were heart rate (HR), HR-corrected ventricular depolarization/repolarization interval (QTc), and ST-segment analysis (STAN) episodic and baseline rise markers, analogue to clinical STAN device alarms. Performance of interval- and waveform-based ECG markers was assessed by correlating predicted and actual hypoxic/normoxic state., Results: Of the markers studied, HR and QTc demonstrated high sensitivity (≥86%), specificity (≥96%), and positive predictive value (PPV) (≥86%) and detected hypoxia in ≥90% of fetuses at 4 min after UCO. In contrast, STAN episodic and baseline rise markers displayed low sensitivity (≤20%) and could not detect severe fetal hypoxia in 65 and 28% of the animals, respectively., Conclusion: Interval-based HR and QTc markers could assess the presence of severe hypoxia. Waveform-based STAN episodic and baseline rise markers were ineffective as markers for hypoxia.
- Published
- 2016
- Full Text
- View/download PDF
11. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Protect the Fetal Brain After Hypoxia-Ischemia.
- Author
-
Ophelders DR, Wolfs TG, Jellema RK, Zwanenburg A, Andriessen P, Delhaas T, Ludwig AK, Radtke S, Peters V, Janssen L, Giebel B, and Kramer BW
- Subjects
- Animals, Brain physiopathology, Brain Injuries physiopathology, Brain Injuries therapy, Cell Proliferation, Disease Models, Animal, Extracellular Vesicles metabolism, Fetus, Humans, Hypoxia-Ischemia, Brain physiopathology, Inflammation physiopathology, Mesenchymal Stem Cells cytology, Sheep, Cell- and Tissue-Based Therapy, Hypoxia-Ischemia, Brain therapy, Inflammation therapy, Mesenchymal Stem Cells metabolism
- Abstract
Unlabelled: Preterm neonates are susceptible to perinatal hypoxic-ischemic brain injury, for which no treatment is available. In a preclinical animal model of hypoxic-ischemic brain injury in ovine fetuses, we have demonstrated the neuroprotective potential of systemically administered mesenchymal stromal cells (MSCs). The mechanism of MSC treatment is unclear but suggested to be paracrine, through secretion of extracellular vesicles (EVs). Therefore, we investigated in this study the protective effects of mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) in a preclinical model of preterm hypoxic-ischemic brain injury. Ovine fetuses were subjected to global hypoxia-ischemia by transient umbilical cord occlusion, followed by in utero intravenous administration of MSC-EVs. The therapeutic effects of MSC-EV administration were assessed by analysis of electrophysiological parameters and histology of the brain. Systemic administration of MSC-EVs improved brain function by reducing the total number and duration of seizures, and by preserving baroreceptor reflex sensitivity. These functional protections were accompanied by a tendency to prevent hypomyelination. Cerebral inflammation remained unaffected by the MSC-EV treatment. Our data demonstrate that MSC-EV treatment might provide a novel strategy to reduce the neurological sequelae following hypoxic-ischemic injury of the preterm brain. Our study results suggest that a cell-free preparation comprising neuroprotective MSC-EVs could substitute MSCs in the treatment of preterm neonates with hypoxic-ischemic brain injury, thereby circumventing the potential risks of systemic administration of living cells., Significance: Bone marrow-derived mesenchymal stromal cells (MSCs) show promise in treating hypoxic-ischemic injury of the preterm brain. Study results suggest administration of extracellular vesicles, rather than intact MSCs, is sufficient to exert therapeutic effects and avoids potential concerns associated with administration of living cells. The therapeutic efficacy of systemically administered mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) on hypoxia-ischemia-induced injury was assessed in the preterm ovine brain. Impaired function and structural injury of the fetal brain was improved following global hypoxia-ischemia. A cell-free preparation of MSC-EVs could substitute for the cellular counterpart in the treatment of preterm neonates with hypoxic-ischemic brain injury. This may open new clinical applications for "off-the-shelf" interventions with MSC-EVs., (©AlphaMed Press.)
- Published
- 2016
- Full Text
- View/download PDF
12. Neuroinflammation and structural injury of the fetal ovine brain following intra-amniotic Candida albicans exposure.
- Author
-
Ophelders DR, Gussenhoven R, Lammens M, Küsters B, Kemp MW, Newnham JP, Payne MS, Kallapur SG, Jobe AH, Zimmermann LJ, Kramer BW, and Wolfs TG
- Subjects
- Animals, Brain Injuries microbiology, Brain Injuries pathology, Calcium-Binding Proteins, Caspase 3 metabolism, DNA-Binding Proteins metabolism, Disease Models, Animal, Encephalitis microbiology, Encephalitis pathology, Enzyme-Linked Immunosorbent Assay, Female, Fluoresceins metabolism, Granulocyte Colony-Stimulating Factor metabolism, Interleukin-3 metabolism, Interleukin-6 metabolism, Ki-67 Antigen metabolism, Male, Microfilament Proteins, Myelin Basic Protein metabolism, Nerve Tissue Proteins metabolism, Pregnancy, Recombinant Fusion Proteins metabolism, Sheep, Brain Injuries etiology, Candida albicans pathogenicity, Candidiasis complications, Encephalitis etiology, Prenatal Exposure Delayed Effects physiopathology
- Abstract
Background: Intra-amniotic Candida albicans (C. Albicans) infection is associated with preterm birth and high morbidity and mortality rates. Survivors are prone to adverse neurodevelopmental outcomes. The mechanisms leading to these adverse neonatal brain outcomes remain largely unknown. To better understand the mechanisms underlying C. albicans-induced fetal brain injury, we studied immunological responses and structural changes of the fetal brain in a well-established translational ovine model of intra-amniotic C. albicans infection. In addition, we tested whether these potential adverse outcomes of the fetal brain were improved in utero by antifungal treatment with fluconazole., Methods: Pregnant ewes received an intra-amniotic injection of 10(7) colony-forming units C. albicans or saline (controls) at 3 or 5 days before preterm delivery at 0.8 of gestation (term ~ 150 days). Fetal intra-amniotic/intra-peritoneal injections of fluconazole or saline (controls) were administered 2 days after C. albicans exposure. Post mortem analyses for fungal burden, peripheral immune activation, neuroinflammation, and white matter/neuronal injury were performed to determine the effects of intra-amniotic C. albicans and fluconazole treatment., Results: Intra-amniotic exposure to C. albicans caused a severe systemic inflammatory response, illustrated by a robust increase of plasma interleukin-6 concentrations. Cerebrospinal fluid cultures were positive for C. albicans in the majority of the 3-day C. albicans-exposed animals whereas no positive cultures were present in the 5-day C. albicans-exposed and fluconazole-treated animals. Although C. albicans was not detected in the brain parenchyma, a neuroinflammatory response in the hippocampus and white matter was seen which was characterized by increased microglial and astrocyte activation. These neuroinflammatory changes were accompanied by structural white matter injury. Intra-amniotic fluconazole reduced fetal mortality but did not attenuate neuroinflammation and white matter injury., Conclusions: Intra-amniotic C. albicans exposure provoked acute systemic and neuroinflammatory responses with concomitant white matter injury. Fluconazole treatment prevented systemic inflammation without attenuating cerebral inflammation and injury.
- Published
- 2016
- Full Text
- View/download PDF
13. Systemic interleukin-2 administration improves lung function and modulates chorioamnionitis-induced pulmonary inflammation in the ovine fetus.
- Author
-
Willems MG, Ophelders DR, Nikiforou M, Jellema RK, Butz A, Delhaas T, Kramer BW, and Wolfs TG
- Subjects
- Animals, Female, Gestational Age, Lipopolysaccharides pharmacology, Pneumonia complications, Pneumonia immunology, Pregnancy, Sheep, Chorioamnionitis drug therapy, Fetus drug effects, Interleukin-2 pharmacology, Pneumonia drug therapy
- Abstract
Chorioamnionitis, an inflammatory reaction of the fetal membranes to microbes, is an important cause of preterm birth and associated with inflammation-driven lung injury. However, inflammation in utero overcomes immaturity of the premature lung by inducing surfactant lipids and lung gas volume. Previously, we found that lipopolysaccharide (LPS)-induced chorioamnionitis resulted in pulmonary inflammation with increased effector T cells and decreased regulatory T cell (Treg) numbers. Because Tregs are crucial for immune regulation, we assessed the effects of interleukin (IL)-2-driven selective Treg expansion on the fetal lung in an ovine chorioamnionitis model. Instrumented fetuses received systemic prophylactic IL-2 treatment [118 days gestational age (dGA)] with or without subsequent exposure to intra-amniotic LPS (122 dGA). Following delivery at 129 dGA (term 147 dGA), pulmonary and systemic inflammation, morphological changes, lung gas volume, and phospholipid concentration were assessed. IL-2 pretreatment increased the FoxP3(+)/CD3(+) ratio, which was associated with reduced CD3-positive cells in the fetal lungs of LPS-exposed animals. Prophylactic IL-2 treatment did not prevent pulmonary accumulation of myeloperoxidase- and PU.1-positive cells or elevation of bronchoalveolar lavage fluid IL-8 and systemic IL-6 concentrations in LPS-exposed animals. Unexpectedly, IL-2 treatment improved fetal lung function of control lambs as indicated by increased disaturated phospholipids and improved lung gas volume. In conclusion, systemic IL-2 treatment in utero preferentially expanded Tregs and improved lung gas volume and disaturated phospholipids. These beneficial effects on lung function were maintained despite the moderate immunomodulatory effects of prophylactic IL-2 in the course of chorioamnionitis., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
14. Selective IL-1α exposure to the fetal gut, lung, and chorioamnion/skin causes intestinal inflammatory and developmental changes in fetal sheep.
- Author
-
Nikiforou M, Kemp MW, van Gorp RH, Saito M, Newnham JP, Reynaert NL, Janssen LE, Jobe AH, Kallapur SG, Kramer BW, and Wolfs TG
- Subjects
- Animals, Cytokines immunology, Cytokines metabolism, Disease Models, Animal, Female, Pregnancy, Sheep, Skin immunology, Chorioamnionitis immunology, Fetus immunology, Interleukin-1alpha immunology, Interleukin-1alpha metabolism, Intestinal Mucosa immunology, Lung immunology
- Abstract
Chorioamnionitis, caused by intra-amniotic exposure to bacteria and their toxic components, is associated with fetal gut inflammation and mucosal injury. In a translational ovine model, we have shown that these adverse intestinal outcomes to chorioamnionitis were the combined result of local gut and pulmonary-driven systemic immune responses. Chorioamnionitis-induced gut inflammation and injury was largely prevented by inhibiting interleukin-1 (IL-1) signaling. Therefore, we investigated whether local (gut-derived) IL-1α signaling or systemic IL-1α-driven immune responses (lung or chorioamnion/skin-derived) were sufficient for intestinal inflammation and mucosal injury in the course of chorioamnionitis. Fetal surgery was performed in sheep to isolate the lung, gastrointestinal tract, and chorioamnion/skin, and IL-1α or saline was given into the trachea, stomach, or amniotic cavity 1 or 6 days before preterm delivery. Selective IL-1α exposure to the lung, gut, or chorioamnion/skin increased the CD3+ cell numbers in the fetal gut. Direct IL-1α exposure to the gut impaired intestinal zonula occludens protein-1 expression, induced villus atrophy, changed the expression pattern of intestinal fatty acid-binding protein along the villus, and increased the CD68, IL-1, and TNF-α mRNA levels in the fetal ileum. With lung or chorioamnion/skin exposure to IL-1α, intestinal inflammation was associated with increased numbers of blood leukocytes without induction of intestinal injury or immaturity. We concluded that local IL-1α signaling was required for intestinal inflammation, disturbed gut maturation, and mucosal injury in the context of chorioamnionitis.
- Published
- 2016
- Full Text
- View/download PDF
15. Multipotent adult progenitor cells for hypoxic-ischemic injury in the preterm brain.
- Author
-
Jellema RK, Ophelders DR, Zwanenburg A, Nikiforou M, Delhaas T, Andriessen P, Mays RW, Deans R, Germeraad WT, Wolfs TG, and Kramer BW
- Subjects
- Animals, Animals, Newborn, Disease Models, Animal, Fetus, Sheep, Adult Stem Cells transplantation, Hypoxia-Ischemia, Brain therapy, Mesenchymal Stem Cell Transplantation methods, Premature Birth
- Abstract
Background: Preterm infants are at risk for hypoxic-ischemic encephalopathy. No therapy exists to treat this brain injury and subsequent long-term sequelae. We have previously shown in a well-established pre-clinical model of global hypoxia-ischemia (HI) that mesenchymal stem cells are a promising candidate for the treatment of hypoxic-ischemic brain injury. In the current study, we investigated the neuroprotective capacity of multipotent adult progenitor cells (MAPC®), which are adherent bone marrow-derived cells of an earlier developmental stage than mesenchymal stem cells and exhibiting more potent anti-inflammatory and regenerative properties., Methods: Instrumented preterm sheep fetuses were subjected to global hypoxia-ischemia by 25 min of umbilical cord occlusion at a gestational age of 106 (term ~147) days. During a 7-day reperfusion period, vital parameters (e.g., blood pressure and heart rate; baroreceptor reflex) and (amplitude-integrated) electroencephalogram were recorded. At the end of the experiment, the preterm brain was studied by histology., Results: Systemic administration of MAPC therapy reduced the number and duration of seizures and prevented decrease in baroreflex sensitivity after global HI. In addition, MAPC cells prevented HI-induced microglial proliferation in the preterm brain. These anti-inflammatory effects were associated with MAPC-induced prevention of hypomyelination after global HI. Besides attenuation of the cerebral inflammatory response, our findings showed that MAPC cells modulated the peripheral splenic inflammatory response, which has been implicated in the etiology of hypoxic-ischemic injury in the preterm brain., Conclusions: In a pre-clinical animal model MAPC cell therapy improved the functional and structural outcome of the preterm brain after global HI. Future studies should establish the mechanism and long-term therapeutic effects of neuroprotection established by MAPC cells in the developing preterm brain exposed to HI. Our study may form the basis for future clinical trials, which will evaluate whether MAPC therapy is capable of reducing neurological sequelae in preterm infants with hypoxic-ischemic encephalopathy.
- Published
- 2015
- Full Text
- View/download PDF
16. Prophylactic Interleukin-2 Treatment Prevents Fetal Gut Inflammation and Injury in an Ovine Model of Chorioamnionitis.
- Author
-
Nikiforou M, Vanderlocht J, Chougnet CA, Jellema RK, Ophelders DR, Joosten M, Kloosterboer N, Senden-Gijsbers BL, Germeraad WT, Kramer BW, and Wolfs TG
- Subjects
- Analgesics, Non-Narcotic pharmacology, Animals, CD3 Complex drug effects, Chorioamnionitis blood, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Female, Ileum immunology, Ileum pathology, Interleukin-2 pharmacology, Intestinal Mucosa injuries, Intestinal Mucosa pathology, Lipopolysaccharides, Lymph Nodes metabolism, Mesentery, Neutrophils drug effects, Pregnancy, Prenatal Injuries etiology, Protective Agents administration & dosage, Protective Agents pharmacology, RNA, Messenger drug effects, Random Allocation, Sheep, T-Lymphocytes, Regulatory metabolism, Analgesics, Non-Narcotic administration & dosage, Chorioamnionitis pathology, Enteritis prevention & control, Interleukin-2 administration & dosage, Prenatal Injuries prevention & control
- Abstract
Background: Chorioamnionitis results from an infection of the fetal membranes and is associated with fetal adverse outcomes notably in the intestine. Using a translational ovine model, we showed that intra-amniotic exposure to inflammatory stimuli decreased the regulatory/effector T (Treg/Teff) cell balance in the gut, which was accompanied by intestinal inflammation and mucosal injury. We thus aimed to augment the Treg/Teff cell ratio in the fetal gut by prophylactic IL-2 treatment and evaluate whether it is sufficient to prevent chorioamnionitis-induced intestinal inflammation and mucosal injury., Methods: Fetal sheep (122 d of gestation) were intra-amniotically exposed to lipopolysaccharide for 2 or 7 days with or without prophylactic IL-2 treatment (4 d). We evaluated the infiltration of inflammatory cells in the ileum and mesenteric lymph nodes. Cytokine gene expression was analyzed in fetal ileum and the inflammatory changes were correlated with gut wall integrity., Results: IL-2 administration preferentially increased intestinal Treg cells and thus the Treg/Teff cell ratio. Prophylactic IL-2 treatment reduced the lipopolysaccharide-induced influx of neutrophils and CD3(+) T cells and decreased the messenger RNA levels of proinflammatory cytokines including IL-6 and IL-17 in the fetal ileum. Importantly, prophylactic IL-2 treatment prevented mucosal damage without inducing fetal adverse treatment outcomes., Conclusions: Our data show that prophylactic IL-2 treatment prevents fetal intestinal inflammation and mucosal injury in the context of experimental chorioamnionitis. Modulation of the Treg/Teff cell balance may contribute to the protective effects of IL-2.
- Published
- 2015
- Full Text
- View/download PDF
17. Are EGF and TLR-4 crucial to understanding the link between milk and NEC?
- Author
-
Derikx JP, Kramer BW, and Wolfs TG
- Subjects
- Animals, Female, Apoptosis immunology, Enterocolitis, Necrotizing prevention & control, Enterocytes immunology, Milk immunology, Signal Transduction immunology, Toll-Like Receptor 4 immunology
- Published
- 2015
- Full Text
- View/download PDF
18. An acute intake of plant stanol esters alters immune-related pathways in the jejunum of healthy volunteers.
- Author
-
De Smet E, Mensink RP, Boekschoten MV, de Ridder R, Germeraad WT, Wolfs TG, and Plat J
- Subjects
- Adult, Anticholesteremic Agents adverse effects, Antigens, Surface blood, Antigens, Surface genetics, Antigens, Surface metabolism, Beverages, Cross-Over Studies, Double-Blind Method, Down-Regulation, Duodenum cytology, Duodenum immunology, Duodenum metabolism, Female, Forkhead Transcription Factors blood, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Humans, Intestinal Mucosa cytology, Intestinal Mucosa metabolism, Jejunum cytology, Jejunum metabolism, Male, Middle Aged, Sitosterols adverse effects, T-Lymphocytes cytology, T-Lymphocytes metabolism, Young Adult, Anticholesteremic Agents administration & dosage, Immunity, Mucosal, Immunomodulation, Intestinal Mucosa immunology, Jejunum immunology, Sitosterols administration & dosage, T-Lymphocytes immunology
- Abstract
Plant sterols and stanols inhibit intestinal cholesterol absorption and consequently lower serum LDL-cholesterol (LDL-C) concentrations. The underlying mechanisms are not yet known. In vitro and animal studies have suggested that changes in intestinal sterol metabolism are attributed to the LDL-C-lowering effects of plant stanol esters. However, similar studies in human subjects are lacking. Therefore, we examined the effects of an acute intake of plant stanol esters on gene expression profiles of the upper small intestine in healthy volunteers. In a double-blind cross-over design, fourteen healthy subjects (eight female and six male; age 21-55 years), with a BMI ranging from 21 to 29 kg/m², received in random order a shake with or without plant stanol esters (4 g). At 5 h after consumption of the shake, biopsies were taken from the duodenum (around the papilla of Vater) and from the jejunum (20 cm distal from the papilla of Vater). Microarray analysis showed that the expression profiles of genes involved in sterol metabolism were not altered. Surprisingly, the pathways involved in T-cell functions were down-regulated in the jejunum. Furthermore, immunohistochemical analysis showed that the number of CD3 (cluster of differentiation number 3), CD4 (cluster of differentiation number 4) and Foxp3⁺ (forkhead box P3-positive) cells was reduced in the plant stanol ester condition compared with the control condition, which is in line with the microarray data. The physiological and functional consequences of the plant stanol ester-induced reduction of intestinal T-cell-based immune activity in healthy subjects deserve further investigation.
- Published
- 2015
- Full Text
- View/download PDF
19. Using trend templates in a neonatal seizure algorithm improves detection of short seizures in a foetal ovine model.
- Author
-
Zwanenburg A, Andriessen P, Jellema RK, Niemarkt HJ, Wolfs TG, Kramer BW, and Delhaas T
- Subjects
- Animals, Asphyxia, Disease Models, Animal, Information Theory, Seizures diagnosis, Sheep, Domestic, Signal Processing, Computer-Assisted, Time Factors, Brain embryology, Brain physiopathology, Electroencephalography methods, Fetal Hypoxia physiopathology, Seizures physiopathology, Support Vector Machine
- Abstract
Seizures below one minute in duration are difficult to assess correctly using seizure detection algorithms. We aimed to improve neonatal detection algorithm performance for short seizures through the use of trend templates for seizure onset and end. Bipolar EEG were recorded within a transiently asphyxiated ovine model at 0.7 gestational age, a common experimental model for studying brain development in humans of 30-34 weeks of gestation. Transient asphyxia led to electrographic seizures within 6-8 h. A total of 3159 seizures, 2386 shorter than one minute, were annotated in 1976 h-long EEG recordings from 17 foetal lambs. To capture EEG characteristics, five features, sensitive to seizures, were calculated and used to derive trend information. Feature values and trend information were used as input for support vector machine classification and subsequently post-processed. Performance metrics, calculated after post-processing, were compared between analyses with and without employing trend information. Detector performance was assessed after five-fold cross-validation conducted ten times with random splits. The use of trend templates for seizure onset and end in a neonatal seizure detection algorithm significantly improves the correct detection of short seizures using two-channel EEG recordings from 54.3% (52.6-56.1) to 59.5% (58.5-59.9) at FDR 2.0 (median (range); p < 0.001, Wilcoxon signed rank test). Using trend templates might therefore aid in detection of short seizures by EEG monitoring at the NICU.
- Published
- 2015
- Full Text
- View/download PDF
20. The effects of dexamethasone and oxygen in ventilated adult sheep with early phase acute respiratory distress syndrome.
- Author
-
Engel M, Nowacki RM, Boden P, Reiss LK, Uhlig S, Reynaert NL, Gopal P, Wouters EF, Willems CH, Kloosterboer N, Wolfs TG, Zimmermann LJ, Vos GD, and Kramer BW
- Subjects
- Acute Lung Injury metabolism, Acute Lung Injury pathology, Acute Lung Injury physiopathology, Adrenal Cortex Hormones toxicity, Age Factors, Animals, Bronchoalveolar Lavage Fluid chemistry, Dexamethasone toxicity, Disease Models, Animal, Female, Inflammation Mediators metabolism, Lung metabolism, Lung pathology, Lung physiopathology, Phospholipids metabolism, Pneumonia metabolism, Pneumonia pathology, Pneumonia physiopathology, Pulmonary Gas Exchange drug effects, Pulmonary Surfactant-Associated Proteins metabolism, Respiratory Distress Syndrome metabolism, Respiratory Distress Syndrome pathology, Respiratory Distress Syndrome physiopathology, Sheep, Time Factors, Acute Lung Injury drug therapy, Adrenal Cortex Hormones pharmacology, Anti-Inflammatory Agents pharmacology, Dexamethasone pharmacology, Lung drug effects, Oxygen Inhalation Therapy adverse effects, Pneumonia therapy, Respiration, Artificial adverse effects, Respiratory Distress Syndrome therapy
- Abstract
Background: Acute respiratory distress syndrome (ARDS) is a life-threating condition with high morbidity and mortality. Inflammation is the main factor in the pathogenesis of ARDS. Therefore systemic corticosteroids are a rational therapeutic approach, but the effect of corticosteroids is still unclear. In this study, we looked at the effects of corticosteroids in ventilated sheep with ARDS, induced by lung lavage., Methods: We performed a prospective, randomised study in 64 ventilated sheep with ARDS, to evaluate the effect of corticosteroids and oxygen concentration on gas exchange and lung injury. Oxygenation index (OI) and ventilation efficacy index (VEI) were calculated to evaluate gas exchange. Lung injury was assessed by inflammatory response in broncho-alveolar lavage fluid (BALF) and plasma and histology of the lung., Results: OI, VEI, lung inflammation, surfactant production, or lung histology was not influenced by corticosteroids. In the 100 % oxygen groups, OI was higher and total number of cells and disaturated phospholipids were lower in BALF., Conclusion: Our study showed that corticosteroids did not influence inflammation in early phase ARDS and that hyperoxia aggravated lung injury which could not be modulated by dexamethasone in early phase ARDS.
- Published
- 2015
- Full Text
- View/download PDF
21. Responses of the spleen to intraamniotic lipopolysaccharide exposure in fetal sheep.
- Author
-
Kuypers E, Willems MG, Jellema RK, Kemp MW, Newnham JP, Delhaas T, Kallapur SG, Jobe AH, Wolfs TG, and Kramer BW
- Subjects
- Amniotic Fluid drug effects, Animals, Apoptosis, CD3 Complex metabolism, Caspase 3 metabolism, Chorioamnionitis physiopathology, Cytokines metabolism, Female, Fetus metabolism, Gestational Age, Immune System, Inflammation, Interleukin-23 metabolism, Models, Animal, Pregnancy, Pregnancy, Animal, RNA, Messenger metabolism, Sheep, Spleen metabolism, Amniotic Fluid metabolism, Lipopolysaccharides chemistry, Spleen immunology
- Abstract
Background: Intrauterine inflammation activates the fetal immune system and can result in organ injury and postnatal complications in preterm infants. As the spleen is an important site for peripheral immune activation, we asked how the fetal spleen would respond to intrauterine inflammation over time. We hypothesized that intraamniotic lipopolysaccharide (IA LPS) exposure induces acute and persistent changes in the splenic cytokine profile and T-cell composition that may contribute to the sustained fetal inflammatory response after chorioamnionitis., Methods: Fetal sheep were exposed to IA LPS 5, 12, and 24 h and 2, 4, 8, or 15 d before delivery at 125 d of gestational age (term = 150 d). Splenic cytokine mRNA levels and cleaved caspase-3, CD3, and Foxp3 expression were evaluated., Results: IA LPS increased interleukin (IL)1, IL4, IL5, and IL10 mRNA by twofold 24 h after injection. Interferon gamma increased by fivefold, whereas IL23 decreased 15 d post-LPS exposure. Cleaved caspase-3-positive cells increased 2 and 8 d after LPS exposure. CD3 immunoreactivity increased within 5 h with increased Foxp3-positive cells at 12 h., Conclusion: Intrauterine inflammation induced a rapid and sustained splenic immune response with persistent changes in the cytokine profile. This altered immune status may drive sustained inflammation and injury in other fetal organs.
- Published
- 2015
- Full Text
- View/download PDF
22. Breast-feeding improves gut maturation compared with formula feeding in preterm babies.
- Author
-
Reisinger KW, de Vaan L, Kramer BW, Wolfs TG, van Heurn LW, and Derikx JP
- Subjects
- Biomarkers urine, Fatty Acid-Binding Proteins urine, Feces chemistry, Female, Gestational Age, Humans, Infant, Newborn, Leukocyte L1 Antigen Complex analysis, Male, Prospective Studies, Breast Feeding, Infant Formula, Infant, Premature growth & development, Intestines growth & development
- Abstract
Objective: The incidence of necrotizing enterocolitis (NEC) is higher in formula-fed babies than in breast-fed babies, which may be caused by breast-feeding-induced gut maturation. The effect of breast-feeding on gut maturation has been widely studied in animal models. This study aimed to assess the effects of breast-feeding on intestinal maturation in prematurely born babies by evaluating postnatal changes in urinary intestinal fatty acid binding protein (I-FABP) levels, a specific enterocyte marker., Methods: Gut maturation in 40 premature babies (<37 weeks of gestation) without gastrointestinal morbidity was studied, of whom 21 were exclusively breast-fed and 19 were formula-fed infants. Urinary I-FABP levels as the measure of gut maturation were measured at 5, 12, 19, and 26 days after birth., Results: In breast-fed infants, there was a significant increase in median urinary I-FABP levels between 5 and 12 days after birth (104 [78-340] pg/mL to 408 [173-1028] pg/mL, P = 0.002), whereas I-FABP concentration in formula-fed infants increased between 12 and 19 days after birth (105 [44-557] pg/mL, 723 [103-1670] pg/mL, P = 0.004). Breast-fed babies had significantly higher median urinary I-FABP levels at postnatal day 12 (P = 0.01)., Conclusions: The time course of the postnatal increase in urinary I-FABP levels reflecting gut maturation was significantly delayed in formula-fed babies, suggesting a delayed physiological response in formula-fed compared with breast-fed infants.
- Published
- 2014
- Full Text
- View/download PDF
23. Intestinal fatty acid-binding protein: a possible marker for gut maturation.
- Author
-
Reisinger KW, Elst M, Derikx JP, Nikkels PG, de Vries B, Adriaanse MP, Jellema RK, Kramer BW, and Wolfs TG
- Subjects
- Animals, Animals, Newborn, Autopsy, Biomarkers metabolism, Enterocytes metabolism, Fatty Acid-Binding Proteins blood, Fatty Acid-Binding Proteins urine, Female, Fetal Blood metabolism, Gestational Age, Humans, Ileum cytology, Ileum growth & development, Infant, Extremely Premature, Intestinal Mucosa cytology, Intestinal Mucosa growth & development, Male, Morphogenesis, Phenotype, Premature Birth, Sheep, Fatty Acid-Binding Proteins metabolism, Ileum metabolism, Intestinal Mucosa metabolism
- Abstract
Background: Gut immaturity is linked with postnatal intestinal disorders. However, biomarkers to assess the intestinal developmental stage around birth are lacking. The aim of this study was to gain more insight on intestinal fatty acid-binding protein (I-FABP) as an indicator of gut maturity., Methods: Antenatal I-FABP distribution and release was investigated in extremely premature, moderately premature, and term lambs, and these findings were verified in human urinary samples. Ileal I-FABP distribution was confirmed in autopsy material within 24 h postnatally., Results: Median (range) serum I-FABP levels were lower in extremely premature lambs compared with moderately premature lambs (156 (50.0-427) vs. 385 (100-1,387) pg/ml; P = 0.02). Contrarily, median early postnatal urine I-FABP levels in human infants were higher in extremely premature compared with moderately premature and term neonates (1,219 (203-15,044) vs. 256 (50-1,453) and 328 (96-1,749) pg/ml; P = 0.008 and P = 0.04, respectively). I-FABP expression was most prominent in nonvacuolated enterocytes and increased with rising gestational age (GA) in ovine and human tissue samples. The epithelial distribution pattern changed from a phenotype displaying I-FABP-positive enterocytes merely in the crypts early in gestation into a phenotype with I-FABP expressing cells exclusively present in the villus tips at term in ovine and human tissue., Conclusion: In this ovine and human study, increasing GA is accompanied by an increase in I-FABP tissue content. Cord I-FABP levels correlate with gestation in ovine fetuses, identifying I-FABP as a marker for gut maturation. Raised postnatal urine I-FABP levels in preterm human infants may indicate intestinal injury and/or inflammation in utero.
- Published
- 2014
- Full Text
- View/download PDF
24. Chorioamnionitis-induced fetal gut injury is mediated by direct gut exposure of inflammatory mediators or by lung inflammation.
- Author
-
Wolfs TG, Kramer BW, Thuijls G, Kemp MW, Saito M, Willems MG, Senthamarai-Kannan P, Newnham JP, Jobe AH, and Kallapur SG
- Subjects
- Amniotic Fluid, Animals, Cell Differentiation, Cell Proliferation, Chorioamnionitis chemically induced, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Epithelial Cells cytology, Epithelial Cells drug effects, Female, Fetal Diseases chemically induced, Gastrointestinal Diseases embryology, Gastrointestinal Diseases pathology, Gene Expression Regulation, Developmental drug effects, Ileitis chemically induced, Ileitis embryology, Ileitis pathology, Ileum embryology, Ileum pathology, Inflammation chemically induced, Inflammation complications, Inflammation pathology, Intestinal Mucosa cytology, Lipopolysaccharides toxicity, Pneumonia chemically induced, Pneumonia pathology, Pregnancy, Random Allocation, Sheep, T-Lymphocytes, Regulatory, Toll-Like Receptors, Chorioamnionitis pathology, Fetal Diseases etiology, Gastrointestinal Diseases etiology, Pneumonia complications
- Abstract
Intra-amniotic exposure to proinflammatory agonists causes chorioamnionitis and fetal gut inflammation. Fetal gut inflammation is associated with mucosal injury and impaired gut development. We tested whether this detrimental inflammatory response of the fetal gut results from a direct local (gut derived) or an indirect inflammatory response mediated by the chorioamnion/skin or lung, since these organs are also in direct contact with the amniotic fluid. The gastrointestinal tract was isolated from the respiratory tract and the amnion/skin epithelia by fetal surgery in time-mated ewes. Lipopolysaccharide (LPS) or saline (controls) was selectively infused in the gastrointestinal tract, trachea, or amniotic compartment at 2 or 6 days before preterm delivery at 124 days gestation (term 150 days). Gastrointestinal and intratracheal LPS exposure caused distinct inflammatory responses in the fetal gut. Inflammatory responses could be distinguished by the influx of leukocytes (MPO(+), CD3(+), and FoxP3(+) cells), tumor necrosis factor-α, and interferon-γ expression and differential upregulation of mRNA levels for Toll-like receptor 1, 2, 4, and 6. Fetal gut inflammation after direct intestinal LPS exposure resulted in severe loss of the tight junctional protein zonula occludens protein 1 (ZO-1) and increased mitosis of intestinal epithelial cells. Inflammation of the fetal gut after selective LPS instillation in the lungs caused only mild disruption of ZO-1, loss in epithelial cell integrity, and impaired epithelial differentiation. LPS exposure of the amnion/skin epithelia did not result in gut inflammation or morphological, structural, and functional changes. Our results indicate that the detrimental consequences of chorioamnionitis on fetal gut development are the combined result of local gut and lung-mediated inflammatory responses.
- Published
- 2014
- Full Text
- View/download PDF
25. Altered canonical Wingless-Int signaling in the ovine fetal lung after exposure to intra-amniotic lipopolysaccharide and antenatal betamethasone.
- Author
-
Kuypers E, Willems MG, Collins JJ, Wolfs TG, Nitsos I, Jane Pillow J, Polglase GR, Kemp MW, Newnham JP, Delhaas T, Jobe AH, Kallapur SG, and Kramer BW
- Subjects
- Animals, Betamethasone chemistry, Female, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Inflammation, Lipopolysaccharides chemistry, Lung metabolism, Lymphoid Enhancer-Binding Factor 1 metabolism, Maternal Exposure, Phosphorylation, Pregnancy, Pregnancy, Animal, Sheep, Sheep, Domestic, Time Factors, Wnt Proteins metabolism, beta Catenin metabolism, Betamethasone administration & dosage, Lipopolysaccharides administration & dosage, Lung pathology, Wnt Signaling Pathway
- Abstract
Background: Antenatal inflammation and maternal corticosteroids induce fetal lung maturation but interfere with late lung development. Canonical Wingless-Int (Wnt) signaling directs lung development and repair. We showed that intra-amniotic (IA) lipopolysaccharide (LPS) exposure disrupted developmental signaling pathways in the preterm lamb lungs. Therefore, we hypothesized that pulmonary Wnt signaling was altered by exposure to IA LPS and/or antenatal corticosteroids., Methods: Ovine fetuses were exposed to IA LPS, maternal intramuscular betamethasone, a control saline injection, or a combination thereof at 107 and/or 114 d gestational age (term = 150 d gestational age) before delivery at 121 d gestational age., Results: IA LPS exposure decreased the lung expression of lymphoid enhancer-binding factor 1 (LEF1), a major Wnt pathway effector. WNT1, WNT4, and downstream messenger β-catenin decreased after LPS exposure. WNT7b mRNA increased fourfold 14 d post-LPS exposure. Betamethasone treatment 7 d before LPS exposure prevented the reduction in LEF1 expression, whereas betamethasone administration after LPS normalized the LPS-induced increase in Wnt7b mRNA., Conclusion: IA LPS exposure decreased canonical Wnt signaling in the developing lung. Antenatal corticosteroids before or after IA inflammation had different effects on pulmonary Wnt signaling. This study provides new insights into possible mechanisms by which prenatal inflammation affects lung development and how corticosteroid can be beneficial in this setting.
- Published
- 2014
- Full Text
- View/download PDF
26. Effects of intra-amniotic lipopolysaccharide and maternal betamethasone on brain inflammation in fetal sheep.
- Author
-
Kuypers E, Jellema RK, Ophelders DR, Dudink J, Nikiforou M, Wolfs TG, Nitsos I, Pillow JJ, Polglase GR, Kemp MW, Saito M, Newnham JP, Jobe AH, Kallapur SG, and Kramer BW
- Subjects
- Amniotic Fluid immunology, Animals, Female, Pregnancy, Sheep, Amniotic Fluid drug effects, Betamethasone pharmacology, Encephalitis immunology, Fetus drug effects, Glucocorticoids pharmacology, Lipopolysaccharides pharmacology
- Abstract
Rationale: Chorioamnionitis and antenatal glucocorticoids are common exposures for preterm infants and can affect the fetal brain, contributing to cognitive and motor deficits in preterm infants. The effects of antenatal glucocorticoids on the brain in the setting of chorioamnionitis are unknown. We hypothesized that antenatal glucocorticoids would modulate inflammation in the brain and prevent hippocampal and white matter injury after intra-amniotic lipopolysaccharide (LPS) exposure., Methods: Time-mated ewes received saline (control), an intra-amniotic injection of 10 mg LPS at 106d GA or 113d GA, maternal intra-muscular betamethasone (0.5 mg/kg maternal weight) alone at 113d GA, betamethasone at 106d GA before LPS or betamethasone at 113d GA after LPS. Animals were delivered at 120d GA (term=150d). Brain structure volumes were measured on T2-weighted MRI images. The subcortical white matter (SCWM), periventricular white matter (PVWM) and hippocampus were analyzed for microglia, astrocytes, apoptosis, proliferation, myelin and pre-synaptic vesicles., Results: LPS and/or betamethasone exposure at different time-points during gestation did not alter brain structure volumes on MRI. Betamethasone alone did not alter any of the measurements. Intra-amniotic LPS at 106d or 113d GA induced inflammation as indicated by increased microglial and astrocyte recruitment which was paralleled by increased apoptosis and hypomyelination in the SCWM and decreased synaptophysin density in the hippocampus. Betamethasone before the LPS exposure at 113d GA prevented microglial activation and the decrease in synaptophysin. Betamethasone after LPS exposure increased microglial infiltration and apoptosis., Conclusion: Intra-uterine LPS exposure for 7d or 14d before delivery induced inflammation and injury in the fetal white matter and hippocampus. Antenatal glucocorticoids aggravated the inflammatory changes in the brain caused by pre-existing intra-amniotic inflammation. Antenatal glucocorticoids prior to LPS reduced the effects of intra-uterine inflammation on the brain. The timing of glucocorticoid administration in the setting of chorioamnionitis can alter outcomes for the fetal brain.
- Published
- 2013
- Full Text
- View/download PDF
27. Systemic G-CSF attenuates cerebral inflammation and hypomyelination but does not reduce seizure burden in preterm sheep exposed to global hypoxia-ischemia.
- Author
-
Jellema RK, Lima Passos V, Ophelders DR, Wolfs TG, Zwanenburg A, De Munter S, Nikiforou M, Collins JJ, Kuypers E, Bos GM, Steinbusch HW, Vanderlocht J, Andriessen P, Germeraad WT, and Kramer BW
- Subjects
- Animals, Disease Models, Animal, Electrocardiography, Electroencephalography, Encephalitis etiology, Fetal Hypoxia pathology, Fetus, Flow Cytometry, Hematopoietic Stem Cell Mobilization, Hypoxia-Ischemia, Brain pathology, Immunohistochemistry, Nerve Fibers, Myelinated drug effects, Seizures etiology, Sheep, Encephalitis pathology, Fetal Hypoxia complications, Granulocyte Colony-Stimulating Factor pharmacology, Hypoxia-Ischemia, Brain complications, Neuroprotective Agents pharmacology
- Abstract
Hypoxic-ischemic encephalopathy (HIE) is common in preterm infants, but currently no curative therapy is available. Cell-based therapy has a great potential in the treatment of hypoxic-ischemic preterm brain injury. Granulocyte-colony stimulating factor (G-CSF) is known to mobilize endogenous hematopoietic stem cells (HSC) and promotes proliferation of endogenous neural stem cells. On these grounds, we hypothesized that systemic G-CSF would be neuroprotective in a large translational animal model of hypoxic-ischemic injury in the preterm brain. Global hypoxia-ischemia (HI) was induced by transient umbilical cord occlusion in instrumented preterm sheep. G-CSF treatment (100μg/kg intravenously, during five consecutive days) was started one day before the global HI insult to ascertain mobilization of endogenous stem cells within the acute phase after global HI. Mobilization of HSC and neutrophils was studied by flow cytometry. Brain sections were stained for microglia (IBA-1), myelin basic protein (MBP) and myeloperoxidase (MPO) to study microglial proliferation, white matter injury and neutrophil invasion respectively. Electrographic seizure activity was analyzed using amplitude-integrated electroencephalogram (aEEG). G-CSF effectively mobilized CD34-positive HSC in the preterm sheep. In addition, G-CSF caused marked mobilization of neutrophils, but did not influence enhanced invasion of neutrophils into the preterm brain after global HI. Microglial proliferation and hypomyelination following global HI were reduced as a result of G-CSF treatment. G-CSF did not cause a reduction of the electrographic seizure activity after global HI. In conclusion, G-CSF induced mobilization of endogenous stem cells which was associated with modulation of the cerebral inflammatory response and reduced white matter injury in an ovine model of preterm brain injury after global HI. G-CSF treatment did not improve neuronal function as shown by seizure analysis. Our study shows that G-CSF treatment has neuroprotective potential following hypoxic-ischemic injury in the preterm brain., (© 2013.)
- Published
- 2013
- Full Text
- View/download PDF
28. Mesenchymal stem cells induce T-cell tolerance and protect the preterm brain after global hypoxia-ischemia.
- Author
-
Jellema RK, Wolfs TG, Lima Passos V, Zwanenburg A, Ophelders DR, Kuypers E, Hopman AH, Dudink J, Steinbusch HW, Andriessen P, Germeraad WT, Vanderlocht J, and Kramer BW
- Subjects
- Animals, Base Sequence, DNA Primers, Disease Models, Animal, Magnetic Resonance Imaging, Polymerase Chain Reaction, Seizures prevention & control, Sheep, Brain embryology, Hypoxia-Ischemia, Brain immunology, Immune Tolerance, Mesenchymal Stem Cells immunology, T-Lymphocytes immunology
- Abstract
Hypoxic-ischemic encephalopathy (HIE) in preterm infants is a severe disease for which no curative treatment is available. Cerebral inflammation and invasion of activated peripheral immune cells have been shown to play a pivotal role in the etiology of white matter injury, which is the clinical hallmark of HIE in preterm infants. The objective of this study was to assess the neuroprotective and anti-inflammatory effects of intravenously delivered mesenchymal stem cells (MSC) in an ovine model of HIE. In this translational animal model, global hypoxia-ischemia (HI) was induced in instrumented preterm sheep by transient umbilical cord occlusion, which closely mimics the clinical insult. Intravenous administration of 2 x 10(6) MSC/kg reduced microglial proliferation, diminished loss of oligodendrocytes and reduced demyelination, as determined by histology and Diffusion Tensor Imaging (DTI), in the preterm brain after global HI. These anti-inflammatory and neuroprotective effects of MSC were paralleled by reduced electrographic seizure activity in the ischemic preterm brain. Furthermore, we showed that MSC induced persistent peripheral T-cell tolerance in vivo and reduced invasion of T-cells into the preterm brain following global HI. These findings show in a preclinical animal model that intravenously administered MSC reduced cerebral inflammation, protected against white matter injury and established functional improvement in the preterm brain following global HI. Moreover, we provide evidence that induction of T-cell tolerance by MSC might play an important role in the neuroprotective effects of MSC in HIE. This is the first study to describe a marked neuroprotective effect of MSC in a translational animal model of HIE.
- Published
- 2013
- Full Text
- View/download PDF
29. Intraamniotic lipopolysaccharide exposure changes cell populations and structure of the ovine fetal thymus.
- Author
-
Kuypers E, Wolfs TG, Collins JJ, Jellema RK, Newnham JP, Kemp MW, Kallapur SG, Jobe AH, and Kramer BW
- Subjects
- Amniotic Fluid, Animals, Apoptosis, Cell Proliferation, Chorioamnionitis chemically induced, Chorioamnionitis pathology, Disease Models, Animal, Female, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Developmental, Gestational Age, Inflammation Mediators metabolism, Injections, Interleukin-17 genetics, Interleukin-17 metabolism, Interleukin-6 genetics, Interleukin-6 metabolism, NF-kappa B metabolism, Pregnancy, RNA, Messenger metabolism, Sheep, Thymus Gland embryology, Thymus Gland pathology, Time Factors, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism, Chorioamnionitis immunology, Lipopolysaccharides administration & dosage, Thymus Gland immunology
- Abstract
Rationale: Chorioamnionitis induces preterm delivery and acute involution of the fetal thymus which is associated with postnatal inflammatory disorders. We studied the immune response, cell composition, and architecture of the fetal thymus following intraamniotic lipopolysaccharide (LPS) exposure., Methods: Time-mated ewes received an intraamniotic injection of LPS 5, 12, or 24 hours or 2, 4, 8, or 15 days before delivery at 125 days gestational age (term = 150 days)., Results: The LPS exposure resulted in decreased blood lymphocytes within 5 hours and decreased thymic corticomedullary ratio within 24 hours. Thymic interleukin 6 (IL6) and IL17 messenger RNA (mRNA) increased 5-fold 24 hours post-LPS exposure. Increased toll-like receptor 4 (TLR4) mRNA and nuclear factor κB positive cells at 24 hours after LPS delivery demonstrated acute thymic activation. Both TLR4 and IL1 mRNA increased by 5-fold and the number of Foxp3-positive cells (Foxp3+ cells) decreased 15 days after exposure., Conclusion: Intraamniotic LPS exposure caused a proinflammatory response, involution, and a persistent depletion of thymic Foxp3+ cells indicating disturbance of the fetal immune homeostasis.
- Published
- 2013
- Full Text
- View/download PDF
30. Antenatal ureaplasma infection impairs development of the fetal ovine gut in an IL-1-dependent manner.
- Author
-
Wolfs TG, Kallapur SG, Knox CL, Thuijls G, Nitsos I, Polglase GR, Collins JJ, Kroon E, Spierings J, Shroyer NF, Newnham JP, Jobe AH, and Kramer BW
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Interleukin 1 Receptor Antagonist Protein administration & dosage, Intestinal Mucosa embryology, Intestinal Mucosa immunology, Intestinal Mucosa microbiology, Intestines immunology, Metagenome immunology, Pregnancy, Prenatal Exposure Delayed Effects immunology, Prenatal Exposure Delayed Effects microbiology, Sheep, Domestic, Chorioamnionitis microbiology, Interleukin-1 immunology, Intestines embryology, Intestines microbiology, Ureaplasma, Ureaplasma Infections complications
- Abstract
Ureaplasma infection of the amniotic cavity is associated with adverse postnatal intestinal outcomes. We tested whether interleukin-1 (IL-1) signaling underlies intestinal pathology following ureaplasma exposure in fetal sheep. Pregnant ewes received intra-amniotic injections of ureaplasma or culture media for controls at 3, 7, and 14 d before preterm delivery at 124 d gestation (term 150 d). Intra-amniotic injections of recombinant human interleukin IL-1 receptor antagonist (rhIL-1ra) or saline for controls were given 3 h before and every 2 d after Ureaplasma injection. Ureaplasma exposure caused fetal gut inflammation within 7 d with damaged villus epithelium and gut barrier loss. Proliferation, differentiation, and maturation of enterocytes were significantly reduced after 7 d of ureaplasma exposure, leading to severe villus atrophy at 14 d. Inflammation, impaired development and villus atrophy of the fetal gut was largely prevented by intra-uterine rhIL-1ra treatment. These data form the basis for a clinical understanding of the role of ureaplasma in postnatal intestinal pathologies.
- Published
- 2013
- Full Text
- View/download PDF
31. Noninvasive measurement of intestinal epithelial damage at time of refeeding can predict clinical outcome after necrotizing enterocolitis.
- Author
-
Reisinger KW, Derikx JP, Thuijls G, van der Zee DC, Brouwers HA, van Bijnen AA, Wolfs TG, van Heurn LW, Buurman WA, and Kramer BW
- Subjects
- Biomarkers urine, Enterocolitis, Necrotizing mortality, Enterocolitis, Necrotizing pathology, Enterocolitis, Necrotizing urine, Epithelial Cells pathology, Fatty Acid-Binding Proteins urine, Female, Humans, Infant, Infant, Newborn, Intestinal Mucosa pathology, Male, Parenteral Nutrition, Total, Predictive Value of Tests, Time Factors, Treatment Outcome, Urinalysis, Enteral Nutrition adverse effects, Enteral Nutrition mortality, Enterocolitis, Necrotizing therapy, Epithelial Cells metabolism, Intestinal Mucosa metabolism
- Abstract
Background: Reintroduction of enteral nutrition in neonates with necrotizing enterocolitis (NEC) should take place when the gut is ready for its normal function. Too early a start of oral feeding might lead to disease relapse, whereas prolonged discontinuation of enteral nutrition is associated with impaired gut function and parenteral nutrition-related complications. This study evaluated whether noninvasive urinary measurement of intestinal fatty acid binding protein (I-FABP) at the time of refeeding can predict clinical outcome in neonates with NEC., Methods: Urinary I-FABP concentrations were measured in 21 infants with NEC just before reintroducing enteral nutrition. Poor outcome was defined as unsuccessful reintroduction of enteral feeding (EF), (re)operation for NEC, or death related to NEC after reintroduction of EF., Results: Median urinary I-FABP levels in neonates with poor outcome (n = 5) were significantly higher as compared with I-FABP levels in neonates with good outcome (n = 16) (P < 0.01). A clinically significant cutoff value of 963 pg/ml was found to discriminate between infants with poor outcome and those with good outcome (sensitivity 80%, specificity 94%)., Conclusion: Noninvasive urinary I-FABP measurement at time of refeeding differentiates neonates with poor outcome from neonates with good outcome in NEC. Urinary I-FABP measurement may therefore be helpful in the timing of EF in neonates with NEC.
- Published
- 2013
- Full Text
- View/download PDF
32. Cerebral inflammation and mobilization of the peripheral immune system following global hypoxia-ischemia in preterm sheep.
- Author
-
Jellema RK, Lima Passos V, Zwanenburg A, Ophelders DR, De Munter S, Vanderlocht J, Germeraad WT, Kuypers E, Collins JJ, Cleutjens JP, Jennekens W, Gavilanes AW, Seehase M, Vles HJ, Steinbusch H, Andriessen P, Wolfs TG, and Kramer BW
- Subjects
- Animals, Animals, Newborn, Female, Fetus immunology, Fetus pathology, Immunity, Innate, Microglia immunology, Microglia pathology, Pregnancy, Sheep, Brain immunology, Brain pathology, Cell Movement immunology, Hypoxia-Ischemia, Brain immunology, Hypoxia-Ischemia, Brain pathology
- Abstract
Background: Hypoxic-ischemic encephalopathy (HIE) is one of the most important causes of brain injury in preterm infants. Preterm HIE is predominantly caused by global hypoxia-ischemia (HI). In contrast, focal ischemia is most common in the adult brain and known to result in cerebral inflammation and activation of the peripheral immune system. These inflammatory responses are considered to play an important role in the adverse outcomes following brain ischemia. In this study, we hypothesize that cerebral and peripheral immune activation is also involved in preterm brain injury after global HI., Methods: Preterm instrumented fetal sheep were exposed to 25 minutes of umbilical cord occlusion (UCO) (n = 8) at 0.7 gestation. Sham-treated animals (n = 8) were used as a control group. Brain sections were stained for ionized calcium binding adaptor molecule 1 (IBA-1) to investigate microglial proliferation and activation. The peripheral immune system was studied by assessment of circulating white blood cell counts, cellular changes of the spleen and influx of peripheral immune cells (MPO-positive neutrophils) into the brain. Pre-oligodendrocytes (preOLs) and myelin basic protein (MBP) were detected to determine white matter injury. Electro-encephalography (EEG) was recorded to assess functional impairment by interburst interval (IBI) length analysis., Results: Global HI resulted in profound activation and proliferation of microglia in the hippocampus, periventricular and subcortical white matter. In addition, non-preferential mobilization of white blood cells into the circulation was observed within 1 day after global HI and a significant influx of neutrophils into the brain was detected 7 days after the global HI insult. Furthermore, global HI resulted in marked involution of the spleen, which could not be explained by increased splenic apoptosis. In concordance with cerebral inflammation, global HI induced severe brain atrophy, region-specific preOL vulnerability, hypomyelination and persistent suppressed brain function., Conclusions: Our data provided evidence that global HI in preterm ovine fetuses resulted in profound cerebral inflammation and mobilization of the peripheral innate immune system. These inflammatory responses were paralleled by marked injury and functional loss of the preterm brain. Further understanding of the interplay between preterm brain inflammation and activation of the peripheral immune system following global HI will contribute to the development of future therapeutic interventions in preterm HIE.
- Published
- 2013
- Full Text
- View/download PDF
33. Inflammation-induced immune suppression of the fetus: a potential link between chorioamnionitis and postnatal early onset sepsis.
- Author
-
Wolfs TG, Jellema RK, Turrisi G, Becucci E, Buonocore G, and Kramer BW
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Infant, Newborn, Infant, Premature, Intestines immunology, Lung immunology, Pregnancy, Respiratory Mucosa immunology, Spleen immunology, Thymus Gland immunology, Chorioamnionitis immunology, Infant, Premature, Diseases immunology, Premature Birth immunology, Sepsis immunology
- Abstract
Chorioamnionitis which results from microbial invasion of the amniotic cavity is the most frequent cause of preterm birth. Chorioamnionitis is associated with an increased risk of early-onset sepsis but the mechanisms underlying this association remain largely unknown. We hypothesize that developmental alterations of fetal organs and the immune system in the course of chorioamnionitis determine the risk of development of early onset sepsis. The purpose of this review is therefore to summarize the consequences of chorioamnionitis on fetal development and speculate how those antenatal changes might predispose to early onset sepsis.
- Published
- 2012
- Full Text
- View/download PDF
34. Increased levels of deleted in malignant brain tumours 1 (DMBT1) in active bacteria-related appendicitis.
- Author
-
Kaemmerer E, Schneider U, Klaus C, Plum P, Reinartz A, Adolf M, Renner M, Wolfs TG, Kramer BW, Wagner N, Mollenhauer J, and Gassler N
- Subjects
- Adolescent, Adult, Aged, Appendicitis genetics, Appendicitis pathology, Appendix pathology, Calcium-Binding Proteins, Child, DNA-Binding Proteins, Enterocytes pathology, Epithelial Cells metabolism, Epithelial Cells pathology, Humans, Male, Middle Aged, Receptors, Cell Surface genetics, Tumor Suppressor Proteins, Appendicitis metabolism, Appendix metabolism, Enterocytes metabolism, Receptors, Cell Surface metabolism
- Abstract
Aims: Deleted in malignant brain tumours 1 (DMBT1; gp340) is a secreted glycoprotein which is found in the surface lining epithelia of human small and large intestine. DMBT1 is suggested to play a role in enterocyte differentiation and surface protection from intestinal bacteria. The aim of this study was to elucidate DMBT1 expression in bacteria-related active intestinal inflammation such as appendicitis., Methods and Results: mRNA and protein levels of DMBT1 were analysed in surgical resections of 50 appendices (active inflammation: n = 25). In non-actively inflamed appendices, inter-individual differences in basal DMBT1 levels of enterocytes and some non-epithelial cells were found. In active appendicitis, enterocytic DMBT1 mRNA expression was increased approximately fivefold, which was paralleled by a corresponding increase of cytoplasmic and secreted DMBT1 protein levels. Increased DMBT1 expression was predominant in enterocytes adjacent to erosive lesions or ulcers., Conclusions: Our data demonstrate that bacteria-related active inflammation results in a sharp increase of DMBT1 levels in enterocytes. These findings substantiate the view that DMBT1 is of functional relevance for host defence and modulation of the course of intestinal bacteria-related inflammatory responses., (© 2012 Blackwell Publishing Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
35. Ovine fetal thymus response to lipopolysaccharide-induced chorioamnionitis and antenatal corticosteroids.
- Author
-
Kuypers E, Collins JJ, Jellema RK, Wolfs TG, Kemp MW, Nitsos I, Pillow JJ, Polglase GR, Newnham JP, Germeraad WT, Kallapur SG, Jobe AH, and Kramer BW
- Subjects
- Adrenal Cortex Hormones therapeutic use, Animals, Apoptosis drug effects, Bone Morphogenetic Protein 4 genetics, Bone Morphogenetic Protein 4 metabolism, CD3 Complex metabolism, Cell Proliferation drug effects, Chorioamnionitis immunology, Chorioamnionitis metabolism, Female, Fetus metabolism, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Gene Expression Regulation drug effects, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Pregnancy, T-Lymphocytes cytology, T-Lymphocytes drug effects, T-Lymphocytes metabolism, Thymus Gland drug effects, Thymus Gland metabolism, Time Factors, Toll-Like Receptors genetics, Toll-Like Receptors metabolism, Adrenal Cortex Hormones pharmacology, Chorioamnionitis chemically induced, Chorioamnionitis drug therapy, Fetus drug effects, Lipopolysaccharides pharmacology, Sheep embryology, Thymus Gland embryology
- Abstract
Rationale: Chorioamnionitis is associated with preterm delivery and involution of the fetal thymus. Women at risk of preterm delivery receive antenatal corticosteroids which accelerate fetal lung maturation and improve neonatal outcome. However, the effects of antenatal corticosteroids on the fetal thymus in the settings of chorioamnionitis are largely unknown. We hypothesized that intra-amniotic exposure to lipopolysaccharide (LPS) causes involution of the fetal thymus resulting in persistent effects on thymic structure and cell populations. We also hypothesized that antenatal corticosteroids may modulate the effects of LPS on thymic development., Methods: Time-mated ewes with singleton fetuses received an intra-amniotic injection of LPS 7 or 14 days before preterm delivery at 120 days gestational age (term = 150 days). LPS and corticosteroid treatment groups received intra-amniotic LPS either preceding or following maternal intra-muscular betamethasone. Gestation matched controls received intra-amniotic and maternal intra-muscular saline. The fetal intra-thoracic thymus was evaluated., Results: Intra-amniotic LPS decreased the cortico-medullary (C/M) ratio of the thymus and increased Toll-like receptor (TLR) 4 mRNA and CD3 expression indicating involution and activation of the fetal thymus. Increased TLR4 and CD3 expression persisted for 14 days but Foxp3 expression decreased suggesting a change in regulatory T-cells. Sonic hedgehog and bone morphogenetic protein 4 mRNA, which are negative regulators of T-cell development, decreased in response to intra-amniotic LPS. Betamethasone treatment before LPS exposure attenuated some of the LPS-induced thymic responses but increased cleaved caspase-3 expression and decreased the C/M ratio. Betamethasone treatment after LPS exposure did not prevent the LPS-induced thymic changes., Conclusion: Intra-amniotic exposure to LPS activated the fetal thymus which was accompanied by structural changes. Treatment with antenatal corticosteroids before LPS partially attenuated the LPS-induced effects but increased apoptosis in the fetal thymus. Corticosteroid administration after the inflammatory stimulus did not inhibit the LPS effects on the fetal thymus.
- Published
- 2012
- Full Text
- View/download PDF
36. IL-1α mediated chorioamnionitis induces depletion of FoxP3+ cells and ileal inflammation in the ovine fetal gut.
- Author
-
Wolfs TG, Kallapur SG, Polglase GR, Pillow JJ, Nitsos I, Newnham JP, Chougnet CA, Kroon E, Spierings J, Willems CH, Jobe AH, and Kramer BW
- Subjects
- Animals, CD4 Lymphocyte Count, CD4-Positive T-Lymphocytes drug effects, Chorioamnionitis immunology, Fatty Acid-Binding Proteins metabolism, Female, Fetus drug effects, Ileum drug effects, Ileum enzymology, Interleukin-1alpha administration & dosage, Membrane Proteins metabolism, Peroxidase metabolism, Phosphoproteins metabolism, Pregnancy, Reverse Transcriptase Polymerase Chain Reaction, Sheep, Domestic, Zonula Occludens-1 Protein, Chorioamnionitis pathology, Fetus pathology, Forkhead Transcription Factors metabolism, Ileum embryology, Ileum pathology, Inflammation pathology, Interleukin-1alpha pharmacology
- Abstract
Background: Endotoxin induced chorioamnionitis increases IL-1 and provokes an inflammatory response in the fetal ileum that interferes with intestinal maturation. In the present study, we tested in an ovine chorioamnionitis model whether IL-1 is a major cytokine driving the inflammatory response in the fetal ileum., Method: Sheep bearing singleton fetuses received a single intraamniotic injection of recombinant ovine IL-1α at 7, 3 or 1 d before caesarian delivery at 125 days gestational age (term = 150 days)., Results: 3 and 7 d after IL-1α administration, intestinal mRNA levels for IL-4, IL-10, IFN-γ and TNF-α were strongly elevated. Numbers of CD3+ and CD4+ T-lymphocytes and myeloidperoxidase+ cells were increased whereas FoxP3+ T-cells were detected at low frequency. This increased proinflammatory state was associated with ileal mucosal barrier loss as demonstrated by decreased levels of the intestinal fatty acid binding protein and disruption of the tight junctional protein ZO-1., Conclusion: Intraamniotic IL-1α causes an acute detrimental inflammatory response in the ileum, suggesting that induction of IL-1 is a critical element in the pathophysiological effects of endotoxin induced chorioamnionitis. A disturbed balance between T-effector and FoxP3+ cells may contribute to this process.
- Published
- 2011
- Full Text
- View/download PDF
37. Localization of the lipopolysaccharide recognition complex in the human healthy and inflamed premature and adult gut.
- Author
-
Wolfs TG, Derikx JP, Hodin CM, Vanderlocht J, Driessen A, de Bruïne AP, Bevins CL, Lasitschka F, Gassler N, van Gemert WG, and Buurman WA
- Subjects
- Adult, Female, Gram-Negative Bacteria pathogenicity, Gram-Negative Bacterial Infections diagnosis, Gram-Negative Bacterial Infections metabolism, Gram-Negative Bacterial Infections microbiology, Humans, Immunoenzyme Techniques, Infant, Newborn, Infant, Premature, Inflammation metabolism, Inflammation microbiology, Inflammatory Bowel Diseases diagnosis, Inflammatory Bowel Diseases microbiology, Lymphocyte Antigen 96 genetics, Male, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Toll-Like Receptor 4 genetics, Gastrointestinal Tract microbiology, Inflammation diagnosis, Inflammatory Bowel Diseases metabolism, Lipopolysaccharides metabolism, Lymphocyte Antigen 96 metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Background: Microbiota in the intestinal lumen provide an abundant source of potentially detrimental antigens, including lipopolysaccharide (LPS), a potent immunostimulatory product of Gram-negative bacteria recognized by the host via TLR-4 and MD-2. An aberrant immune response to LPS or other bacterial antigens has been linked to inflammatory bowel disease (IBD) and necrotizing enterocolitis (NEC)., Methods: We investigated which cells express MD-2 in the normal and inflamed ileum from neonates and adults by immunohistochemistry. Moreover, MD-2 and TLR4 mRNA expression in normal adult ileum was studied by reverse-transcription polymerase chain reaction (RT-PCR) on cells isolated by laser capture microdissection., Results: Premature infants did not show MD-2 expression either in epithelial cells or in the lamina propria. Similarly, MD-2 was absent in epithelial cells and lamina propria inflammatory cells in preterm infants with NEC. MD-2 protein in the healthy term neonatal and adult ileum was predominantly expressed by Paneth cells and some resident inflammatory cells in the lamina propria. MD-2 and TLR-4 mRNA expression was restricted to crypt cells. Also in IBD, Paneth cells were still the sole MD-2-expressing epithelial cells, whereas inflammatory cells (mainly plasma cells) were responsible for the vast majority of the MD-2 expression., Conclusions: The absence of MD-2 in the immature neonatal gut suggests impaired LPS sensing, which could predispose neonates to NEC upon microbial colonization of the immature intestine. The apparent expression of MD-2 by Paneth cells supports the critical concept that these cells respond to luminal bacterial products in order to maintain homeostasis with the intestinal microbiota in vivo.
- Published
- 2010
- Full Text
- View/download PDF
38. Role of arginine in superficial wound healing in man.
- Author
-
Debats IB, Wolfs TG, Gotoh T, Cleutjens JP, Peutz-Kootstra CJ, and van der Hulst RR
- Subjects
- Adult, Aged, Arginase analysis, Arginase metabolism, Citrulline blood, Dietary Supplements, Female, Humans, Male, Middle Aged, Nitrates blood, Nitric Oxide Synthase Type II metabolism, Nitric Oxide Synthase Type III metabolism, Nitrites blood, Ornithine blood, Prospective Studies, Skin cytology, Skin metabolism, Skin Transplantation, Arginine administration & dosage, Arginine metabolism, Skin injuries, Wound Healing drug effects
- Abstract
Arginine supplementation has been identified as advantageous in experimental wound healing. However, the mechanisms underlying this beneficial effect in tissue repair remain unresolved. Animal studies suggest that the beneficial role of arginine supplementation is mediated, at least in part through NO. The latter component mediates processes involved in tissue repair, including angiogenesis, epithelialization and collagen formation. This prospective study is performed to investigate arginine metabolism in acute surgical wounds in man. Expression of enzymes, known to be involved in arginine metabolism, was studied in donor sites of skin grafts of 10 hospitalized patients undergoing skin transplantation. Plasma and wound fluid levels of arginine metabolites (ornithine, citrulline, nitrate and nitrite = NOx) were measured using High Performance Liquid Chromatography. Expression of iNOS, eNOS, arginase-1 and arginase-2 was studied by immunohistochemistry in paraffin sections of skin tissue. Arginase-1 concentration was measured in plasma and wound fluid using ELISA. Arginase-2 was determined using Western blot analysis. We observed increased levels of citrulline, ornithine, NOx and arginase-1 in wound fluid when compared with plasma. Arginase-2 was expressed in both plasma and wound fluid and seemed higher in plasma. iNOS was expressed by neutrophils, macrophages, fibroblasts, keratinocytes and endothelial cells upon wounding, whereas eNOS reactivity was observed in endothelial cells and fibroblasts. Arginase-1 was expressed in neutrophils post-wounding, while arginase-2 staining was observed in endothelial cells, keratinocytes, fibroblasts, macrophages and neutrophils. For the first time, human data support previous animal studies suggesting arginine metabolism for an NO- as well as arginase-mediated reparation of injured skin.
- Published
- 2009
- Full Text
- View/download PDF
39. Endotoxin induced chorioamnionitis prevents intestinal development during gestation in fetal sheep.
- Author
-
Wolfs TG, Buurman WA, Zoer B, Moonen RM, Derikx JP, Thuijls G, Villamor E, Gantert M, Garnier Y, Zimmermann LJ, and Kramer BW
- Subjects
- Animals, Arteries pathology, Female, Immunohistochemistry methods, Inflammation, Microscopy, Fluorescence methods, Nitric Oxide Synthase Type III metabolism, Pregnancy, Pregnancy, Animal, Sheep, Tight Junctions pathology, Time Factors, Chorioamnionitis immunology, Chorioamnionitis veterinary, Endotoxins metabolism, Intestines embryology
- Abstract
Chorioamnionitis is the most significant source of prenatal inflammation and preterm delivery. Prematurity and prenatal inflammation are associated with compromised postnatal developmental outcomes, of the intestinal immune defence, gut barrier function and the vascular system. We developed a sheep model to study how the antenatal development of the gut was affected by gestation and/or by endotoxin induced chorioamnionitis.Chorioamnionitis was induced at different gestational ages (GA). Animals were sacrificed at low GA after 2d or 14d exposure to chorioamnionitis. Long term effects of 30d exposure to chorioamnionitis were studied in near term animals after induction of chorioamnionitis. The cellular distribution of tight junction protein ZO-1 was shown to be underdeveloped at low GA whereas endotoxin induced chorioamnionitis prevented the maturation of tight junctions during later gestation. Endotoxin induced chorioamnionitis did not induce an early (2d) inflammatory response in the gut in preterm animals. However, 14d after endotoxin administration preterm animals had increased numbers of T-lymphocytes, myeloperoxidase-positive cells and gammadelta T-cells which lasted till 30d after induction of chorioamnionitis in then near term animals. At early GA, low intestinal TLR-4 and MD-2 mRNA levels were detected which were further down regulated during endotoxin-induced chorioamnionitis. Predisposition to organ injury by ischemia was assessed by the vascular function of third-generation mesenteric arteries. Endotoxin-exposed animals of low GA had increased contractile response to the thromboxane A2 mimetic U46619 and reduced endothelium-dependent relaxation in responses to acetylcholine. The administration of a nitric oxide (NO) donor completely restored endothelial dysfunction suggesting reduced NO bioavailability which was not due to low expression of endothelial nitric oxide synthase.Our results indicate that the distribution of the tight junctional protein ZO-1, the immune defence and vascular function are immature at low GA and are further compromised by endotoxin-induced chorioamnionitis. This study suggests that both prematurity and inflammation in utero disturb fetal gut development, potentially predisposing to postnatal intestinal pathology.
- Published
- 2009
- Full Text
- View/download PDF
40. Increased release of sMD-2 during human endotoxemia and sepsis: a role for endothelial cells.
- Author
-
Wolfs TG, Dunn-Siegrist I, van't Veer C, Hodin CM, Germeraad WT, van Zoelen MA, van Suylen RJ, Peutz-Kootstra CJ, Elson G, Pugin J, and Buurman WA
- Subjects
- Adult, Case-Control Studies, Endothelial Cells drug effects, Endothelial Cells immunology, Endothelial Cells pathology, Endotoxemia immunology, Female, Hematopoietic System cytology, Hematopoietic System drug effects, Humans, Kinetics, Lipopolysaccharides pharmacology, Liver drug effects, Liver immunology, Liver pathology, Lung drug effects, Lung immunology, Lung pathology, Lymphocyte Antigen 96 blood, Male, Middle Aged, Sepsis blood, Sepsis immunology, Solubility drug effects, Tumor Necrosis Factor-alpha pharmacology, Endothelial Cells metabolism, Endotoxemia metabolism, Lymphocyte Antigen 96 metabolism, Sepsis metabolism
- Abstract
MD-2 is the crucial cofactor of TLR4 in the detection of LPS. Here, we show that soluble MD-2 (sMD-2) circulates in plasma of healthy individuals as a polymeric protein. The total amount of sMD-2 in septic plasma was strongly elevated and contained both sMD-2 polymers and monomers, the latter representing the putative biologically active form of MD-2. Moreover, during experimental human endotoxemia, the monomeric and total sMD-2 content in plasma increased with the kinetics of an acute phase protein. The increase in sMD-2 monomers was paralleled by enhanced TLR4 costimulatory activity. The presence of functional sMD-2 during endotoxemia and sepsis was confirmed by immunodepletion. Immunohistochemistry revealed that MD-2 expression in septic patients was strongly enhanced on endothelium and multiple inflammatory cells in lung and liver. In vitro studies showed that sMD-2 release appears to be restricted to endothelial cells and dendritic cells. Release of sMD-2 by endothelial cells was strongly enhanced by LPS and TNF-alpha stimulation. Taken together, this study demonstrates the increase of both circulating polymeric and functional monomeric sMD-2 during endotoxemia and sepsis, and evidence is provided that the endothelium is involved in this process.
- Published
- 2008
- Full Text
- View/download PDF
41. Toll-like receptor 4 ligation on intrinsic renal cells contributes to the induction of antibody-mediated glomerulonephritis via CXCL1 and CXCL2.
- Author
-
Brown HJ, Lock HR, Wolfs TG, Buurman WA, Sacks SH, and Robson MG
- Subjects
- Albuminuria immunology, Albuminuria metabolism, Albuminuria pathology, Animals, Bone Marrow Cells cytology, Bone Marrow Cells immunology, Bone Marrow Cells metabolism, Cells, Cultured, Chemokine CXCL1 immunology, Chemokine CXCL2 immunology, Female, Glomerulonephritis pathology, Immunoglobulin G metabolism, Immunoglobulin G pharmacology, Ligands, Lipid A pharmacology, Male, Mesangial Cells cytology, Mesangial Cells metabolism, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Neutrophils immunology, Neutrophils pathology, Severity of Illness Index, Sheep, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Chemokine CXCL1 metabolism, Chemokine CXCL2 metabolism, Glomerulonephritis immunology, Glomerulonephritis metabolism, Mesangial Cells immunology, Toll-Like Receptor 4 metabolism
- Abstract
Autoimmune diseases such as glomerulonephritis are exacerbated by infection. This study examined the effect of the Toll-like receptor 4 (TLR4) ligand lipid A on the development of heterologous nephrotoxic nephritis. Administration of nephrotoxic antibody resulted in significant glomerular neutrophil infiltration and albuminuria only when a TLR4 ligand was administered simultaneously. The contribution of TLR4 on renal cells and circulating leukocytes was assessed. Bone marrow chimeras were constructed with TLR4 only on renal cells or bone marrow-derived cells. The administration of nephrotoxic serum and lipid A caused a neutrophil influx in both chimeric groups greater than in sham chimeras that were totally TLR4 deficient but significantly less than in sham chimeras that were totally TLR4 sufficient. Both chimeric groups had greater albuminuria than totally TLR4-deficient sham chimeras; however, the chimeras with TLR4 only on intrinsic renal cells had significantly less than the sham positive group. In situ hybridization showed expression of TLR4 mRNA in mesangial cells and glomerular epithelial cells. For investigation of the potential mechanism by which renal cells could contribute to disease exacerbation, mesangial cells were cultured and found to express mRNA for TLR4, and stimulation of wild-type and TLR4-deficient mesangial cells with LPS caused production of CXC chemokines by wild-type cells only. Treatment of chimeras with TLR4 present only on intrinsic renal cells with anti-CXCL1 and anti-CXCL2 antibody before disease induction significantly reduced renal neutrophil infiltration. These results show that TLR4 on both circulating leukocytes and intrinsic renal cells contributes to the inflammatory effects of antibody deposition within the glomerulus, which depends at least in part on the production of CXC chemokines by intrinsic renal cells.
- Published
- 2007
- Full Text
- View/download PDF
42. Rapid pulmonary expression of acute-phase reactants after local lipopolysaccharide exposure in mice is followed by an interleukin-6 mediated systemic acute-phase response.
- Author
-
Vernooy JH, Reynaert N, Wolfs TG, Cloots RH, Haegens A, de Vries B, Dentener MA, Buurman WA, and Wouters EM
- Subjects
- Animals, Carrier Proteins blood, Carrier Proteins genetics, Liver metabolism, Male, Membrane Glycoproteins blood, Membrane Glycoproteins genetics, Mice, Orosomucoid analysis, Orosomucoid genetics, Trachea drug effects, alpha 1-Antitrypsin analysis, alpha 1-Antitrypsin genetics, Acute-Phase Proteins genetics, Acute-Phase Reaction, Interleukin-6 physiology, Lipopolysaccharides toxicity, Lung metabolism
- Abstract
This study investigated local and systemic innate immune responses in lipopolysaccharide (LPS)-induced lung inflammation in mice. Intratracheal LPS exposure resulted in increased pulmonary mRNA expression for acute-phase reactants (APRs) alpha(1)-antitrypsin (alpha(1)-AT), alpha(1)-acid glycoprotein (AGP), and LPS-binding protein (LBP) from 4 hours post exposure. Although pulmonary serum amyloid P component (SAP) mRNA was not increased, systemic levels of SAP, AGP, and LBP were elevated from 24 hours post exposure. Systemic APRs increase was associated with hepatic mRNA expression. As in vivo neutralization of interleukin (IL)-6, but not tumor necrosis factor (TNF)-alpha, fully ablated hepatic APR mRNA expression, IL-6 may act as signaling molecule between lung and liver. In conclusion, pulmonary LPS exposure induced rapid APR expression in lung, which precedes IL-6-mediated systemic elevation of APRs associated with hepatic APRs expression.
- Published
- 2005
- Full Text
- View/download PDF
43. Apoptotic cell death is initiated during normothermic ischemia in human kidneys.
- Author
-
Wolfs TG, de Vries B, Walter SJ, Peutz-Kootstra CJ, van Heurn LW, Oosterhof GO, and Buurman WA
- Subjects
- Blotting, Western, CASP8 and FADD-Like Apoptosis Regulating Protein, Carcinoma, Renal Cell pathology, Caspase 3, Caspase 8, Caspase 9, Caspases metabolism, Enzyme Activation, Hot Temperature, Humans, Immunohistochemistry, Intracellular Signaling Peptides and Proteins metabolism, Kidney Neoplasms pathology, Microscopy, Fluorescence, Proto-Oncogene Proteins c-bcl-2 metabolism, Time Factors, bcl-2-Associated X Protein, Apoptosis, Ischemia metabolism, Kidney pathology
- Abstract
Ischemic damage plays an important role in post-transplant organ failure. Activation of the apoptotic cascade is crucially involved in post-ischemic inflammation resulting in tissue damage and organ dysfunction. Here we investigate the initiation of the apoptotic cascade during normothermic ischemia in human kidneys using a model for normothermic ischemia with kidneys nephrectomized because of renal cell carcinoma. Ex vivo, kidneys were stored at 37 degrees C, and consecutive biopsies were taken from disease-free tissue. Pro- and anti-apoptotic proteins were assessed by Western blotting and immunofluorescence. During normothermic ischemia the pro-apoptotic proteins Bax and activated caspase-9 increased with ischemia time, whereas caspase-8 was not activated. The anti-apoptotic proteins Bcl-2 and cFLIP decreased in time. Data on Bcl-2 and Bax were supported by immunofluorescence for Bcl-2 and activated Bax. However, activation of the central effector caspase-3, essential for execution of the apoptotic process, was not detected. In conclusion, during normothermic ischemia the apoptotic cascade in the human kidney is initiated, but not fulfilled. Our data show that the duration of ischemia significantly correlates with activation of the apoptotic cascade. These findings provide insight in the initiation of apoptotic cell-death during warm ischemia and may be useful in the assessment of ischemic injury.
- Published
- 2005
- Full Text
- View/download PDF
44. The mannose-binding lectin-pathway is involved in complement activation in the course of renal ischemia-reperfusion injury.
- Author
-
de Vries B, Walter SJ, Peutz-Kootstra CJ, Wolfs TG, van Heurn LW, and Buurman WA
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Biopsy, Complement Activation, Enzyme-Linked Immunosorbent Assay, Humans, Immunohistochemistry, Kidney metabolism, Lectins, Liver metabolism, Lung metabolism, Male, Mice, Neutrophils metabolism, Peroxidase metabolism, Protein Binding, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Kidney pathology, Mannose chemistry, Mannose-Binding Lectin chemistry, Reperfusion Injury pathology
- Abstract
Ischemia-reperfusion (I/R) is an important cause of acute renal failure (ARF). The complement system appears to be essentially involved in I/R injury. However, via which pathway the complement system is activated and in particular whether the mannose-binding lectin (MBL)-pathway is activated is unclear. This tempted us to study the activation and regulation of the MBL-pathway in the course of experimental renal I/R injury and in clinical post-transplant ARF. Mice subjected to renal I/R displayed evident renal MBL-depositions, depending on the duration of warm ischemia, in the early reperfusion phase. Renal deposition of C3, C6 and C9 was observed in the later reperfusion phase. The deposition of MBL-A and -C completely co-localized with the late complement factor C6, showing that MBL is involved in complement activation in the course of renal I/R injury. Moreover, the degree of early MBL-deposition correlated with complement activation, neutrophil-influx, and organ-failure observed in the later reperfusion phase. In serum of mice subjected to renal I/R MBL-A, levels increased in contrast to MBL-C levels, which dropped evidently. In line, liver mRNA levels for MBL-A increased, whereas MBL-C levels decreased. Renal MBL mRNA levels rapidly dropped in the course of renal I/R. Finally, in human biopsies, MBL-depositions were observed early after transplantation of ischemically injured kidneys. In line with our experimental data, in ischemically injured grafts displaying post-transplant organ-failure extensive MBL depositions were observed in peritubular capillaries and tubular epithelial cells. In conclusion, in experimental renal I/R injury and clinical post-transplant ARF the MBL-pathway is activated, followed by activation of the complement system. These data indicate that the MBL-pathway is involved in ischemia-induced complement activation.
- Published
- 2004
- Full Text
- View/download PDF
45. Exogenous alpha-1-acid glycoprotein protects against renal ischemia-reperfusion injury by inhibition of inflammation and apoptosis.
- Author
-
de Vries B, Walter SJ, Wolfs TG, Hochepied T, Räbinä J, Heeringa P, Parkkinen J, Libert C, and Buurman WA
- Subjects
- Animals, Cytoskeleton drug effects, Cytoskeleton ultrastructure, Dose-Response Relationship, Drug, Fucose metabolism, Humans, Mice, Mice, Transgenic, Orosomucoid administration & dosage, Orosomucoid genetics, Orosomucoid metabolism, Rats, Reperfusion Injury pathology, Reperfusion Injury physiopathology, Apoptosis drug effects, Inflammation prevention & control, Orosomucoid pharmacology, Renal Circulation, Reperfusion Injury prevention & control
- Abstract
Background: Although ischemia-reperfusion (I/R) injury represents a major problem in posttransplant organ failure, effective treatment is not available. The acute phase protein alpha-1-acid glycoprotein (AGP) has been shown to be protective against experimental I/R injury. The effects of AGP are thought to be mediated by fucose groups expressed on the AGP protein inhibiting neutrophil infiltration. However, the precise mechanism of protection remains to be established. We therefore studied the effects of exogenous human AGP (hAGP) in a mouse model of ischemic acute renal failure., Methods: Mice were subjected to renal I/R and treated with hAGP, fucose-depleted hAGP, or control treated. Also, transgenic mice over-expressing rat AGP or wild-type controls were subjected to renal I/R., Results: Treatment was with hAGP as well as fucose-depleted hAGP protected mice against I/R-induced acute renal failure. Surprisingly, AGP-over-expressing mice were not protected against I/R injury. Both natural and fucose-depleted hAGP inhibited the activation of the complement system, as determined by renal C3 deposition and influx of neutrophils measured by immunohistochemistry and myeloperoxidase-enzyme-linked immunoadsorbent assay. Tubular epithelial cell structure (actin cytoskeleton) and cell-cell interaction (tight-junction architecture) were completely preserved in AGP-treated mice. Also, epithelial caspase activation and apoptotic DNA cleavage were prevented by AGP treatment., Conclusions: Both natural and fucose-depleted hAGP protect against renal I/R injury by preservation of tubular epithelial structure and inhibition of apoptosis and subsequent inflammation. Therefore, hAGP can be regarded as a potential new therapeutic intervention in the treatment of acute renal failure, as seen after transplantation of ischemically injured kidneys.
- Published
- 2004
- Full Text
- View/download PDF
46. Reduction of circulating redox-active iron by apotransferrin protects against renal ischemia-reperfusion injury.
- Author
-
de Vries B, Walter SJ, von Bonsdorff L, Wolfs TG, van Heurn LW, Parkkinen J, and Buurman WA
- Subjects
- Animals, Apoproteins metabolism, Apoptosis immunology, Complement Activation, Kidney immunology, Kidney metabolism, Kidney pathology, Male, Mice, Nephrectomy, Neutrophils immunology, Oxidation-Reduction, Reactive Oxygen Species blood, Reperfusion Injury immunology, Transferrin metabolism, Transplants adverse effects, Apoproteins pharmacology, Iron blood, Reperfusion Injury drug therapy, Reperfusion Injury metabolism, Transferrin pharmacology
- Abstract
Background: Warm ischemia-reperfusion (I/R) injury plays an important role in posttransplant organ failure. In particular, organs from marginal donors suffer I/R injury. Although iron has been implicated in the pathophysiology of renal I/R injury, the mechanism of iron-mediated injury remains to be established. The authors therefore investigated the role of circulating redox-active iron in an experimental model for renal I/R injury., Methods: Male Swiss mice were subjected to unilateral renal ischemia for 45 min, followed by contralateral nephrectomy and reperfusion. To investigate the role of circulating iron, mice were treated with apotransferrin, an endogenous iron-binding protein, or iron-saturated apotransferrin (holotransferrin)., Results: Renal ischemia induced a significant increase in circulating redox-active iron levels during reperfusion. Apotransferrin, in contrast to holotransferrin, reduced the amount of circulating redox-active iron and abrogated renal superoxide formation. Apotransferrin treatment did not affect I/R-induced renal apoptosis, whereas holotransferrin aggravated apoptotic cell death. Apotransferrin, in contrast to holotransferrin, inhibited the influx of neutrophils. Both apo- and holotransferrin reduced I/R-induced complement deposition, indicating that the effects of transferrin are differentially mediated by its iron and protein moiety. Finally, apotransferrin, in contrast to holotransferrin, dose-dependently inhibited the loss of renal function induced by ischemia., Conclusions: Redox-active iron is released into the circulation in the course of renal I/R. Reducing the amount of circulating redox-active iron by treatment with apotransferrin protects against renal I/R injury, inhibiting oxidative stress, inflammation, and loss of function. Apotransferrin could be used in the treatment of acute renal failure, as seen after transplantation of ischemically damaged organs.
- Published
- 2004
- Full Text
- View/download PDF
47. Lysophosphatidic acid prevents renal ischemia-reperfusion injury by inhibition of apoptosis and complement activation.
- Author
-
de Vries B, Matthijsen RA, van Bijnen AA, Wolfs TG, and Buurman WA
- Subjects
- Animals, Caspase 7, Caspases metabolism, Complement C3 metabolism, Complement C6 metabolism, Disease Models, Animal, Enzyme Activation, Humans, Kidney pathology, Male, Mice, Tumor Necrosis Factor-alpha metabolism, Apoptosis physiology, Complement Activation, Kidney metabolism, Lysophospholipids therapeutic use, Reperfusion Injury drug therapy, Reperfusion Injury prevention & control
- Abstract
Renal ischemia-reperfusion (I/R) injury is an important cause of acute renal failure as observed after renal transplantation, major surgery, trauma, and septic as well as hemorrhagic shock. We previously showed that the inhibition of apoptosis is protective against renal I/R injury, indicating that apoptotic cell-death is an important feature of I/R injury. Lysophosphatidic acid (LPA) is an endogenous phospholipid growth factor with anti-apoptotic properties. This tempted us to investigate the effects of exogenous LPA in a murine model of renal I/R injury. LPA administered at the time of reperfusion dose dependently inhibited renal apoptosis as evaluated by the presence of internucleosomal DNA cleavage. I/R-induced renal apoptosis was only present in tubular epithelial cells with evident disruption of brush border as assessed by immunohistochemistry for active caspase-7 and filamentous actin, respectively. LPA treatment specifically prevented tubular epithelial cell apoptosis but also reduced the I/R-induced loss of brush-border integrity. Besides, LPA showed strong anti-inflammatory effects, inhibiting the renal expression of tumor necrosis factor-alpha and abrogating the influx of neutrophils. Next, LPA dose dependently inhibited activation of the complement system. Moreover, treatment with LPA abrogated the loss of renal function in the course of renal I/R. This study is the first to show that administration of the phospholipid LPA prevents I/R injury, abrogating apoptosis and inflammation. Moreover, exogenous LPA is capable of preventing organ failure because of an ischemic insult and thus may provide new means to treat clinical conditions associated with I/R injury in the kidney and potentially also in other organs.
- Published
- 2003
- Full Text
- View/download PDF
48. Complement factor C5a mediates renal ischemia-reperfusion injury independent from neutrophils.
- Author
-
de Vries B, Köhl J, Leclercq WK, Wolfs TG, van Bijnen AA, Heeringa P, and Buurman WA
- Subjects
- Animals, Antigens, CD biosynthesis, Apoptosis immunology, Chemokine CXCL1, Chemokine CXCL2, Chemokines biosynthesis, Chemokines, CXC biosynthesis, Chemotactic Factors biosynthesis, Complement C5a administration & dosage, Complement C5a antagonists & inhibitors, Complement C5a metabolism, Intercellular Signaling Peptides and Proteins biosynthesis, Kidney pathology, Kidney physiopathology, Male, Mice, Monokines biosynthesis, Neutrophil Infiltration immunology, Neutrophils immunology, Neutrophils pathology, Receptor, Anaphylatoxin C5a, Receptors, Complement antagonists & inhibitors, Receptors, Complement biosynthesis, Reperfusion Injury pathology, Reperfusion Injury physiopathology, Reperfusion Injury prevention & control, Signal Transduction immunology, Tumor Necrosis Factor-alpha biosynthesis, Antigens, CD physiology, Complement C5a physiology, Kidney blood supply, Kidney immunology, Neutrophils physiology, Receptors, Complement physiology, Reperfusion Injury immunology
- Abstract
The complement system has been shown to mediate renal ischemia-reperfusion (I/R) injury. However, the contribution of complement factor C5a to I/R injury, in particular in the kidney, remains to be established. In this study, we investigated the impact of blocking the C5aR pathway on the inflammatory response and on the renal function in a murine model of I/R injury. First, we analyzed C5aR expression in kidneys of healthy mice. Intriguingly, we found expression on mesangial, as well as on tubular epithelial, cells. After I/R injury, C5aR expression was up-regulated in tubular epithelial cells. In addition, mRNA levels of CXC chemokines and TNF-alpha increased significantly and kidneys were heavily infiltrated by neutrophils. Blocking the C5aR pathway by a specific C5a receptor antagonist (C5aRA) abrogated up-regulation of CXC chemokines but not of TNF-alpha and reduced neutrophil infiltration by >50%. Moreover, application of the C5aRA significantly reduced loss of renal function. This improvement of function was independent of the presence of neutrophils because neutrophil depletion by mAb NIMP-R14 did not affect the protective effect of C5aRA treatment. Furthermore, blocking of the C5aR pathway had no influence on renal apoptosis. These data provide evidence that C5a is crucially involved in the pathogenesis of renal I/R injury by modulation of neutrophil-dependent as well as neutrophil-independent pathways, which include the regulation of CXC chemokines but not TNF-alpha or apoptotic pathways.
- Published
- 2003
- Full Text
- View/download PDF
49. Inhibition of complement factor C5 protects against renal ischemia-reperfusion injury: inhibition of late apoptosis and inflammation.
- Author
-
De Vries B, Matthijsen RA, Wolfs TG, Van Bijnen AA, Heeringa P, and Buurman WA
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Blood Urea Nitrogen, Complement Activation, Complement C5 immunology, Complement Membrane Attack Complex analysis, Creatinine blood, Kidney chemistry, Kidney immunology, Kidney pathology, Male, Mice, Nephritis pathology, Neutrophils pathology, Reperfusion Injury pathology, Apoptosis immunology, Complement C5 antagonists & inhibitors, Nephritis drug therapy, Nephritis immunology, Reperfusion Injury drug therapy, Reperfusion Injury immunology
- Abstract
Background: Complement has been implicated in the pathophysiology of renal ischemia-reperfusion (I/R) injury. However, the mechanism underlying complement-mediated renal I/R injury is thus far unknown. To investigate the involvement of complement in I/R injury, we studied the activation and deposition of complement in a murine model of renal I/R injury. Furthermore, we examined the effect of inhibition of complement-factor C5 on renal I/R injury., Methods: Mice were subjected to 45 min of unilateral ischemia and subsequent contralateral nephrectomy and reperfusion for 2, 12, or 24 hr. Mice were control treated or treated with BB5.1, a monoclonal antibody that prevents cleavage of complement factor C5, thereby preventing C5a generation and formation of the membrane attack complex (MAC)., Results: Renal I/R induced extensive deposition of C3 early after reperfusion, whereas C6 and C9 deposition (MAC formation) occurred relatively late. I/R-induced complement deposition was mainly localized to tubular epithelium. Treatment with BB5.1 totally prevented MAC formation but also reduced C3 deposition. Inhibition of C5 strongly inhibited late inflammation, as measured by neutrophil influx and induction of the murine CXC chemokines macrophage inflammatory protein-2, KC, and lipopolysaccharide-induced CXC chemokine. Anti-C5 treatment furthermore abrogated late I/R-induced apoptosis, whereas early apoptosis was not affected. Moreover, BB5.1 treatment significantly protected against I/R-induced renal dysfunction., Conclusions: Renal I/R is followed by activation of the complement system and intrarenal deposition of C3 and MAC. Complement activation plays a crucial role in the regulation of inflammation and late apoptosis. Complement inhibition, by preventing C5 activation, abrogates late apoptosis and inflammation, being strongly protective against renal function loss.
- Published
- 2003
- Full Text
- View/download PDF
50. In vivo expression of Toll-like receptor 2 and 4 by renal epithelial cells: IFN-gamma and TNF-alpha mediated up-regulation during inflammation.
- Author
-
Wolfs TG, Buurman WA, van Schadewijk A, de Vries B, Daemen MA, Hiemstra PS, and van 't Veer C
- Subjects
- Animals, Disease Models, Animal, Epithelial Cells immunology, Gene Expression Regulation immunology, Inflammation immunology, Inflammation pathology, Kidney blood supply, Kidney immunology, Male, Mice, RNA, Messenger biosynthesis, Reperfusion Injury immunology, Reperfusion Injury pathology, Toll-Like Receptor 2, Toll-Like Receptor 4, Toll-Like Receptors, Drosophila Proteins, Epithelial Cells metabolism, Epithelial Cells pathology, Interferon-gamma physiology, Kidney metabolism, Kidney pathology, Membrane Glycoproteins biosynthesis, Receptors, Cell Surface biosynthesis, Tumor Necrosis Factor-alpha physiology, Up-Regulation immunology
- Abstract
The reported requirement of functional Toll-like receptor (TLR)4 for resistance to Gram-negative pyelonephritis prompted us to localize the expression of TLR2 and TLR4 mRNA in the kidney at the cellular level by in situ hybridization. The majority of the constitutive TLR2 and TLR4 mRNA expression was found to be strategically located in the renal epithelial cells. Assuming that the TLR mRNA expression is representative of apical protein expression, this suggests that these cells are able to detect and react with bacteria present in the lumen of the tubules. To gain insight in the regulation of TLR expression during inflammation, we used a model for renal inflammation. Renal inflammation evoked by ischemia markedly enhanced synthesis of TLR2 and TLR4 mRNA in the distal tubular epithelium, the thin limb of Henle's loop, and collecting ducts. The increased renal TLR4 mRNA expression was associated with significant elevation of renal TLR4 protein expression as evaluated by Western blotting. Using RT-PCR, the enhanced TLR2 and TLR4 mRNA expression was shown to be completely dependent on the action of IFN-gamma and TNF-alpha. These results indicate a potential mechanism of increased immunosurveillance during inflammation at the site in which ascending bacteria enter the kidney tissue, i.e., the collecting ducts and the distal part of the nephron.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.