51
|
Oropharyngeal Colostrum Positively Modulates the Inflammatory Response in Preterm Neonates. Nutrients 2020; 12:nu12020413. [PMID: 32033312 PMCID: PMC7071247 DOI: 10.3390/nu12020413] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/01/2020] [Accepted: 02/01/2020] [Indexed: 12/15/2022] Open
Abstract
During the first days of life, premature infants have physiological difficulties swallowing, thereby missing out on the benefits of breastfeeding. The aim of this study is to assess the effects of oropharyngeal mother’s milk administration in the inflammatory signaling of extremely premature infants. Neonates (n = 100) (<32 week’s gestation and/or <1500 g) were divided into two groups: mother’s milk group (n = 48), receiving 0.2 mL of oropharyngeal mother’s milk every 4 h for the first 15 days of life, and a control group (n = 52), not receiving oropharyngeal mother’s milk. Serum concentrations of interleukin (IL) IL-6, IL-8, IL-10, IL-1ra, tumor necrosis factor alpha (TNF-α), and interferón gamma (IFN-γ) were assessed at 1, 3, 15, and 30 days of postnatal life. Maternal and neonatal outcomes were collected. The rate of common neonatal morbidities in both groups was similar. The mother’s milk group achieved full enteral feeding earlier, and showed a decrease in Il-6 on days 15 and 30, in IL-8 on day 30, and in TNF-α and INF-γ on day 15, as well as an increase in IL-1ra on days 3 and 15 and in IL-10 on day 30. Oropharyngeal mother’s milk administration for 15 days decreases the pro-inflammatory state of preterm neonates and provides full enteral nutrition earlier, which could have a positive influence on the development of the immune system and inflammatory response, thereby positively influencing other developmental outcomes.
Collapse
|
52
|
|
53
|
Jackson CM, Mukherjee S, Wilburn AN, Cates C, Lewkowich IP, Deshmukh H, Zacharias WJ, Chougnet CA. Pulmonary Consequences of Prenatal Inflammatory Exposures: Clinical Perspective and Review of Basic Immunological Mechanisms. Front Immunol 2020; 11:1285. [PMID: 32636848 PMCID: PMC7318112 DOI: 10.3389/fimmu.2020.01285] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Chorioamnionitis, a potentially serious inflammatory complication of pregnancy, is associated with the development of an inflammatory milieu within the amniotic fluid surrounding the developing fetus. When chorioamnionitis occurs, the fetal lung finds itself in the unique position of being constantly exposed to the consequent inflammatory meditators and/or microbial products found in the amniotic fluid. This exposure results in significant changes to the fetal lung, such as increased leukocyte infiltration, altered cytokine, and surfactant production, and diminished alveolarization. These alterations can have potentially lasting impacts on lung development and function. However, studies to date have only begun to elucidate the association between such inflammatory exposures and lifelong consequences such as lung dysfunction. In this review, we discuss the pathogenesis of and fetal immune response to chorioamnionitis, detail the consequences of chorioamnionitis exposure on the developing fetal lung, highlighting the various animal models that have contributed to our current understanding and discuss the importance of fetal exposures in regard to the development of chronic respiratory disease. Finally, we focus on the clinical, basic, and therapeutic challenges in fetal inflammatory injury to the lung, and propose next steps and future directions to improve our therapeutic understanding of this important perinatal stress.
Collapse
Affiliation(s)
- Courtney M. Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, United States
| | - Adrienne N. Wilburn
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Chris Cates
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Hitesh Deshmukh
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - William J. Zacharias
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- *Correspondence: Claire A. Chougnet
| |
Collapse
|
54
|
Omer M, Melo AM, Kelly L, Mac Dermott EJ, Leahy TR, Killeen O, Saugstad OD, Savani RC, Molloy EJ. Emerging Role of the NLRP3 Inflammasome and Interleukin-1β in Neonates. Neonatology 2020; 117:545-554. [PMID: 33075792 DOI: 10.1159/000507584] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/28/2020] [Indexed: 11/19/2022]
Abstract
Infection and persistent inflammation have a prominent role in the pathogenesis of brain injury and cerebral palsy, as well as other conditions associated with prematurity such as bronchopulmonary dysplasia. The NLRP3 inflammasome-interleukin (IL)-1β pathway has been extensively studied in adults and pre-clinical models, improving our understanding of innate immunity and offering an attractive therapeutic target that is already contributing to clinical management in many auto-inflammatory disorders. IL-1 blockade has transformed the course and outcome of conditions such as chronic infantile neurological, cutaneous, articular (CINCA/NOMID) syndrome. Inflammasome activation and upregulation has recently been implicated in neonatal brain and lung inflammatory disease and may be a novel therapeutic target.
Collapse
Affiliation(s)
- Murwan Omer
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Ashanty Maggvie Melo
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland
| | - Lynne Kelly
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland
| | - Emma Jane Mac Dermott
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Timothy Ronan Leahy
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Orla Killeen
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Ola Didrik Saugstad
- Department of Pediatric Research, University of Oslo, Oslo, Norway.,Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland, .,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland, .,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland, .,Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland, .,Department of Immunology, Rheumatology, and Neonatology, CHI at Crumlin, Dublin, Ireland,
| |
Collapse
|
55
|
De Lorenzo R, Ramirez GA, Punzo D, Lorioli L, Rovelli R, Canti V, Barera G, Rovere-Querini P. Neonatal outcomes of children born to mothers on biological agents during pregnancy: State of the art and perspectives. Pharmacol Res 2019; 152:104583. [PMID: 31816434 DOI: 10.1016/j.phrs.2019.104583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/29/2019] [Accepted: 11/30/2019] [Indexed: 12/19/2022]
Abstract
Biologic disease-modifying anti-rheumatic drugs (bDMARDs) are used in pregnant patients with rheumatic diseases. Long-term follow-up data about newborns exposed to bDMARDs during pregnancy are however scarce. Here we summarize the published evidence and available recommendations for use of bDMARDs during pregnancy. We analyse clinical features at birth and at follow-up of 84 children, including: 16 consecutive children born to mothers with autoimmune diseases exposed to bDMARDs in utero; 32 children born to mothers with autoimmune diseases who did not receive bDMARDs; 36 children born to healthy mothers. In our monocentric cohort, children born to mothers with autoimmune diseases had lower gestational age at birth compared to those born to healthy mothers, independently of exposure to bDMARDs. At multivariate analysis, prematurity was an independent predictor of the need for antibiotic treatment, but not for hospitalisation or neonatal intensive care unit (ICU) stay during the neonatal period. Exposure to bDMARDs during pregnancy does not seem to interfere with post-natal development up to infancy. Prospective studies are needed in larger cohorts of pregnant patients to confirm that bDMARDs do not have a negative impact on psychomotor achievements in newborns.
Collapse
Affiliation(s)
- Rebecca De Lorenzo
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giuseppe A Ramirez
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniele Punzo
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Laura Lorioli
- Unit of Paediatrics, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Rosanna Rovelli
- Unit of Paediatrics, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valentina Canti
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Graziano Barera
- Unit of Paediatrics, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
56
|
Magalhães RC, Moreira JM, Lauar AO, da Silva AAS, Teixeira AL, E Silva ACS. Inflammatory biomarkers in children with cerebral palsy: A systematic review. RESEARCH IN DEVELOPMENTAL DISABILITIES 2019; 95:103508. [PMID: 31683246 DOI: 10.1016/j.ridd.2019.103508] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/04/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND An exacerbated systemic inflammatory response has been associated with the occurrence of central nervous system injuries that may determine, in long term, motor, sensorial and cognitive disabilities. Persistence of this exacerbated inflammatory response seems to be involved in the pathophysiology of cerebral palsy (CP). METHODS A systematic search was conducted in Bireme, Embase, PubMed and Scopus including studies that were published until August 2019. The key words used were "cerebral palsy", "brain injury", "inflammation", "oxidative stress", "cytokines", "chemokines", "neuropsychomotor development", "neurodevelopment outcomes" and "child". The quality of the eligible studies was determined according to the criteria suggested by the Newcastle-Ottawa Scale (NOS). RESULTS Fourteen eligible studies aimed to investigate the association between peripheral inflammatory molecules and neurodevelopment in infants. The studies differed regarding CP-related risk factors and its classification. Inflammatory proteins were measured in blood, plasma, serum, cerebrospinal fluid or urine. In ten studies, higher circulating levels of cytokines, including IL-1β, IL-6, TNF and CXCL8/IL-8, were associated with abnormal neurological findings. CONCLUSION The investigation of the potential association between inflammatory molecules and neurological development in children with CP requires further original studies in order to clarify the influence of prenatal and perinatal inflammation on neurological outcomes.
Collapse
Affiliation(s)
- Rafael Coelho Magalhães
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Janaina Matos Moreira
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Pediatrics, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Amanda Oliveira Lauar
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ariádna Andrade Saldanha da Silva
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, USA
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Pediatrics, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
57
|
Chin PY, Dorian C, Sharkey DJ, Hutchinson MR, Rice KC, Moldenhauer LM, Robertson SA. Toll-Like Receptor-4 Antagonist (+)-Naloxone Confers Sexually Dimorphic Protection From Inflammation-Induced Fetal Programming in Mice. Endocrinology 2019; 160:2646-2662. [PMID: 31504393 PMCID: PMC6936318 DOI: 10.1210/en.2019-00493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022]
Abstract
Inflammation elicited by infection or noninfectious insults during gestation induces proinflammatory cytokines that can shift the trajectory of development to alter offspring phenotype, promote adiposity, and increase susceptibility to metabolic disease in later life. In this study, we use mice to investigate the utility of a small molecule Toll-like receptor (TLR)4 antagonist (+)-naloxone, the nonopioid isomer of the opioid receptor antagonist (-)-naloxone, for mitigating altered fetal metabolic programming induced by a modest systemic inflammatory challenge in late gestation. In adult progeny exposed to lipopolysaccharide (LPS) challenge in utero, male but not female offspring exhibited elevated adipose tissue, reduced muscle mass, and elevated plasma leptin at 20 weeks of age. Effects were largely reversed by coadministration of (+)-naloxone following LPS. When given alone without LPS, (+)-naloxone elicited accelerated postweaning growth and elevated muscle and fat mass in adult male but not female offspring. LPS induced expression of inflammatory cytokines Il1a, Il1b, Il6, Tnf, and Il10 in fetal brain, placental, and uterine tissues, and (+)-naloxone suppressed LPS-induced cytokine expression. Fetal sex-specific regulation of cytokine expression was evident, with higher Il1a, Il1b, Il6, and Il10 induced by LPS in tissues associated with male fetuses, and greater suppression by (+)-naloxone of Il6 in females. These data demonstrate that modulating TLR4 signaling with (+)-naloxone provides protection from inflammatory diversion of fetal developmental programming in utero, associated with attenuation of gestational tissue cytokine expression in a fetal sex-specific manner. The results suggest that pharmacologic interventions targeting TLR4 warrant evaluation for attenuating developmental programming effects of fetal exposure to maternal inflammatory mediators.
Collapse
Affiliation(s)
- Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Camilla Dorian
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Mark R Hutchinson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, South Australia, Australia
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Correspondence: Sarah A. Robertson, PhD, Robinson Research Institute and the Adelaide Medical School, University of Adelaide, North Terrace, Adelaide, South Australia 5005, Australia. E-mail:
| |
Collapse
|
58
|
Lignelli E, Palumbo F, Myti D, Morty RE. Recent advances in our understanding of the mechanisms of lung alveolarization and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L832-L887. [PMID: 31596603 DOI: 10.1152/ajplung.00369.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common cause of morbidity and mortality in preterm infants. A key histopathological feature of BPD is stunted late lung development, where the process of alveolarization-the generation of alveolar gas exchange units-is impeded, through mechanisms that remain largely unclear. As such, there is interest in the clarification both of the pathomechanisms at play in affected lungs, and the mechanisms of de novo alveoli generation in healthy, developing lungs. A better understanding of normal and pathological alveolarization might reveal opportunities for improved medical management of affected infants. Furthermore, disturbances to the alveolar architecture are a key histopathological feature of several adult chronic lung diseases, including emphysema and fibrosis, and it is envisaged that knowledge about the mechanisms of alveologenesis might facilitate regeneration of healthy lung parenchyma in affected patients. To this end, recent efforts have interrogated clinical data, developed new-and refined existing-in vivo and in vitro models of BPD, have applied new microscopic and radiographic approaches, and have developed advanced cell-culture approaches, including organoid generation. Advances have also been made in the development of other methodologies, including single-cell analysis, metabolomics, lipidomics, and proteomics, as well as the generation and use of complex mouse genetics tools. The objective of this review is to present advances made in our understanding of the mechanisms of lung alveolarization and BPD over the period 1 January 2017-30 June 2019, a period that spans the 50th anniversary of the original clinical description of BPD in preterm infants.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
59
|
Liassides C, Papadopoulos A, Siristatidis C, Damoraki G, Liassidou A, Chrelias C, Kassanos D, Giamarellos-Bourboulis EJ. Single nucleotide polymorphisms of Toll-like receptor-4 and of autophagy-related gene 16 like-1 gene for predisposition of premature delivery: A prospective study. Medicine (Baltimore) 2019; 98:e17313. [PMID: 31577725 PMCID: PMC6783216 DOI: 10.1097/md.0000000000017313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To investigate the impact of carriage of single nucleotide polymorphisms (SNPs) of the Toll-like receptor-4 (TLR4) and of autophagy-related gene 16-like-1 (ATG16L1) in preterm delivery (PTD).A prospective cohort of 145 pregnant women was studied. Women were prospectively followed-up until delivery. Genotyping for rs4986790 (Asp299Gly transition) and rs4986791 (Thr399Ile transition) of TLR4 and for rs2241880 of ATG16L1 was done by PCR-restriction fragment length polymorphism. The primary study endpoint was the impact of carriage of minor alleles of TLR4 on early PTD before gestational week 32. Associations with human chorionic gonadotrophin (hCG) were also analyzed. Peripheral blood mononuclear cells were isolated from 15 healthy women and stimulated for cytokine production.No difference in clinical characteristics was observed between women delivering full term and preterm. The frequency of early PTD was 25% among women carrying minor alleles of TLR4 and 6.8% among women carrying major alleles (P: .032). Odds ratios for PTD were 3.85 among women carrying the GG genotype of rs2241880 and major alleles of TLR4 and 0.26 among carriers of GG genotype and minor alleles of TLR4 (P: .030). The co-presence of GG genotype of rs2241880 and hCG above 70 U/L was an independent variable for PTD. Stimulated production of interleukin-6 was greater among women with GG genotypes of rs2241880.Minor alleles of SNPs of TLR4 predispose to early PTD. The GG genotype of rs2241880 of ATG16L1 is associated with PTD when hCG is supra-elevated.
Collapse
Affiliation(s)
| | | | - Charalampos Siristatidis
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | | | - Charalampos Chrelias
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dimitrios Kassanos
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | |
Collapse
|
60
|
Gomez-Lopez N, Romero R, Xu Y, Miller D, Arenas-Hernandez M, Garcia-Flores V, Panaitescu B, Galaz J, Hsu CD, Para R, Berry SM. Fetal T Cell Activation in the Amniotic Cavity during Preterm Labor: A Potential Mechanism for a Subset of Idiopathic Preterm Birth. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:1793-1807. [PMID: 31492740 PMCID: PMC6799993 DOI: 10.4049/jimmunol.1900621] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/01/2019] [Indexed: 12/17/2022]
Abstract
Prematurity is the leading cause of perinatal morbidity and mortality worldwide. In most cases, preterm birth is preceded by spontaneous preterm labor, a syndrome that is associated with intra-amniotic inflammation, the most studied etiology. However, the remaining etiologies of preterm labor are poorly understood; therefore, most preterm births are categorized as idiopathic. In this study, we provide evidence showing that the fetal immune system undergoes premature activation in women with preterm labor without intra-amniotic inflammation, providing a potential new mechanism of disease for some cases of idiopathic preterm birth. First, we showed that fetal T cells are a predominant leukocyte population in amniotic fluid during preterm gestations. Interestingly, only fetal CD4+ T cells were increased in amniotic fluid of women who underwent idiopathic preterm labor and birth. This increase in fetal CD4+ T cells was accompanied by elevated amniotic fluid concentrations of T cell cytokines such as IL-2, IL-4, and IL-13, which are produced by these cells upon in vitro stimulation, but was not associated with the prototypical cytokine profile observed in women with intra-amniotic inflammation. Also, we found that cord blood T cells, mainly CD4+ T cells, obtained from women with idiopathic preterm labor and birth displayed enhanced ex vivo activation, which is similar to that observed in women with intra-amniotic inflammation. Finally, we showed that the intra-amniotic administration of activated neonatal CD4+ T cells induces preterm birth in mice. Collectively, these findings provide evidence suggesting that fetal T cell activation is implicated in the pathogenesis of idiopathic preterm labor and birth.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201;
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824
- Center for Molecular Obstetrics and Genetics, Wayne State University, Detroit, MI 48201
- Detroit Medical Center, Detroit, MI 48201
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL 33199; and
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Derek Miller
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Bogdan Panaitescu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Jose Galaz
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Robert Para
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Stanley M Berry
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, and Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| |
Collapse
|
61
|
Verstraeten BSE, McCreary JK, Falkenberg EA, Fang X, Weyers S, Metz GAS, Olson DM. Multiple prenatal stresses increase sexual dimorphism in adult offspring behavior. Psychoneuroendocrinology 2019; 107:251-260. [PMID: 31174163 DOI: 10.1016/j.psyneuen.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Maternal gestational stress and immune activation have independently been associated with affective and neurodevelopmental disorders across the lifespan. We investigated whether rats exposed to prenatal maternal stressors (PNMS) consisting of psychological stress, interleukin (IL)-1β or both (two-hit stress) during critical developmental windows displayed a behavioral phenotype representative of these conditions. METHODS Long-Evans dams were exposed to psychological stressors consisting of restraint stress and forced swimming from gestational day (GD)12 to 18 or to no stress (controls). From GD17 until day of delivery, these same animals were injected with saline or IL-1β as a second hit and immune stressor (5 μg/day, intraperitoneally). The behavior of F1 offspring adults was tested on the open field test, elevated plus maze and affective exploration task on postnatal days (P)90, 100 and 110 respectively. RESULTS The effects of PNMS differed depending on the specific testing environment and potentially the age at assessment, especially in female offspring. Both locomotion and anxiety-like behavioral measures were susceptible to PNMS effects. In females, psychological stress increased anxiety-like behavior, whereas IL-1β had an opposite effect, inducing exploration and risk-taking behavior on the open field test and the elevated plus maze. When present, interactions between both stressors limited the anxiogenic effect of psychological stress on its own. In contrast, prenatal psychological stress increased anxiety-like behavior in adult males overall. A similar anxiogenic effect of IL-1β was only found on the open field test while the Stress*IL-1β interaction appeared to limit the effect of either alone. Contrarily, the PNMS effects on anxiety-like behavior on the affective exploration task were highly similar between both sexes. Analysis of males and females together revealed an additive effect of Stress and IL-1β on the number of exits from the refuge, a measure of risk assessment and thus correlated with anxiety. CONCLUSION PNMS affected offspring adult behavior in a sex-dependent manner. Effects on females were more variable, whereas psychological stress mostly induced anxiety-like behavior in males. These data highlight the sexual dimorphism in vulnerability to prenatal stressors. Maternal or stress-induced programming of the stress response and neuroinflammation may play an important role in mediating stress effects on offspring adult behavior.
Collapse
Affiliation(s)
- Barbara S E Verstraeten
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada; Department of Human Structure and Repair, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| | - J Keiko McCreary
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Erin A Falkenberg
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Xin Fang
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada
| | - Steven Weyers
- Department of Human Structure and Repair, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada.
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada.
| |
Collapse
|
62
|
Fedorka CE, Ball BA, Scoggin KE, Loux SC, Troedsson MHT, Adams AA. The feto-maternal immune response to equine placentitis. Am J Reprod Immunol 2019; 82:e13179. [PMID: 31373743 DOI: 10.1111/aji.13179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
PROBLEM Ascending placentitis is one of the leading causes of abortion in the horse. Minimal work has focused on its effect on fetal fluids or the antenatal immune response of the fetus. METHODOLOGY Placentitis was induced via transcervical inoculation of Streptococcus equi ssp Zooepidemicus, and fluids/serum/tissues were collected 4-6 days later following euthanasia. Cytokine concentrations were detected using a multiplex immunoassay within fetal fluids (amniotic and allantoic) and serum (maternal and fetal) in inoculated and control mares. In addition, tissues from fetal (spleen, liver, lung, umbilicus, amnioallantois) and maternal (spleen, liver, lung, chorioallantois, endometrium) origin were analyzed in inoculated and control mares utilizing qPCR for expression of cytokines. RESULTS No difference in cytokine concentrations in maternal or fetal serum was noted between inoculated and control mares. Concentrations of IL-1β, IL-6, IL-10, and GRO were upregulated in the amniotic fluid following inoculation, with a trend toward higher IL-6 concentration in allantoic fluid. The amnioallantoic tissue separating the two fluids had higher expression of IL-1β and IL-6 following inoculation, while chorioallantois and endometrium upregulated IL-1β and IL-8 expression. IL-1β was upregulated in the maternal spleen following inoculation. Fetal spleens were upregulated in expression of IL-1β, GRO, and IL-6, while IL-6 was higher in fetal liver after inoculation than in controls. CONCLUSION The maternal response to placentitis is primarily pro-inflammatory while the fetus appears to play a regulatory role in this inflammation. Additionally, amniotic fluid sampling may be more diagnostic of ascending placentitis than circulating cytokines.
Collapse
Affiliation(s)
- Carleigh E Fedorka
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Barry A Ball
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Kirsten E Scoggin
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Shavahn C Loux
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Mats H T Troedsson
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Amanda A Adams
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
63
|
Exposure to systemic and intrauterine inflammation leads to decreased pup survival via different placental mechanisms. J Reprod Immunol 2019; 133:52-62. [PMID: 31280130 DOI: 10.1016/j.jri.2019.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/21/2019] [Accepted: 06/29/2019] [Indexed: 12/15/2022]
Abstract
PROBLEM Exposure to systemic maternal inflammation (i.e., maternal sepsis, influenza, human immunodeficiency virus, or pyelonephritis) and intrauterine (IU) inflammation (i.e., chorioamnionitis or preterm labor) have been associated with adverse perinatal sequelae. Whether systemic and localized inflammation leading to adverse outcomes have similar placental mechanisms remain unclear. METHOD OF STUDY We conducted a study by murine modeling systemic and localized IU inflammation with injections of either intraperitoneal (IP) or IU interleukin-1β (IL-1β) and compared fetoplacental hemodynamic changes, cytokine/chemokine expression, and fetal loss. RESULTS IU IL-1β exposure reduced offspring survival by 31.1% and IP IL-1β exposure by 34.5% when compared with control pups. Despite this similar outcome in offspring survival, Doppler analysis revealed a stark difference: IU group displayed worsened fetoplacental hemodynamic changes while no differences were found between IP and control groups. While both IU and IP groups had increases in pro-inflammatory cytokines and chemokines, specific gene expression trends differed between the two groups, once again highlighting their mechanistic differences. CONCLUSION While both IP and IU IL-1β exposure similarly affected pup survival, only IU inflammation resulted in fetoplacental hemodynamic changes. We speculate that exposure to maternal systemic and IU inflammation plays a key role in fetal injury by utilizing different placental inflammatory pathways and mechanisms.
Collapse
|
64
|
Chen HJ, Gur TL. Intrauterine Microbiota: Missing, or the Missing Link? Trends Neurosci 2019; 42:402-413. [PMID: 31053242 PMCID: PMC6604064 DOI: 10.1016/j.tins.2019.03.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/21/2019] [Accepted: 03/19/2019] [Indexed: 12/29/2022]
Abstract
The intrauterine environment provides a key interface between the mother and the developing fetus during pregnancy, and is a target for investigating mechanisms of fetal programming. Studies have demonstrated an association between prenatal stress and neurodevelopmental disorders. The role of the intrauterine environment in mediating this effect is still being elucidated. In this review, we discuss emerging preclinical and clinical evidence suggesting the existence of microbial communities in utero. We also outline possible mechanisms of bacterial translocation to the intrauterine environment and immune responses to the presence of microbes or microbial components. Lastly, we overview the effects of intrauterine inflammation on neurodevelopment. We hypothesize that maternal gestational stress leads to disruptions in the maternal oral, gut, and vaginal microbiome that may lead to the translocation of bacteria to the intrauterine environment, eliciting an inflammatory response and resulting in deficits in neurodevelopment.
Collapse
Affiliation(s)
- Helen J Chen
- Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - Tamar L Gur
- Department of Psychiatry and Behavioral Health, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Department of Obstetrics and Gynecology, Wexner Medical Center at The Ohio State University, Columbus, OH, USA; Institute of Behavioral Medicine Research, Wexner Medical Center at The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
65
|
Lim R, Lappas M. Expression and function of macrophage-inducible C-type lectin (Mincle) in inflammation driven parturition in fetal membranes and myometrium. Clin Exp Immunol 2019; 197:95-110. [PMID: 30793298 DOI: 10.1111/cei.13281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2019] [Indexed: 12/31/2022] Open
Abstract
The pivotal role of inflammatory processes in human parturition is well known, but not completely understood. We have performed a study to examine the role of macrophage-inducible C-type lectin (Mincle) in inflammation-associated parturition. Using human samples, we show that spontaneous labour is associated with up-regulated Mincle expression in the myometrium and fetal membranes. Mincle expression was also increased in fetal membranes and myometrium in the presence of pro-labour mediators, the proinflammatory cytokines interleukin (IL)-1B and tumour necrosis factor (TNF), and Toll-like receptor (TLR) ligands fsl-1, poly(I:C), lipopolysaccharide (LPS) and flagellin. These clinical studies are supported by mouse studies, where an inflammatory challenge in a mouse model of preterm birth increased Mincle expression in the uterus. Importantly, elimination of Mincle decreased the effectiveness of proinflammatory cytokines and TLR ligands to induce the expression of pro-labour mediators; namely, proinflammatory cytokines and chemokines, contraction-associated proteins and prostaglandins, and extracellular matrix remodelling enzymes, matrix metalloproteinases. The data presented in this study suggest that Mincle is required when inflammatory activation precipitates parturition.
Collapse
Affiliation(s)
- R Lim
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - M Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| |
Collapse
|
66
|
Lei J, Vermillion MS, Jia B, Xie H, Xie L, McLane MW, Sheffield JS, Pekosz A, Brown A, Klein SL, Burd I. IL-1 receptor antagonist therapy mitigates placental dysfunction and perinatal injury following Zika virus infection. JCI Insight 2019; 4:122678. [PMID: 30944243 DOI: 10.1172/jci.insight.122678] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 02/14/2019] [Indexed: 12/25/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy causes significant adverse sequelae in the developing fetus, and results in long-term structural and neurologic defects. Most preventive and therapeutic efforts have focused on the development of vaccines, antivirals, and antibodies. The placental immunologic response to ZIKV, however, has been largely overlooked as a target for therapeutic intervention. The placental inflammatory response, specifically IL-1β secretion and signaling, is induced by ZIKV infection and represents an environmental factor that is known to increase the risk of perinatal developmental abnormalities. We show in a mouse model that maternally administrated IL-1 receptor antagonist (IRA; Kineret, or anakinra), following ZIKV exposure, can preserve placental function (by improving trophoblast invasion and placental vasculature), increase fetal viability, and reduce neurobehavioral deficits in the offspring. We further demonstrate that while ZIKV RNA is highly detectable in placentas, it is not correlated with fetal viability. Beyond its effects in the placenta, we show that IL-1 blockade may also directly decrease fetal neuroinflammation by mitigating fetal microglial activation in a dose-dependent manner. Our studies distinguish the role of placental inflammation during ZIKV-infected pregnancies, and demonstrate that maternal IRA may attenuate fetal neuroinflammation and improve perinatal outcomes.
Collapse
Affiliation(s)
- Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Meghan S Vermillion
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Molecular and Comparative Pathobiology
| | - Bei Jia
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Han Xie
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Li Xie
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael W McLane
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeanne S Sheffield
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amanda Brown
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
67
|
Geranurimi A, Cheng CWH, Quiniou C, Zhu T, Hou X, Rivera JC, St-Cyr DJ, Beauregard K, Bernard-Gauthier V, Chemtob S, Lubell WD. Probing Anti-inflammatory Properties Independent of NF-κB Through Conformational Constraint of Peptide-Based Interleukin-1 Receptor Biased Ligands. Front Chem 2019; 7:23. [PMID: 30815434 PMCID: PMC6381024 DOI: 10.3389/fchem.2019.00023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
Interleukin-1β (IL-1β) binds to the IL-1 receptor (IL-1R) and is a key cytokine mediator of inflammasome activation. IL-1β signaling leads to parturition in preterm birth (PTB) and contributes to the retinal vaso-obliteration characteristic of oxygen-induced retinopathy (OIR) of premature infants. Therapeutics targeting IL-1β and IL-1R are approved to treat rheumatoid arthritis; however, all are large proteins with clinical limitations including immunosuppression, due in part to inhibition of NF-κB signaling, which is required for immuno-vigilance and cytoprotection. The all-D-amino acid peptide 1 (101.10, H-d-Arg-d-Tyr-d-Thr-d-Val-d-Glu-d-Leu-d-Ala-NH2) is an allosteric IL-1R modulator, which exhibits functional selectivity and conserves NF-κB signaling while inhibiting other IL-1-activated pathways. Peptide 1 has proven effective in experimental models of PTB and OIR. Seeking understanding of the structural requirements for the activity and biased signaling of 1, a panel of twelve derivatives was synthesized employing the various stereochemical isomers of α-amino-γ-lactam (Agl) and α-amino-β-hydroxy-γ-lactam (Hgl) residues to constrain the D-Thr-D-Val dipeptide residue. Using circular dichroism spectroscopy, the peptide conformation in solution was observed to be contingent on Agl, Hgl, and Val stereochemistry. Moreover, the lactam mimic structure and configuration influenced biased IL-1 signaling in an in vitro panel of cellular assays as well as in vivo activity in murine models of PTB and OIR. Remarkably, all Agl and Hgl analogs of peptide 1 did not inhibit NF-κB signaling but blocked other pathways, such as JNK and ROCK2 phosphorylation contingent on structure and configuration. Efficacy in preventing preterm labor correlated with a capacity to block IL-1β-induced IL-1β synthesis. Furthermore, the importance of inhibition of JNK and ROCK2 phosphorylation for enhanced activity was highlighted for prevention of vaso-obliteration in the OIR model. Taken together, lactam mimic structure and stereochemistry strongly influenced conformation and biased signaling. Selective modulation of IL-1 signaling was proven to be particularly beneficial for curbing inflammation in models of preterm labor and retinopathy of prematurity (ROP). A class of biased ligands has been created with potential to serve as selective probes for studying IL-1 signaling in disease. Moreover, the small peptide mimic prototypes are promising leads for developing immunomodulatory therapies with easier administration and maintenance of beneficial effects of NF-κB signaling.
Collapse
Affiliation(s)
- Azade Geranurimi
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
| | - Colin W H Cheng
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.,CHU Sainte-Justine Research Centre, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont Research Centre, Montréal, QC, Canada
| | | | - Tang Zhu
- CHU Sainte-Justine Research Centre, Montréal, QC, Canada
| | - Xin Hou
- CHU Sainte-Justine Research Centre, Montréal, QC, Canada
| | - José Carlos Rivera
- CHU Sainte-Justine Research Centre, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont Research Centre, Montréal, QC, Canada
| | - Daniel J St-Cyr
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
| | - Kim Beauregard
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
| | | | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.,CHU Sainte-Justine Research Centre, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont Research Centre, Montréal, QC, Canada.,Departments of Pediatrics, Pharmacology and Physiology, and Ophthalmology, Université de Montréal, Montréal, QC, Canada
| | - William D Lubell
- Département de Chimie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
68
|
Loering S, Cameron GJM, Starkey MR, Hansbro PM. Lung development and emerging roles for type 2 immunity. J Pathol 2019; 247:686-696. [PMID: 30506724 DOI: 10.1002/path.5211] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Center for Inflammation, Centenary Institute and The School of Life Sciences, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
69
|
Abi Nahed R, Reynaud D, Borg AJ, Traboulsi W, Wetzel A, Sapin V, Brouillet S, Dieudonné MN, Dakouane-Giudicelli M, Benharouga M, Murthi P, Alfaidy N. NLRP7 is increased in human idiopathic fetal growth restriction and plays a critical role in trophoblast differentiation. J Mol Med (Berl) 2019; 97:355-367. [PMID: 30617930 DOI: 10.1007/s00109-018-01737-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 12/13/2022]
Abstract
Fetal growth restriction (FGR) the leading cause of perinatal mortality and morbidity is highly related to abnormal placental development, and placentas from FGR pregnancies are often characterized by increased inflammation. However, the mechanisms of FGR-associated inflammation are far from being understood. NLRP7, a member of a family of receptors involved in the innate immune responses, has been shown to be associated with gestational trophoblastic diseases. Here, we characterized the expression and the functional role of NLRP7 in the placenta and investigated its involvement in the pathogenesis of FGR. We used primary trophoblasts and placental explants that were collected during early pregnancy, and established trophoblast-derived cell lines, human placental villi, and serum samples from early pregnancy (n = 38) and from FGR (n = 40) and age-matched controls (n = 32). Our results show that NLRP7 (i) is predominantly expressed in the trophoblasts during the hypoxic period of placental development and its expression is upregulated by hypoxia and (ii) increases trophoblast proliferation ([3H]-thymidine) and controls the precocious differentiation of trophoblasts towards syncytium (syncytin 1 and 2 and β-hCG production and xCELLigence analysis) and towards invasive extravillous trophoblast (2D and 3D cultures). We have also demonstrated that NLRP7 inflammasome activation in trophoblast cells increases IL-1β, but not IL-18 secretion. In relation to the FGR, we demonstrated that major components of NLRP7 inflammasome machinery are increased and that IL-1β but not IL-18 circulating levels are increased in FGR. Altogether, our results identified NLRP7 as a critical placental factor and provided evidence for its deregulation in FGR. NLRP7 inflammasome is abundantly expressed by trophoblast cells. It is regulated by a key parameter of placental development, hypoxia. It controls trophoblast proliferation, migration, and invasion and exhibits anti-apoptotic role. NLRP7 machinery is deregulated in FGR pregnancies. KEY MESSAGES: NLRP7 inflammasome is abundantly expressed by trophoblast cells. It is regulated by a key parameter of placental development, hypoxia. It controls trophoblast proliferation, migration, and invasion and exhibits anti-apoptotic role. NLRP7 machinery is deregulated in FGR pregnancies.
Collapse
Affiliation(s)
- R Abi Nahed
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - D Reynaud
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - A J Borg
- Department of Medicine, School of Clinical Sciences, Monash University and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - W Traboulsi
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - A Wetzel
- Université Grenoble-Alpes, 38000, Grenoble, France.,Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation-CECOS, Centre Hospitalier Universitaire de Grenoble, 38700, La Tronche, France
| | - V Sapin
- GReD, UMR CNRS 6293 INSERM 1103 Université Clermont Auvergne, CRBC, UFR de Médecine et des Professions Paramédicales, 63000, Clermont-Ferrand, France
| | - S Brouillet
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,Université Grenoble-Alpes, 38000, Grenoble, France.,Hôpital Couple-Enfant, Centre Clinique et Biologique d'Assistance Médicale à la Procréation-CECOS, Centre Hospitalier Universitaire de Grenoble, 38700, La Tronche, France
| | - M N Dieudonné
- GIG - EA 7404 Université de Versailles-Saint-Quentin-en-Yvelines, Université Paris-Saclay, Unité de Formation et de Recherche des Sciences de la Santé Simone Veil, Montigny-le-Bretonneux, France
| | - M Dakouane-Giudicelli
- Institut National de la Santé et de la Recherche Médicale, Unité 1179, Montigny-Le-Bretonneux, France
| | - M Benharouga
- Université Grenoble-Alpes, 38000, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France.,Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Centre National de la Recherche Scientifique, Grenoble, France
| | - P Murthi
- Department of Medicine, School of Clinical Sciences, Monash University and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France. .,Université Grenoble-Alpes, 38000, Grenoble, France. .,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France. .,Unité INSERM U1036, Laboratoire BCI -BIG, CEA Grenoble 17, rue des Martyrs, 38054, Grenoble cedex 9, France.
| |
Collapse
|
70
|
Sow environment during gestation: part I. Influence on maternal physiology and lacteal secretions in relation with neonatal survival. Animal 2019; 13:1432-1439. [DOI: 10.1017/s1751731118002987] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
71
|
Jia X, Cao B, An Y, Zhang X, Wang C. Rapamycin ameliorates lipopolysaccharide-induced acute lung injury by inhibiting IL-1β and IL-18 production. Int Immunopharmacol 2018; 67:211-219. [PMID: 30557824 DOI: 10.1016/j.intimp.2018.12.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 01/06/2023]
Abstract
Interleukin (IL)-1β and IL-18 play central and detrimental roles in the development of acute lung injury (ALI), and mammalian target of rapamycin (mTOR) is involved in regulating IL-1β and IL-18 production. However, it is not clear whether the mTOR specific inhibitor rapamycin can attenuate lipopolysaccharide (LPS)-induced ALI by modulating IL-1β and IL-18 production. In this study, we found that rapamycin ameliorated LPS-induced ALI by inhibiting NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated IL-1β and IL-18 secretion. Mechanistically, elevated autophagy and decreased nuclear factor (NF)-κB activation were associated with downregulated IL-1β and IL-18. Moreover, rapamycin reduced leukocyte infiltration in the lung tissue and bronchoalveolar lavage fluid (BALF), and contributed to the alleviation of LPS-induced ALI. Consistently, rapamycin also significantly inhibited IL-1β and IL-18 production by RAW264.7 cells via increased autophagy and decreased NF-κB signaling in vitro. Our results demonstrated that rapamycin protects mice against LPS-induced ALI partly by inhibiting the production and secretion of IL-1β and IL-18. mTOR and rapamycin might represent an appropriate therapeutic target and strategy for preventing ALI induced by LPS.
Collapse
Affiliation(s)
- Xuehong Jia
- Department of Respiratory Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing 100029, China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, China; Department of Respiratory Medicine, Capital Medical University, Beijing 100069, China
| | - Yunqing An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xulong Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| | - Chen Wang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing 100029, China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, China; Department of Respiratory Medicine, Capital Medical University, Beijing 100069, China; Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
72
|
Brien ME, Baker B, Duval C, Gaudreault V, Jones RL, Girard S. Alarmins at the maternal-fetal interface: involvement of inflammation in placental dysfunction and pregnancy complications 1. Can J Physiol Pharmacol 2018; 97:206-212. [PMID: 30485131 DOI: 10.1139/cjpp-2018-0363] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation is known to be associated with placental dysfunction and pregnancy complications. Infections are well known to be a cause of inflammation but they are frequently undetectable in pregnancy complications. More recently, the focus has been extended to inflammation of noninfectious origin, namely caused by endogenous mediators known as "damage-associated molecular patterns (DAMPs)" or alarmins. In this manuscript, we review the mechanism by which inflammation, sterile or infectious, can alter the placenta and its function. We discuss some classical DAMPs, such as uric acid, high mobility group box 1 (HMGB1), cell-free fetal deoxyribonucleic acid (DNA) (cffDNA), S100 proteins, heat shock protein 70 (HSP70), and adenosine triphosphate (ATP) and their impact on the placenta. We focus on the main placental cells (i.e., trophoblast and Hofbauer cells) and describe the placental response to, and release of, DAMPs. We also covered the current state of knowledge about the role of DAMPs in pregnancy complications including preeclampsia, fetal growth restriction, preterm birth, and stillbirth and possible therapeutic strategies to preserve placental function.
Collapse
Affiliation(s)
- Marie-Eve Brien
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,b Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Bernadette Baker
- c Maternal and Fetal Health Research Centre, University of Manchester, Manchester, M13 9WL, United Kingdom.,d St. Mary's Hospital, Central Manchester University Hospital National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Cyntia Duval
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,e Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Virginie Gaudreault
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,e Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Rebecca L Jones
- c Maternal and Fetal Health Research Centre, University of Manchester, Manchester, M13 9WL, United Kingdom.,d St. Mary's Hospital, Central Manchester University Hospital National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Sylvie Girard
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,b Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.,e Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
73
|
Suff N, Karda R, Diaz JA, Ng J, Baruteau J, Perocheau D, Tangney M, Taylor PW, Peebles D, Buckley SMK, Waddington SN. Ascending Vaginal Infection Using Bioluminescent Bacteria Evokes Intrauterine Inflammation, Preterm Birth, and Neonatal Brain Injury in Pregnant Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2164-2176. [PMID: 30036519 PMCID: PMC6168615 DOI: 10.1016/j.ajpath.2018.06.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
Preterm birth is a serious global health problem and the leading cause of infant death before 5 years of age. At least 40% of cases are associated with infection. The most common way for pathogens to access the uterine cavity is by ascending from the vagina. Bioluminescent pathogens have revolutionized the understanding of infectious diseases. We hypothesized that bioluminescent Escherichia coli can be used to track and monitor ascending vaginal infections. Two bioluminescent strains were studied: E. coli K12 MG1655-lux, a nonpathogenic laboratory strain, and E. coli K1 A192PP-lux2, a pathogenic strain capable of causing neonatal meningitis and sepsis in neonatal rats. On embryonic day 16, mice received intravaginal E. coli K12, E. coli K1, or phosphate-buffered saline followed by whole-body bioluminescent imaging. In both cases, intravaginal delivery of E. coli K12 or E. coli K1 led to bacterial ascension into the uterine cavity, but only E. coli K1 induced preterm parturition. Intravaginal administration of E. coli K1 significantly reduced the proportion of pups born alive compared with E. coli K12 and phosphate-buffered saline controls. However, in both groups of viable pups born after bacterial inoculation, there was evidence of comparable brain inflammation by postnatal day 6. This study ascribes specific mechanisms by which exposure to intrauterine bacteria leads to premature delivery and neurologic inflammation in neonates.
Collapse
Affiliation(s)
- Natalie Suff
- Gene Transfer Technology Group, University College London, London, United Kingdom; Preterm Birth Group, Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London, London, United Kingdom
| | - Rajvinder Karda
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Juan A Diaz
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Joanne Ng
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Julien Baruteau
- Gene Transfer Technology Group, University College London, London, United Kingdom; Department of Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Dany Perocheau
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Mark Tangney
- SynBio Centre, University College Cork, Cork, Ireland
| | - Peter W Taylor
- School of Pharmacy, University College London, London, United Kingdom
| | - Donald Peebles
- Preterm Birth Group, Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London, London, United Kingdom
| | - Suzanne M K Buckley
- Gene Transfer Technology Group, University College London, London, United Kingdom.
| | - Simon N Waddington
- Gene Transfer Technology Group, University College London, London, United Kingdom; MRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
74
|
Beaudry-Richard A, Nadeau-Vallée M, Prairie É, Maurice N, Heckel É, Nezhady M, Pundir S, Madaan A, Boudreault A, Hou X, Quiniou C, Sierra EM, Beaulac A, Lodygensky G, Robertson SA, Keelan J, Adams Waldorf KM, Olson DM, Rivera JC, Lubell WD, Joyal JS, Bouchard JF, Chemtob S. Antenatal IL-1-dependent inflammation persists postnatally and causes retinal and sub-retinal vasculopathy in progeny. Sci Rep 2018; 8:11875. [PMID: 30089839 PMCID: PMC6082873 DOI: 10.1038/s41598-018-30087-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
Antenatal inflammation as seen with chorioamnionitis is harmful to foetal/neonatal organ development including to eyes. Although the major pro-inflammatory cytokine IL-1β participates in retinopathy induced by hyperoxia (a predisposing factor to retinopathy of prematurity), the specific role of antenatal IL-1β associated with preterm birth (PTB) in retinal vasculopathy (independent of hyperoxia) is unknown. Using a murine model of PTB induced with IL-1β injection in utero, we studied consequent retinal and choroidal vascular development; in this process we evaluated the efficacy of IL-1R antagonists. Eyes of foetuses exposed only to IL-1β displayed high levels of pro-inflammatory genes, and a persistent postnatal infiltration of inflammatory cells. This prolonged inflammatory response was associated with: (1) a marked delay in retinal vessel growth; (2) long-lasting thinning of the choroid; and (3) long-term morphological and functional alterations of the retina. Antenatal administration of IL-1R antagonists - 101.10 (a modulator of IL-1R) more so than Kineret (competitive IL-1R antagonist) - prevented all deleterious effects of inflammation. This study unveils a key role for IL-1β, a major mediator of chorioamnionitis, in causing sustained ocular inflammation and perinatal vascular eye injury, and highlights the efficacy of antenatal 101.10 to suppress deleterious inflammation.
Collapse
Affiliation(s)
- Alexandra Beaudry-Richard
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Mathieu Nadeau-Vallée
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada.,Department of Pharmacology, Université de Montréal, Montréal, Canada
| | - Élizabeth Prairie
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Noémie Maurice
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Émilie Heckel
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Mohammad Nezhady
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Sheetal Pundir
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Ankush Madaan
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Amarilys Boudreault
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Xin Hou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Estefania Marin Sierra
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Alexandre Beaulac
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Gregory Lodygensky
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Sarah A Robertson
- Department of Obstetrics and Gynaecology, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Jeffrey Keelan
- Div Obstetrics & Gynaecology, University of Western Australia King Edward Memorial Hospital, Perth, Australia
| | | | - David M Olson
- Departments of Obstetrics and Gynaecology, Pediatrics and Physiology, University of Alberta, Edmonton, AB, Canada
| | - Jose-Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Canada
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada.,Department of Pharmacology, Université de Montréal, Montréal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | | | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada. .,Department of Pharmacology, Université de Montréal, Montréal, Canada. .,Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada.
| |
Collapse
|
75
|
Sittipo P, Lobionda S, Choi K, Sari IN, Kwon HY, Lee YK. Toll-Like Receptor 2-Mediated Suppression of Colorectal Cancer Pathogenesis by Polysaccharide A From Bacteroides fragilis. Front Microbiol 2018; 9:1588. [PMID: 30065713 PMCID: PMC6056687 DOI: 10.3389/fmicb.2018.01588] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/26/2018] [Indexed: 12/24/2022] Open
Abstract
The beneficial role of gut microbiota in intestinal diseases has been highlighted recently. Bacteroides fragilis found in the human gastrointestinal tract is a well-studied example of a beneficial bacterium that protects against intestinal inflammation. Polysaccharide A (PSA) from B. fragilis induces the production of interleukin (IL)-10 from immune cells via Toll-like receptor 2 (TLR2) signaling in animal colitis models. The direct effect of PSA on human colorectal cancer (CRC) cells has not been studied. Here, we report the effect of PSA from B. fragilis on CRC pathogenesis in SW620 and HT29 CRC cells and the molecular signaling underlying these effects. We demonstrated that PSA induced the production of the pro-inflammatory cytokine, IL-8, but not IL-10, in CRC cells. PSA inhibited CRC cell proliferation by controlling the cell cycle and impaired CRC cell migration and invasion by suppressing epithelial mesenchymal transition. Moreover, as in the case of other animal intestinal diseases, the protective role of PSA against CRC pathogenesis was also mediated by TLR2. Our results reveal that PSA from B. fragilis plays a protective role against CRC via TLR2 signaling.
Collapse
Affiliation(s)
| | | | | | | | - Hyog Young Kwon
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, South Korea
| | - Yun Kyung Lee
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, South Korea
| |
Collapse
|
76
|
Vora B, Wang A, Kosti I, Huang H, Paranjpe I, Woodruff TJ, MacKenzie T, Sirota M. Meta-Analysis of Maternal and Fetal Transcriptomic Data Elucidates the Role of Adaptive and Innate Immunity in Preterm Birth. Front Immunol 2018; 9:993. [PMID: 29867970 PMCID: PMC5954243 DOI: 10.3389/fimmu.2018.00993] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/20/2018] [Indexed: 12/27/2022] Open
Abstract
Preterm birth (PTB) is the leading cause of newborn deaths around the world. Spontaneous preterm birth (sPTB) accounts for two-thirds of all PTBs; however, there remains an unmet need of detecting and preventing sPTB. Although the dysregulation of the immune system has been implicated in various studies, small sizes and irreproducibility of results have limited identification of its role. Here, we present a cross-study meta-analysis to evaluate genome-wide differential gene expression signals in sPTB. A comprehensive search of the NIH genomic database for studies related to sPTB with maternal whole blood samples resulted in data from three separate studies consisting of 339 samples. After aggregating and normalizing these transcriptomic datasets and performing a meta-analysis, we identified 210 genes that were differentially expressed in sPTB relative to term birth. These genes were enriched in immune-related pathways, showing upregulation of innate immunity and downregulation of adaptive immunity in women who delivered preterm. An additional analysis found several of these differentially expressed at mid-gestation, suggesting their potential to be clinically relevant biomarkers. Furthermore, a complementary analysis identified 473 genes differentially expressed in preterm cord blood samples. However, these genes demonstrated downregulation of the innate immune system, a stark contrast to findings using maternal blood samples. These immune-related findings were further confirmed by cell deconvolution as well as upstream transcription and cytokine regulation analyses. Overall, this study identified a strong immune signature related to sPTB as well as several potential biomarkers that could be translated to clinical use.
Collapse
Affiliation(s)
- Bianca Vora
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Aolin Wang
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Idit Kosti
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
| | - Hongtai Huang
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Ishan Paranjpe
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Tracey J Woodruff
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Tippi MacKenzie
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States.,Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, CA, United States.,Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
77
|
Soucy-Giguère L, Gasse C, Giguère Y, Demers S, Bujold E, Boutin A. Intra-amniotic inflammation and child neurodevelopment: a systematic review protocol. Syst Rev 2018; 7:12. [PMID: 29357925 PMCID: PMC5778727 DOI: 10.1186/s13643-018-0683-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/12/2018] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Intra-amniotic inflammation is associated with adverse pregnancy and neonatal outcomes. However, the impact on child neurodevelopment remains unclear. We aim to assess the effect of intra-amniotic inflammation on neurodevelopmental outcomes in children. METHODS The databases MEDLINE, Embase, CINAHL, and Cochrane will be searched from their inception until November 2017. Randomized trials and cohort studies in which inflammatory markers were measured in amniotic fluid collected by amniocentesis and in which infant's neurodevelopment was assessed will be eligible. Two reviewers will independently select eligible studies, assess their risk of bias, and extract data. Results will be compared and a third party will be consulted in case of disagreement. Our primary outcome of interest is child neurodevelopment, assessed with either a validated tool or by revision of medical records for specific diagnosis. Secondary outcomes will include abnormal brain imaging. Relative risks will be pooled and sensitivity analyses will be performed for the indication of amniocentesis, gestational age at amniocentesis, gestational age at delivery, and fetal sex. Risk of bias will be assessed using the Cochrane Collaboration's tool for assessing the risk of bias in randomized trials or an adapted version of the ROBINS-1 for the risk of bias in non-randomized studies. DISCUSSION This systematic review will report the current evidence regarding the association between amniotic inflammation and child neurodevelopment, and the modifiers of this association. The review will generate new hypotheses on pathological pathways and will guide future research. SYSTEMATIC REVIEW REGISTRATION PROSPERO 2017 65065.
Collapse
Affiliation(s)
- Laurence Soucy-Giguère
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, 2705, Boul. Laurier, TR-66, Québec, QC G1V 4G2 Canada
| | - Cédric Gasse
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, 2705, Boul. Laurier, TR-66, Québec, QC G1V 4G2 Canada
- Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6 Canada
| | - Yves Giguère
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, 2705, Boul. Laurier, TR-66, Québec, QC G1V 4G2 Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6 Canada
| | - Suzanne Demers
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, 2705, Boul. Laurier, TR-66, Québec, QC G1V 4G2 Canada
- Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6 Canada
| | - Emmanuel Bujold
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, 2705, Boul. Laurier, TR-66, Québec, QC G1V 4G2 Canada
- Department of Gynecology, Obstetrics and Reproduction, Faculty of Medicine, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6 Canada
| | - Amélie Boutin
- Reproduction, Mother and Child Health Unit, CHU de Québec-Université Laval Research Center, Université Laval, 2705, Boul. Laurier, TR-66, Québec, QC G1V 4G2 Canada
| |
Collapse
|
78
|
Glaser K, Silwedel C, Fehrholz M, Waaga-Gasser AM, Henrich B, Claus H, Speer CP. Ureaplasma Species Differentially Modulate Pro- and Anti-Inflammatory Cytokine Responses in Newborn and Adult Human Monocytes Pushing the State Toward Pro-Inflammation. Front Cell Infect Microbiol 2017; 7:484. [PMID: 29234642 PMCID: PMC5712342 DOI: 10.3389/fcimb.2017.00484] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 11/13/2017] [Indexed: 12/22/2022] Open
Abstract
Background:Ureaplasma species have been associated with chorioamnionitis and preterm birth and have been implicated in the pathogenesis of neonatal short and long-term morbidity. However, being mostly commensal bacteria, controversy remains on the pro-inflammatory capacity of Ureaplasma. Discussions are ongoing on the incidence and impact of prenatal, perinatal, and postnatal infection. The present study addressed the impact of Ureaplasma isolates on monocyte-driven inflammation. Methods: Cord blood monocytes of term neonates and adult monocytes, either native or LPS-primed, were cultured with Ureaplasma urealyticum (U. urealyticum) serovar 8 (Uu8) and Ureaplasma parvum serovar 3 (Up3). Using qRT-PCR, cytokine flow cytometry, and multi-analyte immunoassay, we assessed mRNA and protein expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-8, IL-12p40, IL-10, and IL-1 receptor antagonist (IL-1ra) as well as Toll-like receptor (TLR) 2 and TLR4. Results: Uu8 and Up3 induced mRNA expression and protein release of TNF-α, IL-1β and IL-8 in term neonatal and adult monocytes (p < 0.01 and p < 0.05). Intracellular protein expression of TNF-α, IL-1β and IL-8 in Ureaplasma-stimulated cells paralleled those results. Ureaplasma-induced cytokine levels did not significantly differ from LPS-mediated levels except for lower intracellular IL-1β in adult monocytes (Uu8: p < 0.05). Remarkably, ureaplasmas did not induce IL-12p40 response and promoted lower amounts of anti-inflammatory IL-10 and IL-1ra than LPS, provoking a cytokine imbalance more in favor of pro-inflammation (IL-1β/IL-10, IL-8/IL-10 and IL-8/IL-1ra: p < 0.01, vs. LPS). In contrast to LPS, both isolates induced TLR2 mRNA in neonatal and adult cells (p < 0.001 and p < 0.05) and suppressed TLR4 mRNA in adult monocytes (p < 0.05). Upon co-stimulation, Uu8 and Up3 inhibited LPS-induced intracellular IL-1β (p < 0.001 and p < 0.05) and IL-8 in adult monocytes (p < 0.01), while LPS-induced neonatal cytokines were maintained or aggravated (p < 0.05). Conclusion: Our data demonstrate a considerable pro-inflammatory capacity of Ureaplasma isolates in human monocytes. Stimulating pro-inflammatory cytokine responses while hardly inducing immunomodulatory and anti-inflammatory cytokines, ureaplasmas might push monocyte immune responses toward pro-inflammation. Inhibition of LPS-induced cytokines in adult monocytes in contrast to sustained inflammation in term neonatal monocytes indicates a differential modulation of host immune responses to a second stimulus. Modification of TLR2 and TLR4 expression may shape host susceptibility to inflammation.
Collapse
Affiliation(s)
- Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Christine Silwedel
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Markus Fehrholz
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Ana M Waaga-Gasser
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Birgit Henrich
- Institute of Medical Microbiology and Hospital Hygiene, University Clinic of Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Heike Claus
- Institute for Hygiene and Microbiology, University of Wuerzburg, Wuerzburg, Germany
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
79
|
Giraud A, Guiraut C, Chevin M, Chabrier S, Sébire G. Role of Perinatal Inflammation in Neonatal Arterial Ischemic Stroke. Front Neurol 2017; 8:612. [PMID: 29201015 PMCID: PMC5696351 DOI: 10.3389/fneur.2017.00612] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022] Open
Abstract
Based on the review of the literature, perinatal inflammation often induced by infection is the only consistent independent risk factor of neonatal arterial ischemic stroke (NAIS). Preclinical studies show that acute inflammatory processes take place in placenta, cerebral arterial wall of NAIS-susceptible arteries and neonatal brain. A top research priority in NAIS is to further characterize the nature and spatiotemporal features of the inflammatory processes involved in multiple levels of the pathophysiology of NAIS, to adequately design randomized control trials using targeted anti-inflammatory vasculo- and neuroprotective agents.
Collapse
Affiliation(s)
- Antoine Giraud
- EA 4607 SNA EPIS, Jean Monnet University, Saint-Etienne, France.,Child Neurology Division, Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Clémence Guiraut
- Child Neurology Division, Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Mathilde Chevin
- Child Neurology Division, Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Stéphane Chabrier
- French Center for Pediatric Stroke and Pediatric Rehabilitation Unit, Department of Pediatrics, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Guillaume Sébire
- Child Neurology Division, Department of Pediatrics, McGill University, Montréal, QC, Canada
| |
Collapse
|
80
|
Makinson R, Lloyd K, Rayasam A, McKee S, Brown A, Barila G, Grissom N, George R, Marini M, Fabry Z, Elovitz M, Reyes TM. Intrauterine inflammation induces sex-specific effects on neuroinflammation, white matter, and behavior. Brain Behav Immun 2017; 66:277-288. [PMID: 28739513 PMCID: PMC6916731 DOI: 10.1016/j.bbi.2017.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
Exposure to inflammation during pregnancy has been linked to adverse neurodevelopmental consequences for the offspring. One common route through which a developing fetus is exposed to inflammation is with intrauterine inflammation. To that end, we utilized an animal model of intrauterine inflammation (IUI; intrauterine lipopolysaccharide (LPS) administration, 50µg, E15) to assess placental and fetal brain inflammatory responses, white matter integrity, anxiety-related behaviors (elevated zero maze, light dark box, open field), microglial counts, and the CNS cytokine response to an acute injection of LPS in both males and females. These studies revealed that for multiple endpoints (fetal brain cytokine levels, cytokine response to adult LPS challenge) male IUI offspring were uniquely affected by intrauterine inflammation, while for other endpoints (behavior, microglial number) both sexes were similarly affected. These data advance our understanding of sex-specific effects of early life exposure to inflammation in a translationally- relevant model.
Collapse
Affiliation(s)
- Ryan Makinson
- University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Kelsey Lloyd
- University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Aditya Rayasam
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison
| | - Sarah McKee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Amy Brown
- Maternal and Child Health Research Center, Department of OBGYN, University of Pennsylvania, Philadelphia, PA
| | - Guillermo Barila
- Maternal and Child Health Research Center, Department of OBGYN, University of Pennsylvania, Philadelphia, PA
| | - Nicola Grissom
- University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Robert George
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Matt Marini
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison
| | - Michal Elovitz
- Maternal and Child Health Research Center, Department of OBGYN, University of Pennsylvania, Philadelphia, PA
| | - Teresa M. Reyes
- University of Cincinnati, College of Medicine, Cincinnati, OH
| |
Collapse
|
81
|
Inflammatory molecules and neurotrophic factors as biomarkers of neuropsychomotor development in preterm neonates: A Systematic Review. Int J Dev Neurosci 2017; 65:29-37. [PMID: 29051031 DOI: 10.1016/j.ijdevneu.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To provide a systematic review investigating the role of inflammatory molecules and neurotrophic factors as biomarkers of neuropsychomotor development in preterm neonates. DATA SOURCE Databases including PubMed, BIREME, and Scopus were systematically searched. Observational studies, as well as transversal, and cohort studies using human subjects published from 1990 to September 2017 were eligible for inclusion. Two authors independently identified eligible studies and analyzed their characteristics, quality, and accuracy in depth. DATA SYNTHESIS 11 eligible studies clearly investigated the association between peripheral inflammation and motor and/or cognitive development in preterm infants. However, the selected populations differed in relation to the events associated with prematurity and the risk factors to abnormal motor and/or cognitive development. These studies measured circulating levels of cytokines, chemokines, adhesion molecules, acute phase proteins, and growth factors. The most commonly analyzed proteins were IL-1β, IL-6, TNF, CCL5/RANTES, CXCL8/IL-8, IGFBP-1, and VEGF. In seven of the eligible studies, plasma levels of IL-6 correlated with development delay. Two studies reported correlation between CXCL8/IL-8 plasma levels with cognitive and motor delay. In one study, higher levels of MCP-1/CCL2 were associated with better cognitive and motor outcome. CONCLUSION There is preliminary evidence indicating that circulating inflammatory molecules are associated with motor and cognitive development in preterm neonates, even considering different populations.
Collapse
|
82
|
The IL-1β signalling pathway and its role in regulating pro-inflammatory and pro-labour mediators in human primary myometrial cells. Reprod Biol 2017; 17:333-340. [PMID: 28988892 DOI: 10.1016/j.repbio.2017.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/25/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
Abstract
Interleukin (IL)-1β plays a central role in the processes of human labour and delivery. The adaptor proteins involved in the IL-1β signalling pathway in human myometrium are not known. This study sought to determine the role of the adaptor proteins myeloid differentiation primary response 88 (MyD88), tumour necrosis factor receptor-associated factor 6 (TRAF6), IL-1 receptor-associated kinase 4 (IRAK4) and transforming growth factor beta-activated kinase 1 (TAK1) in IL-1β-induced formation of pro-inflammatory and pro-labour mediators in human myometrium. Human primary myometrial cells were transfected with siRNA against MyD88 (siMYD88), TRAF6 (siTRAF6), IRAK4 (siIRAK4) or TAK1 (siTAK1), treated with IL-1β, and assayed for the mRNA expression and or secretion of pro-inflammatory and pro-labour mediators. Transfection of primary myometrial cells with siMYD88, siTRAF6, siIRAK4 and siTAK1 significantly decreased IL-1β-induced IL-1α, IL-6, growth-regulated alpha protein (GRO-α), IL-8, monocyte chemoattractant protein (MCP)-1, intercellular adhesion molecule (ICAM)-1 and cyclooxygenase (COX)-2 mRNA expression and release of IL-6, GRO-α, IL-8, MCP-1, ICAM-1 and prostaglandin PGF2α. The expression and secretion of the extracellular matrix remodelling enzyme matrix metalloproteinase (MMP)-9 was significantly lower with siMYD88 and siTRAF6. Finally, IL-1β-induced nuclear factor κB (NF-κB) transcriptional activity was significantly attenuated by transfection with siMyD88, siTRAF6 and siIRAK4; there was no effect of siTAK1 transfection on NF-κB transcriptional activity. Collectively, these findings suggest that MyD88, TRAF6, IRAK4 and TAK1 are involved in IL-1β signalling in human myometrium. Further studies are required to determine if inhibition of these proteins can prevent preterm birth.
Collapse
|
83
|
Surate Solaligue DE, Rodríguez-Castillo JA, Ahlbrecht K, Morty RE. Recent advances in our understanding of the mechanisms of late lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1101-L1153. [PMID: 28971976 DOI: 10.1152/ajplung.00343.2017] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of lung development is to generate an organ of gas exchange that provides both a thin gas diffusion barrier and a large gas diffusion surface area, which concomitantly generates a steep gas diffusion concentration gradient. As such, the lung is perfectly structured to undertake the function of gas exchange: a large number of small alveoli provide extensive surface area within the limited volume of the lung, and a delicate alveolo-capillary barrier brings circulating blood into close proximity to the inspired air. Efficient movement of inspired air and circulating blood through the conducting airways and conducting vessels, respectively, generates steep oxygen and carbon dioxide concentration gradients across the alveolo-capillary barrier, providing ideal conditions for effective diffusion of both gases during breathing. The development of the gas exchange apparatus of the lung occurs during the second phase of lung development-namely, late lung development-which includes the canalicular, saccular, and alveolar stages of lung development. It is during these stages of lung development that preterm-born infants are delivered, when the lung is not yet competent for effective gas exchange. These infants may develop bronchopulmonary dysplasia (BPD), a syndrome complicated by disturbances to the development of the alveoli and the pulmonary vasculature. It is the objective of this review to update the reader about recent developments that further our understanding of the mechanisms of lung alveolarization and vascularization and the pathogenesis of BPD and other neonatal lung diseases that feature lung hypoplasia.
Collapse
Affiliation(s)
- David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
84
|
Strack M, Billard É, Chatenet D, Lubell WD. Urotensin core mimics that modulate the biological activity of urotensin-II related peptide but not urotensin-II. Bioorg Med Chem Lett 2017. [DOI: 10.1016/j.bmcl.2017.05.088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
85
|
Abstract
Preterm birth (PTB) remains a major obstetric healthcare problem and a significant contributor to perinatal morbidity, mortality, and long-term disability. Over the past few decades, the perinatal outcomes of preterm neonates have improved markedly through research and advances in neonatal care, whereas rates of spontaneous PTB have essentially remained static. However, research into causal pathways and new diagnostic and treatment modalities is now bearing fruit and translational initiatives are beginning to impact upon PTB rates. Successful PTB prevention requires a multifaceted approach, combining public health and educational programs, lifestyle modification, access to/optimisation of obstetric healthcare, effective prediction and diagnostic modalities, and the application of effective, targeted interventions. Progress has been made in some of these areas, although there remain areas of controversy and uncertainty. Attention is now being directed to areas where greater gains can be achieved. In this mini-review, we will briefly and selectively review a range of PTB prevention strategies and initiatives where progress has been made and where exciting opportunities await exploitation, evaluation, and implementation.
Collapse
Affiliation(s)
- Jeff A Keelan
- Division of Obstetrics & Gynaecology, School of Medicine, University of Western Australia King Edward Memorial Hospital, Perth, Australia
| | - John P Newnham
- Division of Obstetrics & Gynaecology, School of Medicine, University of Western Australia King Edward Memorial Hospital, Perth, Australia
| |
Collapse
|