1
|
Takahashi Y, Fee EL, Takahashi T, Usuda H, Ikeda H, Carter SW, Saito Y, Sato S, Mochii N, Chemtob S, Olson DM, Keelan JA, Kumagai Y, Choolani MA, Illanes SE, Saito M, Kemp MW. Interleukin-1 Receptor Antagonists Partially Inhibited Histological Injury but Not Tissue Inflammation in a Sheep Model of Pregnancy. Reprod Sci 2025; 32:1213-1227. [PMID: 39953369 DOI: 10.1007/s43032-024-01781-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/27/2024] [Indexed: 02/17/2025]
Abstract
Intrauterine inflammation is a significant cause of early preterm birth and fetal injury. There is a lack of effective interventions for intrauterine inflammation. This study aimed to determine whether direct fetal treatment with IL-1 receptor antagonists (IL-1RA), specifically anakinra (competitive IL-1RA) or rytvela (allosteric IL-1RA), could reduce intrauterine inflammation caused by intraamniotic injection of E. coli lipopolysaccharides (LPS) in a sheep model of pregnancy. We hypothesized the fetal intramuscular administration of IL1-RA therapy would comprehensively resolve intrauterine inflammation caused by LPS in the pregnant sheep model. Date-mated Merino ewes carrying single fetuses were randomized into four groups: LPS Group (10 mg intraamniotic LPS injection followed by saline), RYTVELA Group (10 mg LPS injection followed by 5 mg rytvela), ANAKINRA Group (LPS injection followed by 100 mg anakinra), and SALINE Group (saline injection followed by saline). All LPS-exposed fetuses had elevated bilirubin levels, leukopenia, and increased inflammatory mediators IL-1β, IL-8, tumour necrosis factor alpha (TNFα), and monocyte chemoattractant protein 1 (MCP-1) in amniotic fluid and lung tissue. Both anakinra and rytvela treatments reduced immunocyte infiltration in chorioamniotic membranes and lungs, and microglial staining, and increased the oligodendrocyte staining, but did not significantly resolve overall inflammation compared to the SALINE Group. In conclusion, fetal intramuscular administration of anakinra and rytvela did not effectively resolve intrauterine inflammation but showed potential in reducing tissue invasion and brain injury markers. These findings suggest that modest inflammation reduction may protect against brain injury and preterm birth, though no additional benefit was observed compared to intraamniotic IL-1RA treatment.
Collapse
Affiliation(s)
- Yuki Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.
- Department of Obstetrics, Tohoku University, 1-1 Seiryomachi Aobaku, Sendai, Miyagi, Japan.
| | - Erin L Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Tsukasa Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Hideyuki Ikeda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sean W Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuya Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shinichi Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Noriyoshi Mochii
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Alberta, Canada
| | - Jeffrey A Keelan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Yusaku Kumagai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics, Tohoku University, 1-1 Seiryomachi Aobaku, Sendai, Miyagi, Japan
| | - Mahesh A Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sebastian E Illanes
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, University of the Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Masatoshi Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Department of Obstetrics, Tohoku University, 1-1 Seiryomachi Aobaku, Sendai, Miyagi, Japan
| | - Matthew W Kemp
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Veterinary and Life Sciences, Murdoch University, Perth, Australia
- Women and Infants Research Foundation, Level 2, Carson House, King Edward Memorial Hospital, Perth, Australia
| |
Collapse
|
2
|
Liu J, Liu D, Sun Q, Su Y, Tang L, Liang H, Ye F, Chen Y, Zhang Q. Plasma proteomic signature of neonates in the context of placental histological chorioamnionitis. BMJ Paediatr Open 2024; 8:e002708. [PMID: 39237269 PMCID: PMC11381644 DOI: 10.1136/bmjpo-2024-002708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/16/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Placental histological chorioamnionitis (HCA) is recognised as a significant risk factor for various adverse neonatal outcomes. This study aims to explore if the inflammatory protein levels in neonates were associated with HCA. METHODS All women with singleton births from February 2020 to November 2022 were selected and divided into three groups based on maternal placental pathology results: the HCA-stage 1 group (n=24), the HCA-stage 2 group (n=16) and the control group (n=17). Olink Target 96 Inflammation Panel was used to detect the levels of 92 inflammation-related proteins in the plasma of newborns from all three groups within 24 hours after birth. We compared the protein profiles through differential protein expression analysis. RESULTS A total of six inflammation-related proteins exhibited significant differences between the HCA-stage 1 and the control group. Specifically, TRANCE and CST5 were significantly upregulated (p=0.006, p=0.025, respectively), whereas the expression of IFN-gamma, CXCL9, CXCL10 and CCL19 was significantly downregulated (p=0.040, p=0.046, p=0.007, p=0.006, respectively). HCA-stage 2 newborns had significantly elevated levels of CD5 and CD6 and decreased IFN-gamma, CXCL10 and CCL19 in comparison to controls. These differential proteins were significantly enriched in positive regulation of cytokine activity, leucocyte chemotaxis and positive regulation of T-cell activation pathway-related Gene Ontology terms. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that viral protein interaction with cytokine and cytokine receptor, interleukin-17/NF-kappa B/toll-like receptor/chemokine signalling pathway, and cytokine-cytokine receptor interaction exhibited significant differences. Spearman analysis demonstrated a significant positive connection between the levels of CD6 and CD5 proteins, not only in neonatal leucocytes but also in maternal leucocytes. Additionally, CD6 was found to be associated with neonatal birth weight. CONCLUSIONS In conclusion, placental histological changes associated with chorioamnionitis appear to influence the expression of inflammatory proteins in offspring. Notably, CD6 and CD5 proteins may potentially contribute to the pathogenesis of HCA-related neonatal diseases.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Die Liu
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Qi Sun
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Yunchao Su
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Lijuan Tang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Haixiao Liang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Fang Ye
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Yuanmei Chen
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Qi Zhang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Yu H, Li D, Zhao X, Fu J. Fetal origin of bronchopulmonary dysplasia: contribution of intrauterine inflammation. Mol Med 2024; 30:135. [PMID: 39227783 PMCID: PMC11373297 DOI: 10.1186/s10020-024-00909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in infants and the most frequent adverse outcome of premature birth, despite major efforts to minimize injury. It is thought to result from aberrant repair response triggered by either prenatal or recurrent postnatal injury to the lungs during development. Intrauterine inflammation is an important risk factor for prenatal lung injury, which is also increasingly linked to BPD. However, the specific mechanisms remain unclear. This review summarizes clinical and animal research linking intrauterine inflammation to BPD. We assess how intrauterine inflammation affects lung alveolarization and vascular development. In addition, we discuss prenatal therapeutic strategies targeting intrauterine inflammation to prevent or treat BPD.
Collapse
Affiliation(s)
- Haoting Yu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Xinyi Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
4
|
Habelrih T, Augustin TL, Mauffette-Whyte F, Ferri B, Sawaya K, Côté F, Gallant M, Olson DM, Chemtob S. Inflammatory mechanisms of preterm labor and emerging anti-inflammatory interventions. Cytokine Growth Factor Rev 2024; 78:50-63. [PMID: 39048393 DOI: 10.1016/j.cytogfr.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Preterm birth is a major public health concern, requiring a deeper understanding of its underlying inflammatory mechanisms and to develop effective therapeutic strategies. This review explores the complex interaction between inflammation and preterm labor, highlighting the pivotal role of the dysregulation of inflammation in triggering premature delivery. The immunological environment of pregnancy, characterized by a fragile balance of immune tolerance and resistance, is disrupted in preterm labor, leading to a pathological inflammatory response. Feto-maternal infections, among other pro-inflammatory stimuli, trigger the activation of toll-like receptors and the production of pro-inflammatory mediators, promoting uterine contractility and cervical ripening. Emerging anti-inflammatory therapeutics offer promising approaches for the prevention of preterm birth by targeting key inflammatory pathways. From TLR-4 antagonists to chemokine and interleukin receptor antagonists, these interventions aim to modulate the inflammatory environment and prevent adverse pregnancy outcomes. In conclusion, a comprehensive understanding of the inflammatory mechanisms leading to preterm labor is crucial for the development of targeted interventions in hope of reducing the incidence of preterm birth and improving neonatal health outcomes.
Collapse
Affiliation(s)
- Tiffany Habelrih
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Thalyssa-Lyn Augustin
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Félix Mauffette-Whyte
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Béatrice Ferri
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Kevin Sawaya
- Research Center, CHU Sainte-Justine, Montreal, QC, Canada; Programmes de cycles supérieurs en sciences biomédicales, Faculté de médecine, Université de Montréal, Montreal, QC, Canada
| | - France Côté
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Mathilde Gallant
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada.
| |
Collapse
|
5
|
Habelrih T, Ferri B, Côté F, Sévigny J, Augustin TL, Sawaya K, Lubell WD, Olson DM, Girard S, Chemtob S. Preventing Preterm Birth: Exploring Innovative Solutions. Clin Perinatol 2024; 51:497-510. [PMID: 38705654 DOI: 10.1016/j.clp.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
This review examines the complexities of preterm birth (PTB), emphasizes the pivotal role of inflammation in the pathogenesis of preterm labor, and assesses current available interventions. Antibiotics, progesterone analogs, mechanical approaches, nonsteroidal anti-inflammatory drugs, and nutritional supplementation demonstrate a limited efficacy. Tocolytic agents, targeting uterine activity and contractility, inadequately prevent PTB by neglecting to act on uteroplacental inflammation. Emerging therapies targeting toll-like receptors, chemokines, and interleukin receptors exhibit promise in mitigating inflammation and preventing PTB.
Collapse
Affiliation(s)
- Tiffany Habelrih
- Université de Montréal, Pavillion Roger-Gaudry, 2900 boul Edouard-Montpetit, H3T 1J4, Montréal, Québec, Canada; Centre de recherche du CHU Sainte-Justine, 3175 ch de la Côte-Sainte-Catherine, H3T 1C5, Montréal, Québec, Canada
| | - Béatrice Ferri
- Université de Montréal, Pavillion Roger-Gaudry, 2900 boul Edouard-Montpetit, H3T 1J4, Montréal, Québec, Canada; Centre de recherche du CHU Sainte-Justine, 3175 ch de la Côte-Sainte-Catherine, H3T 1C5, Montréal, Québec, Canada
| | - France Côté
- Université de Montréal, Pavillion Roger-Gaudry, 2900 boul Edouard-Montpetit, H3T 1J4, Montréal, Québec, Canada; Centre de recherche du CHU Sainte-Justine, 3175 ch de la Côte-Sainte-Catherine, H3T 1C5, Montréal, Québec, Canada
| | - Juliane Sévigny
- Département de Biologie, Université de Sherbrooke, Voie 9, J1X 2X9, Sherbrooke, Québec, Canada
| | - Thalyssa-Lyn Augustin
- Université de Montréal, Pavillion Roger-Gaudry, 2900 boul Edouard-Montpetit, H3T 1J4, Montréal, Québec, Canada; Centre de recherche du CHU Sainte-Justine, 3175 ch de la Côte-Sainte-Catherine, H3T 1C5, Montréal, Québec, Canada
| | - Kevin Sawaya
- Centre de recherche du CHU Sainte-Justine, 3175 ch de la Côte-Sainte-Catherine, H3T 1C5, Montréal, Québec, Canada; Department of Microbiology and Immunology, McGill University, 3775 Rue University, Room 511, H3A 2B4, Montréal, Québec, Canada
| | - William D Lubell
- Département de Chimie, Université de Montréal, Complexe des Sciences, 1375 avenue Thérèse-Lavoie-Roux, Montréal, Québec, H2V 0B3, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, 220 HMRC, T6G 2S2, Edmonton, Alberta, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, 200 First Street SW, Guggenheim Building 3rd floor, Rochester, MN 55905, USA
| | - Sylvain Chemtob
- Université de Montréal, Pavillion Roger-Gaudry, 2900 boul Edouard-Montpetit, H3T 1J4, Montréal, Québec, Canada; Centre de recherche du CHU Sainte-Justine, 3175 ch de la Côte-Sainte-Catherine, H3T 1C5, Montréal, Québec, Canada.
| |
Collapse
|
6
|
Li Q, Li D, Lu J, Zou K, Wang L, Jiao Y, Wang M, Gao R, Song J, Li Y, Li F, Ji J, Wang J, Li L, Ye T, He E, Chen H, Wang Y, Ren J, Bai C, Yang S, Zhang Y. Interface-Stabilized Fiber Sensor for Real-Time Monitoring of Amniotic Fluid During Pregnancy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307726. [PMID: 37775103 DOI: 10.1002/adma.202307726] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Indexed: 10/01/2023]
Abstract
Diseases in pregnancy endanger millions of fetuses worldwide every year. The onset of these diseases can be early warned by the dynamic abnormalities of biochemicals in amniotic fluid, thus requiring real-time monitoring. However, when continuously penetrated by detection devices, the amnion is prone to loss of robustness and rupture, which is difficult to regenerate. Here, an interface-stabilized fiber sensor is presented for real-time monitoring of biochemical dynamics in amniotic fluid during pregnancy. The sensor is seamlessly integrated into the amnion through tissue adhesion, amniotic regeneration, and uniform stress distribution, posing no risk to the amniotic fluid environment. The sensor demonstrates a response performance of less than 0.3% fluctuation under complex dynamic conditions and an accuracy of more than 98% from the second to the third trimester. By applying it to early warning of diseases such as intrauterine hypoxia, intrauterine infection, and fetal growth restriction, fetal survival increases to 95% with timely intervention.
Collapse
Affiliation(s)
- Qianming Li
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Dan Li
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiang Lu
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Kuangyi Zou
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Lie Wang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Yiding Jiao
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Maosen Wang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Rui Gao
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Jie Song
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Yiran Li
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Fangyan Li
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Jianjian Ji
- Key Laboratory of Inflammation and Immunoregulation, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiacheng Wang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Luhe Li
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Tingting Ye
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Er He
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Hao Chen
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Yuanzhen Wang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Junye Ren
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Chenyu Bai
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Shuo Yang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Ye Zhang
- College of Engineering and Applied Sciences, National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Centre, Collaborative Innovation Centre of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
7
|
Lavizzari A, Esposito B, Pesenti N, Shaykhova A, Vizzari G, Ophorst M, Gangi S, Morniroli D, Colnaghi M, Mosca F, Giannì ML. Dose-dependent impact of human milk feeding on tidal breathing flow-volume loop parameters across the first 2 years of life in extremely low-birth-weight infants: a cohort study. Eur J Pediatr 2023; 182:4969-4976. [PMID: 37610435 DOI: 10.1007/s00431-023-05163-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023]
Abstract
The purpose of this study is to test the hypothesis that higher consumption of human milk (HM) in preterm infants with birth weight (BW) <1000 g is associated with improved lung function in a dose-dependent manner over the first 2 years of corrected age (CA). This retrospective study at an academic medical center included infants with BW <1000g. They had lung function assessment by the tidal breathing flow-volume loop (TBFVL) follow-up visits at 0-3-, 3-6-, 6-12-, 12-18-, and 18-24-month CA. One hundred eighty infants were included in the study with a mean (SD) gestational age 26.5 (1.90) weeks and BW 772.4 (147.0) g, 50% were female, and 60% developed BPD. 62.8% of infants received HM during the NICU stay. According to a general linear model (including GA, being small for GA (SGA), sex, human milk percentage, sepsis, and BPD), on average, each week of GA resulted in a higher tPTEF/tE of 1.24 (p = 0.039) and being SGA in a lower tPTEF/tE of 5.75 (p = 0.013) at 0-3-month CA. A higher percentage of human milk out of the total enteral intake was associated with better tPTEF/tE z-scores at 0-3 months (p = 0.004) and 18-24 months of CA (p = 0.041). BPD diagnosis was associated with a relevantly worse tPTEF/tE z-score at 6-12 months of CA (p = 0.003). CONCLUSION Preterm infants with higher consumption of HM had significantly less airway obstruction across the first 2 years, suggesting that human milk may contribute in a dose-dependent manner to improve lung function in early childhood in former preterm infants born ELBW. WHAT IS KNOWN • Human milk feeding reduces the risk of prematurity-related morbidities, including necrotizing enterocolitis, sepsis, lower respiratory tract infections, and BPD. Both exclusive and partial human milk feeding appear to be associated with a lower risk of BPD in preterm infants. WHAT IS NEW • This cohort study of 180 preterm infants with birth weight < 1000 g found that exposure to human milk during hospitalization improves airway obstruction markers tPTEF/tE z-score over the first 2 years of corrected age in a dose-dependent manner.
Collapse
Affiliation(s)
- Anna Lavizzari
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy.
| | - Benedetta Esposito
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Nicola Pesenti
- Revelo Datalabs Srl, Via Spezia 1, 20142, Milan, Italy
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, University of Milan-Bicocca, Milan, Italy
| | - Alina Shaykhova
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giulia Vizzari
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Marijke Ophorst
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Silvana Gangi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Mariarosa Colnaghi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Maria L Giannì
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Takahashi Y, Takahashi T, Usuda H, Carter S, Fee EL, Furfaro L, Chemtob S, Olson DM, Keelan JA, Kallapur S, Kemp MW. Pharmacological blockade of the interleukin-1 receptor suppressed Escherichia coli lipopolysaccharide-induced neuroinflammation in preterm fetal sheep. Am J Obstet Gynecol MFM 2023; 5:101124. [PMID: 37597799 DOI: 10.1016/j.ajogmf.2023.101124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/23/2023] [Accepted: 08/03/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Intraamniotic inflammation is associated with preterm birth, especially in cases occurring before 32 weeks' gestation, and is causally linked with an increased risk for neonatal mortality and morbidity. Targeted anti-inflammatory interventions may assist in improving the outcomes for pregnancies impacted by intrauterine inflammation. Interleukin-1 is a central upstream mediator of inflammation. Accordingly, interleukin-1 is a promising candidate target for intervention therapies and has been targeted previously using the interleukin-1 receptor antagonist, anakinra. Recent studies have shown that the novel, noncompetitive, allosteric interleukin-1 receptor inhibitor, rytvela, partially resolved inflammation associated with preterm birth and fetal injury. In this study, we used a preterm sheep model of chorioamnionitis to investigate the anti-inflammatory efficacy of rytvela and anakinra, administered in the amniotic fluid in the setting of intraamniotic Escherichia coli lipopolysaccharide exposure. OBJECTIVE We hypothesized that both rytvela and anakinra would reduce lipopolysaccharide-induced intrauterine inflammation and protect the fetal brain. STUDY DESIGN Ewes with a singleton fetus at 105 days of gestation (term is ∼150 days) were randomized to one of the following groups: (1) intraamniotic injections of 2 mL saline at time=0 and time=24 hours as a negative control group (saline group, n=12); (2) intraamniotic injection of 10 mg Escherichia coli lipopolysaccharide in 2 mL saline and intraamniotic injections of 2 mL saline at time=0 hours and time=24 hours as an inflammation positive control group (lipopolysaccharide group, n=11); (3) intraamniotic injection of Escherichia coli lipopolysaccharide in 2 mL saline and intraamniotic injections of 2.5 mg rytvela at time=0 hours and time=24 hours to test the anti-inflammatory efficacy of rytvela (lipopolysaccharide + rytvela group, n=10); or (4) intraamniotic injection of Escherichia coli lipopolysaccharide in 2 mL saline and intraamniotic injections of 100 mg anakinra at time=0 hours and time=24 hours to test the anti-inflammatory efficacy of anakinra (lipopolysaccharide + anakinra group, n=12). Amniotic fluid was sampled at time 0, 24, and 48 hours (ie, at each intervention and at delivery). Fetal umbilical cord blood was collected at delivery for differential blood counts and chemical studies. Inflammation was characterized by the analysis of fetal tissue cytokine and chemokine levels using quantitative polymerase chain reaction, enzyme-linked inmmunosorbent assay, and histology. The primary study outcome of interest was the assessment of anakinra and rytvela brain-protective effects in the setting of Escherichia coli lipopolysaccharide-induced intrauterine inflammation. Secondary outcomes of interest were to assess protection from fetal and intrauterine (ie, amniotic fluid, chorioamnion) inflammation. RESULTS Intraamniotic administration of lipopolysaccharide caused inflammation of the fetal lung, brain, and chorioamnionitis in preterm fetal sheep. Relative to treatment with saline only in the setting of lipopolysaccharide exposure, intraamniotic administration of both rytvela and anakinra both significantly prevented periventricular white matter injury, microglial activation, and histologic chorioamnionitis. Anakinra showed additional efficacy in inhibiting fetal lung myeloperoxidase activity, but its use was associated with metabolic acidaemia and reduced fetal plasma insulin-like growth factor-1 levels at delivery. CONCLUSION Intraamniotic administration of rytvela or anakinra significantly inhibited fetal brain inflammation and chorioamnionitis in preterm fetal sheep exposed to intraamniotic lipopolysaccharide. In addition, anakinra treatment was associated with potential negative impacts on the developing fetus.
Collapse
Affiliation(s)
- Yuki Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp); Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan (Drs Y Takahashi, T Takahashi, Usuda, and Kemp).
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp); Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan (Drs Y Takahashi, T Takahashi, Usuda, and Kemp)
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp); Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan (Drs Y Takahashi, T Takahashi, Usuda, and Kemp)
| | - Sean Carter
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp); Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Dr S Carter, and Kemp)
| | - Erin L Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp)
| | - Lucy Furfaro
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp)
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Canada (Dr Chemtob)
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Alberta, Canada (Dr Olson)
| | - Jeffrey A Keelan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp)
| | - Suhas Kallapur
- Department of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA (Dr Kallapur)
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia (Drs Y Takahashi, T Takahashi, M Usuda, and Carter, Ms Fee, and Drs Furfaro, Keelan, and Kemp); Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan (Drs Y Takahashi, T Takahashi, Usuda, and Kemp); School of Veterinary and Life Sciences, Murdoch University, Perth, Australia (Dr Kemp); Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Dr S Carter, and Kemp)
| |
Collapse
|
9
|
Ahmed M, Casanova NG, Zaghloul N, Gupta A, Rodriguez M, Robbins IR, Kempf CL, Sun X, Song JH, Hernon VR, Sammani S, Camp SM, Moreira A, Hsu CD, Garcia JGN. The eNAMPT/TLR4 inflammatory cascade drives the severity of intra-amniotic inflammation in pregnancy and predicts infant outcomes. Front Physiol 2023; 14:1129413. [PMID: 37415908 PMCID: PMC10319582 DOI: 10.3389/fphys.2023.1129413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction: Intra-amniotic inflammation (IAI) or chorioamnionitis is a common complication of pregnancy producing significant maternal morbidity/mortality, premature birth and neonatal risk of chronic lung diseases such as bronchopulmonary dysplasia (BPD). We examined eNAMPT (extracellular nicotinamide phosphoribosyltransferase), a critical inflammatory DAMP and TLR4 ligand, as a potential therapeutic target to reduce IAI severity and improve adverse fetal/neonatal outcomes. Methods: Blood/tissue samples were examined in: 1) women with histologically-proven chorioamnionitis, 2) very low birth weight (VLBW) neonates, and 3) a preclinical murine pregnancy model of IAI. Groups of pregnant IAI-exposed mice and pups were treated with an eNAMPT-neutralizing mAb. Results: Human placentas from women with histologically-proven chorioamnionitis exhibited dramatic NAMPT expression compared to placentas without chorioamnionitis. Increased NAMPT expression in whole blood from VLBW neonates (day 5) significantly predicted BPD development. Compared to untreated LPS-challenged murine dams (gestational day 15), pups born to eNAMPT mAb-treated dams (gestational days 15/16) exhibited a > 3-fold improved survival, reduced neonate lung eNAMPT/cytokine levels, and reduced development and severity of BPD and pulmonary hypertension (PH) following postnatal exposure to 100% hyperoxia days 1-14. Genome-wide gene expression studies of maternal uterine and neonatal cardiac tissues corroborated eNAMPT mAb-induced reductions in inflammatory pathway genes. Discussion: The eNAMPT/TLR4 inflammatory pathway is a highly druggable contributor to IAI pathobiology during pregnancy with the eNAMPT-neutralizing mAb a novel therapeutic strategy to decrease premature delivery and improve short- and long-term neonatal outcomes. eNAMPT blood expression is a potential biomarker for early prediction of chronic lung disease among premature neonates.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nancy G. Casanova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nahla Zaghloul
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Akash Gupta
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Marisela Rodriguez
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Ian R. Robbins
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L. Kempf
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Xiaoguang Sun
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Jin H. Song
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Vivian Reyes Hernon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Alvaro Moreira
- Department of Pediatrics, UT Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| |
Collapse
|
10
|
Patel S, Ciechanowicz S, Blumenfeld YJ, Sultan P. Epidural-related maternal fever: incidence, pathophysiology, outcomes, and management. Am J Obstet Gynecol 2023; 228:S1283-S1304.e1. [PMID: 36925412 DOI: 10.1016/j.ajog.2022.06.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 03/18/2023]
Abstract
Epidural-related maternal fever affects 15% to 25% of patients who receive a labor epidural. Two meta-analyses demonstrated that epidural-related maternal fever is a clinical phenomenon, which is unlikely to be caused by selection bias. All commonly used neuraxial techniques, local anesthetics with or without opioids, and maintenance regimens are associated with epidural-related maternal fever, however, the impact of each component is unknown. Two major theories surrounding epidural-related maternal fever development have been proposed. First, labor epidural analgesia may lead to the development of hyperthermia through a sterile (noninfectious) inflammatory process. This process may involve reduced activation of caspase-1 (a protease involved in cell apoptosis and activation of proinflammatory pathways) secondary to bupivacaine, which impairs the release of the antipyrogenic cytokine, interleukin-1-receptor antagonist, from circulating leucocytes. Detailed mechanistic processes of epidural-related maternal fever remain to be determined. Second, thermoregulatory mechanisms secondary to neuraxial blockade have been proposed, which may also contribute to epidural-related maternal fever development. Currently, there is no prophylactic strategy that can safely prevent epidural-related maternal fever from occurring nor can it easily be distinguished clinically from other causes of intrapartum fever, such as chorioamnionitis. Because intrapartum fever (of any etiology) is associated with adverse outcomes for both the mother and baby, it is important that all parturients who develop intrapartum fever are investigated and treated appropriately, irrespective of labor epidural utilization. Institution of treatment with appropriate antimicrobial therapy is recommended if an infectious cause of fever is suspected. There is currently insufficient evidence to warrant a change in recommendations regarding provision of labor epidural analgesia and the benefits of good quality labor analgesia must continue to be reiterated to expectant mothers.
Collapse
Affiliation(s)
- Selina Patel
- Department of Anesthesia, Pain and Perioperative Medicine, University of Miami, Miller School of Medicine, Miami, FL
| | - Sarah Ciechanowicz
- Department of Anaesthesia, University College London Hospital, London, United Kingdom
| | - Yair J Blumenfeld
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA
| | - Pervez Sultan
- Department of Anesthesia, Critical Care and Pain Medicine, Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
11
|
Owen JC, Garrick SP, Peterson BM, Berger PJ, Nold MF, Sehgal A, Nold-Petry CA. The role of interleukin-1 in perinatal inflammation and its impact on transitional circulation. Front Pediatr 2023; 11:1130013. [PMID: 36994431 PMCID: PMC10040554 DOI: 10.3389/fped.2023.1130013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/31/2023] Open
Abstract
Preterm birth is defined as delivery at <37 weeks of gestational age (GA) and exposes 15 million infants worldwide to serious early life diseases. Lowering the age of viability to 22 weeks GA entailed provision of intensive care to a greater number of extremely premature infants. Moreover, improved survival, especially at extremes of prematurity, comes with a rising incidence of early life diseases with short- and long-term sequelae. The transition from fetal to neonatal circulation is a substantial and complex physiologic adaptation, which normally happens rapidly and in an orderly sequence. Maternal chorioamnionitis or fetal growth restriction (FGR) are two common causes of preterm birth that are associated with impaired circulatory transition. Among many cytokines contributing to the pathogenesis of chorioamnionitis-related perinatal inflammatory diseases, the potent pro-inflammatory interleukin (IL)-1 has been shown to play a central role. The effects of utero-placental insufficiency-related FGR and in-utero hypoxia may also be mediated, in part, via the inflammatory cascade. In preclinical studies, blocking such inflammation, early and effectively, holds great promise for improving the transition of circulation. In this mini-review, we outline the mechanistic pathways leading to abnormalities in transitional circulation in chorioamnionitis and FGR. In addition, we explore the therapeutic potential of targeting IL-1 and its influence on perinatal transition in the context of chorioamnionitis and FGR.
Collapse
Affiliation(s)
- Josephine C. Owen
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Steven P. Garrick
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Briana M. Peterson
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Philip J. Berger
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Marcel F. Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Claudia A. Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Correspondence: Claudia A. Nold-Petry
| |
Collapse
|
12
|
Yao D, Zhao J, Zhang Q, Wang T, Ni M, Qi S, Shen Q, Li W, Li B, Ding X, Liu Z. Aberrant methylation of Serpine1 mediates lung injury in neonatal mice prenatally exposed to intrauterine inflammation. Cell Biosci 2022; 12:164. [PMID: 36183130 PMCID: PMC9526974 DOI: 10.1186/s13578-022-00901-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/17/2022] [Indexed: 11/05/2022] Open
Abstract
Background Intrauterine inflammation (IUI) alters epigenetic modifications in offspring, leading to lung injury. However, the epigenetic mechanism underlying IUI-induced lung injury remains uncertain. In the present study, we aim to investigate the effect of IUI on lung development, and to identify the key molecule involved in this process and its epigenetic regulatory mechanism. Results Serpine1 was upregulated in the lung tissue of neonatal mice with IUI. Intranasal delivery of Serpine1 siRNA markedly reversed IUI-induced lung injury. Serpine1 overexpression substantially promoted cell senescence of both human and murine lung epithelial cells, reflected by decreased cell proliferation and increased senescence-associated β-galactosidase activity, G0/G1 cell fraction, senescence marker, and oxidative and DNA damage marker expression. IUI decreased the methylation level of the Serpine1 promoter, and methylation of the promoter led to transcriptional repression of Serpine1. Furthermore, IUI promoted the expression of Tet1 potentially through TNF-α, while Tet1 facilitated the demethylation of Serpine1 promoter. DNA pull-down and ChIP assays revealed that the Serpine1 promoter was regulated by Rela and Hdac2. DNA demethylation increased the recruitment of Rela to the Serpine1 promoter and induced the release of Hdac2. Conclusion Increased Serpine1 expression mediated by DNA demethylation causes lung injury in neonatal mice with IUI. Therefore, therapeutic interventions targeting Serpine1 may effectively prevent IUI-induced lung injury. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00901-8.
Collapse
Affiliation(s)
- Dongting Yao
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China ,grid.411480.80000 0004 1799 1816Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiuru Zhao
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Qianqian Zhang
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Wang
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Ni
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Sudong Qi
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Qianwen Shen
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Li
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Baihe Li
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Xiya Ding
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiwei Liu
- grid.16821.3c0000 0004 0368 8293Departments of Neonatology, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, 20030 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Pavlidis I, Stock SJ. Preterm Birth Therapies to Target Inflammation. J Clin Pharmacol 2022; 62 Suppl 1:S79-S93. [PMID: 36106783 PMCID: PMC9545799 DOI: 10.1002/jcph.2107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022]
Abstract
Preterm birth (PTB; defined as delivery before 37 weeks of pregnancy) is the leading cause of morbidity and mortality in infants and children aged <5 years, conferring potentially devastating short- and long-term complications. Despite extensive research in the field, there is currently a paucity of medications available for PTB prevention and treatment. Over the past few decades, inflammation in gestational tissues has emerged at the forefront of PTB pathophysiology. Even in the absence of infection, inflammation alone can prematurely activate the main components of parturition resulting in uterine contractions, cervical ripening and dilatation, membrane rupture, and subsequent PTB. Mechanistic studies have identified critical elements of the complex inflammatory molecular pathways involved in PTB. Here, we discuss therapeutic options that target such key mediators with an aim to prevent, postpone, or treat PTB. We provide an overview of more traditional therapies that are currently used or being tested in humans, and we highlight recent advances in preclinical studies introducing novel approaches with therapeutic potential. We conclude that urgent collaborative action is required to address the unmet need of developing effective strategies to tackle the challenge of PTB and its complications.
Collapse
Affiliation(s)
- Ioannis Pavlidis
- University of Warwick Biomedical Research Unit in Reproductive HealthCoventryUK
| | | |
Collapse
|
14
|
Pang R, Mujuni BM, Martinello KA, Webb EL, Nalwoga A, Ssekyewa J, Musoke M, Kurinczuk JJ, Sewegaba M, Cowan FM, Cose S, Nakakeeto M, Elliott AM, Sebire NJ, Klein N, Robertson NJ, Tann CJ. Elevated serum IL-10 is associated with severity of neonatal encephalopathy and adverse early childhood outcomes. Pediatr Res 2022; 92:180-189. [PMID: 33674741 PMCID: PMC9411052 DOI: 10.1038/s41390-021-01438-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neonatal encephalopathy (NE) contributes substantially to child mortality and disability globally. We compared cytokine profiles in term Ugandan neonates with and without NE, with and without perinatal infection or inflammation and identified biomarkers predicting neonatal and early childhood outcomes. METHODS In this exploratory biomarker study, serum IL-1α, IL-6, IL-8, IL-10, TNFα, and VEGF (<12 h) were compared between NE and non-NE infants with and without perinatal infection/inflammation. Neonatal (severity of NE, mortality) and early childhood (death or neurodevelopmental impairment to 2.5 years) outcomes were assessed. Predictors of outcomes were explored with multivariable linear and logistic regression and receiver-operating characteristic analyses. RESULTS Cytokine assays on 159 NE and 157 non-NE infants were performed; data on early childhood outcomes were available for 150 and 129, respectively. NE infants had higher IL-10 (p < 0.001), higher IL-6 (p < 0.017), and lower VEGF (p < 0.001) levels. Moderate and severe NE was associated with higher IL-10 levels compared to non-NE infants (p < 0.001). Elevated IL-1α was associated with perinatal infection/inflammation (p = 0.013). Among NE infants, IL-10 predicted neonatal mortality (p = 0.01) and adverse early childhood outcome (adjusted OR 2.28, 95% CI 1.35-3.86, p = 0.002). CONCLUSIONS Our findings support a potential role for IL-10 as a biomarker for adverse outcomes after neonatal encephalopathy. IMPACT Neonatal encephalopathy is a common cause of child death and disability globally. Inflammatory cytokines are potential biomarkers of encephalopathy severity and outcome. In this Ugandan health facility-based cohort, neonatal encephalopathy was associated with elevated serum IL-10 and IL-6, and reduced VEGF at birth. Elevated serum IL-10 within 12 h after birth predicted severity of neonatal encephalopathy, neonatal mortality, and adverse early childhood developmental outcomes, independent of perinatal infection or inflammation, and provides evidence to the contribution of the inflammatory processes. Our findings support a role for IL-10 as a biomarker for adverse outcomes after neonatal encephalopathy in a sub-Saharan African cohort.
Collapse
Affiliation(s)
- Raymand Pang
- Institute for Women's Health, University College London, London, UK
| | - Brian M Mujuni
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | | | - Emily L Webb
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Angela Nalwoga
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Julius Ssekyewa
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Margaret Musoke
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | | | - Margaret Sewegaba
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Frances M Cowan
- Department of Pediatrics, Imperial College London, London, UK
| | - Stephen Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Margaret Nakakeeto
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Alison M Elliott
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Neil J Sebire
- UCL Institute of Child Health and GOSH BRC, UCL, London, UK
| | - Nigel Klein
- UCL Institute of Child Health and GOSH BRC, UCL, London, UK
| | - Nicola J Robertson
- Institute for Women's Health, University College London, London, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Cally J Tann
- Institute for Women's Health, University College London, London, UK.
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda.
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
15
|
Jackson CM, Demmert M, Mukherjee S, Isaacs T, Thompson R, Chastain C, Gray J, Senthamaraikannan P, Presicce P, Chetal K, Salomonis N, Miller LA, Jobe AH, Kallapur SG, Zacharias WJ, Lewkowich IP, Deshmukh H, Chougnet CA. A potent myeloid response is rapidly activated in the lungs of premature Rhesus macaques exposed to intra-uterine inflammation. Mucosal Immunol 2022; 15:730-744. [PMID: 35314757 PMCID: PMC9259482 DOI: 10.1038/s41385-022-00495-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023]
Abstract
Up to 40% of preterm births are associated with histological chorioamnionitis (HCA), which leads to elevated levels of pro-inflammatory mediators and microbial products in the amniotic fluid, which come in contact with fetal lungs. Yet, fetal pulmonary immune responses to such exposure remain poorly characterized. To address this gap, we used our established HCA model, in which pregnant Rhesus macaques receive intraamniotic (IA) saline or LPS. IA LPS induced a potent and rapid myeloid cell response in fetal lungs, dominated by neutrophils and monocytes/macrophages. Infiltrating and resident myeloid cells exhibited transcriptional profiles consistent with exposure to TLR ligands, as well as cytokines, notably IL-1 and TNFα. Although simultaneous, in vivo blockade of IL-1 and TNFα signaling did not prevent the inflammatory cell recruitment, it blunted the lung overall inflammatory state reducing communication between, and activation of, infiltrating immune cells. Our data indicate that the fetal innate immune system can mount a rapid multi-faceted pulmonary immune response to in utero exposure to inflammation. These data provide mechanistic insights into the association between HCA and the postnatal lung morbidities of the premature infant and highlight therapeutic potential of inflammatory blockade in the fetus.
Collapse
Affiliation(s)
- Courtney M Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Martin Demmert
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Department of Pediatrics, Institute for Systemic Inflammation Research, University of Lϋbeck, Lϋbeck, Germany.
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Travis Isaacs
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ravyn Thompson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chase Chastain
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jerilyn Gray
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paranth Senthamaraikannan
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lisa A Miller
- California National Primate Research Center, University of California Davis, Davis, CA, USA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - William J Zacharias
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ian P Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hitesh Deshmukh
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
16
|
Ali A, Weber V, Thompson SJ, EF Abbott T, Harmston R, David AL, Ackland GL. IL1-ra polymorphisms and risk of epidural-related maternal fever (EPIFEVER-2): study protocol for a multicentre, observational mechanistic cohort study. Int J Obstet Anesth 2022; 50:103540. [DOI: 10.1016/j.ijoa.2022.103540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/28/2022] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
|
17
|
Amniotic LPS-Induced Apoptosis in the Fetal Brain Is Suppressed by Vaginal LPS Preconditioning but Is Promoted by Continuous Ischemic Reperfusion. Int J Mol Sci 2022; 23:ijms23031787. [PMID: 35163709 PMCID: PMC8836254 DOI: 10.3390/ijms23031787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Chorioamnionitis (CAM) is an increasingly common disease affecting pregnant women which derives from bacterial vaginosis. In different clinical cases, it has been shown that CAM can cause multiple risk factors for fetal brain damage, such as infection, and intra-uterine asphyxia. However, the molecular mechanism remains unknown. In this study, we established a novel CAM mouse model by exposing pregnant mice to a combination of three risk factors: vaginal lipopolysaccharides (LPS), amniotic LPS, and ischemic reperfusion. We found amniotic LPS caused Parkinson's disease-like fetal brain damage, in a dose and time-dependent manner. Moreover, the mechanism of this fetal brain damage is apoptosis induced by amniotic LPS but it was inhibited by being pretreated with a vaginal LPS challenge before amniotic LPS injection. In contrast, amniotic LPS with continuous ischemic reperfusion caused a higher level of apoptotic cell death than amniotic LPS alone. In particular, a potential neuroprotective biomarker phosphorylation (p)-CREB (ser133) appeared in only vaginal LPS preconditioned before amniotic LPS, whereas ischemic reperfusion triggered IKK phosphorylation after amniotic LPS. Despite the need for many future investigations, this study also discussed a developed understanding of the molecular mechanism of how these phenotypes occurred.
Collapse
|
18
|
Verma J, Sankar MJ, Atmakuri K, Agarwal R, Das B. Gut microbiome dysbiosis in neonatal sepsis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:125-147. [DOI: 10.1016/bs.pmbts.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
19
|
Ackland GL, Van Duijvenboden S, Abbott TE, Gutierrez del Arroyo A, Wilson MJ, David AL. Interleukin-1 receptor antagonist, mode of analgesia and risk of Caesarean delivery after onset of labour: a Mendelian randomisation analysis. Br J Anaesth 2022; 128:89-97. [PMID: 34802694 PMCID: PMC8787778 DOI: 10.1016/j.bja.2021.09.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Lower circulating levels of the anti-inflammatory cytokine interleukin-1 receptor antagonist (IL-1ra) are associated with intrapartum inflammation and epidural analgesia-related maternal fever, both of which increase the rate of obstetric interventions. We hypothesised that genetic variants determining IL-1ra levels would be associated with Caesarean delivery rates after the onset of labour. METHODS We performed Mendelian randomisation analyses in parous women ≥16 yr old who received either non-neuraxial or neuraxial analgesia for their first two labours (UK Biobank). We used an established genetic score (calculated as 0-4, determined by the presence/absence of rs6743376 and rs1542176 alleles), in which the complete absence of both alleles causes the lowest IL-1ra levels. The primary outcome was Caesarean delivery after the onset of labour (odds ratio [OR]: 95% confidence intervals). RESULTS There were 7731 women (mean [standard deviation] age at first birth: 25 [5] yr) who had complete genetic scores and delivery data. For women who received non-neuraxial analgesia, Caesarean delivery rates were different across allele scores (χ2=12.4; P=0.015): 104/596 (17.4%) women with zero allele score underwent Caesarean delivery, compared with 654/5015 (13.0%) with allele score ≥1 (OR 1.41; 1.12-1.77). For women who had neuraxial analgesia, Caesarean delivery was not different across allele scores, ranging from 18.1% to 20.8% (χ2=0.29; P=0.99). Caesarean delivery was independent of type of analgesia for 818/7731 (10.6%) women with zero allele scores (OR 0.93; 0.63-1.39), but was higher in women receiving neuraxial analgesia with allele scores ≥1 (OR 1.55; 1.35-1.79; P<0.001). CONCLUSIONS Mendelian randomisation analysis suggests that higher IL-1ra levels are associated with reduced Caesarean delivery rate. Neuraxial analgesia appears to disrupt this link. CLINICAL TRIAL REGISTRATION UK Biobank study 62745.
Collapse
Affiliation(s)
- Gareth L. Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK,Corresponding author.
| | | | - Tom E.F. Abbott
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Ana Gutierrez del Arroyo
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Matthew J. Wilson
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Anna L. David
- Elizabeth Garret Anderson Institute for Women's Health, University College London, London, UK
| | | |
Collapse
|
20
|
Ayash TA, Vancolen SY, Segura M, Allard MJ, Sebire G. Protective Effects of Interleukin-1 Blockade on Group B Streptococcus-Induced Chorioamnionitis and Subsequent Neurobehavioral Impairments of the Offspring. Front Endocrinol (Lausanne) 2022; 13:833121. [PMID: 35846278 PMCID: PMC9283950 DOI: 10.3389/fendo.2022.833121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
Group B Streptococcus (GBS) is one of the most common bacteria isolated in human chorioamnionitis. Placental infection due to GBS is a major risk factor for fetal organ injuries, preterm birth, perinatal morbidity and mortality, and life-long multiorgan morbidities. Preclinical and clinical studies have shown that GBS-induced infection drives polymorphonuclear (PMN) cell infiltration within the placenta, the hallmark of human chorioamnionitis. In preclinical and clinical studies, the upregulation of interleukin(IL)-1β in the placenta and maternal/fetal blood was associated with a high risk of neurodevelopmental impairments in the progeny. We hypothesized that targeted IL-1 blockade administered to the dam alleviates GBS-induced chorioamnionitis and the downstream fetal inflammatory response syndrome (FIRS). IL-1 receptor antagonist (IL-1Ra) improved the gestational weight gain of GBS-infected dams and did not worsen the infectious manifestations. IL-1Ra reduced the IL-1β titer in the maternal sera of GBS-infected dams. IL-1Ra decreased the levels of IL-1β, IL-6, chemokine (C-X-C motif) ligand 1 (CXCL1), and polymorphonuclear (PMN) infiltration in GBS-infected placenta. IL-1Ra treatment reduced the IL-1β titer in the fetal sera of GBS-exposed fetuses. IL-1 blockade also alleviated GBS-induced FIRS and subsequent neurobehavioral impairments of the offspring without worsening the outcome of GBS infection. Altogether, these results showed that IL-1 plays a key role in the physiopathology of live GBS-induced chorioamnionitis and consequent neurobehavioral impairments.
Collapse
Affiliation(s)
| | | | - Mariela Segura
- Faculty of Veterinary Medicine, Université de Montreal, St-Hyacinthe, QC, Canada
| | | | - Guillaume Sebire
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- *Correspondence: Guillaume Sebire,
| |
Collapse
|
21
|
Kelly SB, Stojanovska V, Zahra VA, Moxham A, Miller SL, Moss TJM, Hooper SB, Nold MF, Nold-Petry CA, Dean JM, Bennet L, Polglase GR, Gunn AJ, Galinsky R. Interleukin-1 blockade attenuates white matter inflammation and oligodendrocyte loss after progressive systemic lipopolysaccharide exposure in near-term fetal sheep. J Neuroinflammation 2021; 18:189. [PMID: 34465372 PMCID: PMC8408978 DOI: 10.1186/s12974-021-02238-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Background Increased systemic and tissue levels of interleukin (IL)-1β are associated with greater risk of impaired neurodevelopment after birth. In this study, we tested the hypothesis that systemic IL-1 receptor antagonist (Ra) administration would attenuate brain inflammation and injury in near-term fetal sheep exposed to lipopolysaccharide (LPS). Methods Chronically instrumented near-term fetal sheep at 0.85 of gestation were randomly assigned to saline infusion (control, n = 9), repeated LPS infusions (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng, n = 8) or repeated LPS plus IL-1Ra infusions (13 mg/kg infused over 4 h) started 1 h after each LPS infusion (n = 9). Sheep were euthanized 4 days after starting infusions for histology. Results LPS infusions increased circulating cytokines and were associated with electroencephalogram (EEG) suppression with transiently reduced mean arterial blood pressure, and increased carotid artery perfusion and fetal heart rate (P < 0.05 vs. control for all). In the periventricular and intragyral white matter, LPS-exposure increased IL-1β immunoreactivity, numbers of caspase 3+ cells and microglia, reduced astrocyte and olig-2+ oligodendrocyte survival but did not change numbers of mature CC1+ oligodendrocytes, myelin expression or numbers of neurons in the cortex and subcortical regions. IL-1Ra infusions reduced circulating cytokines and improved recovery of EEG activity and carotid artery perfusion. Histologically, IL-1Ra reduced microgliosis, IL-1β expression and caspase-3+ cells, and improved olig-2+ oligodendrocyte survival. Conclusion IL-1Ra improved EEG activity and markedly attenuated systemic inflammation, microgliosis and oligodendrocyte loss following LPS exposure in near-term fetal sheep. Further studies examining the long-term effects on brain maturation are now needed. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02238-4.
Collapse
Affiliation(s)
- Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Vanesa Stojanovska
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Alison Moxham
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Timothy J M Moss
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Marcel F Nold
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Paediatrics, Monash University, Melbourne, Victoria, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Australia
| | - Claudia A Nold-Petry
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| | - Justin M Dean
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright street, Melbourne, Victoria, 3168, Australia. .,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
22
|
Liu G, Särén L, Douglasson H, Zhou XH, Åberg PM, Ollerstam A, Betts CJ, Balogh Sivars K. Precision cut lung slices: an ex vivo model for assessing the impact of immunomodulatory therapeutics on lung immune responses. Arch Toxicol 2021; 95:2871-2877. [PMID: 34191076 DOI: 10.1007/s00204-021-03096-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/15/2021] [Indexed: 11/28/2022]
Abstract
Chronic inflammatory diseases of the respiratory tract, such as chronic obstructive pulmonary disease (COPD) and asthma, are severe lung diseases that require effective treatments. In search for new medicines for these diseases, there is an unmet need for predictive and translatable disease-relevant in vitro/ex vivo models to determine the safety and efficacy of novel drug candidates. Here, we report the use of precision cut lung slices (PCLS) as a potential ex vivo platform to study compound effects in a physiologically relevant environment. PCLS derived from an elastase-challenged mouse model display key characteristics of increased inflammation ex vivo, which is exacerbated further upon challenge with LPS, mimicking the immune insult of a pathogen triggering disease exacerbation. Such LPS-induced inflammatory conditions are significantly abrogated by immunomodulatory agents targeting specific inflammatory signaling pathways in the absence of cytotoxic effects in lung slices. Thus, an ex vivo model of PCLS with a simulated pathogenic insult can replicate proposed in vivo pharmacological effects and thus could potentially act as a valuable tool to investigate the underlying mechanisms associated with lung safety, therapeutic efficacy and exacerbations with infection.
Collapse
Affiliation(s)
- Guanghui Liu
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Linnea Särén
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Helena Douglasson
- Bioscience Cough and in Vivo, Early Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Hong Zhou
- Patient Safety, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Per M Åberg
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Ollerstam
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine J Betts
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Kinga Balogh Sivars
- Clinical Testing, Global Procurement, Operations, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
23
|
Coler BS, Shynlova O, Boros-Rausch A, Lye S, McCartney S, Leimert KB, Xu W, Chemtob S, Olson D, Li M, Huebner E, Curtin A, Kachikis A, Savitsky L, Paul JW, Smith R, Adams Waldorf KM. Landscape of Preterm Birth Therapeutics and a Path Forward. J Clin Med 2021; 10:2912. [PMID: 34209869 PMCID: PMC8268657 DOI: 10.3390/jcm10132912] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Preterm birth (PTB) remains the leading cause of infant morbidity and mortality. Despite 50 years of research, therapeutic options are limited and many lack clear efficacy. Tocolytic agents are drugs that briefly delay PTB, typically to allow antenatal corticosteroid administration for accelerating fetal lung maturity or to transfer patients to high-level care facilities. Globally, there is an unmet need for better tocolytic agents, particularly in low- and middle-income countries. Although most tocolytics, such as betamimetics and indomethacin, suppress downstream mediators of the parturition pathway, newer therapeutics are being designed to selectively target inflammatory checkpoints with the goal of providing broader and more effective tocolysis. However, the relatively small market for new PTB therapeutics and formidable regulatory hurdles have led to minimal pharmaceutical interest and a stagnant drug pipeline. In this review, we present the current landscape of PTB therapeutics, assessing the history of drug development, mechanisms of action, adverse effects, and the updated literature on drug efficacy. We also review the regulatory hurdles and other obstacles impairing novel tocolytic development. Ultimately, we present possible steps to expedite drug development and meet the growing need for effective preterm birth therapeutics.
Collapse
Affiliation(s)
- Brahm Seymour Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Oksana Shynlova
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (O.S.); (A.B.-R.); (S.L.)
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5G 1E2, Canada
| | - Adam Boros-Rausch
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (O.S.); (A.B.-R.); (S.L.)
| | - Stephen Lye
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (O.S.); (A.B.-R.); (S.L.)
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5G 1E2, Canada
| | - Stephen McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Kelycia B. Leimert
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada; (K.B.L.); (W.X.); (D.O.)
| | - Wendy Xu
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada; (K.B.L.); (W.X.); (D.O.)
| | - Sylvain Chemtob
- Departments of Pediatrics, Université de Montréal, Montréal, QC H3T 1J4, Canada;
| | - David Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R7, Canada; (K.B.L.); (W.X.); (D.O.)
- Departments of Pediatrics and Physiology, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
- Department of Biological Sciencies, Columbia University, New York, NY 10027, USA
| | - Emily Huebner
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Anna Curtin
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Alisa Kachikis
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Leah Savitsky
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
| | - Jonathan W. Paul
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (J.W.P.); (R.S.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (J.W.P.); (R.S.)
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA; (B.S.C.); (S.M.); (M.L.); (E.H.); (A.C.); (A.K.); (L.S.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Gomez-Lopez N, Romero R, Varrey A, Leng Y, Miller D, Done B, Xu Y, Bhatti G, Motomura K, Gershater M, Pique-Regi R, Tarca AL. RNA Sequencing Reveals Diverse Functions of Amniotic Fluid Neutrophils and Monocytes/Macrophages in Intra-Amniotic Infection. J Innate Immun 2020; 13:63-82. [PMID: 33152737 DOI: 10.1159/000509718] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
Intra-amniotic infection, the invasion of microbes into the amniotic cavity resulting in inflammation, is a clinical condition that can lead to adverse pregnancy outcomes for the mother and fetus as well as severe long-term neonatal morbidities. Despite much research focused on the consequences of intra-amniotic infection, there remains little knowledge about the innate immune cells that respond to invading microbes. We performed RNA-seq of sorted amniotic fluid neutrophils and monocytes/macrophages from women with intra-amniotic infection to determine the transcriptomic differences between these innate immune cells. Further, we sought to identify specific transcriptomic pathways that were significantly altered by the maternal or fetal origin of amniotic fluid neutrophils and monocytes/macrophages, the presence of a severe fetal inflammatory response, and pregnancy outcome (i.e., preterm or term delivery). We show that significant transcriptomic differences exist between amniotic fluid neutrophils and monocytes/macrophages from women with intra-amniotic infection, indicating the distinct roles these cells play. The transcriptome of amniotic fluid immune cells varies based on their maternal or fetal origin, and the significant transcriptomic differences between fetal and maternal monocytes/macrophages imply that those of fetal origin exhibit impaired functions. Notably, transcriptomic changes in amniotic fluid monocytes/macrophages suggest that these immune cells collaborate with neutrophils in the trafficking of fetal leukocytes throughout the umbilical cord (i.e., funisitis). Finally, amniotic fluid neutrophils and monocytes/macrophages from preterm deliveries display enhanced transcriptional activity compared to those from term deliveries, highlighting the protective role of these cells during this vulnerable period. Collectively, these findings demonstrate the underlying complexity of local innate immune responses in women with intra-amniotic infection and provide new insights into the functions of neutrophils and monocytes/macrophages in the amniotic cavity.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA, .,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA, .,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA,
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA.,Detroit Medical Center, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Aneesha Varrey
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Meyer Gershater
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| |
Collapse
|
25
|
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, Collins JJP. Early origins of lung disease: towards an interdisciplinary approach. Eur Respir Rev 2020; 29:29/157/200191. [PMID: 33004528 DOI: 10.1183/16000617.0191-2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/18/2020] [Indexed: 12/27/2022] Open
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).
Collapse
Affiliation(s)
- Niki D J Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland.,Authors are listed alphabetically except for N.D.J. Ubags and J.J.P. Collins
| | - Miguel A Alejandre Alcazar
- Dept of Paediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, Translational Experimental Paediatrics, Experimental Pulmonology, University of Cologne, Cologne, Germany.,Centre of Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Suhas G Kallapur
- Neonatal-Perinatal Medicine, Dept of Pediatrics, David Geffen School of Medicine, UCLA Mattel Children's Hospital, Los Angeles, CA, USA
| | - Sylvia Knapp
- Dept of Medicine I/Research Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria.,CeMM, Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Sophie Lanone
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Clare M Lloyd
- Inflammation, Repair and Development, National Heart & Lung Institute, Imperial College London, London, UK
| | - Rory E Morty
- Dept of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Dept of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Centre, Member of the German Centre for Lung Research, Giessen, Germany
| | - Céline Pattaroni
- Dept of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Niki L Reynaert
- Dept of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Robbert J Rottier
- Dept of Paediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Hermelijn H Smits
- Dept of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Wouter A A de Steenhuijsen Piters
- Dept of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Centre Utrecht, Utrecht, The Netherlands.,National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | - Jennifer J P Collins
- Dept of Paediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands .,Authors are listed alphabetically except for N.D.J. Ubags and J.J.P. Collins
| |
Collapse
|
26
|
Xiong Y, Wintermark P. Therapeutic interventions for fetal inflammatory response syndrome (FIRS). Semin Fetal Neonatal Med 2020; 25:101112. [PMID: 32303464 DOI: 10.1016/j.siny.2020.101112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fetal inflammatory response syndrome (FIRS) is a condition defined by systemic inflammation in the fetus, a rapid increase of pro-inflammatory cytokines into the fetal circulation (including interleukin-1 and interleukin-6), as well as a cellular response (such as increased neutrophils, monocyte/macrophages, and T cells) and the presence of funisitis. FIRS can lead to death and multisystem organ damage in the fetus and newborn. Brain injuries and subsequent risk of cerebral palsy and cognitive impairments are the most threatening long-term complications. This paper reviews the definition of FIRS, summarizes its associated complications, briefly describes the available methods to study FIRS, and discusses in more detail the potential therapeutic candidates that have been so far studied to protect the fetus/newborn from FIRS and to alleviate its associated complications and sequelae.
Collapse
Affiliation(s)
- Ying Xiong
- Zhongnan Hospital of Wuhan University, Department of Anesthesiology, Wuhan University, Wuhan, China; Research Institute of the McGill University Health Centre, Montreal, Canada.
| | - Pia Wintermark
- Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, Montreal, Canada; Research Institute of the McGill University Health Centre, Montreal, Canada.
| |
Collapse
|
27
|
Jung E, Romero R, Yeo L, Diaz-Primera R, Marin-Concha J, Para R, Lopez AM, Pacora P, Gomez-Lopez N, Yoon BH, Kim CJ, Berry SM, Hsu CD. The fetal inflammatory response syndrome: the origins of a concept, pathophysiology, diagnosis, and obstetrical implications. Semin Fetal Neonatal Med 2020; 25:101146. [PMID: 33164775 PMCID: PMC10580248 DOI: 10.1016/j.siny.2020.101146] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetus can deploy a local or systemic inflammatory response when exposed to microorganisms or, alternatively, to non-infection-related stimuli (e.g., danger signals or alarmins). The term "Fetal Inflammatory Response Syndrome" (FIRS) was coined to describe a condition characterized by evidence of a systemic inflammatory response, frequently a result of the activation of the innate limb of the immune response. FIRS can be diagnosed by an increased concentration of umbilical cord plasma or serum acute phase reactants such as C-reactive protein or cytokines (e.g., interleukin-6). Pathologic evidence of a systemic fetal inflammatory response indicates the presence of funisitis or chorionic vasculitis. FIRS was first described in patients at risk for intraamniotic infection who presented preterm labor with intact membranes or preterm prelabor rupture of the membranes. However, FIRS can also be observed in patients with sterile intra-amniotic inflammation, alloimmunization (e.g., Rh disease), and active autoimmune disorders. Neonates born with FIRS have a higher rate of complications, such as early-onset neonatal sepsis, intraventricular hemorrhage, periventricular leukomalacia, and death, than those born without FIRS. Survivors are at risk for long-term sequelae that may include bronchopulmonary dysplasia, neurodevelopmental disorders, such as cerebral palsy, retinopathy of prematurity, and sensorineuronal hearing loss. Experimental FIRS can be induced by intra-amniotic administration of bacteria, microbial products (such as endotoxin), or inflammatory cytokines (such as interleukin-1), and animal models have provided important insights about the mechanisms responsible for multiple organ involvement and dysfunction. A systemic fetal inflammatory response is thought to be adaptive, but, on occasion, may become dysregulated whereby a fetal cytokine storm ensues and can lead to multiple organ dysfunction and even fetal death if delivery does not occur ("rescued by birth"). Thus, the onset of preterm labor in this context can be considered to have survival value. The evidence so far suggests that FIRS may compound the effects of immaturity and neonatal inflammation, thus increasing the risk of neonatal complications and long-term morbidity. Modulation of a dysregulated fetal inflammatory response by the administration of antimicrobial agents, anti-inflammatory agents, or cell-based therapy holds promise to reduce infant morbidity and mortality.
Collapse
Affiliation(s)
- Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA; Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA.
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Julio Marin-Concha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ashley M Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chong Jai Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
28
|
Sahoo D, Zaramela LS, Hernandez GE, Mai U, Taheri S, Dang D, Stouch AN, Medal RM, McCoy AM, Aschner JL, Blackwell TS, Zengler K, Prince LS. Transcriptional profiling of lung macrophages identifies a predictive signature for inflammatory lung disease in preterm infants. Commun Biol 2020; 3:259. [PMID: 32444859 PMCID: PMC7244484 DOI: 10.1038/s42003-020-0985-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Lung macrophages mature after birth, placing newborn infants, particularly those born preterm, within a unique window of susceptibility to disease. We hypothesized that in preterm infants, lung macrophage immaturity contributes to the development of bronchopulmonary dysplasia (BPD), the most common serious complication of prematurity. By measuring changes in lung macrophage gene expression in preterm patients at risk of BPD, we show here that patients eventually developing BPD had higher inflammatory mediator expression even on the first day of life. Surprisingly, the ex vivo response to LPS was similar across all samples. Our analysis did however uncover macrophage signature genes whose expression increased in the first week of life specifically in patients resilient to disease. We propose that these changes describe the dynamics of human lung macrophage differentiation. Our study therefore provides new mechanistic insight into both neonatal lung disease and human developmental immunology.
Collapse
Affiliation(s)
- Debashis Sahoo
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Livia S Zaramela
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gilberto E Hernandez
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Uyen Mai
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sahar Taheri
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Dharanidhar Dang
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ashley N Stouch
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rachel M Medal
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Alyssa M McCoy
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Judy L Aschner
- Department of Pediatrics, Joseph M Sanzari Children's Hospital, Hackensack University Medical Center, Hackensack Meridian School of Medicine at Seton Hall, Hackensack, NJ, 07110, USA
| | - Timothy S Blackwell
- Departments of Medicine, Cancer Biology, and Developmental Cell Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Karsten Zengler
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Lawrence S Prince
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
29
|
Mandell E, Ryan S, Seedorf GJ, Gonzalez T, Abman SH, Fleet JC. Maternal vitamin D deficiency induces transcriptomic changes in newborn rat lungs. J Steroid Biochem Mol Biol 2020; 199:105613. [PMID: 32007564 DOI: 10.1016/j.jsbmb.2020.105613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/14/2019] [Accepted: 01/29/2020] [Indexed: 12/24/2022]
Abstract
Vitamin D deficiency (VDD) during pregnancy is common and related to several maternal and fetal morbidities. Vitamin D (VD) plays a role in normal lung development and VDD causes abnormal airway, alveolar, and vascular growth in newborn rats. Here we use an unbiased transcriptomic approach to identify pathways altered in the lungs of offspring from VDD dams. The lungs of newborn offspring from VD replete and VDD dams were removed and RNA from these samples were analyzed using Affymetrix microarrays. Data were RMA normalized, differential gene expression was determined using Significance Analysis of Microarrays (5 % FDR) and pathway enrichment analysis was assessed. There were 2233 differentially expressed transcripts between the VDD and control lungs (1889 up, 344 down). Consistent with the suppression of lung growth in the VDD group, there were significant suppression of signal transduction pathways related to vascular biology and anabolic signaling pathways, e.g. the insulin-like growth factor-1 receptor (IGF-1R), fibroblast growth factor (FGF), cell cycle control. A major, enriched functional category was upregulation of pathways related to the innate immune system, including pathways for granulocyte and macrophage development, chemotaxis, and activation of cytokine signaling through Jak/Stat (e.g. resulting in higher IL1 α and β). We conclude that VDD during fetal development alters multiple pathways beyond the predicted angiogeneic alterations. These changes either contribute to, or reflect, the abnormal airway, alveolar, and vascular growth seen in the neonatal lung resulting from maternal VDD. The pattern also suggests abnormal lung development caused by maternal VDD creates a proinflammatory milieu that could contribute to the suppression of lung growth and development.
Collapse
Affiliation(s)
- Erica Mandell
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado, Denver Anschutz Medical Center, Aurora, CO, USA.
| | - Sharon Ryan
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado, Denver Anschutz Medical Center, Aurora, CO, USA
| | - Gregory J Seedorf
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado, Denver Anschutz Medical Center, Aurora, CO, USA
| | - Tania Gonzalez
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado, Denver Anschutz Medical Center, Aurora, CO, USA
| | - Steven H Abman
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado, Denver Anschutz Medical Center, Aurora, CO, USA
| | - James C Fleet
- Department of Nutrition Science, Purdue University Center for Cancer Research, West Lafayette, IN USA
| |
Collapse
|
30
|
Cappelletti M, Presicce P, Kallapur SG. Immunobiology of Acute Chorioamnionitis. Front Immunol 2020; 11:649. [PMID: 32373122 PMCID: PMC7177011 DOI: 10.3389/fimmu.2020.00649] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
31
|
Mustafa SB, Hernandez TF, Johnson-Pais TL, Kumar PA, Petershack JA, Henson BM, Seidner SR. IL-1 promotes α-epithelial Sodium Channel (α-ENaC) expression in murine lung epithelial cells: involvement of NF-κB. J Cell Commun Signal 2019; 14:303-314. [PMID: 31659629 DOI: 10.1007/s12079-019-00533-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 10/09/2019] [Indexed: 01/13/2023] Open
Abstract
Intra-amniotic exposure to proinflammatory cytokines such as interleukin-1 (IL-1) correlates with a decreased incidence of respiratory distress syndrome (RDS) in infants following premature birth. At birth, inadequate absorption of fluid from the fetal lung contributes to the onset RDS. Lung fluid clearance is coupled to Na+ transport via epithelial sodium channels (ENaC). In this study, we assessed the effects of IL-1 on the expression of ENaC, particularly the α-subunit which is critical for fetal lung fluid clearance at birth. Cultured mouse lung epithelial (MLE-12) cells were treated with either IL-1α or IL-1β to determine their effects on α-ENaC expression. Changes in IL-1-induced α-ENaC levels in the presence of IL-1 receptor antagonist (IL-1ra), cycloheximide, NF-κB inhibitor, and MAP kinase inhibitors were investigated. IL-1α and IL-1β independently induced a significant increase of α-ENaC mRNA and protein after 24 h compared to untreated cells. IL-1-dependent increases in α-ENaC protein were mitigated by IL-1ra and cycloheximide. IL-1 exposure induced NF-κB binding activity. Attenuation of IL-1-induced NF-κB activation by its inhibitor SN50 decreased α-ENaC protein abundance. Inhibition of ERK 1,2 MAPK significantly decreased both IL-1α and β-induced α-ENaC protein expression whereas inhibition of p38 MAPK only blocked IL-1β-induced α-ENaC protein levels. In contrast, IL-1-induced α-ENaC protein levels were unaffected by a c-Jun N-terminal kinase (JNK) inhibitor. Our results suggest that in MLE-12 cells, IL-1-induced elevation of α-ENaC is mediated via NF-κB activation and in part involves stimulation of the ERK 1,2 and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Shamimunisa B Mustafa
- Department of Pediatrics/Division of Neonatology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA.
| | - Tania F Hernandez
- Department of Pediatrics/Division of Neonatology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA
| | - Teresa L Johnson-Pais
- Department of Urology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA
| | - Pratap A Kumar
- Department of Urology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA
| | - Jean A Petershack
- Department of Pediatrics/Division of Neonatology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA
| | - Barbara M Henson
- Department of Pediatrics/Division of Neonatology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA
| | - Steven R Seidner
- Department of Pediatrics/Division of Neonatology, University of Texas Health Science Center, 7703 Floyd Curl Drive, MSC 7812, San Antonio, TX, 78229-3900, USA
| |
Collapse
|
32
|
Umbilical cord blood versus mesenchymal stem cells for inflammation-induced preterm brain injury in fetal sheep. Pediatr Res 2019; 86:165-173. [PMID: 30858474 DOI: 10.1038/s41390-019-0366-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/17/2019] [Accepted: 02/27/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Chorioamnionitis and fetal inflammation are principal causes of neuropathology detected after birth, particularly in very preterm infants. Preclinical studies show that umbilical cord blood (UCB) cells are neuroprotective, but it is uncertain if allogeneic UCB cells are a feasible early intervention for preterm infants. In contrast, mesenchymal stem cells (MSCs) are more readily accessible and show strong anti-inflammatory benefits. We aimed to compare the neuroprotective benefits of UCB versus MSCs in a large animal model of inflammation-induced preterm brain injury. We hypothesized that MSCs would afford greater neuroprotection. METHODS Chronically instrumented fetal sheep at 0.65 gestation received intravenous lipopolysaccharide (150 ng; 055:B5, n = 8) over 3 consecutive days; or saline for controls (n = 8). Cell-treated animals received 108 UCB mononuclear cells (n = 7) or 107 umbilical cord MSCs (n = 8), intravenously, 6 h after the final lipopolysaccharide dose. Seven days later, cerebrospinal fluid and brain tissue was collected for analysis. RESULTS Lipopolysaccharide induced neuroinflammation and apoptosis, and reduced the number of mature oligodendrocytes. MSCs reduced astrogliosis, but UCB did not have the same effect. UCB significantly decreased cerebral apoptosis and protected mature myelinating oligodendrocytes, but MSCs did not. CONCLUSION UCB appears to better protect white matter development in the preterm brain in response to inflammation-induced brain injury in fetal sheep.
Collapse
|
33
|
Butler B, De Dios R, Nguyen L, McKenna S, Ghosh S, Wright CJ. Developmentally Regulated Innate Immune NFκB Signaling Mediates IL-1α Expression in the Perinatal Murine Lung. Front Immunol 2019; 10:1555. [PMID: 31354715 PMCID: PMC6637303 DOI: 10.3389/fimmu.2019.01555] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/21/2019] [Indexed: 12/17/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common morbidity complicating premature birth. Importantly, preclinical models have demonstrated that IL-1 receptor antagonism prevents the lung injury and subsequent abnormal development that typically results following perinatal exposure to inflammatory stresses. This receptor is activated by two pro-inflammatory cytokines, IL-1α and IL-1β. While many studies have linked IL-1β to BPD development, IL-1α is relatively under-studied. The objective of our study was to determine whether systemic inflammatory stress induces IL-1α expression in the neonatal lung, and if so, whether this expression is mediated by innate immune NFκB signaling. We found that endotoxemia induced IL-1α expression during the saccular stage of neonatal lung development and was not present in the other neonatal organs or the adult lung. This IL-1α expression was dependent upon sustained pulmonary NFκB activation, which was specific to the neonatal lung. Using in vivo and in vitro approaches, we found that pharmacologic and genetic inhibition of NFκB signaling attenuated IL-1α expression. These findings demonstrate that innate immune regulation of IL-1α expression is developmentally regulated and occurs via an NFκB dependent mechanism. Importantly, the specific role of developmentally regulated pulmonary IL-1α expression remains unknown. Future studies must determine the effect of attenuating innate immune IL-1α expression in the developing lung before adopting broad IL-1 receptor antagonism as an approach to prevent neonatal lung injury.
Collapse
Affiliation(s)
- Brittany Butler
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Robyn De Dios
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Leanna Nguyen
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sarah McKenna
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
34
|
Papagianis PC, Pillow JJ, Moss TJ. Bronchopulmonary dysplasia: Pathophysiology and potential anti-inflammatory therapies. Paediatr Respir Rev 2019; 30:34-41. [PMID: 30201135 DOI: 10.1016/j.prrv.2018.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/17/2018] [Indexed: 12/26/2022]
Abstract
Inflammation of the preterm lungs is key to the pathogenesis of bronchopulmonary dysplasia (BPD), whether it arises as a consequence of intrauterine inflammation or postnatal respiratory management. This review explores steroidal and non-steroidal therapies for reducing neonatal pulmonary inflammation, aimed at treating or preventing BPD.
Collapse
Affiliation(s)
- Paris C Papagianis
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics and Gynecology, Monash University, Clayton, VIC 3168, Australia; Human Sciences and Centre for Neonatal Research and Education, The University of Western Australia, Crawley, WA, Australia.
| | - J J Pillow
- Human Sciences and Centre for Neonatal Research and Education, The University of Western Australia, Crawley, WA, Australia.
| | - Timothy J Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics and Gynecology, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
35
|
Shrestha AK, Bettini ML, Menon RT, Gopal VYN, Huang S, Edwards DP, Pammi M, Barrios R, Shivanna B. Consequences of early postnatal lipopolysaccharide exposure on developing lungs in mice. Am J Physiol Lung Cell Mol Physiol 2019; 316:L229-L244. [PMID: 30307313 PMCID: PMC6383495 DOI: 10.1152/ajplung.00560.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of infants that is characterized by interrupted lung development. Postnatal sepsis causes BPD, yet the contributory mechanisms are unclear. To address this gap, studies have used lipopolysaccharide (LPS) during the alveolar phase of lung development. However, the lungs of infants who develop BPD are still in the saccular phase of development, and the effects of LPS during this phase are poorly characterized. We hypothesized that chronic LPS exposure during the saccular phase disrupts lung development by mechanisms that promote inflammation and prevent optimal lung development and repair. Wild-type C57BL6J mice were intraperitoneally administered 3, 6, or 10 mg/kg of LPS or a vehicle once daily on postnatal days (PNDs) 3-5. The lungs were collected for proteomic and genomic analyses and flow cytometric detection on PND6. The impact of LPS on lung development, cell proliferation, and apoptosis was determined on PND7. Finally, we determined differences in the LPS effects between the saccular and alveolar lungs. LPS decreased the survival and growth rate and lung development in a dose-dependent manner. These effects were associated with a decreased expression of proteins regulating cell proliferation and differentiation and increased expression of those mediating inflammation. While the lung macrophage population of LPS-treated mice increased, the T-regulatory cell population decreased. Furthermore, LPS-induced inflammatory and apoptotic response and interruption of cell proliferation and alveolarization was greater in alveolar than in saccular lungs. Collectively, the data support our hypothesis and reveal several potential therapeutic targets for sepsis-mediated BPD in infants.
Collapse
Affiliation(s)
- Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Matthew L Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, McNair Medical Institute, Baylor College of Medicine , Houston, Texas
| | - Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Vashisht Y N Gopal
- Department of Melanoma Medical Oncology and Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, Texas
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine , Houston, Texas
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine , Houston, Texas
| | - Mohan Pammi
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital , Houston, Texas
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
36
|
del Arroyo A, Sanchez J, Patel S, Phillips S, Reyes A, Cubillos C, Fernando R, David A, Sultan P, Ackland G, Reeve A, Sodha S, Ciechanowicz S, Olearo E, Dick J, Stewart A. Role of leucocyte caspase-1 activity in epidural-related maternal fever: a single-centre, observational, mechanistic cohort study. Br J Anaesth 2019; 122:92-102. [DOI: 10.1016/j.bja.2018.09.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 09/03/2018] [Accepted: 09/24/2018] [Indexed: 01/19/2023] Open
|
37
|
Schüller SS, Kramer BW, Villamor E, Spittler A, Berger A, Levy O. Immunomodulation to Prevent or Treat Neonatal Sepsis: Past, Present, and Future. Front Pediatr 2018; 6:199. [PMID: 30073156 PMCID: PMC6060673 DOI: 10.3389/fped.2018.00199] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Despite continued advances in neonatal medicine, sepsis remains a leading cause of death worldwide in neonatal intensive care units. The clinical presentation of sepsis in neonates varies markedly from that in older children and adults, and distinct acute inflammatory responses results in age-specific inflammatory and protective immune response to infection. This review first provides an overview of the neonatal immune system, then covers current mainstream, and experimental preventive and adjuvant therapies in neonatal sepsis. We also discuss how the distinct physiology of the perinatal period shapes early life immune responses and review strategies to reduce neonatal sepsis-related morbidity and mortality. A summary of studies that characterize immune ontogeny and neonatal sepsis is presented, followed by discussion of clinical trials assessing interventions such as breast milk, lactoferrin, probiotics, and pentoxifylline. Finally, we critically appraise future treatment options such as stem cell therapy, other antimicrobial protein and peptides, and targeting of pattern recognition receptors in an effort to prevent and/or treat sepsis in this highly vulnerable neonatal population.
Collapse
Affiliation(s)
- Simone S. Schüller
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Boris W. Kramer
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Andreas Spittler
- Department of Surgery, Research Labs & Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Boston, MA, United States
| |
Collapse
|
38
|
Kothe TB, Royse E, Kemp MW, Usuda H, Saito M, Musk GC, Jobe AH, Hillman NH. Epidermal growth factor receptor inhibition with Gefitinib does not alter lung responses to mechanical ventilation in fetal, preterm lambs. PLoS One 2018; 13:e0200713. [PMID: 30005089 PMCID: PMC6044532 DOI: 10.1371/journal.pone.0200713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/02/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is important for airway branching and lung maturation. Mechanical ventilation of preterm lambs causes increases in EGFR and EGFR ligand mRNA in the lung. Abnormal EGFR signaling may contribute to the development of bronchopulmonary dysplasia. HYPOTHESIS Inhibition of EGFR signaling will decrease airway epithelial cell proliferation and lung inflammation caused by mechanical ventilation in preterm, fetal sheep. METHODS Following exposure of the fetal head and chest at 123±1 day gestational age and with placental circulation intact, fetal lambs (n = 4-6/group) were randomized to either: 1) Gefitinib 15 mg IV and 1 mg intra-tracheal or 2) saline IV and IT. Lambs were further assigned to 15 minutes of either: a) Injurious mechanical ventilation (MV) or b) Continuous positive airway pressure (CPAP) 5 cmH2O. After the 15 minute intervention, the animals were returned to the uterus and delivered after i) 6 or ii) 24 hours in utero. RESULTS MV caused lung injury and inflammation, increased lung mRNA for cytokines and EGFR ligands, caused airway epithelial cell proliferation, and decreased airway epithelial phosphorylated ERK1/2. Responses to MV were unchanged by Gefitinib. Gefitinib altered expression of EGFR mRNA in the lung and liver of both CPAP and MV animals. Gefitinib decreased the liver SAA3 mRNA response to MV at 6 hours. There were no differences in markers of lung injury or inflammation between CPAP animals receiving Gefitinib or saline. CONCLUSION Inhibition of the EGFR pathway did not alter acute lung inflammation or injury from mechanical ventilation in preterm sheep.
Collapse
Affiliation(s)
- T. Brett Kothe
- Division of Neonatology, Cardinal Glennon Children’s Hospital, Saint Louis University, Saint Louis, Missouri, United States of America
| | - Emily Royse
- Division of Neonatology, Cardinal Glennon Children’s Hospital, Saint Louis University, Saint Louis, Missouri, United States of America
| | - Matthew W. Kemp
- School of Women’s and Infants’ Health, University of Western Australia, Perth, Western Australia, Australia
| | - Haruo Usuda
- School of Women’s and Infants’ Health, University of Western Australia, Perth, Western Australia, Australia
| | - Masatoshi Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Gabrielle C. Musk
- Animal Care Services, University of Western Australia, Perth, Western Australia, Australia
| | - Alan H. Jobe
- School of Women’s and Infants’ Health, University of Western Australia, Perth, Western Australia, Australia
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Noah H. Hillman
- Division of Neonatology, Cardinal Glennon Children’s Hospital, Saint Louis University, Saint Louis, Missouri, United States of America
| |
Collapse
|
39
|
Pan J, Zhan C, Yuan T, Wang W, Shen Y, Sun Y, Wu T, Gu W, Chen L, Yu H. Effects and molecular mechanisms of intrauterine infection/inflammation on lung development. Respir Res 2018; 19:93. [PMID: 29747649 PMCID: PMC5946538 DOI: 10.1186/s12931-018-0787-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/23/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Intrauterine infection/inflammation plays an important role in the development of lung injury and bronchopulmonary dysplasia (BPD) in preterm infants, While a multifactorial genesis is likely, mechanisms involved in BPD after intrauterine infection/inflammation are largely unknown. Recent studies have suggested microRNAs (miRNAs) are likely to play a role. Therefore, this study aimed to study the effects and mechanisms of intrauterine infection/inflammation on lung development, and to identify miRNAs related to lung injury and BPD. METHODS An animal model of intrauterine infection/inflammation was established with pregnant SD rats endocervically inoculated with E.coli. The fetal and neonatal rats were observed at embryonic day (E) 17, 19, 21 and postnatal day (P) 1, 3, 7, 14, respectively. Body weight, lung weight, the expression levels of NLRP3, TNF-α, IL-lβ, IL-6, VEGF, Collagen I, SP-A, SP-B and SP-C in the lung tissues of fetal and neonatal rats were measured. Expression profiles of 1218 kinds of miRNAs in the lungs of neonatal rats were detected by miRNA microarray technique. Target genes of the identified miRNAs were predicted through online software. RESULTS Intrauterine infection/inflammation compromised not only weight development but also lung development of the fetal and neonatal rats. The results showed significantly increased expression of NLRP3, TNF-α, IL-1β, IL-6, Collagen I, and significantly decreased expression of VEGF, SP-A, SP-B and SP-C in the fetal and neonatal rat lung tissues in intrauterine infection group compared to the control group at different observation time point (P < 0.05). Forty-three miRNAs with significant differential expression were identified. Possible target genes regulated by the identified miRNAs are very rich. CONCLUSIONS Intrauterine infection/inflammation results in lung histological changes which are very similar to those observed in BPD. Possible mechanisms may include NLRP3 inflammasome activation followed by inflammatory cytokines expression up-regulated, inhibiting the expression of pulmonary surfactant proteins, interfering with lung interstitial development. There are many identified miRNAs which target a wide range of genes and may play an important role in the processes of lung injury and BPD.
Collapse
Affiliation(s)
- Jiarong Pan
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Canyang Zhan
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Tianming Yuan
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Weiyan Wang
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Ying Shen
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Yi Sun
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Tai Wu
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Weizhong Gu
- Zhejiang Key Laboratory for Diagnosis and Therapy of Neonatal Disease, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Lihua Chen
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| | - Huimin Yu
- Department of Neonatology, Children’s Hospital, Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310052 People’s Republic of China
| |
Collapse
|
40
|
Kothe TB, Royse E, Kemp MW, Schmidt A, Salomone F, Saito M, Usuda H, Watanabe S, Musk GC, Jobe AH, Hillman NH. Effects of budesonide and surfactant in preterm fetal sheep. Am J Physiol Lung Cell Mol Physiol 2018; 315:L193-L201. [PMID: 29671605 DOI: 10.1152/ajplung.00528.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanical ventilation causes lung injury and systemic inflammatory responses in preterm sheep and is associated with bronchopulmonary dysplasia (BPD) in preterm infants. Budesonide added to surfactant decreased BPD by 20% in infants. We wanted to determine the effects of budesonide and surfactant on injury from high tidal volume (VT) ventilation in preterm lambs. Ewes at 125 ± 1 days gestational age had fetal surgery to expose fetal head and chest with placental circulation intact. Lambs were randomized to 1) mechanical ventilation with escalating VT to target 15 ml/kg by 15 min or 2) continuous positive airway pressure (CPAP) of 5 cmH2O. After the 15-min intervention, lambs were given surfactant 100 mg/kg with saline, budesonide 0.25 mg/kg, or budesonide 1 mg/kg. The fetuses were returned to the uterus for 24 h and then delivered and ventilated for 30 min to assess lung function. Budesonide levels were low in lung and plasma. CPAP groups had improved oxygenation, ventilation, and decreased injury markers compared with fetal VT lambs. Budesonide improved ventilation in CPAP lambs. Budesonide decreased lung weights and lung liquid and increased lung compliance and surfactant protein mRNA. Budesonide decreased proinflammatory and acute-phase responses in lung. Airway thickness increased in animals not receiving budesonide. Systemically, budesonide decreased monocyte chemoattractant protein-1 mRNA and preserved glycogen in liver. Results with 0.25 and 1 mg/kg budesonide were similar. We concluded that budesonide with surfactant matured the preterm lung and decreased the liver responses but did not improve lung function after high VT injury in fetal sheep.
Collapse
Affiliation(s)
- T Brett Kothe
- Division of Neonatology, Cardinal Glennon Children's Hospital, Saint Louis University , Saint Louis, Missouri
| | - Emily Royse
- Division of Neonatology, Cardinal Glennon Children's Hospital, Saint Louis University , Saint Louis, Missouri
| | - Matthew W Kemp
- School of Women's and Infants' Health, University of Western Australia , Perth, Western Australia , Australia
| | - Augusto Schmidt
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, Ohio
| | - Fabrizio Salomone
- Department of Preclinical Pharmacology Research and Development, Chiesi Farmaceutici, Parma , Italy
| | - Masatoshi Saito
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital , Sendai , Japan
| | - Haruo Usuda
- School of Women's and Infants' Health, University of Western Australia , Perth, Western Australia , Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital , Sendai , Japan
| | - Shimpei Watanabe
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital , Sendai , Japan
| | - Gabrielle C Musk
- Animal Care Services, University of Western Australia , Perth, Western Australia , Australia
| | - Alan H Jobe
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, Ohio.,School of Women's and Infants' Health, University of Western Australia , Perth, Western Australia , Australia
| | - Noah H Hillman
- Division of Neonatology, Cardinal Glennon Children's Hospital, Saint Louis University , Saint Louis, Missouri
| |
Collapse
|
41
|
Presicce P, Park CW, Senthamaraikannan P, Bhattacharyya S, Jackson C, Kong F, Rueda CM, DeFranco E, Miller LA, Hildeman DA, Salomonis N, Chougnet CA, Jobe AH, Kallapur SG. IL-1 signaling mediates intrauterine inflammation and chorio-decidua neutrophil recruitment and activation. JCI Insight 2018; 3:98306. [PMID: 29563340 DOI: 10.1172/jci.insight.98306] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/13/2018] [Indexed: 12/31/2022] Open
Abstract
Neutrophil infiltration of the chorioamnion-decidua tissue at the maternal-fetal interface (chorioamnionitis) is a leading cause of prematurity, fetal inflammation, and perinatal mortality. We induced chorioamnionitis in preterm rhesus macaques by intraamniotic injection of LPS. Here, we show that, during chorioamnionitis, the amnion upregulated phospho-IRAK1-expressed neutrophil chemoattractants CXCL8 and CSF3 in an IL-1-dependent manner. IL-1R blockade decreased chorio-decidua neutrophil accumulation, neutrophil activation, and IL-6 and prostaglandin E2 concentrations in the amniotic fluid. Neutrophils accumulating in the chorio-decidua had increased survival mediated by BCL2A1, and IL-1R blockade also decreased BCL2A1+ chorio-decidua neutrophils. Readouts for inflammation in a cohort of women with preterm delivery and chorioamnionitis were similar to findings in the rhesus macaques. IL-1 is a potential therapeutic target for chorioamnionitis and associated morbidities.
Collapse
Affiliation(s)
| | | | | | | | - Courtney Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Cesar M Rueda
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily DeFranco
- Department of Obstetrics/Gynecology, Maternal-Fetal Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Lisa A Miller
- California National Primate Research Center, Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UCD, Davis, California, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nathan Salomonis
- Division of Biomedical informatics, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
42
|
Visconti K, Senthamaraikannan P, Kemp MW, Saito M, Kramer BW, Newnham JP, Jobe AH, Kallapur SG. Extremely preterm fetal sheep lung responses to antenatal steroids and inflammation. Am J Obstet Gynecol 2018; 218:349.e1-349.e10. [PMID: 29274832 DOI: 10.1016/j.ajog.2017.12.207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/27/2017] [Accepted: 12/14/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND The efficacy of antenatal steroids for fetal lung maturation in the periviable period is not fully understood. OBJECTIVE We sought to determine the lung maturational effects of antenatal steroids and inflammation in early gestation sheep fetuses, similar to the periviable period in human beings. STUDY DESIGN Date-mated ewes with singleton fetuses were randomly assigned to 1 of 4 treatment groups (n = 8/group): (1) maternal intramuscular injection of betamethasone; (2) intraamniotic lipopolysaccharide; (3) betamethasone + lipopolysaccharide; and (4) intraamniotic + intramuscular saline (controls). Fetuses were delivered surgically 48 hours later at 94 days' gestation (63% term gestation) for comprehensive evaluations of lung maturation, and lung and systemic inflammation. RESULTS Relative to controls, first, betamethasone increased the fetal lung air space to mesenchymal area ratio by 47% but did not increase the messenger RNAs for the surfactant proteins-B and -C that are important for surfactant function or increase the expression of pro-surfactant protein-C in the alveolar type II cells. Second, betamethasone increased expression of 1 of the 4 genes in surfactant lipid synthetic pathways. Third, betamethasone increased genes involved in epithelium sodium channel transport, but not sodium-potassium adenosine triphosphatase or Aquaporin 5. Fourth, lipopolysaccharide increased proinflammatory genes in the lung but did not effectively recruit activated inflammatory cells. Last, betamethasone incompletely suppressed lipopolysaccharide-induced lung inflammation. In the liver, betamethasone when given alone increased the expression of serum amyloid A3 and C-reactive protein messenger RNAs. CONCLUSION Compared the more mature 125-day gestation sheep, antenatal steroids do not induce pulmonary surfactants during the periviable period, indicating a different response.
Collapse
|
43
|
Vagus nerve stimulation in pregnant rats and effects on inflammatory markers in the brainstem of neonates. Pediatr Res 2018; 83:514-519. [PMID: 29053705 PMCID: PMC5866172 DOI: 10.1038/pr.2017.265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/25/2017] [Indexed: 01/29/2023]
Abstract
BackgroundVagus nerve stimulation (VNS) is an Food and Drug Administration-approved method delivering electrical impulses for treatment of depression and epilepsy in adults. The vagus nerve innervates the majority of visceral organs and cervix, but potential impacts of VNS on the progress of pregnancy and the fetus are not well studied.MethodsWe tested the hypothesis that VNS in pregnant dams does not induce inflammatory changes in the cardio-respiratory control regions of the pups' brainstem, potentially impacting the morbidity and mortality of offspring. Pregnant dams were implanted with stimulators providing intermittent low or high frequency electrical stimulation of the sub-diaphragmatic esophageal segment of the vagus nerve for 6-7 days until delivery. After birth, we collected pup brainstems that included cardio-respiratory control regions and counted the cells labeled for pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, tumor necrosis factor-α) and high mobility group box 1.ResultsNeither pup viability nor number of cells labeled for pro-inflammatory cytokines in nucleus tractus solitarii or hypoglossal motor nucleus was impaired by VNS. We provide evidence suggesting that chronic VNS of pregnant mothers does not impede the progress or outcome of pregnancy.ConclusionVNS does not cause preterm birth, affect well-being of progeny, or impact central inflammatory processes that are critical for normal cardiovascular and respiratory function in newborns.
Collapse
|
44
|
McKenna S, Butler B, Jatana L, Ghosh S, Wright CJ. Inhibition of IκBβ/NFκB signaling prevents LPS-induced IL1β expression without increasing apoptosis in the developing mouse lung. Pediatr Res 2017; 82:1064-1072. [PMID: 28753596 PMCID: PMC5761659 DOI: 10.1038/pr.2017.182] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023]
Abstract
BackgroundThe pro-inflammatory consequences of IL1β expression contribute to the pathogenesis of bronchopulmonary dysplasia. Selectively targeting Lipopolysaccharide (LPS)-induced IκBβ/NFκB signaling attenuates IL1β mRNA expression in macrophages. Whether targeting IκBβ/NFκB signaling affects the anti-apoptotic gene expression, a known consequence of global LPS-induced NFκB inhibition, is unknown.MethodsMacrophages (RAW 264.7, bone marrow-derived macrophage) were assessed for LPS-induced IL1β mRNA/protein expression, anti-apoptotic gene expression, cell viability (trypan blue exclusion), and activation of apoptosis (caspase-3 and PARP cleavage) following pharmacologic and genetic attenuation of IκBβ/NFκB signaling. Expressions of IL1β and anti-apoptotic genes were assessed in endotoxemic newborn mice (P0) with intact (WT), absent (IκBβ KO), and attenuated (IκBβ overexpressing) IκBβ/NFκB signaling.ResultsIn cultured macrophages, pharmacologic and genetic inhibition of LPS-induced IκBβ/NFκB signaling significantly attenuated IL1β mRNA and protein expression. Importantly, targeting IκBβ/NFκB signaling did not attenuate LPS-induced expression of anti-apoptotic genes or result in cell death. In endotoxemic neonatal mice, targeting LPS-induced IκBβ/NFκB signaling significantly attenuated pulmonary IL1β expression without affecting the anti-apoptotic gene expression.ConclusionTargeting IκBβ/NFκB signaling prevents LPS-induced IL1β expression without inducing apoptosis in cultured macrophages and in the lungs of endotoxemic newborn mice. Inhibiting this pathway may prevent inflammatory injury without affecting the protective role of NFκB activity in the developing lung.
Collapse
Affiliation(s)
- Sarah McKenna
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Brittany Butler
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Laurie Jatana
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Sankar Ghosh
- Department of Microbiology & Immunology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Clyde J. Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| |
Collapse
|
45
|
Boonkasidecha S, Kannan PS, Kallapur SG, Jobe AH, Kemp MW. Fetal skin as a pro-inflammatory organ: Evidence from a primate model of chorioamnionitis. PLoS One 2017; 12:e0184938. [PMID: 28957335 PMCID: PMC5619751 DOI: 10.1371/journal.pone.0184938] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/01/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Intrauterine infection is a primary cause of preterm birth and fetal injury. The pro-inflammatory role of the fetal skin in the setting of intrauterine infection remains poorly characterized. Whether or not inflammation of the fetal skin occurs in primates remains unstudied. Accordingly, we hypothesized that: i) the fetal primate skin would mount a pro-inflammatory response to preterm birth associated pro-inflammatory agents (lipopolysaccharides from Escherichia coli, live Ureaplasma parvum, interleukin-1β) and; ii) that inhibiting interleukin-1 signaling would decrease the skin inflammatory response. METHODS Rhesus macaques with singleton pregnancies received intraamniotic injections of either sterile saline (control) or one of three pro-inflammatory agonists: E. coli lipopolysaccharides, interluekin-1β or live U. parvum under ultrasound guidance. A fourth group of animals received both E. coli lipopolysaccharide and interleukin-1 signaling inhibitor interleukin-1 receptor antagonist (Anakinra) prior to delivery. Animals were surgically delivered at approximately 130 days' gestational age. RESULTS Intraamniotic lipopolysaccharide caused an inflammatory skin response characterized by increases in interluekin-1β,-6 and -8 mRNA at 16 hours. There was a modest inflammatory response to U. parvum, but interleukin-1β alone caused no inflammatory response in the fetal skin. Intraamniotic Anakinra treatment of lipopolysaccharide-exposed animals significantly reduced skin inflammation. CONCLUSIONS Intraamniotic lipopolysaccharide and U. parvum were associated with modest increases in the expression of inflammatory mediators in primate fetal skin. Although administration of Interleukin-1β alone did not elicit an inflammatory response, lipopolysaccharide-driven skin inflammation was decreased following intraamniotic Anakinra therapy. These findings provide support for the role of the fetal skin in the development of the fetal inflammatory response.
Collapse
Affiliation(s)
- Suppawat Boonkasidecha
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio, United States of America.,Division of Neonatology, Department of Pediatrics, Queen Sirikit National Institute of Child Health, College of Medicine, Rangsit University, Bangkok, Thailand
| | - Paranthaman Senthamarai Kannan
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio, United States of America
| | - Suhas G Kallapur
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio, United States of America.,School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Alan H Jobe
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio, United States of America.,School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Matthew W Kemp
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| |
Collapse
|
46
|
Ribeiro A, Mayer C, Wilson C, Martin R, MacFarlane P. Intratracheal LPS administration attenuates the acute hypoxic ventilatory response: Role of brainstem IL-1β receptors. Respir Physiol Neurobiol 2017; 242:45-51. [DOI: 10.1016/j.resp.2017.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 01/01/2023]
|
47
|
Collins JJP, Tibboel D, de Kleer IM, Reiss IKM, Rottier RJ. The Future of Bronchopulmonary Dysplasia: Emerging Pathophysiological Concepts and Potential New Avenues of Treatment. Front Med (Lausanne) 2017; 4:61. [PMID: 28589122 PMCID: PMC5439211 DOI: 10.3389/fmed.2017.00061] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
Yearly more than 15 million babies are born premature (<37 weeks gestational age), accounting for more than 1 in 10 births worldwide. Lung injury caused by maternal chorioamnionitis or preeclampsia, postnatal ventilation, hyperoxia, or inflammation can lead to the development of bronchopulmonary dysplasia (BPD), one of the most common adverse outcomes in these preterm neonates. BPD patients have an arrest in alveolar and microvascular development and more frequently develop asthma and early-onset emphysema as they age. Understanding how the alveoli develop, and repair, and regenerate after injury is critical for the development of therapies, as unfortunately there is still no cure for BPD. In this review, we aim to provide an overview of emerging new concepts in the understanding of perinatal lung development and injury from a molecular and cellular point of view and how this is paving the way for new therapeutic options to prevent or treat BPD, as well as a reflection on current treatment procedures.
Collapse
Affiliation(s)
- Jennifer J P Collins
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Ismé M de Kleer
- Division of Pediatric Pulmonology, Department of Pediatrics, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Division of Neonatology, Department of Pediatrics, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
48
|
Nadeau-Vallée M, Chin PY, Belarbi L, Brien MÈ, Pundir S, Berryer MH, Beaudry-Richard A, Madaan A, Sharkey DJ, Lupien-Meilleur A, Hou X, Quiniou C, Beaulac A, Boufaied I, Boudreault A, Carbonaro A, Doan ND, Joyal JS, Lubell WD, Olson DM, Robertson SA, Girard S, Chemtob S. Antenatal Suppression of IL-1 Protects against Inflammation-Induced Fetal Injury and Improves Neonatal and Developmental Outcomes in Mice. THE JOURNAL OF IMMUNOLOGY 2017; 198:2047-2062. [PMID: 28148737 DOI: 10.4049/jimmunol.1601600] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/30/2016] [Indexed: 01/08/2023]
Abstract
Preterm birth (PTB) is commonly accompanied by in utero fetal inflammation, and existing tocolytic drugs do not target fetal inflammatory injury. Of the candidate proinflammatory mediators, IL-1 appears central and is sufficient to trigger fetal loss. Therefore, we elucidated the effects of antenatal IL-1 exposure on postnatal development and investigated two IL-1 receptor antagonists, the competitive inhibitor anakinra (Kineret) and a potent noncompetitive inhibitor 101.10, for efficacy in blocking IL-1 actions. Antenatal exposure to IL-1β induced Tnfa, Il6, Ccl2, Pghs2, and Mpges1 expression in placenta and fetal membranes, and it elevated amniotic fluid IL-1β, IL-6, IL-8, and PGF2α, resulting in PTB and marked neonatal mortality. Surviving neonates had increased Il1b, Il6, Il8, Il10, Pghs2, Tnfa, and Crp expression in WBCs, elevated plasma levels of IL-1β, IL-6, and IL-8, increased IL-1β, IL-6, and IL-8 in fetal lung, intestine, and brain, and morphological abnormalities: e.g., disrupted lung alveolarization, atrophy of intestinal villus and colon-resident lymphoid follicle, and degeneration and atrophy of brain microvasculature with visual evoked potential anomalies. Late gestation treatment with 101.10 abolished these adverse outcomes, whereas Kineret exerted only modest effects and no benefit for gestation length, neonatal mortality, or placental inflammation. In a LPS-induced model of infection-associated PTB, 101.10 prevented PTB, neonatal mortality, and fetal brain inflammation. There was no substantive deviation in postnatal growth trajectory or adult body morphometry after antenatal 101.10 treatment. The results implicate IL-1 as an important driver of neonatal morbidity in PTB and identify 101.10 as a safe and effective candidate therapeutic.
Collapse
Affiliation(s)
- Mathieu Nadeau-Vallée
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Peck-Yin Chin
- Department of Obstetrics and Gynecology, Adelaide Medical School and Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Lydia Belarbi
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Marie-Ève Brien
- Department of Obstetrics and Gynecology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada.,Department of Microbiology, Infectiology, and Immunology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada
| | - Sheetal Pundir
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Martin H Berryer
- Department of Neurosciences, CHU Sainte-Justine Research Center, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Alexandra Beaudry-Richard
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Ankush Madaan
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - David J Sharkey
- Department of Obstetrics and Gynecology, Adelaide Medical School and Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Alexis Lupien-Meilleur
- Department of Neurosciences, CHU Sainte-Justine Research Center, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Xin Hou
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Christiane Quiniou
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Alexandre Beaulac
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Ines Boufaied
- Department of Obstetrics and Gynecology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada.,Department of Microbiology, Infectiology, and Immunology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada
| | - Amarilys Boudreault
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada
| | - Adriana Carbonaro
- Department of Obstetrics and Gynecology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada.,Department of Microbiology, Infectiology, and Immunology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada
| | - Ngoc-Duc Doan
- Department of Chemistry, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Jean-Sebastien Joyal
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - William D Lubell
- Department of Chemistry, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - David M Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R3, Canada; and.,Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Sarah A Robertson
- Department of Obstetrics and Gynecology, Adelaide Medical School and Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia;
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada; .,Department of Microbiology, Infectiology, and Immunology, University of Montreal, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1J4, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada; .,Department of Ophthalmology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec H3T 1C5, Canada.,Department of Pharmacology, University of Montreal, Montreal, Quebec H3T 1J4, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
49
|
Jackson CM, Wells CB, Tabangin ME, Meinzen-Derr J, Jobe AH, Chougnet CA. Pro-inflammatory immune responses in leukocytes of premature infants exposed to maternal chorioamnionitis or funisitis. Pediatr Res 2017; 81:384-390. [PMID: 27814345 PMCID: PMC5309139 DOI: 10.1038/pr.2016.232] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Acute chorioamnionitis contributes to premature birth, and is associated with postbirth complications. How chorioamnionitis impacts neonate's developing immune system has not been well defined. METHODS Blood from extremely preterm infants (≤28 wk gestation) was drawn at the first, second, and fourth week of life. Blood was either left unstimulated or stimulated for 4 h with PMA/ionomycin. mRNA expression of transcription factors in unstimulated cells (RORC, TBET, GATA3, and Forkhead box protein 3 (FOXP3)) and inflammatory cytokines (IFN-γ, TNF-α, IL-2, IL-4, IL-5, and IL-6) in unstimulated and stimulated cells were analyzed. Data were analyzed based on the diagnosis of chorioamnionitis, funisitis and bronchopulmonary dysplasia (BPD). RESULTS At 1 wk of life, exposure to funisitis, but not maternal chorioamnionitis was associated with an increased expression of RORC and RORC/FOXP3 ratio. These increases in RORC and RORC/FOXP3 ratio were sustained over the 4 wk of follow-up. Leukocytes from infants who developed BPD had increased stimulated and unstimulated IL-4 at the first week of life, but these increases were not sustained over time. In contrast, infants with mild BPD had a sustained decrease in stimulated IL-2. CONCLUSION Chorioamnionitis exposure, in particular to funisitis, lead to enhanced Th17-like responses that persist for 4 wk after birth. Infants who later developed BPD did not exhibit a strikingly distinct immune profile.
Collapse
Affiliation(s)
- Courtney M. Jackson
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio. USA
| | - Casey B. Wells
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio. USA
| | - Meredith E. Tabangin
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jareen Meinzen-Derr
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Alan H. Jobe
- Division of Neonatology/Pulmonary Biology, the Perinatal Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, OH, USA
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio. USA
| |
Collapse
|
50
|
Petersen RY, Royse E, Kemp MW, Miura Y, Noe A, Jobe AH, Hillman NH. Distending Pressure Did Not Activate Acute Phase or Inflammatory Responses in the Airways and Lungs of Fetal, Preterm Lambs. PLoS One 2016; 11:e0159754. [PMID: 27463520 PMCID: PMC4962990 DOI: 10.1371/journal.pone.0159754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/06/2016] [Indexed: 11/18/2022] Open
Abstract
Background Mechanical ventilation at birth causes airway injury and lung inflammation in preterm sheep. Continuous positive airway pressure (CPAP) is being increasingly used clinically to transition preterm infants at birth. Objective To test if distending pressures will activate acute phase reactants and inflammatory changes in the airways of fetal, preterm lambs. Methods The head and chest of fetal lambs at 128±1 day GA were surgically exteriorized. With placental circulation intact, fetal lambs were then randomized to one of five 15 minute interventions: PEEP of 0, 4, 8, 12, or 16 cmH2O. Recruitment volumes were recorded. Fetal lambs remained on placental support for 30 min after the intervention. The twins of each 0 cmH2O animal served as controls. Fetal lung fluid (FLF), bronchoalveolar lavage fluid (BAL), right mainstem bronchi and peripheral lung tissue were evaluated for inflammation. Results Recruitment volume increased from 0.4±0.04 mL/kg at 4 cmH2O to 2.4±0.3 mL/kg at 16 cmH2O. The lambs were surfactant deficient, and all pressures were below the opening inflection pressure on pressure-volume curve. mRNA expression of early response genes and pro-inflammatory cytokines did not increase in airway tissue or lung tissue at any pressure compared to controls. FLF and BAL also did not have increases in early response proteins. No histologic changes or Egr-1 activation was present at the pressures used. Conclusion Distending pressures as high as 16 cmH2O did not recruit lung volume at birth and did not increase markers of injury in the lung or airways in non-breathing preterm fetal sheep.
Collapse
Affiliation(s)
- Rebecca Y. Petersen
- Division of Neonatology, Cardinal Glennon Children’s Hospital, Saint Louis University, Saint Louis, MO, 63104, United States of America
| | - Emily Royse
- Division of Neonatology, Cardinal Glennon Children’s Hospital, Saint Louis University, Saint Louis, MO, 63104, United States of America
| | - Matthew W. Kemp
- School of Women and Infants’ Health, University of Western Australia, Perth, WA, 6009, Australia
| | - Yuichiro Miura
- School of Women and Infants’ Health, University of Western Australia, Perth, WA, 6009, Australia
| | - Andres Noe
- School of Women and Infants’ Health, University of Western Australia, Perth, WA, 6009, Australia
| | - Alan H. Jobe
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, United States of America
- School of Women and Infants’ Health, University of Western Australia, Perth, WA, 6009, Australia
| | - Noah H. Hillman
- Division of Neonatology, Cardinal Glennon Children’s Hospital, Saint Louis University, Saint Louis, MO, 63104, United States of America
- * E-mail:
| |
Collapse
|