46 results on '"Collins, Jennifer J. P."'
Search Results
2. Intraamniotic Lipopolysaccharide Exposure Changes Cell Populations and Structure of the Ovine Fetal Thymus
- Author
-
Kuypers, Elke, Wolfs, Tim G. A. M., Collins, Jennifer J. P., Jellema, Reint K., Newnham, John P., Kemp, Matthew W., Kallapur, Suhas G., Jobe, Alan H., and Kramer, Boris W.
- Published
- 2013
- Full Text
- View/download PDF
3. Comparison of Recruitment Manoeuvres in Ventilated Sheep with Acute Respiratory Distress Syndrome
- Author
-
Engel, Monique, Nowacki, Relana M. E., Reiss, Lucy K., Uhlig, Stefan, Willems, Coen H. M. P., Kloosterboer, Nico, van Iwaarden, J. Freek, Sewing, Alide C. P., Seehase, Matthias, Lambermont, Verena A. C., Collins, Jennifer J. P., Zimmermann, Luc J. I., Vos, Gijs D., and Kramer, Boris W.
- Published
- 2013
- Full Text
- View/download PDF
4. Hypoxia-Inducible Factors Promote Alveolar Development and Regeneration
- Author
-
Vadivel, Arul, Alphonse, Rajesh S., Etches, Nicholas, van Haaften, Timothy, Collins, Jennifer J. P., O’Reilly, Megan, Eaton, Farah, and Thébaud, Bernard
- Published
- 2014
- Full Text
- View/download PDF
5. Early origins of lung disease: towards an interdisciplinary approach
- Author
-
Ubags, Niki D. J., Alcazar, Miguel A. Alejandre, Kallapur, Suhas G., Knapp, Sylvia, Lanone, Sophie, Lloyd, Clare M., Morty, Rory E., Pattaroni, Celine, Reynaert, Niki L., Rottier, Robbert J., Smits, Hermelijn H., Piters, Wouter A. A. de Steenhuijsen, Strickland, Deborah H., Collins, Jennifer J. P., Ubags, Niki D. J., Alcazar, Miguel A. Alejandre, Kallapur, Suhas G., Knapp, Sylvia, Lanone, Sophie, Lloyd, Clare M., Morty, Rory E., Pattaroni, Celine, Reynaert, Niki L., Rottier, Robbert J., Smits, Hermelijn H., Piters, Wouter A. A. de Steenhuijsen, Strickland, Deborah H., and Collins, Jennifer J. P.
- Abstract
The prenatal and perinatal environments can have profound effects on the development of chronic inflammatory diseases. However, mechanistic insight into how the early-life microenvironment can impact upon development of the lung and immune system and consequent initiation and progression of respiratory diseases is still emerging. Recent studies investigating the developmental origins of lung diseases have started to delineate the effects of early-life changes in the lung, environmental exposures and immune maturation on the development of childhood and adult lung diseases. While the influencing factors have been described and studied in mostly animal models, it remains challenging to pinpoint exactly which factors and at which time point are detrimental in lung development leading to respiratory disease later in life. To advance our understanding of early origins of chronic lung disease and to allow for proper dissemination and application of this knowledge, we propose four major focus areas: 1) policy and education; 2) clinical assessment; 3) basic and translational research; and 4) infrastructure and tools, and discuss future directions for advancement. This review is a follow-up of the discussions at the European Respiratory Society Research Seminar Early origins of lung disease: towards an interdisciplinary approach (Lisbon, Portugal, November 2019).
- Published
- 2020
6. Antenatal Inflammation Reduces Expression of Caveolin-1 and Influences Multiple Signaling Pathways in Preterm Fetal Lungs
- Author
-
Kunzmann, Steffen, Collins, Jennifer J. P., Yang, Yang, Uhlig, Stefan, Kallapur, Suhar G., Speer, Christian P., Jobe, Alan H., and Kramer, Boris W.
- Published
- 2011
- Full Text
- View/download PDF
7. The Future of Bronchopulmonary Dysplasia: Emerging Pathophysiological Concepts and Potential New Avenues of Treatment
- Author
-
Collins, Jennifer J. P., primary, Tibboel, Dick, additional, de Kleer, Ismé M., additional, Reiss, Irwin K. M., additional, and Rottier, Robbert J., additional
- Published
- 2017
- Full Text
- View/download PDF
8. Propofol administration to the fetal–maternal unit reduces cardiac oxidative stress in preterm lambs subjected to prenatal asphyxia and cardiac arrest
- Author
-
Seehase, Matthias, primary, Houthuizen, Patrick, additional, Collins, Jennifer J. P., additional, Zimmermann, Luc J., additional, and Kramer, Boris W., additional
- Published
- 2016
- Full Text
- View/download PDF
9. First Neuromuscular Contact Correlates with Onset of Primary Myogenesis in Rat and Mouse Limb Muscles
- Author
-
Hurren, Bradley, primary, Collins, Jennifer J. P., additional, Duxson, Marilyn J., additional, and Deries, Marianne, additional
- Published
- 2015
- Full Text
- View/download PDF
10. The Axonal Guidance Cue Semaphorin 3C Contributes to Alveolar Growth and Repair
- Author
-
Vadivel, Arul, primary, Alphonse, Rajesh S., additional, Collins, Jennifer J. P., additional, van Haaften, Tim, additional, O’Reilly, Megan, additional, Eaton, Farah, additional, and Thébaud, Bernard, additional
- Published
- 2013
- Full Text
- View/download PDF
11. Antenatal glucocorticoids counteract LPS changes in TGF-β pathway and caveolin-1 in ovine fetal lung
- Author
-
Collins, Jennifer J. P., primary, Kunzmann, Steffen, additional, Kuypers, Elke, additional, Kemp, Matthew W., additional, Speer, Christian P., additional, Newnham, John P., additional, Kallapur, Suhas G., additional, Jobe, Alan H., additional, and Kramer, Boris W., additional
- Published
- 2013
- Full Text
- View/download PDF
12. Comparison of Recruitment Manoeuvres in Ventilated Sheep with Acute Respiratory Distress Syndrome
- Author
-
Engel, Monique, primary, Nowacki, Relana M. E., additional, Reiss, Lucy K., additional, Uhlig, Stefan, additional, Willems, Coen H. M. P., additional, Kloosterboer, Nico, additional, van Iwaarden, J. Freek, additional, Sewing, Alide C. P., additional, Seehase, Matthias, additional, Lambermont, Verena A. C., additional, Collins, Jennifer J. P., additional, Zimmermann, Luc J. I., additional, Vos, Gijs D., additional, and Kramer, Boris W., additional
- Published
- 2012
- Full Text
- View/download PDF
13. LPS-induced chorioamnionitis and antenatal corticosteroids modulate Shh signaling in the ovine fetal lung
- Author
-
Collins, Jennifer J. P., primary, Kuypers, Elke, additional, Nitsos, Ilias, additional, Jane Pillow, J., additional, Polglase, Graeme R., additional, Kemp, Matthew W., additional, Newnham, John P., additional, Cleutjens, Jack P., additional, Frints, Suzanna G. M., additional, Kallapur, Suhas G., additional, Jobe, Alan H., additional, and Kramer, Boris W., additional
- Published
- 2012
- Full Text
- View/download PDF
14. New Surfactant with SP-B and C Analogs Gives Survival Benefit after Inactivation in Preterm Lambs
- Author
-
Seehase, Matthias, primary, Collins, Jennifer J. P., additional, Kuypers, Elke, additional, Jellema, Reint K., additional, Ophelders, Daan R. M. G., additional, Ospina, Olga L., additional, Perez-Gil, J., additional, Bianco, Federico, additional, Garzia, Raffaella, additional, Razzetti, Roberta, additional, and Kramer, Boris W., additional
- Published
- 2012
- Full Text
- View/download PDF
15. Ovine Fetal Thymus Response to Lipopolysaccharide-Induced Chorioamnionitis and Antenatal Corticosteroids
- Author
-
Kuypers, Elke, primary, Collins, Jennifer J. P., additional, Jellema, Reint K., additional, Wolfs, Tim G. A. M., additional, Kemp, Matthew W., additional, Nitsos, Ilias, additional, Pillow, J. Jane, additional, Polglase, Graeme R., additional, Newnham, John P., additional, Germeraad, Wilfred T. V., additional, Kallapur, Suhas G., additional, Jobe, Alan H., additional, and Kramer, Boris W., additional
- Published
- 2012
- Full Text
- View/download PDF
16. Intra-amniotic LPS and antenatal betamethasone: inflammation and maturation in preterm lamb lungs
- Author
-
Kuypers, Elke, primary, Collins, Jennifer J. P., additional, Kramer, Boris W., additional, Ofman, Gaston, additional, Nitsos, Ilias, additional, Pillow, J. Jane, additional, Polglase, Graeme R., additional, Kemp, Matthew W., additional, Newnham, John P., additional, Gavilanes, Antonio W. D., additional, Nowacki, Relana, additional, Ikegami, Machiko, additional, Jobe, Alan H., additional, and Kallapur, Suhas G., additional
- Published
- 2012
- Full Text
- View/download PDF
17. Chronic Fetal Exposure to Ureaplasma parvum Suppresses Innate Immune Responses in Sheep
- Author
-
Kallapur, Suhas G., primary, Kramer, Boris W., additional, Knox, Christine L., additional, Berry, Clare A., additional, Collins, Jennifer J. P., additional, Kemp, Matthew W., additional, Nitsos, Ilias, additional, Polglase, Graeme R., additional, Robinson, James, additional, Hillman, Noah H., additional, Newnham, John P., additional, Chougnet, Claire, additional, and Jobe, Alan H., additional
- Published
- 2011
- Full Text
- View/download PDF
18. Pulmonary and systemic inflammatory responses to intra-amniotic IL-1α in fetal sheep
- Author
-
Kallapur, Suhas G., primary, Kramer, Boris W., additional, Nitsos, Ilias, additional, Pillow, J. Jane, additional, Collins, Jennifer J. P., additional, Polglase, Graeme R., additional, Newnham, John P., additional, and Jobe, Alan H., additional
- Published
- 2011
- Full Text
- View/download PDF
19. Inflammation in fetal sheep from intra-amniotic injection of Ureaplasma parvum
- Author
-
Collins, Jennifer J. P., primary, Kallapur, Suhas G., additional, Knox, Christine L., additional, Nitsos, Ilias, additional, Polglase, Graeme R., additional, Pillow, J. Jane, additional, Kuypers, Elke, additional, Newnham, John P., additional, Jobe, Alan H., additional, and Kramer, Boris W., additional
- Published
- 2010
- Full Text
- View/download PDF
20. The mammalian myotome: a muscle with no innervation
- Author
-
Deries, Marianne, primary, Collins, Jennifer J. P., additional, and Duxson, Marilyn J., additional
- Published
- 2008
- Full Text
- View/download PDF
21. Cerebral inflammation and mobilization of the peripheral immune system following global hypoxia-ischemia in preterm sheep.
- Author
-
Jellema, Reint K., Passos, Valéria Lima, Zwanenburg, Alex, Ophelders, Daan RMG, De Munter, Stephanie, Vanderlocht, Joris, Germeraad, Wilfred TV, Kuypers, Elke, Collins, Jennifer J. P., Cleutjens, Jack P. M., Jennekens, Ward, Gavilanes, Antonio W. D., Seehase, Matthias, Vles, Hans J., Steinbusch, Harry, Andriessen, Peter, Wolfs, Tim GAM, and Kramer, Boris W.
- Subjects
INFLAMMATION ,IMMUNE system ,HYPOXEMIA ,ISCHEMIA ,BRAIN injuries ,PREMATURE infants ,SHEEP as laboratory animals - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
22. Pulmonary and systemic inflammatory responses to intra-amniotic IL-1 α in fetal sheep.
- Author
-
Kallapur, Suhas G., Kramer, Boris W., Nitsos, Ilias, Pillow, J. Jane, Collins, Jennifer J. P., Polglase, Graeme R., Newnham, John P., and Jobe, Alan H.
- Subjects
AMNIOTIC liquid examination ,BRONCHOALVEOLAR lavage ,SHEEP as laboratory animals ,GESTATIONAL age ,ANIMAL models in research ,PULMONARY pharmacology ,EPIDEMIOLOGY - Abstract
Clinical and epidemiological studies implicate IL-1 as an important mediator of perinatal inflammation. We tested the hypothesis that intra-amniotic IL-1α would induce pulmonary and systemic fetal inflammatory responses. Sheep with singleton fetuses were given an intra-amniotic injection of recombinant sheep IL-1α (100 μg) and were delivered 1, 3, or 7 days later, at 124 ± 1 days gestation (n=5-8/group). A separate group of sheep were given two intra-amniotic IL-1α injections (100 μg dose each): 7 days and again 1 day prior to delivery. IL-1α induced a robust increase in monocytes, neutrophils, lymphocytes, and IL-8 protein in bronchoalveolar lavage fluid. H
2 O2 secretion was increased in inflammatory cells isolated from lungs of IL-1α-exposed lambs upon LPS challenge in vitro compared with control monocytes. T lymphocytes were recruited to the lung. IL-1β, cyclooxygenase-1, and cyclooxygenase-2 mRNA expression increased in the lung 1 day after intra-amniotic IL-1α exposure. Lung volumes increased 7 days after intra-amniotic IL-1α exposure, with minimal anatomic changes in air space morphology. The weight of the posterior mediastinal lymph node draining the lung and the gastrointestinal tract doubled, inducible nitric oxide synthase (NOSII)-positive cells increased, and Foxp3-positive T-regulatory lymphocytes decreased in the lymph node after IL-1α exposure. In the blood, neutrophil counts and plasma haptoglobin increased after IL-1α exposure. Compared with a single exposure, exposure to intra-amniotic IL-1α 7 days and again 1 day before delivery had a variable effect (increases in some inflammatory markers, but not pulmonary cytokines). IL-1α is a potent mediator of the fetal inflammatory response syndrome. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
23. The Dog Can’t Eat Your Homework: 9 Benefits to E-filing.
- Author
-
COLLINS, JENNIFER J.
- Subjects
ELECTRONIC filing systems ,COST control - Abstract
The article presents the benefits of electronic filing which includes immediate access to the court's file, time saving, and cost reduction.
- Published
- 2017
24. Key paediatric messages from the 2018 European Respiratory Society International Congress
- Author
-
Deborah Snijders, Graham L. Hall, Jürg Barben, Bulent Karadag, Jonathan Grigg, Jennifer J. P. Collins, Andrew Prayle, Heidi Makrinioti, Bart L. Rottier, Bruna Rubbo, Myrofora Goutaki, Giuseppe Fabio Parisi, Raffaella Nenna, Norrice M. Liu, Katie A. Hunt, Ricardo M. Fernandes, Robbert J. Rottier, Mariëlle W. Pijnenburg, Theodore Dassios, Pediatric Surgery, Pediatrics, Nenna, Raffaella, Hunt, Katie A., Dassios, Theodore, Collins, Jennifer J. P., Rottier, Robbert J., Liu, Norrice M., Rottier, Bart, Goutaki, Myrofora, Karadag, Bulent, Prayle, Andrew, Fernandes, Ricardo M., Parisi, Giuseppe Fabio, Barben, Jurg, Rubbo, Bruna, Snijders, Deborah, Makrinioti, Heidi, Hall, Graham, Pijnenburg, Marielle W., Grigg, Jonathan, and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,education ,AZITHROMYCIN ,Congress Highlights ,lcsh:Medicine ,CHILDREN ,610 Medicine & health ,DIAGNOSIS ,03 medical and health sciences ,0302 clinical medicine ,360 Social problems & social services ,PRETERM INFANTS ,International congress ,medicine ,030212 general & internal medicine ,Early career ,Lung function ,health care economics and organizations ,MUTATIONS ,business.industry ,lcsh:R ,BRONCHOPULMONARY DYSPLASIA ,PREVENTION ,humanities ,LUNG-FUNCTION ,030228 respiratory system ,Family medicine ,ASTHMA ,PRIMARY CILIARY DYSKINESIA ,business - Abstract
In this article, the Group Chairs and early career members of the European Respiratory Society (ERS) Paediatric Assembly highlight some of the most interesting findings in the field of paediatrics which were presented at the 2018 international ERS Congress., The 2018 international #ERSCongress in Paris, France reflected the @ERSTalk's commitment to support the continuing professional and academic development of the 1500 members of the Paediatric Assembly. http://ow.ly/P2Ck50pDRy6
- Published
- 2019
25. Early origins of lung disease: towards an interdisciplinary approach.
- Author
-
Ubags NDJ, Alejandre Alcazar MA, Kallapur SG, Knapp S, Lanone S, Lloyd CM, Morty RE, Pattaroni C, Reynaert NL, Rottier RJ, Smits HH, de Steenhuijsen Piters WAA, Strickland DH, and Collins JJP
- Subjects
- Animals, Chronic Disease, Environmental Exposure, Female, Humans, Lung, Pregnancy, Lung Diseases diagnosis, Lung Diseases epidemiology, Lung Diseases etiology, Respiratory Tract Diseases
- Abstract
The prenatal and perinatal environments can have profound effects on the development of chronic inflammatory diseases. However, mechanistic insight into how the early-life microenvironment can impact upon development of the lung and immune system and consequent initiation and progression of respiratory diseases is still emerging. Recent studies investigating the developmental origins of lung diseases have started to delineate the effects of early-life changes in the lung, environmental exposures and immune maturation on the development of childhood and adult lung diseases. While the influencing factors have been described and studied in mostly animal models, it remains challenging to pinpoint exactly which factors and at which time point are detrimental in lung development leading to respiratory disease later in life. To advance our understanding of early origins of chronic lung disease and to allow for proper dissemination and application of this knowledge, we propose four major focus areas: 1) policy and education; 2) clinical assessment; 3) basic and translational research; and 4) infrastructure and tools, and discuss future directions for advancement. This review is a follow-up of the discussions at the European Respiratory Society Research Seminar "Early origins of lung disease: towards an interdisciplinary approach" (Lisbon, Portugal, November 2019)., Competing Interests: Conflict of interest: N.D.J. Ubags has nothing to disclose. Conflict of interest: M.A. Alejandre Alcazar reports grants from Deutsche Forschungsgemeinschaft, Marga and Walter Boll Stiftung and Stiftung Oskar-Helene-Heim, during the conduct of the study. Conflict of interest: S.G. Kallapur has nothing to disclose. Conflict of interest: S. Knapp reports grants from Austrian Science Fund, during the conduct of the study. Conflict of interest: S. Lanone has nothing to disclose. Conflict of interest: C.M. Lloyd has nothing to disclose. Conflict of interest: R.E. Morty has nothing to disclose. Conflict of interest: C. Pattaroni has nothing to disclose. Conflict of interest: N.L. Reynaert reports grants from Lung Foundation Netherlands, during the conduct of the study. Conflict of interest: R.J. Rottier has nothing to disclose. Conflict of interest: H.H. Smits has nothing to disclose. Conflict of interest: W.A.A. de Steenhuijsen Piters has nothing to disclose. Conflict of interest: D.H. Strickland has nothing to disclose. Conflict of interest: J.J.P. Collins reports grants from Lung Foundation Netherlands, during the conduct of the study., (Copyright ©ERS 2020.)
- Published
- 2020
- Full Text
- View/download PDF
26. A sneak peek into the Early Career Members' session at the ERS International Congress and the experience of organising an ERS Research Seminar.
- Author
-
Collins JJP, Bartel S, Ubags ND, Gompelmann D, and De Brandt J
- Abstract
A sneak peek into the @EarlyCareerERS session at #ERSCongress 2020 and the experience of organising an @EuroRespSoc Research Seminar http://bit.ly/39yncgO., Competing Interests: Conflict of interest: J.J.P. Collins has nothing to disclose. Conflict of interest: S. Bartel reports grants and personal fees from Bencard Allergie GmbH outside the submitted work. Conflict of interest: N.D. Ubags has nothing to disclose. Conflict of interest: D. Gompelmannreports personal fees from Pulmonx, Novartis, Boehringer Ingelheim, Chiesi, AstraZeneca, Berlin Chemie, Uptake Medical, Grifols and Olympus, outside the submitted work. Conflict of interest: J. De Brandt has nothing to disclose., (Copyright ©ERS 2020.)
- Published
- 2020
- Full Text
- View/download PDF
27. Key paediatric messages from the 2018 European Respiratory Society International Congress.
- Author
-
Nenna R, Hunt KA, Dassios T, Collins JJP, Rottier RJ, Liu NM, Rottier B, Goutaki M, Karadag B, Prayle A, Fernandes RM, Parisi GF, Barben J, Rubbo B, Snijders D, Makrinioti H, Hall G, Pijnenburg MW, and Grigg J
- Abstract
In this article, the Group Chairs and early career members of the European Respiratory Society (ERS) Paediatric Assembly highlight some of the most interesting findings in the field of paediatrics which were presented at the 2018 international ERS Congress., Competing Interests: Conflict of interest: R. Nenna has nothing to disclose. Conflict of interest: K.A. Hunt reports personal fees from Chiesi and Medimmune, and grants from SLE Ltd, outside the submitted work. Conflict of interest: T. Dassios has nothing to disclose. Conflict of interest: J.J.P. Collins has nothing to disclose. Conflict of interest: R.J. Rottier has nothing to disclose. Conflict of interest: N.M. Liu has nothing to disclose. Conflict of interest: B. Rottier has nothing to disclose. Conflict of interest: M. Goutaki has nothing to disclose. Conflict of interest: B. Karadag has nothing to disclose. Conflict of interest: A. Prayle has nothing to disclose. Conflict of interest: R.M. Fernandes has served on independent data monitoring committees for studies sponsored by Ablynx, and participated in clinical trials sponsored by Janssen. Conflict of interest: G.F. Parisi has nothing to disclose. Conflict of interest: J. Barben has nothing to disclose. Conflict of interest: B. Rubbo has nothing to disclose. Conflict of interest: D. Snijders has nothing to disclose. Conflict of interest: H. Makrinoti has nothing to disclose. Conflict of interest: G. Hall has nothing to disclose. Conflict of interest: M.W. Pijnenburg has nothing to disclose. Conflict of interest: J. Grigg reports personal fees from GSK, Vifor Pharmaceuticals, AstraZeneca and Respiri UK, outside the submitted work.
- Published
- 2019
- Full Text
- View/download PDF
28. Impaired Angiogenic Supportive Capacity and Altered Gene Expression Profile of Resident CD146 + Mesenchymal Stromal Cells Isolated from Hyperoxia-Injured Neonatal Rat Lungs.
- Author
-
Collins JJP, Lithopoulos MA, Dos Santos CC, Issa N, Möbius MA, Ito C, Zhong S, Vadivel A, and Thébaud B
- Subjects
- Animals, Animals, Newborn, Bronchopulmonary Dysplasia etiology, Bronchopulmonary Dysplasia physiopathology, CD146 Antigen genetics, Cell Proliferation drug effects, Epithelial Cells drug effects, Epithelial Cells pathology, Gene Expression Regulation, Developmental drug effects, Humans, Lung metabolism, Lung pathology, Lung Injury chemically induced, Lung Injury pathology, Mesenchymal Stem Cells pathology, Oxygen administration & dosage, Rats, T-Lymphocytes metabolism, T-Lymphocytes pathology, Bronchopulmonary Dysplasia metabolism, Lung Injury metabolism, Mesenchymal Stem Cells metabolism, Oxygen adverse effects
- Abstract
Bronchopulmonary dysplasia (BPD), the most common complication of extreme preterm birth, can be caused by oxygen-related lung injury and is characterized by impaired alveolar and vascular development. Mesenchymal stromal cells (MSCs) have lung protective effects. Conversely, BPD is associated with increased MSCs in tracheal aspirates. We hypothesized that endogenous lung (L-)MSCs are perturbed in a well-established oxygen-induced rat model mimicking BPD features. Rat pups were exposed to 21% or 95% oxygen from birth to postnatal day 10. On day 12, CD146
+ L-MSCs were isolated and characterized according to the International Society for Cellular Therapy criteria. Epithelial and vascular repair potential were tested by scratch assay and endothelial network formation, respectively, immune function by mixed lymphocyte reaction assay. Microarray analysis was performed using the Affymetrix GeneChip and gene set enrichment analysis software. CD146+ L-MSCs isolated from rat pups exposed to hyperoxia had decreased CD73 expression and inhibited lung endothelial network formation. CD146+ L-MSCs indiscriminately promoted epithelial wound healing and limited T cell proliferation. Expression of potent antiangiogenic genes of the axonal guidance cue and CDC42 pathways was increased after in vivo hyperoxia, whereas genes of the anti-inflammatory Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and lung/vascular growth-promoting fibroblast growth factor (FGF) pathways were decreased. In conclusion, in vivo hyperoxia exposure alters the proangiogenic effects and FGF expression of L-MSCs. In addition, decreased CD73 and JAK/STAT expression suggests decreased immune function. L-MSC function may be perturbed and contribute to BPD pathogenesis. These findings may lead to improvements in manufacturing exogenous MSCs with superior repair capabilities.- Published
- 2018
- Full Text
- View/download PDF
29. Early Career Members at the ERS Lung Science Conference: cell-matrix interactions in lung disease and regeneration: Early career forum.
- Author
-
Almendros I, Alsafadi HN, Bölükbas D, Collins JJP, Duch P, Garrido-Martin EM, Kahn N, Karampitsakos T, Mahmutovic Persson I, Tzouvelekis A, Uhl FE, and Bartel S
- Abstract
.@EarlyCareerERS looks back on #LSC2018 http://ow.ly/6hjS30jB6P9., Competing Interests: Conflict of interest: S. Bartel has received personal fees from Bencard Allergie GmbH for serving as a member of an advisory board and a project grant from Bencard Allergie GmbH, outside the submitted work.
- Published
- 2018
- Full Text
- View/download PDF
30. Human induced pluripotent stem cell-derived lung progenitor and alveolar epithelial cells attenuate hyperoxia-induced lung injury.
- Author
-
Shafa M, Ionescu LI, Vadivel A, Collins JJP, Xu L, Zhong S, Kang M, de Caen G, Daneshmand M, Shi J, Fu KZ, Qi A, Wang Y, Ellis J, Stanford WL, and Thébaud B
- Subjects
- Animals, Animals, Newborn, Cell Differentiation, Disease Models, Animal, Humans, Induced Pluripotent Stem Cells ultrastructure, Lung Injury pathology, Mice, Mice, Inbred NOD, Mice, SCID, Oxygen, Teratoma pathology, Alveolar Epithelial Cells cytology, Hyperoxia complications, Induced Pluripotent Stem Cells cytology, Lung Injury etiology, Lung Injury therapy
- Abstract
Background Aims: Bronchopulmonary dysplasia (BPD), a chronic lung disease characterized by disrupted lung growth, is the most common complication in extreme premature infants. BPD leads to persistent pulmonary disease later in life. Alveolar epithelial type 2 cells (AEC2s), a subset of which represent distal lung progenitor cells (LPCs), promote normal lung growth and repair. AEC2 depletion may contribute to persistent lung injury in BPD. We hypothesized that induced pluripotent stem cell (iPSC)-derived AECs prevent lung damage in experimental oxygen-induced BPD., Methods: Mouse AECs (mAECs), miPSCs/mouse embryonic stem sells, human umbilical cord mesenchymal stromal cells (hUCMSCs), human (h)iPSCs, hiPSC-derived LPCs and hiPSC-derived AECs were delivered intratracheally to hyperoxia-exposed newborn mice. Cells were pre-labeled with a red fluorescent dye for in vivo tracking., Results: Airway delivery of primary mAECs and undifferentiated murine pluripotent cells prevented hyperoxia-induced impairment in lung function and alveolar growth in neonatal mice. Similar to hUCMSC therapy, undifferentiated hiPSCs also preserved lung function and alveolar growth in hyperoxia-exposed neonatal NOD/SCID mice. Long-term assessment of hiPSC administration revealed local teratoma formation and cellular infiltration in various organs. To develop a clinically relevant cell therapy, we used a highly efficient method to differentiate hiPSCs into a homogenous population of AEC2s. Airway delivery of hiPSC-derived AEC2s and hiPSC-derived LPCs, improved lung function and structure and resulted in long-term engraftment without evidence of tumor formation., Conclusions: hiPSC-derived AEC2 therapy appears effective and safe in this model and warrants further exploration as a therapeutic option for BPD and other lung diseases characterized by AEC injury., (Copyright © 2017 International Society for Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Human Umbilical Cord Mesenchymal Stromal Cells Improve Survival and Bacterial Clearance in Neonatal Sepsis in Rats.
- Author
-
Zhu Y, Xu L, Collins JJP, Vadivel A, Cyr-Depauw C, Zhong S, Mense L, Möbius MA, and Thébaud B
- Subjects
- Animals, Antimicrobial Cationic Peptides, Cathelicidins blood, Escherichia coli physiology, Humans, Inflammation pathology, Lung pathology, Macrophages metabolism, Neonatal Sepsis blood, Neutrophils metabolism, Phagocytosis, Rats, Spleen pathology, Survival Analysis, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells cytology, Neonatal Sepsis microbiology, Neonatal Sepsis therapy, Umbilical Cord cytology
- Abstract
Sepsis is the main cause of morbidity and mortality in neonates. Mesenchymal stromal cells (MSCs) are potent immune-modulatory cells. Their effect in neonatal sepsis has never been explored. We hypothesized that human umbilical cord-derived MSCs (hUC-MSCs) improve survival in experimental neonatal sepsis. Sepsis was induced in 3-day-old rats by intravenous injection of Escherichia coli (5 × 10
5 /rat). One hour after infection, rats were treated intravenously with normal saline, hUC-MSCs, or with interferon-γ preconditioned hUC-MSCs (107 cells/kg). Eighteen hours after infection, survival, bacterial counts, lung neutrophil and macrophage influx, phagocytosis and apoptosis of splenocytes plasma, and LL-37 concentration were evaluated. Animals were observed for survival for 72 h after E. coli injection. Treatment with either hUC-MSCs or preconditioned hUC-MSCs significantly increased survival (hUC-MSCs, 81%; preconditioned hUC-MSCs, 89%; saline, 51%; P < 0.05). Both hUC-MSCs and preconditioned hUC-MSCs enhanced bacterial clearance. Lung neutrophil influx was decreased with preconditioned hUC-MSCs. The number of activated macrophages (CD206+ ) in the spleen was increased with hUC-MSCs and preconditioned hUC-MSCs; preconditioned hUC-MSCs increased the phagocytic activity of CD206+ macrophages. hUC-MSCs and preconditioned hUC-MSCs decreased splenocyte apoptosis in E. coli infected rats. Finally, LL-37 plasma levels were elevated in neonatal rats treated with hUC-MSCs or preconditioned hUC-MSCs. hUC-MSCs enhance survival and bacterial clearance in experimental neonatal sepsis. hUC-MSCs may be an effective adjunct therapy to reduce neonatal sepsis-related morbidity and mortality.- Published
- 2017
- Full Text
- View/download PDF
32. Isolation of CD146+ Resident Lung Mesenchymal Stromal Cells from Rat Lungs.
- Author
-
Collins JJ, Möbius MA, and Thébaud B
- Subjects
- Animals, Biomarkers, Cell Differentiation, Cell- and Tissue-Based Therapy, Rats, Lung, Mesenchymal Stem Cells
- Abstract
Mesenchymal stromal cells (MSCs) are increasingly recognized for their therapeutic potential in a wide range of diseases, including lung diseases. Besides the use of bone marrow and umbilical cord MSCs for exogenous cell therapy, there is also increasing interest in the repair and regenerative potential of resident tissue MSCs. Moreover, they likely have a role in normal organ development, and have been attributed roles in disease, particularly those with a fibrotic nature. The main hurdle for the study of these resident tissue MSCs is the lack of a clear marker for the isolation and identification of these cells. The isolation technique described here applies multiple characteristics of lung resident MSCs (L-MSCs). Upon sacrifice of the rats, lungs are removed and rinsed multiple times to remove blood. Following mechanical dissociation by scalpel, the lungs are digested for 2-3 hr using a mix of collagenase type I, neutral protease and DNase type I. The obtained single cell suspension is subsequently washed and layered over density gradient medium (density 1.073 g/ml). After centrifugation, cells from the interphase are washed and plated in culture-treated flasks. Cells are cultured for 4-7 days in physiological 5% O2, 5% CO2 conditions. To deplete fibroblasts (CD146(-)) and to ensure a population of only L-MSCs (CD146(+)), positive selection for CD146(+) cells is performed through magnetic bead selection. In summary, this procedure reliably produces a population of primary L-MSCs for further in vitro study and manipulation. Because of the nature of the protocol, it can easily be translated to other experimental animal models.
- Published
- 2016
- Full Text
- View/download PDF
33. Lung mesenchymal stromal cells in development and disease: to serve and protect?
- Author
-
Collins JJ and Thébaud B
- Subjects
- Animals, Animals, Newborn, Humans, Infant, Newborn, Infant, Premature, Bronchopulmonary Dysplasia pathology, Lung pathology, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells pathology
- Abstract
Significance: Bronchopulmonary dysplasia (BPD) is a disease of the developing lung that afflicts extreme preterm infants in the neonatal intensive care unit. Follow-up studies into adulthood show that BPD is not merely a problem of the neonatal period, as it also may predispose to early-onset emphysema and poor lung function in later life., Recent Advances: The increasing promise of bone marrow- or umbilical cord-derived mesenchymal stromal cells (MSCs) to repair neonatal and adult lung diseases may for the first time offer the chance to make substantial strides in improving the outcome of extreme premature infants at risk of developing BPD. As more knowledge has been obtained on MSCs over the past decades, it has become clear that each organ has its own reservoir of endogenous MSCs, including the lung., Critical Issues: We have only barely scratched the surface on what resident lung MSCs exactly are and what their role and function in lung development may be. Moreover, what happens to these putative repair cells in BPD when alveolar development goes awry and why do their counterparts from the bone marrow and umbilical cord succeed in restoring normal alveolar development when they themselves do not?, Future Directions: Much work remains to be carried out to validate lung MSCs, but with the high potential of MSC-based treatment for BPD and other lung diseases, a thorough understanding of the endogenous lung MSC will be pivotal to get to the bottom of these diseases.
- Published
- 2014
- Full Text
- View/download PDF
34. Effects of intra-amniotic lipopolysaccharide exposure on the fetal lamb lung as gestation advances.
- Author
-
Lambermont VA, Kuypers E, Collins JJ, Pillow JJ, Newnham JP, Polglase GR, Nitsos I, Kemp MW, Jobe AH, Kallapur SG, and Kramer BW
- Subjects
- Animals, Body Weight, Female, Lung drug effects, Organ Size, Pregnancy, Lipopolysaccharides pharmacology, Lung embryology, Pregnancy, Animal, Sheep embryology
- Abstract
Background: Intra-amniotic lipopolysaccharide (LPS) exposure may affect neonatal outcome by altering fetal lung and immune system development. We hypothesized that intra-amniotic LPS exposure would cause persistent fetal pulmonary responses as the lungs develop in utero., Methods: Fetal lambs were exposed to intra-amniotic LPS at 118 or at 118 and 123 d of gestational age (GA) with delivery at 125, 133, or 140 d (term = 147 d). Immune responses, PU.1 expression, Toll-like receptor (TLR)-1,2,4,6 mRNA levels, mast cell levels, and pulmonary elastin deposition were evaluated., Results: After a single dose of LPS, pulmonary inflammatory responses were observed with increases of (i) PU.1 and TLR1 at 125 d GA and (ii) monocytes, lymphocytes, TLR2, and TLR6 at 133 d GA. Repetitive LPS exposure resulted in (i) increases of neutrophils, monocytes, PU.1, and TLR1 at 125 d GA; (ii) increases of neutrophils, PU.1, and TLR2 at 133 d GA; and (iii) decreases of mast cells, elastin foci, TLR4, and TLR6 at early gestation. At 140 d GA, only PU.1 was increased after repetitive LPS exposure., Conclusion: The preterm fetal lung can respond to a single exposure or repeated exposures from intra-amniotic LPS in multiple ways, but the absence of inflammatory and structural changes in LPS-exposed fetuses delivered near term suggest that the fetus can resolve an inflammatory stimulus in utero with time.
- Published
- 2014
- Full Text
- View/download PDF
35. Progenitor cells of the distal lung and their potential role in neonatal lung disease.
- Author
-
Collins JJ and Thébaud B
- Subjects
- Humans, Infant, Newborn, Bronchopulmonary Dysplasia metabolism, Bronchopulmonary Dysplasia pathology, Bronchopulmonary Dysplasia physiopathology, Infant, Extremely Premature, Lung metabolism, Lung pathology, Lung physiopathology, Stem Cells
- Abstract
Bronchopulmonary dysplasia (BPD) is the most common adverse outcome in extreme preterm neonates (born before 28 weeks gestation). BPD is characterized by interrupted lung growth and may predispose to early-onset emphysema and poor lung function in later life. At present, there is no treatment for BPD. Recent advances in stem/progenitor cell biology have enabled the exploration of endogenous lung progenitor populations in health and disease. In parallel, exogenous stem/progenitor cell administration has shown promise in protecting the lung from injury in the experimental setting. This review will provide an outline of the progenitor populations that have currently been identified in all tissue compartments of the distal lung and how they may be affected in BPD. A thorough understanding of the lung's endogenous progenitor populations during normal development, injury and repair may one day allow us to harness their regenerative capacity., (Copyright © 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
36. 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
37. 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
38. Thrown off balance: the effect of antenatal inflammation on the developing lung and immune system.
- Author
-
Kunzmann S, Collins JJ, Kuypers E, and Kramer BW
- Subjects
- Animals, Female, Humans, Immune Tolerance physiology, Pregnancy, Chorioamnionitis physiopathology, Immune System physiology, Inflammation physiopathology, Lung physiology, Peripartum Period
- Abstract
In recent years, translational research with various animal models has been helpful to answer basic questions about the effect of antenatal inflammation on maturation and development of the fetal lung and immune system. The fetal lung and immune systems are very plastic and their development can be conditioned and influenced by both endogenous and/or exogenous factors. Antenatal inflammation can induce pulmonary inflammation, leading to lung injury and remodeling in the fetal lung. Exposure to antenatal inflammation can induce interleukin-1α production, which enhances surfactant protein and lipid synthesis thereby promoting lung maturation. Interleukin-1α is therefore a candidate for the link between lung inflammation and lung maturation, preventing respiratory distress syndrome in preterm infants. Antenatal inflammation can, however, cause structural changes in the fetal lung and affect the expression of growth factors, such as transforming growth factor-beta, connective tissue growth factor, fibroblast growth factor-10, or bone morphogenetic protein-4, which are essential for branching morphogenesis. These alterations cause alveolar and microvascular simplification resembling the histology of bronchopulmonary dysplasia. Antenatal inflammation may also affect neonatal outcome by modulating the responsiveness of the immune system. Lipopolysaccharide-tolerance (endotoxin hyporesponsiveness/immunoparalysis), induced by exposure to inflammation in utero, may prevent fetal lung damage, but increases susceptibility to postnatal infections. Moreover, prenatal exposure to inflammation appears to be a predisposition for the development of adverse neonatal outcomes, like bronchopulmonary dysplasia, if the preterm infant is exposed to a second postnatal hit, such as mechanical ventilation oxygen exposure, infections, or steroids., (Copyright © 2013 Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
39. Propofol administration to the fetal-maternal unit reduces cardiac injury in late-preterm lambs subjected to severe prenatal asphyxia and cardiac arrest.
- Author
-
Seehase M, Houthuizen P, Jellema RK, Collins JJ, Bekers O, Breuer J, and Kramer BW
- Subjects
- Anesthetics, Inhalation administration & dosage, Animals, Animals, Newborn, Asphyxia Neonatorum blood, Asphyxia Neonatorum physiopathology, Biomarkers blood, Caspase 3 metabolism, Disease Models, Animal, Enzyme Activation, Female, Gestational Age, Heart Arrest blood, Heart Arrest physiopathology, Humans, Infant, Newborn, Isoflurane administration & dosage, Ligation, Myocardium metabolism, Phosphorylation, Pregnancy, Proto-Oncogene Proteins c-akt metabolism, Respiration, Artificial, Resuscitation, STAT3 Transcription Factor metabolism, Sheep, Stroke Volume drug effects, Time Factors, Troponin T blood, Ultrasonography, Umbilical Cord surgery, Ventricular Dysfunction, Left blood, Ventricular Dysfunction, Left diagnostic imaging, Ventricular Dysfunction, Left physiopathology, Ventricular Function, Left drug effects, Anesthetics, Intravenous administration & dosage, Asphyxia Neonatorum therapy, Cesarean Section, Heart Arrest therapy, Myocardium pathology, Premature Birth, Propofol administration & dosage, Ventricular Dysfunction, Left prevention & control
- Abstract
Background: Cardiac dysfunction is reported to occur after severe perinatal asphyxia. We hypothesized that anesthesia of the mother with propofol during emergency cesarean section (c-section) would result in less cardiac injury (troponin T) in preterm fetuses exposed to global severe asphyxia in utero than anesthesia with isoflurane. We tested whether propofol decreases the activity of proapoptotic caspase-3 by activating the antiapoptotic AKT kinase family and the signal transducer and activator of transcription-3 (STAT-3)., Methods: Pregnant ewes were randomized to receive either propofol or isoflurane anesthesia. A total of 44 late-preterm lambs were subjected to in utero umbilical cord occlusion (UCO), resulting in asphyxia and cardiac arrest, or sham treatment. After emergency c-section, each fetus was resuscitated, mechanically ventilated, and supported under anesthesia for 8 h using the same anesthetic as the one received by its mother., Results: At 8 h after UCO, the fetuses whose mothers had received propofol anesthesia had lower plasma troponin T levels, and showed a trend toward a higher median left ventricular ejection fraction (LVEF) of 84% as compared with 74% for those whose mothers had received isoflurane. Postasphyxia activation of caspase-3 was lower in association with propofol anesthesia than with isoflurane. Postasphyxia levels of STAT-3 and the AKT kinase family rose 655% and 500%, respectively with the use of propofol anesthesia for the mother., Conclusion: The use of propofol for maternal anesthesia results in less cardiac injury in late-preterm lambs subjected to asphyxia than the use of isoflurane anesthesia. The underlying mechanism may be activation of the antiapoptotic STAT-3 and AKT pathways.
- Published
- 2013
- Full Text
- View/download PDF
40. Antenatal glucocorticoids counteract LPS changes in TGF-β pathway and caveolin-1 in ovine fetal lung.
- Author
-
Collins JJ, Kunzmann S, Kuypers E, Kemp MW, Speer CP, Newnham JP, Kallapur SG, Jobe AH, and Kramer BW
- Subjects
- Animals, Caveolin 1 genetics, Connective Tissue Growth Factor genetics, Connective Tissue Growth Factor metabolism, Female, Fetus drug effects, Fetus immunology, Fetus metabolism, Gene Expression, Gene Expression Regulation drug effects, Injections, Intramuscular, Lipopolysaccharides pharmacology, Lung drug effects, Lung immunology, Maternal-Fetal Exchange, Phosphorylation, Pregnancy, Protein Processing, Post-Translational, Sheep, Smad2 Protein metabolism, Transforming Growth Factor beta metabolism, Betamethasone administration & dosage, Caveolin 1 metabolism, Glucocorticoids administration & dosage, Lung metabolism, Transforming Growth Factor beta physiology
- Abstract
Inflammation and antenatal glucocorticoids, the latter given to mothers at risk for preterm birth, affect lung development and may contribute to the development of bronchopulmonary dysplasia (BPD). The effects of the combined exposures on inflammation and antenatal glucocorticoids on transforming growth factor (TGF)-β signaling are unknown. TGF-β and its downstream mediators are implicated in the etiology of BPD. Therefore, we asked whether glucocorticoids altered intra-amniotic lipopolysaccharide (LPS) effects on TGF-β expression, its signaling molecule phosphorylated sma and mothers against decapentaplegic homolog 2 (pSmad2), and the downstream mediators connective tissue growth factor (CTGF) and caveolin-1 (Cav-1). Ovine singleton fetuses were randomized to receive either an intra-amniotic injection of LPS and/or maternal betamethasone (BTM) intramuscularly 7 and/or 14 days before delivery at 120 days gestational age (GA; term = 150 days GA). Saline was used for controls. Protein levels of TGF-β1 and -β2 were measured by ELISA. Smad2 phosphorylation was assessed by immunohistochemistry and Western blot. CTGF and Cav-1 mRNA and protein levels were determined by RT-PCR and Western blot. Free TGF-β1 and -β2 and total TGF-β1 levels were unchanged after LPS and/or BTM exposure, although total TGF-β2 increased in animals exposed to BTM 7 days before LPS. pSmad2 immunostaining increased 7 days after LPS exposure although pSmad2 protein expression did not increase. Similarly, CTGF mRNA and protein levels increased 7 days after LPS exposure as Cav-1 mRNA and protein levels decreased. BTM exposure before LPS prevented CTGF induction and Cav-1 downregulation. This study demonstrated that the intrauterine inflammation-induced TGF-β signaling can be inhibited by antenatal glucocorticoids in fetal lungs.
- Published
- 2013
- Full Text
- View/download PDF
41. Repeated intrauterine exposures to inflammatory stimuli attenuated transforming growth factor-β signaling in the ovine fetal lung.
- Author
-
Collins JJ, Kallapur SG, Knox CL, Kemp MW, Kuypers E, Zimmermann LJ, Newnham JP, Jobe AH, and Kramer BW
- Subjects
- Amnion drug effects, Animals, Chorioamnionitis, Disease Models, Animal, Elastin analysis, Female, Gestational Age, Lipopolysaccharides administration & dosage, Lung chemistry, Phosphorylation, Pregnancy, Receptors, Transforming Growth Factor beta analysis, Smad2 Protein metabolism, Transforming Growth Factor beta1 analysis, Transforming Growth Factor beta2 analysis, Transforming Growth Factor beta3 analysis, Ureaplasma, Inflammation physiopathology, Lung embryology, Pregnancy Complications physiopathology, Sheep embryology, Signal Transduction, Transforming Growth Factor beta physiology
- Abstract
Background: Bronchopulmonary dysplasia (BPD) is one of the most common complications after preterm birth and is associated with intrauterine exposure to bacteria. Transforming growth factor-β (TGFβ) is implicated in the development of BPD., Objectives: We hypothesized that different and/or multiple bacterial signals could elicit divergent TGFβ signaling responses in the developing lung., Methods: Time-mated pregnant Merino ewes received an intra-amniotic injection of lipopolysaccharide (LPS) and/or Ureaplasma parvum serovar 3 (UP) at 117 days' and/or 121/122 days' gestational age (GA). Controls received an equivalent injection of saline and or media. Lambs were euthanized at 124 days' GA (term = 150 days' GA). TGFβ1, TGFβ2, TGFβ3, TGFβ receptor (R)1 and TGFβR2 protein levels, Smad2 phosphorylation and elastin deposition were evaluated in lung tissue., Results: Total TGFβ1 and TGFβ2 decreased by 24 and 51% after combined UP+LPS exposure, whereas total TGFβ1 increased by 31% after 7 days' LPS exposure but not after double exposures. Alveolar expression of TGFβR2 decreased 75% after UP, but remained unaltered after double exposures. Decreased focal elastin deposition after single LPS exposure was prevented by double exposures., Conclusions: TGFβ signaling components and elastin responded differently to intrauterine LPS and UP exposure. Multiple bacterial exposures attenuated TGFβ signaling and normalized elastin deposition., (Copyright © 2013 S. Karger AG, Basel.)
- Published
- 2013
- Full Text
- View/download PDF
42. LPS-induced chorioamnionitis and antenatal corticosteroids modulate Shh signaling in the ovine fetal lung.
- Author
-
Collins JJ, Kuypers E, Nitsos I, Jane Pillow J, Polglase GR, Kemp MW, Newnham JP, Cleutjens JP, Frints SG, Kallapur SG, Jobe AH, and Kramer BW
- Subjects
- Animals, Betamethasone therapeutic use, Bone Morphogenetic Protein 4 genetics, Bone Morphogenetic Protein 4 metabolism, Cell Proliferation, Chorioamnionitis drug therapy, Chorioamnionitis immunology, Collagen Type I metabolism, Elastin metabolism, Female, Fetus drug effects, Fetus embryology, Fetus pathology, Fibroblast Growth Factor 10 genetics, Fibroblast Growth Factor 10 metabolism, Gene Expression, Gene Expression Regulation, Developmental, Glucocorticoids therapeutic use, HSP70 Heat-Shock Proteins metabolism, Oncogene Proteins genetics, Oncogene Proteins metabolism, Pregnancy, Pulmonary Alveoli drug effects, Pulmonary Alveoli embryology, Pulmonary Alveoli pathology, Sheep, Signal Transduction, Trans-Activators genetics, Trans-Activators metabolism, Zinc Finger Protein GLI1, Betamethasone pharmacology, Chorioamnionitis metabolism, Fetus metabolism, Glucocorticoids pharmacology, Hedgehog Proteins metabolism, Lipopolysaccharides pharmacology, Pulmonary Alveoli metabolism
- Abstract
Chorioamnionitis and antenatal corticosteroids mature the fetal lung functionally but disrupt late-gestation lung development. Because Sonic Hedgehog (Shh) signaling is a major pathway directing lung development, we hypothesized that chorioamnionitis and antenatal corticosteroids modulated Shh signaling, resulting in an altered fetal lung structure. Time-mated ewes with singleton ovine fetuses received an intra-amniotic injection of lipopolysaccharide (LPS) and/or maternal intramuscular betamethasone 7 and/or 14 days before delivery at 120 days gestational age (GA) (term = 150 days GA). Intra-amniotic LPS exposure decreased Shh mRNA levels and Gli1 protein expression, which was counteracted by both betamethasone pre- or posttreatment. mRNA and protein levels of fibroblast growth factor 10 and bone morphogenetic protein 4, which are important mediators of lung development, increased 2-fold and 3.5-fold, respectively, 14 days after LPS exposure. Both 7-day and 14-day exposure to LPS changed the mRNA levels of elastin (ELN) and collagen type I alpha 1 (Col1A1) and 2 (Col1A2), which resulted in fewer elastin foci and increased collagen type I deposition in the alveolar septa. Corticosteroid posttreatment prevented the decrease in ELN mRNA and increased elastin foci and decreased collagen type I deposition in the fetal lung. In conclusion, fetal lung exposure to LPS was accompanied by changes in key modulators of lung development resulting in abnormal lung structure. Betamethasone treatment partially prevented the changes in developmental processes and lung structure. This study provides new insights into clinically relevant prenatal exposures and fetal lung development.
- Published
- 2012
- Full Text
- View/download PDF
43. Intra-amniotic LPS and antenatal betamethasone: inflammation and maturation in preterm lamb lungs.
- Author
-
Kuypers E, Collins JJ, Kramer BW, Ofman G, Nitsos I, Pillow JJ, Polglase GR, Kemp MW, Newnham JP, Gavilanes AW, Nowacki R, Ikegami M, Jobe AH, and Kallapur SG
- Subjects
- Amnion, Animals, Bronchoalveolar Lavage Fluid cytology, Chorioamnionitis etiology, Chorioamnionitis immunology, Cytokines genetics, Cytokines metabolism, Female, Fetal Organ Maturity drug effects, Fetal Organ Maturity immunology, Gene Expression, Inflammation drug therapy, Inflammation immunology, Lipopolysaccharides pharmacology, Lung drug effects, Lung enzymology, Lung immunology, Male, Medroxyprogesterone Acetate therapeutic use, Peroxidase metabolism, Phosphatidylcholines metabolism, Pregnancy, Premature Birth immunology, Premature Birth prevention & control, Pulmonary Surfactant-Associated Protein C genetics, Pulmonary Surfactant-Associated Protein C metabolism, Pulmonary Surfactant-Associated Protein D genetics, Pulmonary Surfactant-Associated Protein D metabolism, Random Allocation, Sheep, Toll-Like Receptors genetics, Toll-Like Receptors metabolism, Betamethasone therapeutic use, Chorioamnionitis drug therapy, Fetal Development drug effects, Glucocorticoids therapeutic use, Lung embryology
- Abstract
The proinflammatory stimulus of chorioamnionitis is commonly associated with preterm delivery. Women at risk of preterm delivery receive antenatal glucocorticoids to functionally mature the fetal lung. However, the effects of the combined exposures of chorioamnionitis and antenatal glucocorticoids on the fetus are poorly understood. Time-mated ewes with singleton fetuses received an intra-amniotic injection of lipopolysaccharide (LPS) either preceding or following maternal intramuscular betamethasone 7 or 14 days before delivery, and the fetuses were delivered at 120 days gestational age (GA) (term = 150 days GA). Gestation matched controls received intra-amniotic and maternal intramuscular saline. Compared with saline controls, intra-amniotic LPS increased inflammatory cells in the bronchoalveolar lavage and myeloperoxidase, Toll-like receptor 2 and 4 mRNA, PU.1, CD3, and Foxp3-positive cells in the fetal lung. LPS-induced lung maturation measured as increased airway surfactant and improved lung gas volumes. Intra-amniotic LPS-induced inflammation persisted until 14 days after exposure. Betamethasone treatment alone induced modest lung maturation but, when administered before intra-amniotic LPS, suppressed lung inflammation. Interestingly, betamethasone treatment after LPS did not counteract inflammation but enhanced lung maturation. We conclude that the order of exposures of intra-amniotic LPS or maternal betamethasone had large effects on fetal lung inflammation and maturation.
- Published
- 2012
- Full Text
- View/download PDF
44. Lipopolysaccharide-induced chorioamnionitis is confined to one amniotic compartment in twin pregnant sheep.
- Author
-
Gantert M, Jellema RK, Heineman H, Gantert J, Collins JJ, Seehase M, Lambermont VA, Keck A, Garnier Y, Zimmermann LJ, Kadyrov M, Gavilanes AW, and Kramer BW
- Subjects
- Amnion immunology, Analysis of Variance, Animals, Bronchoalveolar Lavage Fluid immunology, Chorioamnionitis chemically induced, Chorioamnionitis immunology, Disease Models, Animal, Female, Gestational Age, Leukocyte Count, Lung embryology, Lung immunology, Lung Compliance, Neutrophils immunology, Placenta immunology, Pregnancy, Pregnancy, Multiple, Sheep, Domestic, Amnion pathology, Chorioamnionitis pathology, Lipopolysaccharides, Lung pathology, Placenta pathology
- Abstract
Background: Chorioamnionitis is a major risk factor for preterm birth in multifetal pregnancies. However, there is little clinical data whether chorioamnionitis is restricted to one amniotic compartment in multifetal pregnancies., Objective: To explore whether chorioamnionitis is confined to the exposed compartment and does not cross to the unaffected fetus in twin pregnancy., Methods: In twin pregnant sheep, one of the twins was exposed to either 2 or 14 days of intra-amniotic lipopolysaccharide (LPS) while the co-twin was exposed to either 2 or 14 days of intra-amniotic saline (n = 3 for each exposure). Singletons were included in this study to compare the grade of inflammation with twins. All fetuses were delivered at 125 days of gestation (term = 150 days). Chorioamnionitis was confirmed by histological examination. Lung inflammation was assessed by cell count in bronchoalveolar lavage. Lung compliance was assessed at 40 cm H(2)O. Results were compared using analysis of variance (ANOVA) with a post-hoc Tukey analysis., Results: Inflammation in placenta, membranes and lung of LPS-exposed twins was significantly higher after 2 and 14 days of exposure when compared to the saline-exposed co-twins. Lung compliance in LPS-exposed twins was significantly increased after 14 days when compared to saline-exposed co-twins. Intrauterine LPS exposure increased lung compliance and inflammation in the membranes, placenta and lung to the same extent in twins as in singletons., Conclusion: In twin pregnant sheep, inflammation of the membranes, placenta and fetal lung was strictly limited to the exposed fetus in the amniotic compartment in which the LPS was injected., (Copyright © 2012 S. Karger AG, Basel.)
- Published
- 2012
- Full Text
- View/download PDF
45. Pulmonary and systemic inflammatory responses to intra-amniotic IL-1α in fetal sheep.
- Author
-
Kallapur SG, Kramer BW, Nitsos I, Pillow JJ, Collins JJ, Polglase GR, Newnham JP, and Jobe AH
- Subjects
- Amnion, Animals, Bronchoalveolar Lavage Fluid immunology, Female, Fetus drug effects, Forkhead Transcription Factors biosynthesis, Lung, Lymph Nodes metabolism, Mediastinitis pathology, Pregnancy, Sheep, Domestic, Fetal Diseases immunology, Fetus immunology, Inflammation immunology, Interleukin-1alpha pharmacology
- Abstract
Clinical and epidemiological studies implicate IL-1 as an important mediator of perinatal inflammation. We tested the hypothesis that intra-amniotic IL-1α would induce pulmonary and systemic fetal inflammatory responses. Sheep with singleton fetuses were given an intra-amniotic injection of recombinant sheep IL-1α (100 μg) and were delivered 1, 3, or 7 days later, at 124 ± 1 days gestation (n=5-8/group). A separate group of sheep were given two intra-amniotic IL-1α injections (100 μg dose each): 7 days and again 1 day prior to delivery. IL-1α induced a robust increase in monocytes, neutrophils, lymphocytes, and IL-8 protein in bronchoalveolar lavage fluid. H(2)O(2) secretion was increased in inflammatory cells isolated from lungs of IL-1α-exposed lambs upon LPS challenge in vitro compared with control monocytes. T lymphocytes were recruited to the lung. IL-1β, cyclooxygenase-1, and cyclooxygenase-2 mRNA expression increased in the lung 1 day after intra-amniotic IL-1α exposure. Lung volumes increased 7 days after intra-amniotic IL-1α exposure, with minimal anatomic changes in air space morphology. The weight of the posterior mediastinal lymph node draining the lung and the gastrointestinal tract doubled, inducible nitric oxide synthase (NOSII)-positive cells increased, and Foxp3-positive T-regulatory lymphocytes decreased in the lymph node after IL-1α exposure. In the blood, neutrophil counts and plasma haptoglobin increased after IL-1α exposure. Compared with a single exposure, exposure to intra-amniotic IL-1α 7 days and again 1 day before delivery had a variable effect (increases in some inflammatory markers, but not pulmonary cytokines). IL-1α is a potent mediator of the fetal inflammatory response syndrome.
- Published
- 2011
- Full Text
- View/download PDF
46. Inflammation in fetal sheep from intra-amniotic injection of Ureaplasma parvum.
- Author
-
Collins JJ, Kallapur SG, Knox CL, Nitsos I, Polglase GR, Pillow JJ, Kuypers E, Newnham JP, Jobe AH, and Kramer BW
- Subjects
- Animals, Bronchoalveolar Lavage Fluid cytology, Cytokines genetics, Cytokines metabolism, Female, Gestational Age, Humans, Male, Pregnancy, Ureaplasma Infections metabolism, Ureaplasma Infections pathology, Fetus immunology, Fetus microbiology, Fetus physiopathology, Pneumonia immunology, Pneumonia microbiology, Pneumonia pathology, Sheep, Ureaplasma pathogenicity, Ureaplasma Infections immunology
- Abstract
Bronchopulmonary dysplasia is associated with chorioamnionitis and fetal lung inflammation. Ureaplasma species are the bacteria most frequently isolated from chorioamnionitis. Very chronic ureaplasma colonization of amniotic fluid causes low-grade lung inflammation and functional lung maturation in fetal sheep. Less is known about shorter exposures of the fetal lung. Therefore, we hypothesized that ureaplasmas would cause an acute inflammatory response that would alter lung development. Singleton ovine fetuses received intra-amniotic Ureaplasma parvum serovar 3 or control media at 110, 117, or 121 days and were delivered at 124 days gestational age (term = 150 days). Inflammation was assessed by 1) cell counts in bronchoalveolar lavage fluid (BALF), and 2) cytokine mRNA measurements, immunohistochemistry, and flow cytometry for inflammatory cells and elastin and α-smooth muscle actin (α-SMA) staining in lung tissue. Neutrophils were increased in BALF 3 days after exposure to ureaplasmas (P = 0.01). Myeloperoxidase-positive cells increased after 3 days (P = 0.03), and major histocompatibility complex (MHC) class II-positive cells increased after 14 days of ureaplasma exposure (P = 0.001). PU.1 (macrophage marker)- or CD3 (T lymphocyte marker)-positive cells were not induced by ureaplasmas. CD3-positive cells in the posterior mediastinal lymph node increased in ureaplasma-exposed animals at 3, 7, and 14 days (P = 0.002). Focal elastin depositions decreased in alveolar septa at 14 days (P = 0.002), whereas α-SMA increased in arteries and bronchioli. U. parvum induced a mild acute inflammatory response and changed elastin and α-SMA deposition in the lung, which may affect lung structure and subsequent development.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.