101
|
Abstract
Infections during pregnancy may affect a developing fetus. If left untreated, these infections can lead to the death of the mother, fetus, or neonate and other adverse sequelae. There are many factors that impact infection during pregnancy, such as the immune system changes during pregnancy, hormonal flux, stress, and the microbiome. We review some of the outcomes of infection during pregnancy, such as preterm birth, chorioamnionitis, meningitis, hydrocephaly, developmental delays, microcephaly, and sepsis. Transmission routes are discussed regarding how a pregnant woman may pass her infection to her fetus. This is followed by examples of infection during pregnancy: bacterial, viral, parasitic, and fungal infections. There are many known organisms that are capable of producing similar congenital defects during pregnancy; however, whether these infections share common mechanisms of action is yet to be determined. To protect the health of pregnant women and their offspring, additional research is needed to understand how these intrauterine infections adversely affect pregnancies and/or neonates in order to develop prevention strategies and treatments.
Collapse
|
102
|
Ali A, Cui X, Eyles D. Developmental vitamin D deficiency and autism: Putative pathogenic mechanisms. J Steroid Biochem Mol Biol 2018; 175:108-118. [PMID: 28027915 DOI: 10.1016/j.jsbmb.2016.12.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/31/2016] [Accepted: 12/23/2016] [Indexed: 01/08/2023]
Abstract
Autism is a neurodevelopmental disease that presents in early life. Despite a considerable amount of studies, the neurobiological mechanisms underlying autism remain obscure. Both genetic and environmental factors are involved in the development of autism. Vitamin D deficiency is emerging as a consistently reported risk factor in children. One reason for the prominence now being given to this risk factor is that it would appear to interact with several other epidemiological risk factors for autism. Vitamin D is an active neurosteroid and plays crucial neuroprotective roles in the developing brain. It has important roles in cell proliferation and differentiation, immunomodulation, regulation of neurotransmission and steroidogenesis. Animal studies have suggested that transient prenatal vitamin D deficiency is associated with altered brain development. Here we review the potential neurobiological mechanisms linking prenatal vitamin D deficiency and autism and also discuss what future research targets must now be addressed.
Collapse
Affiliation(s)
- Asad Ali
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia
| | - Xiaoying Cui
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia
| | - Darryl Eyles
- Queensland Brain Institute, University of Queensland, Qld 4072, Australia; Queensland Centre for Mental Health Research, Wacol, Qld 4076, Australia.
| |
Collapse
|
103
|
The microbiota influences cell death and microglial colonization in the perinatal mouse brain. Brain Behav Immun 2018; 67:218-229. [PMID: 28890156 PMCID: PMC5696094 DOI: 10.1016/j.bbi.2017.08.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022] Open
Abstract
The mammalian fetus develops in a largely sterile environment, and direct exposure to a complex microbiota does not occur until birth. We took advantage of this to examine the effect of the microbiota on brain development during the first few days of life. The expression of anti- and pro-inflammatory cytokines, developmental cell death, and microglial colonization in the brain were compared between newborn conventionally colonized mice and mice born in sterile, germ-free (GF) conditions. Expression of the pro-inflammatory cytokines interleukin 1β and tumor necrosis factor α was markedly suppressed in GF newborns. GF mice also had altered cell death, with some regions exhibiting higher rates (paraventricular nucleus of the hypothalamus and the CA1 oriens layer of the hippocampus) and other regions exhibiting no change or lower rates (arcuate nucleus of the hypothalamus) of cell death. Microglial labeling was elevated in GF mice, due to an increase in both microglial cell size and number. The changes in cytokine expression, cell death and microglial labeling were evident on the day of birth, but were absent on embryonic day 18.5, approximately one-half day prior to expected delivery. Taken together, our results suggest that direct exposure to the microbiota at birth influences key neurodevelopmental events and does so within hours. These findings may help to explain some of the behavioral and neurochemical alterations previously seen in adult GF mice.
Collapse
|
104
|
Zhang Z, Jyoti A, Balakrishnan B, Williams M, Singh S, Chugani DC, Kannan S. Trajectory of inflammatory and microglial activation markers in the postnatal rabbit brain following intrauterine endotoxin exposure. Neurobiol Dis 2017; 111:153-162. [PMID: 29274431 DOI: 10.1016/j.nbd.2017.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/17/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Maternal infection is a risk factor for periventricular leukomalacia and cerebral palsy (CP) in neonates. We have previously demonstrated hypomyelination and motor deficits in newborn rabbits, as seen in patients with cerebral palsy, following maternal intrauterine endotoxin administration. This was associated with increased microglial activation, primarily involving the periventricular region (PVR). In this study we hypothesized that maternal intrauterine inflammation leads to a pro-inflammatory environment in the PVR that is associated with microglial activation in the first 2 postnatal weeks. METHODS Timed pregnant New Zealand white rabbits underwent laparotomy on gestational day 28 (G28). They were randomly divided to receive lipopolysaccharide (LPS; 20μg/kg in 1mL saline) (Endotoxin group) or saline (1mL) (control saline, CS group), administrated along the wall of the uterus. The PVR from the CS and Endotoxin kits were harvested at G29 (1day post-injury), postnatal day1 (PND1, 3day post-injury) and PND5 (7days post-injury) for real-time PCR, ELISA and immunohistochemistry. Kits from CS and Endotoxin groups underwent longitudinal MicroPET imaging, with [11C]PK11195, a tracer for microglial activation. RESULTS We found that intrauterine endotoxin exposure resulted in pro-inflammatory microglial activation in the PVR of rabbits in the first postnatal week. This was evidenced by increased TSPO (translocator protein) expression co-localized with microglia/macrophages in the PVR, and changes in the microglial morphology (ameboid soma and retracted processes). In addition, CD11b level significantly increased with a concomitant decline in the CD45 level in the PVR at G29 and PND1. There was a significant elevation of pro-inflammatory cytokines and iNOS, and decreased anti-inflammatory markers in the Endotoxin kits at G29, PND1 and PND5. Increased [11C]PK11195 binding to the TSPO measured in vivo by PET imaging in the brain of Endotoxin kits was present up to PND14-17. CONCLUSIONS Our results indicate that a robust pro-inflammatory microglial phenotype/brain milieu commenced within 24h after LPS exposure and persisted through PND5 and in vivo TSPO binding was found at PND14-17. This suggests that there may be a window of opportunity to treat after birth. Therapies aimed at inducing an anti-inflammatory phenotype in microglia might promote recovery in maternal inflammation induced neonatal brain injury.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, United States
| | - Amar Jyoti
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, United States
| | - Bindu Balakrishnan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, United States
| | - Monica Williams
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, United States
| | - Sarabdeep Singh
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, United States
| | - Diane C Chugani
- Nemours/AI duPont Hospital for Children, Wilmington, DE, United States; Communication Sciences and Disorders Department, University of Delaware, Newark, DE, United States
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| |
Collapse
|
105
|
Zhang X, Rocha-Ferreira E, Li T, Vontell R, Jabin D, Hua S, Zhou K, Nazmi A, Albertsson AM, Sobotka K, Ek J, Thornton C, Hagberg H, Mallard C, Leavenworth JW, Zhu C, Wang X. γδT cells but not αβT cells contribute to sepsis-induced white matter injury and motor abnormalities in mice. J Neuroinflammation 2017; 14:255. [PMID: 29262837 PMCID: PMC5738716 DOI: 10.1186/s12974-017-1029-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/08/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Infection and sepsis are associated with brain white matter injury in preterm infants and the subsequent development of cerebral palsy. METHODS In the present study, we used a neonatal mouse sepsis-induced white matter injury model to determine the contribution of different T cell subsets (αβT cells and γδT cells) to white matter injury and consequent behavioral changes. C57BL/6J wild-type (WT), T cell receptor (TCR) δ-deficient (Tcrd -/-, lacking γδT cells), and TCRα-deficient (Tcra -/-, lacking αβT cells) mice were administered with lipopolysaccharide (LPS) at postnatal day (PND) 2. Brain myelination was examined at PNDs 12, 26, and 60. Motor function and anxiety-like behavior were evaluated at PND 26 or 30 using DigiGait analysis and an elevated plus maze. RESULTS White matter development was normal in Tcrd -/- and Tcrα -/- compared to WT mice. LPS exposure induced reductions in white matter tissue volume in WT and Tcrα -/- mice, but not in the Tcrd -/- mice, compared with the saline-treated groups. Neither LPS administration nor the T cell deficiency affected anxiety behavior in these mice as determined with the elevated plus maze. DigiGait analysis revealed motor function deficiency after LPS-induced sepsis in both WT and Tcrα -/- mice, but no such effect was observed in Tcrd -/- mice. CONCLUSIONS Our results suggest that γδT cells but not αβT cells contribute to sepsis-induced white matter injury and subsequent motor function abnormalities in early life. Modulating the activity of γδT cells in the early stages of preterm white matter injury might represent a novel therapeutic strategy for the treatment of perinatal brain injury.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Li
- Henan Key Laboratory of Child Brain Injury, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden
| | - Regina Vontell
- Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Darakhshan Jabin
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Sha Hua
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden.,Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University Medical School, Luwan Branch, Shanghai, China
| | - Kai Zhou
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Arshed Nazmi
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Anna-Maj Albertsson
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Kristina Sobotka
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Joakim Ek
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Claire Thornton
- Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Henrik Hagberg
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Carina Mallard
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Jianmei W Leavenworth
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA.,Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden.
| | - Xiaoyang Wang
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden. .,Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
106
|
Albertsson AM, Zhang X, Vontell R, Bi D, Bronson RT, Supramaniam V, Baburamani AA, Hua S, Nazmi A, Cardell S, Zhu C, Cantor H, Mallard C, Hagberg H, Leavenworth JW, Wang X. γδ T Cells Contribute to Injury in the Developing Brain. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:757-767. [PMID: 29248460 PMCID: PMC5840494 DOI: 10.1016/j.ajpath.2017.11.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/11/2017] [Accepted: 11/07/2017] [Indexed: 01/04/2023]
Abstract
Brain injury in premature infants, especially periventricular leukomalacia, is an important cause of neurologic disabilities. Inflammation contributes to perinatal brain injury development, but the essential mediators that lead to early-life brain injury remain largely unknown. Neonates have reduced capacity for mounting conventional αβT-cell responses. However, γδT cells are already functionally competent during early development and are important in early-life immunity. We investigated the potential contribution of γδT cells to preterm brain injury using postmortem brains from human preterm infants with periventricular leukomalacia and two animal models of preterm brain injury—the hypoxic-ischemic mouse model and a fetal sheep asphyxia model. Large numbers of γδT cells were observed in the brains of mice, sheep, and postmortem preterm infants after injury, and depletion of γδT cells provided protection in the mouse model. The common γδT-cell–associated cytokines interferon-γ and IL-17A were not detectable in the brain. Although there were increased mRNA levels of Il17f and Il22 in the mouse brains after injury, neither IL-17F nor IL-22 cytokines contributed to preterm brain injury. These findings highlight unique features of injury in the developing brain, where, unlike injury in the mature brain, γδT cells function as initiators of injury independently of common γδT-cell–associated cytokines. This finding will help to identify therapeutic targets for preventing or treating preterm infants with brain injury.
Collapse
Affiliation(s)
- Anna-Maj Albertsson
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoli Zhang
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Provincial Key Laboratory of Child Brain Injury, Zhengzhou, China
| | - Regina Vontell
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Dan Bi
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Roderick T Bronson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts; Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Veena Supramaniam
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Ana A Baburamani
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Sha Hua
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Cardiovascular Medicine, Luwan Branch of Shanghai Jiaotong University Medical School Ruijin Hospital, Shanghai, China
| | - Arshed Nazmi
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanna Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Provincial Key Laboratory of Child Brain Injury, Zhengzhou, China; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Harvey Cantor
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts; Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Carina Mallard
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom; Department of Clinical Sciences, East Hospital, Gothenburg, Sweden
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama; Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Xiaoyang Wang
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
107
|
Kohli-Lynch M, Russell NJ, Seale AC, Dangor Z, Tann CJ, Baker CJ, Bartlett L, Cutland C, Gravett MG, Heath PT, Ip M, Le Doare K, Madhi SA, Rubens CE, Saha SK, Schrag S, Sobanjo-ter Meulen A, Vekemans J, O’Sullivan C, Nakwa F, Ben Hamouda H, Soua H, Giorgakoudi K, Ladhani S, Lamagni T, Rattue H, Trotter C, Lawn JE. Neurodevelopmental Impairment in Children After Group B Streptococcal Disease Worldwide: Systematic Review and Meta-analyses. Clin Infect Dis 2017; 65:S190-S199. [PMID: 29117331 PMCID: PMC5848372 DOI: 10.1093/cid/cix663] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Survivors of infant group B streptococcal (GBS) disease are at risk of neurodevelopmental impairment (NDI), a burden not previously systematically quantified. This is the 10th of 11 articles estimating the burden of GBS disease. Here we aimed to estimate NDI in survivors of infant GBS disease. METHODS We conducted systematic literature reviews (PubMed/Medline, Embase, Latin American and Caribbean Health Sciences Literature [LILACS], World Health Organization Library Information System [WHOLIS], and Scopus) and sought unpublished data on the risk of NDI after invasive GBS disease in infants <90 days of age. We did meta-analyses to derive pooled estimates of the percentage of infants with NDI following GBS meningitis. RESULTS We identified 6127 studies, of which 18 met eligibility criteria, all from middle- or high-income contexts. All 18 studies followed up survivors of GBS meningitis; only 5 of these studies also followed up survivors of GBS sepsis and were too few to pool in a meta-analysis. Of meningitis survivors, 32% (95% CI, 25%-38%) had NDI at 18 months of follow-up, including 18% (95% CI, 13%-22%) with moderate to severe NDI. CONCLUSIONS GBS meningitis is an important risk factor for moderate to severe NDI, affecting around 1 in 5 survivors. However, data are limited, and we were unable to estimate NDI after GBS sepsis. Comparability of studies is difficult due to methodological differences including variability in timing of clinical reviews and assessment tools. Follow-up of clinical cases and standardization of methods are essential to fully quantify the total burden of NDI associated with GBS disease, and inform program priorities.
Collapse
Affiliation(s)
- Maya Kohli-Lynch
- Maternal, Adolescent, Reproductive and Child Health Centre, London School of Hygiene & Tropical Medicine, United Kingdom
- Centre for Child and Adolescent Health, School of Social and Community Medicine, University of Bristol, United Kingdom
| | - Neal J Russell
- Maternal, Adolescent, Reproductive and Child Health Centre, London School of Hygiene & Tropical Medicine, United Kingdom
- King’s College London, United Kingdom
| | - Anna C Seale
- Maternal, Adolescent, Reproductive and Child Health Centre, London School of Hygiene & Tropical Medicine, United Kingdom
- College of Health and Medical Sciences, Haramaya University, Dire Dawa, Ethiopia
| | - Ziyaad Dangor
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, and
- Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cally J Tann
- Maternal, Adolescent, Reproductive and Child Health Centre, London School of Hygiene & Tropical Medicine, United Kingdom
- Neonatal Medicine, University College London Hospitals NHS Foundation Trust, United Kingdom
| | - Carol J Baker
- Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Linda Bartlett
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Clare Cutland
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, and
| | - Michael G Gravett
- Global Alliance to Prevent Prematurity and Stillbirth, Seattle, Washington
- Department of Obstetrics and Gynecology, University of Washington, Seattle
| | - Paul T Heath
- Vaccine Institute, Institute for Infection and Immunity, St George’s Hospital, University of London and St George’s University Hospitals NHS Foundation Trust, United Kingdom;
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, Chinese University of Hong Kong
| | - Kirsty Le Doare
- Vaccine Institute, Institute for Infection and Immunity, St George’s Hospital, University of London and St George’s University Hospitals NHS Foundation Trust, United Kingdom;
- Centre for International Child Health, Imperial College London, United Kingdom
| | - Shabir A Madhi
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, and
- National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa
| | - Craig E Rubens
- Maternal, Adolescent, Reproductive and Child Health Centre, London School of Hygiene & Tropical Medicine, United Kingdom
- Department of Global Health, University of Washington, Seattle
| | | | - Stephanie Schrag
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | - Catherine O’Sullivan
- Vaccine Institute, Institute for Infection and Immunity, St George’s Hospital, University of London and St George’s University Hospitals NHS Foundation Trust, United Kingdom;
| | - Firdose Nakwa
- Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Hechmi Ben Hamouda
- Department of Neonatology, University Hospital Tahar Sfar, Mahdia, Tunisia
| | - Habib Soua
- Department of Neonatology, University Hospital Tahar Sfar, Mahdia, Tunisia
| | | | | | | | - Hilary Rattue
- Vaccine Institute, Institute for Infection and Immunity, St George’s Hospital, University of London and St George’s University Hospitals NHS Foundation Trust, United Kingdom;
| | | | - Joy E Lawn
- Maternal, Adolescent, Reproductive and Child Health Centre, London School of Hygiene & Tropical Medicine, United Kingdom
| |
Collapse
|
108
|
Domínguez Rubio AP, Correa F, Aisemberg J, Dorfman D, Bariani MV, Rosenstein RE, Zorrilla Zubilete M, Franchi AM. Maternal administration of melatonin exerts short- and long-term neuroprotective effects on the offspring from lipopolysaccharide-treated mice. J Pineal Res 2017; 63. [PMID: 28776755 DOI: 10.1111/jpi.12439] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/31/2017] [Indexed: 01/02/2023]
Abstract
Preterm birth is a major contributor to early and delayed physical and cognitive impairment. Epidemiological and experimental data indicate that maternal infections are a significant and preventable cause of preterm birth. Recently, melatonin has been suggested to exert neuroprotective effects in several models of brain injury. Here, we sought to investigate whether the administration of melatonin is able to prevent lipopolysaccharide (LPS)-induced fetal brain damage in a model of LPS-induced preterm labor. For this purpose, 15-day pregnant BALB/c mice received intraperitoneally 2 doses of LPS or vehicle: the first one at 10:00 hours (0.26 mg/kg) and the second at 13:00 hours (0.52 mg/kg). On day 14 of pregnancy, a group of mice was subcutaneously implanted with a pellet of 25 mg melatonin. This experimental protocol resulted in 100% of preterm birth and pup death in the LPS group and a 50% of term birth and pup survival in the melatonin + LPS group. In the absence of melatonin, fetuses from LPS-treated mothers showed histological signs of brain damage, microglial/macrophage activation, and higher levels of IL-1β, inducible nitric oxide synthase (NOS), and neuronal NOS mRNAs as well as increased histone acetyltransferase activity and histone H3 hyperacetylation. In contrast, antenatal administration of melatonin prevented LPS-induced fetal brain damage. Moreover, when behavioral traits were analyzed in the offspring from control, melatonin, and melatonin + LPS, no significant differences were found, suggesting that melatonin prevented LPS-induced long-term neurodevelopmental impairments. Collectively, our results suggest that melatonin could be a new therapeutic tool to prevent fetal brain damage and its long-term consequences induced by maternal inflammation.
Collapse
Affiliation(s)
- Ana Paula Domínguez Rubio
- Laboratorio de Fisiopatología de la Preñez y el Parto, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernando Correa
- Laboratorio de Fisiopatología de la Preñez y el Parto, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julieta Aisemberg
- Laboratorio de Fisiopatología de la Preñez y el Parto, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Damián Dorfman
- Laboratorio de Neuroquimíca Retiniana y Oftalmología Experimental, Departamento de Bioquímica Humana, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Victoria Bariani
- Laboratorio de Fisiopatología de la Preñez y el Parto, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ruth Estela Rosenstein
- Laboratorio de Neuroquimíca Retiniana y Oftalmología Experimental, Departamento de Bioquímica Humana, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Zorrilla Zubilete
- Laboratorio de Neuropsicofarmacología del Estrés, Departamento de Farmacología, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana María Franchi
- Laboratorio de Fisiopatología de la Preñez y el Parto, Facultad de Medicina, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
109
|
Huang J, Zhang L, Kang B, Zhu T, Li Y, Zhao F, Qu Y, Mu D. Association between perinatal hypoxic-ischemia and periventricular leukomalacia in preterm infants: A systematic review and meta-analysis. PLoS One 2017; 12:e0184993. [PMID: 28931047 PMCID: PMC5607162 DOI: 10.1371/journal.pone.0184993] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/04/2017] [Indexed: 11/18/2022] Open
Abstract
Background Although investigators have implicated hypoxic-ischemia (HI) as a potential cause of periventricular leukomalacia (PVL), the role of clinical risk factors or markers for HI in the development of PVL remains controversial. The aim of this study was to identify perinatal HI-related factors associated with PVL. Method The PubMed, EMBASE, and Cochrane Library databases were searched. The last search was performed on January 2017. Summary effect estimates (pooled odds ratios [ORs]) were calculated for each risk factor using fixed or random effects models with tests for heterogeneity and publication bias. Results Fifteen studies with a total of 12,851 participants were included in this meta-analysis, and 14 potential risk factors were analyzed. The pooled results showed that mothers with oligohydramnios (OR, 1.55; 95% confidence interval [CI], 1.05 to 2.30), preterm infants with acidemia (OR, 1.87; 95% CI, 1.18 to 2.97), 1-minute Apgar score <7 (OR 2.69; 95% CI, 1.13 to 6.41), 5-minute Apgar score <7 (OR, 1.89; 95% CI, 1.39 to 2.56), apnea (OR, 1.76; 95% CI, 1.07 to 2.90), respiratory distress syndrome (OR, 1.46; 95% CI, 1.04 to 2.03), and seizures (OR, 4.60; 95% CI, 2.84 to 7.46) were associated with increased risk of PVL. Conclusion This study identified perinatal HI-related risk factors for the development of PVL in preterm infants. Future large-scale prospective clinical studies are required to validate and extend these findings.
Collapse
Affiliation(s)
- Jichong Huang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Bingyao Kang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Tingting Zhu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yafei Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Fengyan Zhao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
- * E-mail:
| |
Collapse
|
110
|
Shiow LR, Favrais G, Schirmer L, Schang AL, Cipriani S, Andres C, Wright JN, Nobuta H, Fleiss B, Gressens P, Rowitch DH. Reactive astrocyte COX2-PGE2 production inhibits oligodendrocyte maturation in neonatal white matter injury. Glia 2017; 65:2024-2037. [PMID: 28856805 DOI: 10.1002/glia.23212] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/12/2017] [Accepted: 08/08/2017] [Indexed: 12/19/2022]
Abstract
Inflammation is a major risk factor for neonatal white matter injury (NWMI), which is associated with later development of cerebral palsy. Although recent studies have demonstrated maturation arrest of oligodendrocyte progenitor cells (OPCs) in NWMI, the identity of inflammatory mediators with direct effects on OPCs has been unclear. Here, we investigated downstream effects of pro-inflammatory IL-1β to induce cyclooxygenase-2 (COX2) and prostaglandin E2 (PGE2) production in white matter. First, we assessed COX2 expression in human fetal brain and term neonatal brain affected by hypoxic-ischemic encephalopathy (HIE). In the developing human brain, COX2 was expressed in radial glia, microglia, and endothelial cells. In human term neonatal HIE cases with subcortical WMI, COX2 was strongly induced in reactive astrocytes with "A2" reactivity. Next, we show that OPCs express the EP1 receptor for PGE2, and PGE2 acts directly on OPCs to block maturation in vitro. Pharmacologic blockade with EP1-specific inhibitors (ONO-8711, SC-51089), or genetic deficiency of EP1 attenuated effects of PGE2. In an IL-1β-induced model of NWMI, astrocytes also exhibit "A2" reactivity and induce COX2. Furthermore, in vivo inhibition of COX2 with Nimesulide rescues hypomyelination and behavioral impairment. These findings suggest that neonatal white matter astrocytes can develop "A2" reactivity that contributes to OPC maturation arrest in NWMI through induction of COX2-PGE2 signaling, a pathway that can be targeted for neonatal neuroprotection.
Collapse
Affiliation(s)
- Lawrence R Shiow
- Department of Pediatrics and Division of Neonatology.,Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California
| | - Geraldine Favrais
- INSERM U930, Universite Francois Rabelais, Tours, France.,Neonatal intensive care unit, CHRU de Tours, Universite Francois Rabelais, Tours, France.,PROTECT, INSERM, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Lucas Schirmer
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California.,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Anne-Laure Schang
- PROTECT, INSERM, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France.,PremUP, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Sara Cipriani
- PROTECT, INSERM, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France.,PremUP, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France
| | | | - Jaclyn N Wright
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California
| | - Hiroko Nobuta
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California
| | - Bobbi Fleiss
- PROTECT, INSERM, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France.,PremUP, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France.,Department of Perinatal Imaging and Health, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas Hospital, London, United Kingdom
| | - Pierre Gressens
- PROTECT, INSERM, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France.,PremUP, Universite Paris Diderot, Sorbonne Paris Cite, Paris, France.,Department of Perinatal Imaging and Health, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas Hospital, London, United Kingdom
| | - David H Rowitch
- Department of Pediatrics and Division of Neonatology.,Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California.,Department of Paediatrics, and Wellcome Trust-MRC Stem Cell Institute, Cambridge University, Cambridge, United Kingdom
| |
Collapse
|
111
|
Wang X, Ma N, Sun Q, Huang C, Liu Y, Luo X. Elevated NF-κB signaling in Asherman syndrome patients and animal models. Oncotarget 2017; 8:15399-15406. [PMID: 28148903 PMCID: PMC5362494 DOI: 10.18632/oncotarget.14853] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/09/2017] [Indexed: 12/26/2022] Open
Abstract
Asherman syndrome (intrauterine adhesion) is often associated with menstrual abnormalities, infertility and recurrent miscarriage in female. Currently the molecular mechanism regulating the pathogenesis of Asherman syndrome is not known. Here we revealed that the inflammatory factor NF-κB expression is significantly elevated in the endometrial samples of Asherman syndrome patients. To further study the molecular mechanisms, we established an Asherman syndrome rat model and confirmed the important role of NF-κB in the pathogenesis of Asherman syndrome. In addition, our rat model provided direct evidence that intrauterine adhesion results in impaired pregnancy, supporting the clinical association between intrauterine adhesion and mis-regulated pregnancy. Our result identified NF-κB as a novel pathogenesis factor of Asherman syndrome and provided new insights for the prevention and treatment of intrauterine adhesions in Asherman syndrome patients.
Collapse
Affiliation(s)
- Xiangzhen Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China, 510630.,Nanshan Maternity and Child Healthcare Hospital of Shenzhen, Shenzhen, Guangdong, China, 518052
| | - Nana Ma
- The First Affiliated Hospital of Xinxiang medical college of Henan, Xinxiang, Henan, China, 453100
| | - Qiannan Sun
- Changzhi City People's Hospital of Shanxin Medical University Affiliated Hospital, Changzhi, Shanxi, China, 046000
| | - Chenlingzi Huang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China, 510630
| | - Yanmei Liu
- Huadu District, Guangzhou City People's Hospital, Guangzhou, Guangdong, China, 510630
| | - Xin Luo
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China, 510630
| |
Collapse
|
112
|
Modulatory Mechanism of Polyphenols and Nrf2 Signaling Pathway in LPS Challenged Pregnancy Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8254289. [PMID: 29138679 PMCID: PMC5613688 DOI: 10.1155/2017/8254289] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/16/2017] [Indexed: 12/16/2022]
Abstract
Early embryonic loss and adverse birth outcomes are the major reproductive disorders that affect both human and animals. The LPS induces inflammation by interacting with robust cellular mechanism which was considered as a plethora of numerous reproductive disorders such as fetal resorption, preterm birth, teratogenicity, intrauterine growth restriction, abortion, neural tube defects, fetal demise, and skeletal development retardation. LPS-triggered overproduction of free radicals leads to oxidative stress which mediates inflammation via stimulation of NF-κB and PPARγ transcription factors. Flavonoids, which exist in copious amounts in nature, possess a wide array of functions; their supplementation during pregnancy activates Nrf2 signaling pathway which encounters pregnancy disorders. It was further presumed that the development of strong antioxidant uterine environment during gestation can alleviate diseases which appear at adult stages. The purpose of this review is to focus on modulatory properties of flavonoids on oxidative stress-mediated pregnancy insult and abnormal outcomes and role of Nrf2 activation in pregnancy disorders. These findings would be helpful for providing new insights in ameliorating oxidative stress-induced pregnancy disorders.
Collapse
|
113
|
Threlkeld SW, Lim YP, La Rue M, Gaudet C, Stonestreet BS. Immuno-modulator inter-alpha inhibitor proteins ameliorate complex auditory processing deficits in rats with neonatal hypoxic-ischemic brain injury. Brain Behav Immun 2017; 64:173-179. [PMID: 28286301 PMCID: PMC5482760 DOI: 10.1016/j.bbi.2017.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/08/2017] [Accepted: 03/08/2017] [Indexed: 10/20/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is recognized as a significant problem in the perinatal period, contributing to life-long language-learning and other cognitive impairments. Central auditory processing deficits are common in infants with hypoxic-ischemic encephalopathy and have been shown to predict language learning deficits in other at risk infant populations. Inter-alpha inhibitor proteins (IAIPs) are a family of structurally related plasma proteins that modulate the systemic inflammatory response to infection and have been shown to attenuate cell death and improve learning outcomes after neonatal brain injury in rats. Here, we show that systemic administration of IAIPs during the early HI injury cascade ameliorates complex auditory discrimination deficits as compared to untreated HI injured subjects, despite reductions in brain weight. These findings have significant clinical implications for improving central auditory processing deficits linked to language learning in neonates with HI related brain injury.
Collapse
Affiliation(s)
- Steven W. Threlkeld
- Department of Neuroscience, Regis College, 235 Wellesley street, Weston MA, 02493, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02903, USA,Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI 02912, USA
| | - Molly La Rue
- Departments of Psychology and Biology, Rhode Island College, 600 Mount Pleasant Ave. Providence, RI, 02904, USA
| | - Cynthia Gaudet
- Departments of Psychology and Biology, Rhode Island College, 600 Mount Pleasant Ave. Providence, RI, 02904, USA
| | - Barbara S. Stonestreet
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI 02905, USA
| |
Collapse
|
114
|
McDougall ARA, Hale N, Rees S, Harding R, De Matteo R, Hooper SB, Tolcos M. Erythropoietin Protects Against Lipopolysaccharide-Induced Microgliosis and Abnormal Granule Cell Development in the Ovine Fetal Cerebellum. Front Cell Neurosci 2017; 11:224. [PMID: 28804448 PMCID: PMC5532439 DOI: 10.3389/fncel.2017.00224] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 07/13/2017] [Indexed: 11/13/2022] Open
Abstract
Erythropoietin (EPO) ameliorates inflammation-induced injury in cerebral white matter (WM). However, effects of inflammation on the cerebellum and neuroprotective effects of EPO are unknown. Our aims were to determine: (i) whether lipopolysaccharide (LPS)-induced intrauterine inflammation causes injury to, and/or impairs development of the cerebellum; and (ii) whether recombinant human EPO (rhEPO) mitigates these changes. At 107 ± 1 days gestational age (DGA; ~0.7 of term), fetal sheep received LPS (~0.9 μg/kg; i.v.) or an equivalent volume of saline, followed 1 h later with 5000 IU/kg rhEPO (i.v.) or an equivalent volume of saline (i.v.). This generated the following experimental groups: control (saline + saline; n = 6), LPS (LPS + saline, n = 8) and LPS + rhEPO (n = 8). At necropsy (116 ± 1 DGA; ~0.8 of term) the brain was perfusion-fixed and stained histologically (H&E) and immunostained to identify granule cells (Neuronal Nuclei, NeuN), granule cell proliferation (Ki67), Bergmann glia (glial fibrillary acidic protein, GFAP), astrogliosis (GFAP) and microgliosis (Iba-1). In comparison to controls, LPS fetuses had an increased density of Iba-1-positive microglia (p < 0.005) in the lobular WM; rhEPO prevented this increase (p < 0.05). The thickness of both the proliferative (Ki67-positive) and post-mitotic zones (Ki67-negative) of the EGL were increased in LPS-exposed fetuses compared to controls (p < 0.05), but were not different between controls and LPS + rhEPO fetuses. LPS also increased (p < 0.001) the density of granule cells (NeuN-positive) in the internal granule layer (IGL); rhEPO prevented the increase (p < 0.01). There was no difference between groups in the areas of the vermis (total cross-section), molecular layer (ML), IGL or WM, the density of NeuN-positive granule cells in the ML, the linear density of Bergmann glial fibers, the areal density or somal area of the Purkinje cells, the areal coverage of GFAP-positive astrocytes in the lobular and deep WM, the density of Iba-1-positive microglia in the deep WM or the density of apopotic cells in the cerebellum. LPS-induced intrauterine inflammation caused microgliosis and abnormal development of granule cells. rhEPO ameliorated these changes, suggesting that it is neuroprotective against LPS-induced inflammatory effects in the cerebellum.
Collapse
Affiliation(s)
- Annie R A McDougall
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Sandra Rees
- Department of Anatomy and Neuroscience, University of MelbourneParkville, VIC, Australia
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash UniversityClayton, VIC, Australia
| | - Robert De Matteo
- Department of Anatomy and Developmental Biology, Monash UniversityClayton, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT UniversityMelbourne, VIC, Australia
| |
Collapse
|
115
|
Maternal dendrimer-based therapy for inflammation-induced preterm birth and perinatal brain injury. Sci Rep 2017; 7:6106. [PMID: 28733619 PMCID: PMC5522481 DOI: 10.1038/s41598-017-06113-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 06/08/2017] [Indexed: 01/06/2023] Open
Abstract
Preterm birth is a major risk factor for adverse neurological outcomes in ex-preterm children, including motor, cognitive, and behavioral disabilities. N-acetyl-L-cysteine therapy has been used in clinical studies; however, it requires doses that cause significant side effects. In this study, we explore the effect of low dose N-acetyl-L-cysteine therapy, delivered using a targeted, systemic, maternal, dendrimer nanoparticle (DNAC), in a mouse model of intrauterine inflammation. Our results demonstrated that intraperitoneal maternal DNAC administration significantly reduced the preterm birth rate and altered placental immune profile with decreased CD8+ T-cell infiltration. Furthermore, we demonstrated that DNAC improved neurobehavioral outcomes and reduced fetal neuroinflammation and long-term microglial activation in offspring. Our study is the first to provide evidence for the role of CD8+ T-cell in the maternal-fetal interface during inflammation and further support the efficacy of DNAC in preventing preterm birth and prematurity-related outcomes.
Collapse
|
116
|
Teo JD, Morris MJ, Jones NM. Maternal obesity increases inflammation and exacerbates damage following neonatal hypoxic-ischaemic brain injury in rats. Brain Behav Immun 2017; 63:186-196. [PMID: 27746186 DOI: 10.1016/j.bbi.2016.10.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE In humans, maternal obesity is associated with an increase in the incidence of birth related difficulties. However, the impact of maternal obesity on the severity of brain injury in offspring is not known. Recent studies have found evidence of increased glial response and inflammatory mediators in the brains as a result of obesity in humans and rodents. We hypothesised that hypoxic-ischaemic (HI) brain injury is greater in neonatal offspring from obese rat mothers compared to lean controls. METHODS Female Sprague Dawley rats were randomly allocated to high fat (HFD, n=8) or chow (n=4) diet and mated with lean male rats. On postnatal day 7 (P7), male and female pups were randomly assigned to HI injury or control (C) groups. HI injury was induced by occlusion of the right carotid artery followed by 3h exposure to 8% oxygen, at 37°C. Control pups were removed from the mother for the same duration under ambient conditions. Righting behaviour was measured on day 1 and 7 following HI. The extent of brain injury was quantified in brain sections from P14 pups using cresyl violet staining and the difference in volume between brain hemispheres was measured. RESULTS Before mating, HFD mothers were 11% heavier than Chow mothers (p<0.05, t-test). Righting reflex was delayed in offspring from HFD-fed mothers compared to the Chow mothers. The Chow-HI pups showed a loss in ipsilateral brain tissue, while the HFD-HI group had significantly greater loss. No significant difference was detected in brain volume between the HFD-C and Chow-C pups. When analysed on a per litter basis, the size of the injury was significantly correlated with maternal weight. Similar observations were made with neuronal staining showing a greater loss of neurons in the brain of offspring from HFD-mothers following HI compared to Chow. Astrocytes appeared to more hypertrophic and a greater number of microglia were present in the injured hemisphere in offspring from mothers on HFD. HI caused an increase in the proportion of amoeboid microglia and exposure to maternal HFD exacerbated this response. In the contralateral hemisphere, offspring exposed to maternal HFD displayed a reduced proportion of ramified microglia. CONCLUSIONS Our data clearly demonstrate that maternal obesity can exacerbate the severity of brain damage caused by HI in neonatal offspring. Given that previous studies have shown enhanced inflammatory responses in offspring of obese mothers, these factors including gliosis and microglial infiltration are likely to contribute to enhanced brain injury.
Collapse
Affiliation(s)
- Jonathan D Teo
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, New South Wales, Australia
| | - Margaret J Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, New South Wales, Australia
| | - Nicole M Jones
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, New South Wales, Australia.
| |
Collapse
|
117
|
Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun 2017; 63:210-223. [PMID: 27865947 DOI: 10.1016/j.bbi.2016.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
The perinatal period has often been described as immune deficient. However, it has become clear that immune responses in the neonate following exposure to microbes or as a result of tissue injury may be substantial and play a role in perinatal brain injury. In this article we will review the immune cell composition under normal physiological conditions in the perinatal period, both in the human and rodent. We will summarize evidence of the inflammatory responses to stimuli and discuss how neonatal immune activation, both in the central nervous system and in the periphery, may contribute to perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden.
| |
Collapse
|
118
|
Lu HY, Ma JL, Shan JY, Zhang J, Wang QX, Zhang Q. High-mobility group box-1 and receptor for advanced glycation end products in preterm infants with brain injury. World J Pediatr 2017; 13:228-235. [PMID: 27995540 DOI: 10.1007/s12519-016-0077-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 07/15/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND High-mobility group box-1 (HMGB1) protein acts as an important pro-infl ammatory mediator, which is capable of activating inflammation and tissue repair. HMGB1 can bind to its receptor such as advanced glycation end products (RAGE). RAGE, in turn, can promote the production of pro-inflammatory cytokines. Soluble RAGE (sRAGE) is a truncated form of the receptor comprising the extracellular domain of RAGE and can inhibit RAGE-activation. The objective of this study was to investigate whether HMGB1 and RAGE are involved in the development of brain injury in preterm infants. METHODS In total, 108 infants ≤34 weeks gestation at birth were divided into 3 groups according to cranial altrasound scan: mild brain damage (n=33), severe brain damage (n=8) and no brain damage (n=67). All the placentas were submitted for pathologic evaluation. Histological chorioamnionitis (HCA) was defined as neutrophil infi ltration of amniotic membranes, umbilical cord or chorionic plate. Expressions of HMGB1 and RAGE proteins were assessed by immunohistochemical analysis. The concentration of HMGB1 and sRAGE in umbilical cord blood were measured by enzyme-linked immunosorbent assay. RESULTS The frequency of HCA was 30.12%. HCA was associated with elevated concentrations of HMGB1 and decreased sRAGE in umbilical cord blood. The severe brain injury group demonstrated higher cord blood HMGB1 concentrations (P<0.001) and lower sRAGE concentrations (P<0.001) than both other groups. Brain injury in the premature infants was linked to intense staining for HMGB1/RAGE, particularly in infl ammatory cells. CONCLUSIONS Changes of cord blood HMGB1 and sRAGE of premature infants had direct relationship with the degree of infl ammation and severity of brain damage. Monitoring sRAGE and HMGB1 levels may be helpful to predict intrauterine infection and brain injury in premature infants.
Collapse
Affiliation(s)
- Hong-Yan Lu
- Department of Pediatrics, the Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, China.
| | - Jiang-Lin Ma
- Department of Pediatrics, the Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, China
| | - Ji-Yan Shan
- Department of Pediatrics, the Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, China
| | - Jie Zhang
- Department of Pediatrics, the Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, China
| | - Qiu-Xia Wang
- Department of Pediatrics, the Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, China
| | - Qiang Zhang
- Department of Pediatrics, the Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, China
| |
Collapse
|
119
|
Pironkova RP, Giamelli J, Seiden H, Parnell VA, Gruber D, Sison CP, Kowal C, Ojamaa K. Brain injury with systemic inflammation in newborns with congenital heart disease undergoing heart surgery. Exp Ther Med 2017; 14:228-238. [PMID: 28672919 PMCID: PMC5488503 DOI: 10.3892/etm.2017.4493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/13/2017] [Indexed: 12/03/2022] Open
Abstract
The potential role of systemic inflammation on brain injury in newborns with congenital heart disease (CHD) was assessed by measuring levels of central nervous system (CNS)-derived proteins in serum prior to and following cardiac surgery. A total of 23 newborns (gestational age, 39±1 weeks) with a diagnosis of CHD that required cardiac surgery with cardiopulmonary bypass (CPB) were enrolled in the current study. Serum samples were collected immediately prior to surgery and 2, 24 and 48 h following CPB, and serum levels of phosphorylated neurofilament-heavy subunit (pNF-H), neuron-specific enolase (NSE) and S100B were analyzed. Systemic inflammation was assessed by measuring serum concentrations of complement C5a and complement sC5b9, and the following cytokines: Interleukin (IL)-1β, IL-6, IL-8, IL-10, IL12p70, interferon γ and tumor necrosis factor (TNF)-α. Analysis of cord blood from normal term deliveries (n=26) provided surrogate normative values for newborns. pNF-H and S100B were 2.4- to 2.8-fold higher (P<0.0001) in patient sera than in cord blood prior to surgery and remained elevated following CPB. Pre-surgical serum pNF-H and S100B levels directly correlated with interleukin (IL)-12p70 (ρ=0.442, P<0.05). pNF-H was inversely correlated with arterial pO2 prior to surgery (ρ=−0.493, P=0.01) and directly correlated with arterial pCO2 post-CPB (ρ=0.426, P<0.05), suggesting that tissue hypoxia and inflammation contribute to blood brain barrier (BBB) dysfunction and neuronal injury. Serum IL12p70, IL-6, IL-8, IL-10 and TNF-α levels were significantly higher in patients than in normal cord blood and levels of these cytokines increased following CPB (P<0.001). Activation of complement was observed in all patients prior to surgery, and serum C5a and sC5b9 remained elevated up to 48 h post-surgery. Furthermore, they were correlated (P<0.05) with low arterial pO2, high pCO2 and elevated arterial pressure in the postoperative period. Length of mechanical ventilation was associated directly with post-surgery serum IL-12p70 and IL-8 concentrations (P<0.05). Elevated serum concentrations of pNF-H and S100B in neonates with CHD suggest BBB dysfunction and CNS injury, with concurrent hypoxemia and an activated inflammatory response potentiating this effect.
Collapse
Affiliation(s)
- Rossitza P Pironkova
- Division of Pediatric Cardiology, Department of Pediatrics, Cohen Children's Medical Center of New York at Northwell Health, New Hyde Park, NY 11040, USA
| | - Joseph Giamelli
- Division of Pediatric Cardiology, Department of Pediatrics, Cohen Children's Medical Center of New York at Northwell Health, New Hyde Park, NY 11040, USA
| | - Howard Seiden
- Division of Pediatric Cardiology, Department of Pediatrics, Cohen Children's Medical Center of New York at Northwell Health, New Hyde Park, NY 11040, USA
| | - Vincent A Parnell
- Division of Pediatric Cardiothoracic Surgery, Department of Pediatrics, Cohen Children's Medical Center of New York at Northwell Health, New Hyde Park, NY 11040, USA
| | - Dorota Gruber
- Division of Pediatric Cardiology, Department of Pediatrics, Cohen Children's Medical Center of New York at Northwell Health, New Hyde Park, NY 11040, USA.,Department of Pediatrics, Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA
| | - Cristina P Sison
- Biostatistics Unit, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA.,Department of Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA
| | - Czeslawa Kowal
- Center for Musculoskeletal and Autoimmune Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Kaie Ojamaa
- Division of Pediatric Cardiology, Department of Pediatrics, Cohen Children's Medical Center of New York at Northwell Health, New Hyde Park, NY 11040, USA.,Department of Molecular Medicine, Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA.,Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
120
|
Huang HC, Yu HR, Hsu TY, Chen IL, Huang HC, Chang JC, Yang KD. MicroRNA-142-3p and let-7g Negatively Regulates Augmented IL-6 Production in Neonatal Polymorphonuclear Leukocytes. Int J Biol Sci 2017; 13:690-700. [PMID: 28655995 PMCID: PMC5485625 DOI: 10.7150/ijbs.17030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/13/2017] [Indexed: 11/05/2022] Open
Abstract
Neonatal PMN are qualitatively impaired in functions, yet they frequently reveal augmented inflammatory reactions during sepsis. Here, we hypothesized that PMN from newborns produce more IL-6 than those from adults under LPS stimulation, in which transcriptional or posttranscriptional regulation is involved in the altered expression. We found that neonatal PMN produced significantly higher IL-6 mRNA and protein than adult PMN. The higher IL-6 expression was not related to transcriptional but posttranscriptional regulation as the IL-6 expression was affected by the addition of cycloheximide but not actinomycin. To examine whether miRNA was involved in the IL-6 regulation of neonatal PMN, we surveyed differential displays of miRNAs that could potentially regulate IL-6 expression before and after LPS stimulation. Four miRNAs: hsa-miR-26a, hsa-miR-26b, hsa-miR-142-3p and hsa-let 7g decreased or increased after LPS treatment for 4 h. Further validation by qRT-PCR identified miR-26b, miR-142-3p and let-7g significantly changed in neonatal PMN after LPS stimulation. The functional verification by transfection of miR-142-3p and let-7g precursors into neonatal PMN significantly repressed the IL-6 mRNA and protein expression, suggesting that miR-142-3p and let-7g negatively regulate IL-6 expression in neonatal PMN. Modulation of miRNA expression may be used to regulate IL-6 production in newborns with altered inflammatory reactions.
Collapse
Affiliation(s)
- Hsin-Chun Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Kaohsiung, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Linkou, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Kaohsiung, Taiwan
| | - Te-Yao Hsu
- Department of Obstetrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Kaohsiung, Taiwan
| | - I-Lun Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Kaohsiung, Taiwan
| | - Jen-Chieh Chang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Kaohsiung, Taiwan
| | - Kuender D Yang
- Department of Pediatrics, Mackay Memorial Hospital, Tamshui, Taiwan.,Institute of Clinical Medical Sciences, National Yang Ming University, Taipei, Taiwan
| |
Collapse
|
121
|
Paton MCB, McDonald CA, Allison BJ, Fahey MC, Jenkin G, Miller SL. Perinatal Brain Injury As a Consequence of Preterm Birth and Intrauterine Inflammation: Designing Targeted Stem Cell Therapies. Front Neurosci 2017; 11:200. [PMID: 28442989 PMCID: PMC5385368 DOI: 10.3389/fnins.2017.00200] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/24/2017] [Indexed: 12/20/2022] Open
Abstract
Chorioamnionitis is a major cause of preterm birth and brain injury. Bacterial invasion of the chorion and amnion, and/or the placenta, can lead to a fetal inflammatory response, which in turn has significant adverse consequences for the developing fetal brain. Accordingly, there is a strong causal link between chorioamnionitis, preterm brain injury and the pathogenesis of severe postnatal neurological deficits and cerebral palsy. Currently there are no treatments to protect or repair against brain injury in preterm infants born after pregnancy compromised by intrauterine infection. This review describes the injurious cascade of events in the preterm brain in response to a severe fetal inflammatory event. We will highlight specific periods of increased vulnerability, and the potential effects of therapeutic intervention with cell-based therapies. Many clinical trials are underway to investigate the efficacy of stem cells to treat patients with cerebral palsy. Stem cells, obtained from umbilical cord tissue and cord blood, normally discarded after birth, are emerging as a safe and potentially effective therapy. It is not yet known, however, which stem cell type(s) are the most efficacious for administration to preterm infants to treat brain injury-mediated inflammation. Individual stem cell populations found in cord blood and tissue, such as mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs), have a number of potential benefits that may specifically target preterm inflammatory-induced brain injury. MSCs have strong immunomodulatory potential, protecting against global and local neuroinflammatory cascades triggered during infection to the fetus. EPCs have angiogenic and vascular reparative qualities that make them ideal for neurovascular repair. A combined therapy using both MSCs and EPCs to target inflammation and promote angiogenesis for re-establishment of vital vessel networks is a treatment concept that warrants further investigation.
Collapse
Affiliation(s)
- Madison C B Paton
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| | - Courtney A McDonald
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Beth J Allison
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Michael C Fahey
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Paediatrics, Monash UniversityClayton, VIC, Australia
| | - Graham Jenkin
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| | - Suzanne L Miller
- Neurodevelopment and Neuroprotection Research Group, The Ritchie Centre, Hudson Institute of Medical Research, Monash UniversityClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash Medical Centre, Monash UniversityClayton, VIC, Australia
| |
Collapse
|
122
|
Maternal Immunoreactivity to Herpes Simplex Virus 2 and Risk of Autism Spectrum Disorder in Male Offspring. mSphere 2017; 2:mSphere00016-17. [PMID: 28251181 PMCID: PMC5322345 DOI: 10.1128/msphere.00016-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/02/2017] [Indexed: 02/03/2023] Open
Abstract
The cause (or causes) of most cases of autism spectrum disorder is unknown. Evidence from epidemiological studies and work in animal models of neurodevelopmental disorders suggest that both genetic and environmental factors may be implicated. The latter include gestational infection and immune activation. In our cohort, high levels of antibodies to herpes simplex virus 2 at midpregnancy were associated with an elevated risk of autism spectrum disorder in male offspring. These findings provide support for the hypothesis that gestational infection may contribute to the pathogenesis of autism spectrum disorder and have the potential to drive new efforts to monitor women more closely for cryptic gestational infection and to implement suppressive therapy during pregnancy. Maternal infections during pregnancy are associated with risk of neurodevelopmental disorders, including autism spectrum disorders (ASDs). Proposed pathogenetic mechanisms include fetal infection, placental inflammation, and maternal cytokines or antibodies that cross the placenta. The Autism Birth Cohort comprises mothers, fathers, and offspring recruited in Norway in 1999 to 2008. Through questionnaire screening, referrals, and linkages to a national patient registry, 442 mothers of children with ASD were identified, and 464 frequency-matched controls were selected. Immunoglobulin G (IgG) antibodies to Toxoplasma gondii, rubella virus, cytomegalovirus (CMV), herpes simplex virus 1 (HSV-1), and HSV-2 in plasma collected at midpregnancy and after delivery were measured by multiplexed immunoassays. High levels of HSV-2 IgG antibodies in maternal midpregnancy plasma were associated with increased risk of ASD in male offspring (an increase in HSV-2 IgG levels from 240 to 640 arbitrary units/ml was associated with an odds ratio of 2.07; 95% confidence interval, 1.06 to 4.06; P = 0.03) when adjusted for parity and child’s birth year. No association was found between ASD and the presence of IgG antibodies to Toxoplasma gondii, rubella virus, CMV, or HSV-1. Additional studies are needed to test for replicability of risk and specificity of the sex effect and to examine risk associated with other infections. IMPORTANCE The cause (or causes) of most cases of autism spectrum disorder is unknown. Evidence from epidemiological studies and work in animal models of neurodevelopmental disorders suggest that both genetic and environmental factors may be implicated. The latter include gestational infection and immune activation. In our cohort, high levels of antibodies to herpes simplex virus 2 at midpregnancy were associated with an elevated risk of autism spectrum disorder in male offspring. These findings provide support for the hypothesis that gestational infection may contribute to the pathogenesis of autism spectrum disorder and have the potential to drive new efforts to monitor women more closely for cryptic gestational infection and to implement suppressive therapy during pregnancy.
Collapse
|
123
|
De Amici M, Perotti F, Marseglia G, Ierullo A, Bollani L, Decembrino L, Licari A, Quaglini S, Stronati M, Spinillo A. Cord and blood levels of newborn IgE: Correlation, role and influence of maternal IgE. Immunobiology 2017; 222:450-453. [DOI: 10.1016/j.imbio.2016.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 07/11/2016] [Accepted: 08/05/2016] [Indexed: 11/30/2022]
|
124
|
Ronovsky M, Berger S, Molz B, Berger A, Pollak DD. Animal Models of Maternal Immune Activation in Depression Research. Curr Neuropharmacol 2017; 14:688-704. [PMID: 26666733 PMCID: PMC5050397 DOI: 10.2174/1570159x14666151215095359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/24/2015] [Accepted: 11/09/2015] [Indexed: 01/17/2023] Open
Abstract
Abstract: Background Depression and schizophrenia are debilitating mental illnesses with significant socio-economic impact. The high degree of comorbidity between the two disorders, and shared symptoms and risk factors, suggest partly common pathogenic mechanisms. Supported by human and animal studies, maternal immune activation (MIA) has been intimately associated with the development of schizophrenia. However, the link between MIA and depression has remained less clear, in part due to the lack of appropriate animal models. Objective Here we aim to summarize findings obtained from studies using MIA animal models and discuss their relevance for preclinical depression research. Methods Results on molecular, cellular and behavioral phenotypes in MIA animal models were collected by literature search (PubMed) and evaluated for their significance for depression. Results Several reports on offspring depression-related behavioral alterations indicate an involvement of MIA in the development of depression later in life. Depression-related behavioral phenotypes were frequently paralleled by neurogenic and neurotrophic deficits and modulated by several genetic and environmental factors. Conclusion Literature evidence analyzed in this review supports a relevance of MIA as animal model for a specific early life adversity, which may prime an individual for the development of distinct psychopathologies later life. MIA animal models may present a unique tool for the identification of additional exogenous and endogenous factors, which are required for the manifestation of a specific neuropsychiatric disorder, such as depression, later in life. Hereby, novel insights into the molecular mechanisms involved in the pathophysiology of depression may be obtained, supporting the identification of alternative therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| |
Collapse
|
125
|
Patra A, Chen X, Sadowska GB, Zhang J, Lim YP, Padbury JF, Banks WA, Stonestreet BS. Neutralizing anti-interleukin-1β antibodies reduce ischemia-related interleukin-1β transport across the blood-brain barrier in fetal sheep. Neuroscience 2017; 346:113-125. [PMID: 28089577 DOI: 10.1016/j.neuroscience.2016.12.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
Hypoxic ischemic insults predispose to perinatal brain injury. Pro-inflammatory cytokines are important in the evolution of this injury. Interleukin-1β (IL-1β) is a key mediator of inflammatory responses and elevated IL-1β levels in brain correlate with adverse neurodevelopmental outcomes after brain injury. Impaired blood-brain barrier (BBB) function represents an important component of hypoxic-ischemic brain injury in the fetus. In addition, ischemia-reperfusion increases cytokine transport across the BBB of the ovine fetus. Reducing pro-inflammatory cytokine entry into brain could represent a novel approach to attenuate ischemia-related brain injury. We hypothesized that infusions of neutralizing IL-1β monoclonal antibody (mAb) reduce IL-1β transport across the BBB after ischemia in the fetus. Fetal sheep were studied 24-h after 30-min of carotid artery occlusion. Fetuses were treated with placebo- or anti-IL-1β mAb intravenously 15-min and 4-h after ischemia. Ovine IL-1β protein expressed from IL-1β pGEX-2T vectors in Escherichia coli (E. coli) BL-21 cells was produced, purified, and radiolabeled with 125I. BBB permeability was quantified using the blood-to-brain transfer constant (Ki) with 125I-radiolabeled-IL-1β. Increases in anti-IL-1β mAb were observed in the brain of the mAb-treated group (P<0.001). Blood-to-brain transport of 125I-IL-1β was lower (P<0.04) across brain regions in the anti-IL-1β mAb-treated than placebo-treated ischemic fetuses. Plasma 125I-IL-1β counts were higher (P<0.001) in the anti-IL-1β mAb- than placebo-treated ischemic fetuses. Systemic infusions of anti-IL-1β mAb reduce IL-1β transport across the BBB after ischemia in the ovine fetus. Our findings suggest that conditions associated with increases in systemic pro-inflammatory cytokines and neurodevelopmental impairment could benefit from an anti-cytokine therapeutic strategy.
Collapse
Affiliation(s)
- Aparna Patra
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States.
| | - Xiaodi Chen
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - Grazyna B Sadowska
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - Jiyong Zhang
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - Yow-Pin Lim
- ProThera Biologics, Providence, RI 02903, United States
| | - James F Padbury
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, United States
| | - Barbara S Stonestreet
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States.
| |
Collapse
|
126
|
Chen G, Sun W, Liang Y, Chen T, Guo W, Tian W. Maternal diabetes modulates offspring cell proliferation and apoptosis during odontogenesis via the TLR4/NF-κB signalling pathway. Cell Prolif 2016; 50. [PMID: 27981756 DOI: 10.1111/cpr.12324] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/05/2016] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES Maternal gestational diabetes leads to an adverse in utero environment and increases the risk of malformations during embryo organogenesis. In the present study, we analysed the effects of maternal diabetes on tooth germ cell proliferation and apoptosis in offspring, and investigated their underlying mechanisms. MATERIALS AND METHODS A rat model of maternal diabetes was induced by intraperitoneal injection of streptozotocin and the pregnant rats were divided into three groups: controls, the diabetic group and diabetic group with insulin treatment. Offspring of the three groups were collected and cell proliferation and apoptosis in tooth germs were analysed. Primary dental papilla cells and dental epithelial stem cells were isolated and treated with high glucose in vitro, in an attempt to simulate maternal diabetes-induced hyperglycaemia in vivo. RESULTS Maternal diabetes significantly affected cell proliferation and apoptosis in offspring tooth germs. The TLR4/NF-ĸB signalling pathway was activated in the tooth germs of offspring of diabetic dams. High glucose treatment activated the TLR4/NF-ĸB signalling pathway in primary dental papilla cells and dental epithelial stem cells in vitro, resulting in suppression of cell proliferation and enhancement of apoptosis. TLR4 knockdown significantly reduced adverse effects induced by high glucose treatment. CONCLUSIONS Maternal gestational diabetes significantly impaired dental epithelial and mesenchymal cell proliferation and apoptosis in offspring, possibly by activation of the TLR4/NF-ĸB signalling pathway.
Collapse
Affiliation(s)
- Guoqing Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Wenhua Sun
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Yan Liang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China College of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Tian Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China College of Stomatology, Sichuan University, Chengdu, China.,Department of Pedodontics, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China College of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
127
|
Faden M, Holm M, Allred E, Fichorova R, Dammann O, Leviton A. Antenatal glucocorticoids and neonatal inflammation-associated proteins. Cytokine 2016; 88:199-208. [PMID: 27668972 PMCID: PMC5067239 DOI: 10.1016/j.cyto.2016.09.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/02/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND To date, studies of the relationship between antenatal glucocorticoids (AGC) and neonatal inflammation in preterm newborns have been largely limited to umbilical cord blood specimens. AIM To explore the association between exposure to antenatal glucocorticoids and concentrations of inflammation-related proteins in whole blood collected from very preterm newborns at multiple times during the first postnatal month. METHODS We measured the protein concentrations on postnatal day 1 (N=1118), day 7 (N=1138), day 14 (N=1030), day 21 (N=936) and day 28 (N=877) from infants born before the 28th week of gestation and explored the relationship between antenatal steroid receipt and protein concentrations in the highest and lowest quartiles. The creation of multinomial logistic regression models (adjusted for potential confounders) allowed us calculate odds ratios and 95% confidence intervals. RESULTS Twenty of 420 assessments [21 (proteins)×2 (exposure levels: partial and full)×2 (quartile levels: top and bottom)×5 (days)] were statistically significant without any cohesive pattern. CONCLUSION Among infants born before 28 weeks of gestational age, neither full, nor partial courses of antenatal glucocorticoids have a sustained anti-inflammatory effect.
Collapse
Affiliation(s)
- Maheer Faden
- Division of Neonatology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada; Department of Newborn Medicine, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia.
| | - Mari Holm
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Elizabeth Allred
- Department of Neurology, Harvard Medical School, Boston, MA, United States; Neuroepidemiology Unit, Department of Neurology, Boston Children's Hospital, Boston, MA, United States
| | - Raina Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Olaf Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, United States; Perinatal Neuropidemiology Unit, Hannover Medical School, 30625 Hannover, Germany
| | - Alan Leviton
- Department of Neurology, Harvard Medical School, Boston, MA, United States; Neuroepidemiology Unit, Department of Neurology, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
128
|
Schmidt AF, Kannan PS, Chougnet CA, Danzer SC, Miller LA, Jobe AH, Kallapur SG. Intra-amniotic LPS causes acute neuroinflammation in preterm rhesus macaques. J Neuroinflammation 2016; 13:238. [PMID: 27596440 PMCID: PMC5011884 DOI: 10.1186/s12974-016-0706-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023] Open
Abstract
Background Chorioamnionitis is associated with an increased risk of brain injury in preterm neonates. Inflammatory changes in brain could underlie this injury. Here, we evaluated whether neuroinflammation is induced by chorioamnionitis in a clinically relevant model. Methods Rhesus macaque fetuses were exposed to either intra-amniotic (IA) saline, or IA lipopolysaccharide (LPS) (1 mg) 16 or 48 h prior to delivery at 130 days (85 % of gestation) (n = 4–5 animals/group). We measured cytokines in the cerebrospinal fluid (CSF), froze samples from the left brain for molecular analysis, and immersion fixed the right brain hemisphere for immunohistology. We analyzed the messenger RNA (mRNA) levels of the pro-inflammatory cytokines IL-1β, CCL2, TNF-α, IL-6, IL-8, IL-10, and COX-2 in the periventricular white matter (PVWM), cortex, thalamus, hippocampus, and cerebellum by RT-qPCR. Brain injury was assessed by immunohistology for myelin basic protein (MBP), IBA1 (microglial marker), GFAP (astrocyte marker), OLIG2 (oligodendrocyte marker), NeuN (neuronal marker), CD3 (T cells), and CD14 (monocytes). Microglial proliferation was assessed by co-immunostaining for IBA1 and Ki67. Data were analyzed by ANOVA with Tukey’s post-test. Results IA LPS increased mRNA expression of pro-inflammatory cytokines in the PVWM, thalamus, and cerebellum, increased IL-6 concentration in the CSF, and increased apoptosis in the periventricular area after 16 h. Microglial proliferation in the white matter was increased 48 h after IA LPS. Conclusions LPS-induced chorioamnionitis caused neuroinflammation, microglial proliferation, and periventricular apoptosis in a clinically relevant model of chorioamnionitis in fetal rhesus macaques. These findings identify specific responses in the fetal brain and support the hypothesis that neuroinflammatory changes may mediate the adverse neurodevelopmental outcomes associated with chorioamnionitis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0706-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Augusto F Schmidt
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Paranthaman S Kannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lisa A Miller
- California National Primate Research Center and Department of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - Alan H Jobe
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Suhas G Kallapur
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
129
|
Berger R, Garnier Y, Jensen A. Perinatal Brain Damage: Underlying Mechanisms and Neuroprotective Strategies. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760200900601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Richard Berger
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Universitätsfrauenklinik Bochum, Knappschaftskrankenhaus, In der Schornau 23-25, 44982 Bochum, Germany
| | | | - Arne Jensen
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany
| |
Collapse
|
130
|
Garnier Y, Coumans A, Berger R, Jensen A, Hasaart THM. Endotoxemia Severely Affects Circulation During Normoxia and Asphyxia in Immature Fetal Sheep. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760100800303] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - A. Coumans
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of Obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands
| | - R. Berger
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of Obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands; Universitätsfrauenklinik Bochum, Knappschaftskrankenhaus, In der Schornau 23 - 25, D-44892 Bochum, Germany
| | | | - T. H. M. Hasaart
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of Obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands
| |
Collapse
|
131
|
Levy O. Innate immunity of the human newborn: distinct cytokine responses to LPS and other Toll-like receptor agonists. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110020701] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Human newborns are at increased risk of microbial invasion and display diminished responses to many vaccines indicating a functional immaturity of the immune system at birth. Such altered immune reactivity may reflect the demands of in utero existence, including the need to avoid potentially harmful inflammatory immune reactions. Despite normal basal expression of Toll-like receptors and membrane CD14, innate immune responses of neonatal mononuclear cells to lipopolysaccharide are characterized by markedly reduced release of the pro-inflammatory Th1-polarizing cytokines TNF-α and interferon-γ with relative preservation of anti-inflammatory Th2-polarizing cytokines. Differences between newborns and adults with respect to TLR-induced TNF-α release extend to a range of TLR agonists, including bacterial lipopeptides, and are due to differences in soluble factors present in blood plasma. Soluble factors in neonatal blood plasma suppress TLR-induced TNF-α release from monocytes and efforts to identify and characterize these inhibitors are on-going. Such altered immunity to TLR agonists is likely to alter both innate and adaptive immune responses in newborns profoundly. Definition of the mechanisms underlying distinct neonatal immunity promises to identify novel ways to prevent and treat infection in this relatively high-risk population.
Collapse
Affiliation(s)
- Ofer Levy
- Division of Infectious Diseases, Children's Hospital, Harvard Medical School; Boston, Massachusetts USA,
| |
Collapse
|
132
|
Watkins CC, Andrews SR. Clinical studies of neuroinflammatory mechanisms in schizophrenia. Schizophr Res 2016; 176:14-22. [PMID: 26235751 DOI: 10.1016/j.schres.2015.07.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/08/2015] [Accepted: 07/08/2015] [Indexed: 12/27/2022]
Abstract
Schizophrenia is a pervasive neurodevelopmental disorder that appears to result from genetic and environmental factors. Although the dopamine hypothesis is the driving theory behind the majority of translation research in schizophrenia, emerging evidence suggests that aberrant immune mechanisms in the peripheral and central nervous system influence the etiology of schizophrenia and the pathophysiology of psychotic symptoms that define the illness. The initial interest in inflammatory processes comes from epidemiological data and historical observations, dating back several decades. A growing body of research on developmental exposure to infection, stress-induced inflammatory response, glial cell signaling, structural and functional brain changes and therapeutic trials demonstrates the impact that inflammation has on the onset and progression of schizophrenia. Research in animal models of psychosis has helped to advance clinical and basic science investigations of the immune mechanisms disrupted in schizophrenia. However, they are limited by the inability to recapitulate the human experience of hallucinations, delusions and thought disorder that define psychosis. To date, translational studies of inflammatory mechanisms in human subjects have not been reviewed in great detail. Here, we critically review clinical studies that focus on inflammatory mechanisms in schizophrenia. Understanding the neuroinflammatory mechanisms involved in schizophrenia may be essential in identifying potential therapeutic targets to minimize the morbidity and mortality of schizophrenia by interrupting disease development.
Collapse
Affiliation(s)
- Crystal C Watkins
- Memory Center in Neuropsychiatry, Sheppard Pratt Health Systems, Baltimore, MD, United States; Department of Psychiatry, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, United States.
| | - Sarah Ramsay Andrews
- Department of Psychiatry, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, United States
| |
Collapse
|
133
|
Wedekind L, Belkacemi L. Altered cytokine network in gestational diabetes mellitus affects maternal insulin and placental-fetal development. J Diabetes Complications 2016; 30:1393-400. [PMID: 27230834 DOI: 10.1016/j.jdiacomp.2016.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/16/2016] [Accepted: 05/09/2016] [Indexed: 12/15/2022]
Abstract
Pregnancy is characterized by an altered inflammatory profile, compared to the non-pregnant state with an adequate balance between pro-and anti-inflammatory cytokines needed for normal development. Cytokines are small secreted proteins expressed mainly in immunocompetent cells in the reproductive system. From early developmental stages onward, the secretory activity of placenta cells clearly contributes to increase local as well as systemic levels of cytokines. The placental production of cytokines may affect mother and fetus independently. In turn because of this unique position at the maternal fetal interface, the placenta is also exposed to the regulatory influence of cytokines from maternal and fetal circulations, and hence, may be affected by changes in any of these. Gestational diabetes mellitus (GDM) is associated with an overall alteration of the cytokine network. This review discusses the changes that occur in cytokines post GDM and their negative effects on maternal insulin and placental-fetal development.
Collapse
Affiliation(s)
- Lauren Wedekind
- Stanford University, Program in Human Biology, Stanford, CA, 94305, USA
| | - Louiza Belkacemi
- University of Houston, Departments of Biology and Biochemistry, Houston, TX, 77204, USA.
| |
Collapse
|
134
|
Feldhaus B, Dietzel ID, Heumann R, Berger R. Effects of Interferon-γ and Tumor Necrosis Factor-α on Survival and Differentiation of Oligodendrocyte Progenitors. ACTA ACUST UNITED AC 2016; 11:89-96. [PMID: 14980310 DOI: 10.1016/j.jsgi.2003.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE There is strong evidence from recent clinical studies that ascending intrauterine infection is associated with an increased incidence of periventricular leukomalacia in very premature fetuses. Periventricular leukomalacia is characterized by disrupted myelination from a loss of oligodendrocyte progenitors. We investigated the effects of proinflammatory cytokines on the survival and differentiation of this cell type. METHODS Cultures of more than 90% A2B5-positive progenitors were prepared from neonatal rats and kept for 3 days in medium supplemented with factors that stimulate cell proliferation. After 1 day in proliferation medium, cells were treated with interferon-gamma (100 U/mL) and tumor necrosis factor-alpha (100 ng/mL) for 48 hours triggering an increase in apoptotic A2B5 progenitor cells from 3.2 +/- 2.3% to 11.0 +/- 2.6%. After cytokine treatment cultures were transferred to medium containing factors to promote differentiation of progenitors into the myelinating phenotype. RESULTS In cytokine pretreated cultures, only 2.6 +/- 1.1% of total cells survived after a total of 9 days in vitro, whereas in untreated cultures most cells differentiated as shown by expression of myelin basic protein, myelin-associated glycoprotein, 2,3-cyclic nucleotide 3-phosphodiesterase, and myelin oligodendrocyte-specific protein. Using ten-fold reduced concentrations of combined interferon-gamma (10 U/mL) and tumor necrosis factor-alpha (10 ng/mL) pretreatment resulted in a survival to 11.2 +/- 4.9% of total cells with 36.3 +/- 11.6% A2B5-positive cells at day 9. This indicates a major enrichment of undifferentiated cells compared with untreated controls which harbored only 1.0 +/- 0.3% A2B5-positive cells. CONCLUSION Inflammatory cytokines not only induced apoptotic cell death but also prevented the differentiation of immature A2B5 oligodendrocyte progenitors into the myelinating phenotype.
Collapse
Affiliation(s)
- Beatrix Feldhaus
- Departments ofDepartment of Obstetrics and Gynecology, University of Bochum, Bochum, Germany
| | | | | | | |
Collapse
|
135
|
Nitsos I, Rees SM, Duncan J, Kramer BW, Harding R, Newnham JP, Moss TJM. Chronic Exposure to Intra-Amniotic Lipopolysaccharide Affects the Ovine Fetal Brain. ACTA ACUST UNITED AC 2016; 13:239-47. [PMID: 16697939 DOI: 10.1016/j.jsgi.2006.02.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Fetal brain injury is associated with chorioamnionitis, which is often present without signs of overt infection or fetal compromise. We aimed to determine if prolonged exposure to intrauterine inflammation caused by intra-amniotic infusion of lipopolysaccharide (LPS) would affect the fetal brain. METHODS At 80 days of pregnancy ewes bearing singletons had osmotic pumps implanted intra-amniotically to infuse Escherichia coli LPS (055:B5; n = 8) or saline (n = 7) for 28 days. At delivery (110 days), umbilical arterial blood and chorioamnion were assessed for inflammation; cytokine concentrations (interleukin [IL]-6 and IL-8) in amniotic fluid and fetal and maternal plasma were measured. The fetal cerebral hemispheres were examined for gross anatomical changes and the number of activated microglia/macrophages, astrocytes, and oligodendrocytes estimated after immunohistochemical staining. RESULTS Intra-amniotic administration of LPS caused chorioamnionitis, fetal leucocytosis, and a moderate to extensive infiltration of activated microglia/macrophages in the subcortical white matter in six of eight fetuses; the remaining two fetuses were less affected. Within these focal regions of damage there was an attenuation of astrocytic processes, axonal injury, and a reduction in the number of 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) immunoreactive oligodendrocytes in areas of extensive focal damage. In control fetuses there was mild (3/7) or no infiltration of activated microglia/macrophages in the subcortical white matter. Overall the infiltration of activated microglia/macrophages in the white matter was significantly greater in LPS-exposed fetuses compared to controls. In regions devoid of injury, the number of oligodendrocytes and astrocytes was not different between groups, nor was there a difference in the volume of cerebral white matter or density of blood vessels within the white matter. Amniotic fluid IL-6 and IL-8, and maternal plasma IL-8 concentrations were significantly increased by LPS infusion. CONCLUSIONS An increase in inflammatory cells and axonal disruption in the subcortical white matter of the fetal brain can accompany chorioamnionitis induced by intra-amniotic administration of LPS, but cystic lesions do not occur. Thus, the effect on the fetal brain is milder than that reported from animal models of acute fetal/intrauterine infection.
Collapse
Affiliation(s)
- Ilias Nitsos
- School of Women's and Infants' Health, The University of Western Australia, Crawley, Western Australia, Australia.
| | | | | | | | | | | | | |
Collapse
|
136
|
Garnier Y, Coumans ABC, Jensen A, Hasaart THM, Berger R. Infection-Related Perinatal Brain Injury: The Pathogenic Role of Impaired Fetal Cardiovascular Control. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/s1071-55760300150-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | - Tom H. M. Hasaart
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands
| | - Richard Berger
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany; Department of obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands; Universitätsfrauenklinik Bochum, Knappschaftskrankenhaus, In der Schornau 23-25, 44982 Bochum
| |
Collapse
|
137
|
Coumans ABC, Garnier Y, Supçun S, Jensen A, Berger R, Hasaart THM. The Effects of Low-Dose Endotoxin on the Umbilicoplacental Circulation in Preterm Sheep. ACTA ACUST UNITED AC 2016; 11:289-93. [PMID: 15219882 DOI: 10.1016/j.jsgi.2003.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE In the present study we examined the effects of low-dose endotoxin (lipopolysaccharides, LPS) on continuously recorded umbilical blood flow. METHODS Twenty fetal sheep were catheterized at a gestational age of 107 +/- 1 days. A flow probe was placed around either the common umbilical artery or one single umbilical artery. Three days later fetuses received either 100 or 500 nanograms of LPS (n = 14) or 2 mL saline (n = 6) intravenously. Six fetuses died within 12 hours after LPS. Fetal heart rate (FHR), mean arterial pressure (MAP), and umbilical blood flow (Q(umb)) were monitored for 3 days. RESULTS FHR increased by 25 +/- 4% at 4-5 hours after LPS (P <.01) and was elevated for 15 hours after LPS. MAP increased by 18 +/- 5% 1 hour after LPS (P <.01) and returned to control value 4-5 hours after LPS. Q(umb) began to decrease 1 hour after LPS and was minimal (-30 +/- 7%, P <.001) at 4-5 hours after LPS. Q(umb) slowly returned to the control value at 12 hours after LPS. Placental vascular resistance increased by 73 +/- 37% (P <.01), whereas pH did not appreciably change. CONCLUSION Intravenous application of endotoxin caused a substantial and long-lasting decrease in umbilical blood flow resulting in fetal hypoxemia without acidemia. These effects may be of significance in the development of fetal brain damage associated with intrauterine infection.
Collapse
Affiliation(s)
- A B C Coumans
- Department of Obstetrics and Gynecology, University of Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
138
|
Lu HY, Zhang Q, Wang QX, Lu JY. Contribution of Histologic Chorioamnionitis and Fetal Inflammatory Response Syndrome to Increased Risk of Brain Injury in Infants With Preterm Premature Rupture of Membranes. Pediatr Neurol 2016; 61:94-98.e1. [PMID: 27353694 DOI: 10.1016/j.pediatrneurol.2016.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND To determine the association of histologic chorioamnionitis (HCA) and fetal inflammatory response syndrome (FIRS) with brain injuries in infants born to mothers with preterm premature rupture of membranes. METHODS A total of 103 singleton infants born to mothers with preterm premature rupture of membranes were enrolled. The placental inflammation was confirmed by HCA, and FIRS was defined in fetuses with preterm labor and an elevation of the fetal plasma interleukin-6 concentration. Examination of brain images was conducted to confirm the existence of brain injuries. Based on placental HCA and umbilical cord blood interleukin-6 level, all patients were divided into three groups: HCA(-)FIRS(+), HCA(+)FIRS(-), and HCA(+)FIRS(+). RESULTS Among all infants with preterm premature rupture of membranes, 53.40% were exposed to HCA, 20.38% experienced FIRS, and the overall incidence of brain injuries was 38.83%. The incidence of brain injury in HCA(-)FIRS(+), HCA(+)FIRS(-), and HCA(+)FIRS(+) groups were 20.83%, 41.18%, and 76.19%, respectively. HCA at the advanced grades and stages was associated with increased risk of brain injury. Umbilical cord blood levels of interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor alpha (TNF-α), and granulocyte-colony stimulating factor (G-CSF) in premature infants with brain injuries were significantly higher than in those without brain injuries. Infants diagnosed with both HCA and FIRS showed significantly higher levels of IL-8, TNF-α, and G-CSF than those with HCA alone. CONCLUSIONS Preterm infants exposed to severe chorioamnionitis had an increased risk of brain injury. IL-6, IL-8, TNF-α, and G-CSF in cord blood were associated with brain injuries in preterm infants and may be used as extradiagnostic criteria.
Collapse
Affiliation(s)
- Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Qiang Zhang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiu-Xia Wang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jun-Ying Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
139
|
Graham EM, Burd I, Everett AD, Northington FJ. Blood Biomarkers for Evaluation of Perinatal Encephalopathy. Front Pharmacol 2016; 7:196. [PMID: 27468268 PMCID: PMC4942457 DOI: 10.3389/fphar.2016.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022] Open
Abstract
Recent research in identification of brain injury after trauma shows many possible blood biomarkers that may help identify the fetus and neonate with encephalopathy. Traumatic brain injury shares many common features with perinatal hypoxic-ischemic encephalopathy. Trauma has a hypoxic component, and one of the 1st physiologic consequences of moderate-severe traumatic brain injury is apnea. Trauma and hypoxia-ischemia initiate an excitotoxic cascade and free radical injury followed by the inflammatory cascade, producing injury in neurons, glial cells and white matter. Increased excitatory amino acids, lipid peroxidation products, and alteration in microRNAs and inflammatory markers are common to both traumatic brain injury and perinatal encephalopathy. The blood-brain barrier is disrupted in both leading to egress of substances normally only found in the central nervous system. Brain exosomes may represent ideal biomarker containers, as RNA and protein transported within the vesicles are protected from enzymatic degradation. Evaluation of fetal or neonatal brain derived exosomes that cross the blood-brain barrier and circulate peripherally has been referred to as the "liquid brain biopsy." A multiplex of serum biomarkers could improve upon the current imprecise methods of identifying fetal and neonatal brain injury such as fetal heart rate abnormalities, meconium, cord gases at delivery, and Apgar scores. Quantitative biomarker measurements of perinatal brain injury and recovery could lead to operative delivery only in the presence of significant fetal risk, triage to appropriate therapy after birth and measure the effectiveness of treatment.
Collapse
Affiliation(s)
- Ernest M. Graham
- Division of Maternal-Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Irina Burd
- Division of Maternal-Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Allen D. Everett
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Division of Cardiology, Department of Pediatrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Frances J. Northington
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
140
|
Zhang S, Wang Y, Li D, Wu J, Si W, Wu Y. Necrostatin-1 Attenuates Inflammatory Response and Improves Cognitive Function in Chronic Ischemic Stroke Mice. MEDICINES (BASEL, SWITZERLAND) 2016; 3:E16. [PMID: 28930126 PMCID: PMC5456247 DOI: 10.3390/medicines3030016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/18/2016] [Accepted: 06/21/2016] [Indexed: 12/27/2022]
Abstract
Multiple cell death is involved in ischemic brain injury. Necroptosis, a recently reported cell death, may be the most suitable cell death mechanism in a subpopulation of neurons under ischemic injury. It reported that a small molecule, necrostatin-1 (Nec-1), has a potent inhibitory effect on necroptotic cell death in vivo and in vitro. The aim of the current study was to investigate the role of Nec-1 on cognitive function in chronic ischemic stroke mice induced by bilateral common carotid artery stenosis (BCAS). Here, 12-week-old C57BL/6 mice received intragastric administration with Nec-1 or vehicle for two weeks after stroke, and then, the effect and possible mechanism were determined. We demonstrated that inhibition of necroptosis prevented cognitive impairment and reduced inflammatory response in the ischemic brain injury mouse model. These data suggested that inhibition of necroptosis provided a potential therapeutic option for cognitive rehabilitation in chronic ischemic stroke.
Collapse
Affiliation(s)
- Shehong Zhang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yuyang Wang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Dake Li
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Junfa Wu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Wen Si
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yi Wu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
141
|
Parker SE, Lijewski VA, Janulewicz PA, Collett BR, Speltz ML, Werler MM. Upper respiratory infection during pregnancy and neurodevelopmental outcomes among offspring. Neurotoxicol Teratol 2016; 57:54-59. [PMID: 27343815 PMCID: PMC5056812 DOI: 10.1016/j.ntt.2016.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/02/2016] [Accepted: 06/21/2016] [Indexed: 11/21/2022]
Abstract
Objective Maternal infection during pregnancy is associated with psychiatric disorders among offspring. The aim of this study was to investigate associations between upper respiratory infection (URI) in pregnancy and measures of cognitive and behavioral outcomes in child offspring. Materials and methods A longitudinal study of 534 mother-child pairs with information regarding prenatal exposures collected through an interview conducted on average one year after delivery and subsequent participation in a childhood cognitive and psychosocial assessment between the ages 5–12 years. Childhood cognition was measured using the Peabody Picture Vocabulary Test (PPVT-III) and the Beery-Buktenica Test of Visual Motor Integration-Fifth Edition (VMI-5) and behavioral function measured using the Child Behavior Checklist (CBCL) and teacher-report using the Teacher Report Form (TRF). Adjusted mean differences (adjMD) in outcome measures were calculated between mothers reporting the presence or absence of a URI during pregnancy. Results URI during pregnancy was not associated with the two measures of cognition given to offspring, but was associated with modest increases in total behavioral problems reported by mothers (adjMD: 3.72; CI: 1.91–5.54) and teachers (adjMD: 2.74; CI: 0.97–4.50). We observed differences in CBCL and TRF scores based on timing of URI: infections in mid-pregnancy (lunar months 4–5) were associated with poorer scores than were infections in early pregnancy (lunar months 2–3). Conclusions In general, URI in pregnancy was not associated with decrements in childhood cognition, but may be associated with behavior problems. URI in early pregnancy was not associated with childhood cognitive outcomes. URI in pregnancy was associated with increases in behavior problems among offspring. Associations with behavior were varied by the timing and duration of reported URI. Both treated and untreated URI were associated with increases in behavior problems.
Collapse
Affiliation(s)
- Samantha E Parker
- Department of Epidemiology, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, United States.
| | - Virginia A Lijewski
- Department of Epidemiology, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, United States
| | - Patricia A Janulewicz
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, United States
| | - Brent R Collett
- Department of Psychiatry and Behavioral Sciences, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, United States
| | - Matthew L Speltz
- Department of Psychiatry and Behavioral Sciences, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, United States
| | - Martha M Werler
- Department of Epidemiology, Boston University School of Public Health, 715 Albany Street, Boston, MA 02118, United States
| |
Collapse
|
142
|
Preterm white matter brain injury is prevented by early administration of umbilical cord blood cells. Exp Neurol 2016; 283:179-87. [PMID: 27317990 DOI: 10.1016/j.expneurol.2016.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 12/29/2022]
Abstract
Infants born very preterm are at high risk for neurological deficits including cerebral palsy. In this study we assessed the neuroprotective effects of umbilical cord blood cells (UCBCs) and optimal administration timing in a fetal sheep model of preterm brain injury. 50 million allogeneic UCBCs were intravenously administered to fetal sheep (0.7 gestation) at 12h or 5d after acute hypoxia-ischemia (HI) induced by umbilical cord occlusion. The fetal brains were collected at 10d after HI. HI (n=7) was associated with reduced number of oligodendrocytes (Olig2+) and myelin density (CNPase+), and increased density of activated microglia (Iba-1+) in cerebral white matter compared to control fetuses (P<0.05). UCBCs administered at 12h, but not 5d after HI, significantly protected white matter structures and suppressed cerebral inflammation. Activated microglial density showed a correlation with decreasing oligodendrocyte number (P<0.001). HI caused cell death (TUNEL+) in the internal capsule and cell proliferation (Ki-67+) in the subventricular zone compared to control (P<0.05), while UCBCs at 12h or 5d ameliorated these effects. Additionally, UCBCs at 12h induced a significant systemic increase in interleukin-10 at 10d, and reduced oxidative stress (malondialdehyde) following HI (P<0.05). UCBC administration at 12h after HI reduces preterm white matter injury, via anti-inflammatory and antioxidant actions.
Collapse
|
143
|
Rodríguez-Trujillo A, Cobo T, Vives I, Bosch J, Kacerovsky M, Posadas DE, Ángeles MA, Gratacós E, Jacobsson B, Palacio M. Gestational age is more important for short-term neonatal outcome than microbial invasion of the amniotic cavity or intra-amniotic inflammation in preterm prelabor rupture of membranes. Acta Obstet Gynecol Scand 2016; 95:926-33. [PMID: 27061307 DOI: 10.1111/aogs.12905] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 03/30/2016] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The aim of this study was to evaluate, in women with preterm prelabor rupture of membranes (PPROM), the impact on short-term neonatal outcome of microbial invasion of the amniotic cavity (MIAC), intra-amniotic inflammation (IAI), and the microorganisms isolated in women with MIAC, when gestational age is taken into account. MATERIAL AND METHODS Prospective cohort study. We included women with PPROM (22.0-34.0 weeks of gestation) with available information about MIAC, IAI and short-term neonatal outcome. MIAC was defined as positive aerobic/anaerobic/genital Mycoplasma culture in amniotic fluid. Definition of IAI was based on interleukin-6 levels in amniotic fluid. Main outcome measures were Apgar score <7 at 5 min, umbilical artery pH ≤7.0, days in the neonatal intensive care unit, and composite neonatal morbidity, including any of the following: intraventricular hemorrhage grade III-IV, respiratory distress syndrome, early-onset neonatal sepsis, periventricular leukomalacia, necrotizing enterocolitis, and fetal or neonatal death. Labor was induced after 32.0 weeks if lung maturity was confirmed; and otherwise after 34.0 weeks. RESULTS MIAC and IAI were found in 38% (72/190) and 67% (111/165), respectively. After adjustment for gestational age at delivery, no differences in short-term neonatal outcome were found between women with either MIAC or IAI, compared with the non-infection/non-inflammation ("No-MIAC/No-IAI") group. Furthermore, short-term neonatal outcome did not differ between the MIAC caused by Ureaplasma spp. group, the MIAC caused by other microorganisms group and the "No-MIAC/No-IAI" group. CONCLUSIONS Gestational age at delivery seems to be more important for short-term neonatal outcome than MIAC or IAI in PPROM.
Collapse
Affiliation(s)
- Adriano Rodríguez-Trujillo
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Teresa Cobo
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Irene Vives
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Jordi Bosch
- Microbiology, Biomedical Diagnostic Center, Hospital Clínic and ISGlobal Barcelona Institute for Global Health, University of Barcelona, Barcelona, Spain
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - David E Posadas
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Martina A Ángeles
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Eduard Gratacós
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Genetics and Bioinformatics, Area of Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Montse Palacio
- BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, Fetal i+D Fetal Medicine Research Center, IDIBAPS, University of Barcelona, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
144
|
Rumajogee P, Bregman T, Miller SP, Yager JY, Fehlings MG. Rodent Hypoxia-Ischemia Models for Cerebral Palsy Research: A Systematic Review. Front Neurol 2016; 7:57. [PMID: 27199883 PMCID: PMC4843764 DOI: 10.3389/fneur.2016.00057] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 04/03/2016] [Indexed: 12/28/2022] Open
Abstract
Cerebral palsy (CP) is a complex multifactorial disorder, affecting approximately 2.5-3/1000 live term births, and up to 22/1000 prematurely born babies. CP results from injury to the developing brain incurred before, during, or after birth. The most common form of this condition, spastic CP, is primarily associated with injury to the cerebral cortex and subcortical white matter as well as the deep gray matter. The major etiological factors of spastic CP are hypoxia/ischemia (HI), occurring during the last third of pregnancy and around birth age. In addition, inflammation has been found to be an important factor contributing to brain injury, especially in term infants. Other factors, including genetics, are gaining importance. The classic Rice-Vannucci HI model (in which 7-day-old rat pups undergo unilateral ligation of the common carotid artery followed by exposure to 8% oxygen hypoxic air) is a model of neonatal stroke that has greatly contributed to CP research. In this model, brain damage resembles that observed in severe CP cases. This model, and its numerous adaptations, allows one to finely tune the injury parameters to mimic, and therefore study, many of the pathophysiological processes and conditions observed in human patients. Investigators can recreate the HI and inflammation, which cause brain damage and subsequent motor and cognitive deficits. This model further enables the examination of potential approaches to achieve neural repair and regeneration. In the present review, we compare and discuss the advantages, limitations, and the translational value for CP research of HI models of perinatal brain injury.
Collapse
Affiliation(s)
- Prakasham Rumajogee
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Tatiana Bregman
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Steven P Miller
- Department of Pediatrics, Hospital for Sick Children , Toronto, ON , Canada
| | - Jerome Y Yager
- Division of Pediatric Neurosciences, Stollery Children's Hospital, University of Alberta , Edmonton, AB , Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada; Division of Neurosurgery, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
145
|
Preventive effects of minocycline in a neurodevelopmental two-hit model with relevance to schizophrenia. Transl Psychiatry 2016; 6:e772. [PMID: 27045842 PMCID: PMC4872396 DOI: 10.1038/tp.2016.38] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/23/2015] [Accepted: 02/22/2016] [Indexed: 12/25/2022] Open
Abstract
Maternal immune activation can increase the vulnerability of the offspring to develop neuroimmune and behavioral abnormalities in response to stress in puberty. In offspring of immune-challenged mothers, stress-induced inflammatory processes precede the adult onset of multiple behavioral dysfunctions. Here, we explored whether an early anti-inflammatory intervention during peripubertal stress exposure might prevent the subsequent emergence of adult behavioral pathology. We used an environmental two-hit model in mice, in which prenatal maternal administration of the viral mimetic poly(I:C) served as the first hit, and exposure to sub-chronic unpredictable stress during peripubertal maturation as the second hit. Using this model, we examined the effectiveness of the tetracycline antibiotic minocycline (MINO) given during stress exposure to block stress-induced inflammatory responses and to prevent subsequent behavioral abnormalities. We found that combined exposure to prenatal immune activation and peripubertal stress caused significant deficits in prepulse inhibition and increased sensitivity to the psychotomimetic drugs amphetamine and dizocilpine in adulthood. MINO treatment during stress exposure prevented the emergence of these behavioral dysfunctions. In addition, the pharmacological intervention blocked hippocampal and prefrontal microglia activation and interleukin-1β expression in offspring exposed to prenatal infection and peripubertal stress. Together, these findings demonstrate that presymptomatic MINO treatment can prevent the subsequent emergence of multiple behavioral abnormalities relevant to human neuropsychiatric disorders with onset in early adulthood, including schizophrenia. Our epidemiologically informed two-hit model may thus encourage attempts to explore the use of anti-inflammatory agents in the early course of brain disorders that are characterized by signs of central nervous system inflammation during development.
Collapse
|
146
|
Xie D, Shen F, He S, Chen M, Han Q, Fang M, Zeng H, Chen C, Deng Y. IL-1β induces hypomyelination in the periventricular white matter through inhibition of oligodendrocyte progenitor cell maturation via FYN/MEK/ERK signaling pathway in septic neonatal rats. Glia 2016; 64:583-602. [PMID: 26678483 DOI: 10.1002/glia.22950] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 11/08/2015] [Accepted: 11/18/2015] [Indexed: 02/05/2023]
Abstract
Neuroinflammation elicited by microglia plays a key role in periventricular white matter (PWM) damage (PWMD) induced by infectious exposure. This study aimed to determine if microglia-derived interleukin-1β (IL-1β) would induce hypomyelination through suppression of maturation of oligodendrocyte progenitor cells (OPCs) in the developing PWM. Sprague-Dawley rats (1-day old) were injected with lipopolysaccharide (LPS) (1 mg/kg) intraperitoneally, following which upregulated expression of IL-1β and IL-1 receptor 1 (IL-1R1 ) was observed. This was coupled with enhanced apoptosis and suppressed proliferation of OPCs in the PWM. The number of PDGFR-α and NG2-positive OPCs was significantly decreased in the PWM at 24 h and 3 days after injection of LPS, whereas it was increased at 14 days and 28 days. The protein expression of Olig1, Olig2, and Nkx2.2 was significantly reduced, and mRNA expression of Tcf4 and Axin2 was upregulated in the developing PWM after LPS injection. The expression of myelin basic protein (MBP) and 2',3'-cyclic-nucleotide 3"-phosphodiesterase (CNPase) was downregulated in the PWM at 14 days and 28 days after LPS injection; this was linked to reduction of the proportion of myelinated axons and thinner myelin sheath as revealed by electron microscopy. Primary cultured OPCs treated with IL-1β showed the failure of maturation and proliferation. Furthermore, FYN/MEK/ERK signaling pathway was involved in suppression of maturation of primary OPCs induced by IL-1β administration. Our results suggest that following LPS injection, microglia are activated and produce IL-1β in the PWM in the neonatal rats. Excess IL-1β inhibits the maturation of OPCs via suppression of FYN/MEK/ERK phosphorylation thereby leading to axonal hypomyelination.
Collapse
Affiliation(s)
- Di Xie
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Fengcai Shen
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Shantou University Medical College (FCS), Shantou, Guangdong, People's Republic of China. 515063
| | - Shaoru He
- Department of Neonatology, Guangdong General Hospital, Guangzhou, People's Republic of China
| | - Mengmeng Chen
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Shantou University Medical College (FCS), Shantou, Guangdong, People's Republic of China. 515063
| | - Qianpeng Han
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Ming Fang
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Hongke Zeng
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Chunbo Chen
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yiyu Deng
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| |
Collapse
|
147
|
Marangoni C, Hernandez M, Faedda GL. The role of environmental exposures as risk factors for bipolar disorder: A systematic review of longitudinal studies. J Affect Disord 2016; 193:165-74. [PMID: 26773919 DOI: 10.1016/j.jad.2015.12.055] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/19/2015] [Accepted: 12/26/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND The role of environmental risk factors in the development of bipolar disorder (BD) is not well characterized. We evaluate the prevalence, duration, and predictive value of environmental exposures for BD in longitudinal studies. METHODS We conducted a systematic search of PubMed, Scopus and PsychINFO databases until April 01, 2015, using the following words in combination: prenatal exposure; maternal exposure; trauma; childhood abuse; alcoholism; cannabis; smoking; cocaine; central stimulants; opioids; uv light; pollution; global warming; vitamin d AND bipolar disorder. Additional references were obtained through cross-referencing. We included (1) longitudinal cohort studies or case-control studies nested within longitudinal designs; (2) studies of subjects without lifetime BD diagnoses at initial assessment and a diagnosis of BD at follow-up by clinical or structured assessment. Familial-risk studies were excluded. We tabulated details of study-design, exposure, diagnostic criteria, risk of bipolar disorder expressed as odd ratio (OR), relative risk (RR) or hazard ratio (HR). RESULTS Of 2119 studies found, 22 met inclusion criteria. Risk factors identified can be grouped in 3 clusters: neurodevelopment (maternal influenza during pregnancy; indicators of fetal development), substances (cannabis, cocaine, other drugs - opioids, tranquilizers, stimulants, sedatives), physical/psychological stress (parental loss, adversities, abuses, brain injury). LIMITATIONS Heterogeneity of designs and methodology prevented the use of meta-analysis of the findings; studies did not provide sensitivity, specificity and predictive value of the risk factors identified; case-control studies classify cases based on diagnostic membership, but do not control for familial or genetic liability; methods for determining the exposures varied among studies. CONCLUSION Only preliminary evidence exists that exposure to viral infection, substances or trauma increase the likelihood of BD. Given the limited data available, the specificity, sensitivity and predictive value could not be computed. As exposures are sometimes amenable to prevention, further research is needed.
Collapse
|
148
|
Kurahashi H, Okumura A, Kubota T, Kidokoro H, Maruyama K, Hayakawa M, Itakura A, Matsuzawa K, Yamamoto H, Kato T, Hayakawa F, Watanabe K. Increased fetal heart rate variability in periventricular leukomalacia. Brain Dev 2016; 38:196-203. [PMID: 26338690 DOI: 10.1016/j.braindev.2015.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/15/2015] [Accepted: 08/24/2015] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This study used quantitative analysis to determine whether increased variability in fetal heart rate (FHR) is related to the risk of developing periventricular leukomalacia (PVL). METHODS We analyzed 124 FHR traces of neonates delivered preterm at 27-33 weeks' gestation to 105 mothers. FHR traces 1-3h before delivery were translated into power-spectrum curves using a fast Fourier transformation. The total power (the area under the curve of 1-10 cycles per minute), segmental power of every cycle per minute, peak power, and frequency edges were calculated, and their relationship with the subsequent development of PVL was examined. RESULTS Total power was significantly higher in the PVL group (n=9, median 1813, range 1064-2426) compared to the non-PVL group (n=114, median 1383, range 381-3324, p=0.029). Infants in the PVL group had greater segmental power in segments with 1-2, 2-3, and 9-10 cycles per minute, than those in the non-PVL group. Total power of ⩾1550 was significantly correlated with the subsequent development of PVL and premature rupture of membranes. Furthermore, the frequency of pregnancy-induced hypertension was significantly reduced in the fetuses with a total power of ⩾1550. CONCLUSION Our study suggests that a fetus with increased FHR variability is at risk of developing PVL. This study provides additional evidence supporting the contribution of antenatal factors to the subsequent development of PVL.
Collapse
Affiliation(s)
- Hirokazu Kurahashi
- Department of Pediatrics, Aichi Medical University, Nagakute, Aichi, Japan; Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihisa Okumura
- Department of Pediatrics, Aichi Medical University, Nagakute, Aichi, Japan; Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan.
| | - Tetsuo Kubota
- Department of Pediatrics, Anjo Kosei Hospital, Anjo, Aichi, Japan
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Pediatrics, Anjo Kosei Hospital, Anjo, Aichi, Japan
| | - Koichi Maruyama
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Hayakawa
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Maternity and Perinatal Care Center, Nagoya University Hospital, Nagoya, Japan
| | - Atsuo Itakura
- Maternity and Perinatal Care Center, Nagoya University Hospital, Nagoya, Japan; Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Katsuji Matsuzawa
- Department of Gynecology and Obstetrics, Anjo Kosei Hospital, Anjo, Aichi, Japan
| | - Hiroyuki Yamamoto
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toru Kato
- Department of Pediatrics, Okazaki City Hospital, Okazaki, Aichi, Japan
| | - Fumio Hayakawa
- Department of Pediatrics, Okazaki City Hospital, Okazaki, Aichi, Japan
| | | |
Collapse
|
149
|
Sciaky-Tamir Y, Hershkovitz R, Mazor M, Shelef I, Erez O. The use of imaging technology in the assessment of the fetal inflammatory response syndrome-imaging of the fetal thymus. Prenat Diagn 2016; 35:413-9. [PMID: 25601186 DOI: 10.1002/pd.4560] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 12/07/2014] [Accepted: 01/09/2015] [Indexed: 12/28/2022]
Abstract
The fetal inflammatory response syndrome (FIRS) describes a state of extensive fetal multi organ involvement during chorioamnionitis, and is associated with grave implications on perinatal outcome. The syndrome has been linked to the preterm parturition syndrome and is associated with inflammation/infection processes in most of the fetal organs. The fetal thymus, a major organ in the developing immune system involutes during severe neonatal disease and has been shown to be smaller in fetuses with FIRS. Various methods for imaging of the fetal thymus and measurement are described. Currently the only method to diagnose FIRS prenatally is through amniocentesis. We suggest that women who are admitted with preterm labor with intact membranes and those with PPROM should have a detailed sonographic examination of the fetal thymus as a surrogate marker of fetal involvement in intrauterine infection/inflammation processes.
Collapse
Affiliation(s)
- Yael Sciaky-Tamir
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, School of Medicine, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
150
|
Abstract
Cerebral palsy is the most common cause of childhood-onset, lifelong physical disability in most countries, affecting about 1 in 500 neonates with an estimated prevalence of 17 million people worldwide. Cerebral palsy is not a disease entity in the traditional sense but a clinical description of children who share features of a non-progressive brain injury or lesion acquired during the antenatal, perinatal or early postnatal period. The clinical manifestations of cerebral palsy vary greatly in the type of movement disorder, the degree of functional ability and limitation and the affected parts of the body. There is currently no cure, but progress is being made in both the prevention and the amelioration of the brain injury. For example, administration of magnesium sulfate during premature labour and cooling of high-risk infants can reduce the rate and severity of cerebral palsy. Although the disorder affects individuals throughout their lifetime, most cerebral palsy research efforts and management strategies currently focus on the needs of children. Clinical management of children with cerebral palsy is directed towards maximizing function and participation in activities and minimizing the effects of the factors that can make the condition worse, such as epilepsy, feeding challenges, hip dislocation and scoliosis. These management strategies include enhancing neurological function during early development; managing medical co-morbidities, weakness and hypertonia; using rehabilitation technologies to enhance motor function; and preventing secondary musculoskeletal problems. Meeting the needs of people with cerebral palsy in resource-poor settings is particularly challenging.
Collapse
|