151
|
van der Burg JW, Sen S, Chomitz VR, Seidell JC, Leviton A, Dammann O. The role of systemic inflammation linking maternal BMI to neurodevelopment in children. Pediatr Res 2016; 79:3-12. [PMID: 26375474 PMCID: PMC4888781 DOI: 10.1038/pr.2015.179] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/01/2015] [Indexed: 11/09/2022]
Abstract
Children of obese mothers are at increased risk of developmental adversities. Maternal obesity is linked to an inflammatory in utero environment, which, in turn, is associated with neurodevelopmental impairments in the offspring. This is an integrated mechanism review of animal and human literature related to the hypothesis that maternal obesity causes maternal and fetal inflammation, and that this inflammation adversely affects the neurodevelopment of children. We propose integrative models in which several aspects of inflammation are considered along the causative pathway linking maternal obesity with neurodevelopmental limitations.
Collapse
Affiliation(s)
- Jelske W. van der Burg
- Department of Health and Life Sciences, Faculty of Earth and Life Sciences, VU University Amsterdam, Amsterdam, The Netherlands
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Sarbattama Sen
- Department of Pediatrics and Mother Infant Research Institute, Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, USA
- Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Virginia R. Chomitz
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Jaap C. Seidell
- Department of Health and Life Sciences, Faculty of Earth and Life Sciences, VU University Amsterdam, Amsterdam, The Netherlands
| | - Alan Leviton
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Olaf Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Perinatal Epidemiology Unit, Hannover Medical School, Hannover, Germany
| |
Collapse
|
152
|
Abstract
Preterm newborns are highly susceptible to brain injury. White matter injury is among the dominant patterns of brain injury in preterm newborns. the purpose of this review is to discuss the pathogenesis, diagnosis, management, and prevention of white matter injury in premature newborns. the long-term outcome of white matter injury in children born prematurely is also addressed.
Collapse
Affiliation(s)
- Dawn Gano
- UCSF Benioff Children's Hospital, 550 16th Street, Box 0137, San Francisco, CA 94158, USA
| |
Collapse
|
153
|
Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, Kusanovic JP, Dong Z, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L, Kim YM, Kim YM. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med 2016; 44:53-76. [PMID: 26360486 PMCID: PMC5625297 DOI: 10.1515/jpm-2015-0121] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/02/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Microbial invasion of the fetus due to intra-amniotic infection can lead to a systemic inflammatory response characterized by elevated concentrations of cytokines in the umbilical cord plasma/serum. Clinical chorioamnionitis represents the maternal syndrome often associated with intra-amniotic infection, although other causes of this syndrome have been recently described. The objective of this study was to characterize the umbilical cord plasma cytokine profile in neonates born to mothers with clinical chorioamnionitis at term, according to the presence or absence of bacteria and/or intra-amniotic inflammation. MATERIALS AND METHODS A cross-sectional study was conducted, including patients with clinical chorioamnionitis at term (n=38; cases) and those with spontaneous term labor without clinical chorioamnionitis (n=77; controls). Women with clinical chorioamnionitis were classified according to the results of amniotic fluid culture, broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry (PCR/ESI-MS) and amniotic fluid interleukin (IL)-6 concentration into three groups: 1) no intra-amniotic inflammation; 2) intra-amniotic inflammation without detectable microorganisms; or 3) microbial-associated intra-amniotic inflammation. A fetal inflammatory response syndrome (FIRS) was defined as an umbilical cord plasma IL-6 concentration >11 pg/mL. The umbilical cord plasma concentrations of 29 cytokines were determined with sensitive and specific V-PLEX immunoassays. Nonparametric statistical methods were used for analysis, adjusting for a false discovery rate of 5%. RESULTS 1) Neonates born to mothers with clinical chorioamnionitis at term (considered in toto) had significantly higher median umbilical cord plasma concentrations of IL-6, IL-12p70, IL-16, IL-13, IL-4, IL-10 and IL-8, but significantly lower interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF)-α concentrations than neonates born to mothers with spontaneous term labor without clinical chorioamnionitis; 2) neonates born to mothers with clinical chorioamnionitis at term but without intra-amniotic inflammation had higher concentrations of IL-6, IL-12p70, IL-13, IL-4, IL-5, and IL-8, but lower IFN-γ, than neonates not exposed to clinical chorioamnionitis, suggesting that maternal fever in the absence of intra-amniotic inflammation leads to a change in the fetal cytokine network; 3) there were significant, positive correlations between maternal and umbilical cord plasma IL-6 and IL-8 concentrations (IL-6: Spearman correlation=0.53; P<0.001; IL-8: Spearman correlation=0.42; P<0.001), consistent with placental transfer of cytokines; 4) an elevated fetal plasma IL-6 (>11 pg/mL), the diagnostic criterion for FIRS, was present in 21% of cases (8/38), and all these neonates were born to mothers with proven intra-amniotic infection; and 5) FIRS was associated with a high concentration of umbilical cord plasma IL-8, IL-10 and monocyte chemoattractant protein (MCP)-1. CONCLUSIONS Neonates born to mothers with clinical chorioamnionitis at term had higher concentrations of umbilical cord plasma cytokines than those born to mothers without clinical chorioamnionitis. Even neonates exposed to clinical chorioamnionitis but not to intra-amniotic inflammation had elevated concentrations of multiple cytokines, suggesting that intrapartum fever alters the fetal immune response.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA,Department of Molecular Obstetrics and Genetics, Wayne State University, Detroit, MI, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikolina Docheva
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhonghui Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Juan P. Kusanovic
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Center for Research and Innovation in Maternal-Fetal Medicine (CIMAF). Department of Obstetrics and Gynecology, Sótero del Río Hospital, Santiago, Chile,Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Zhong Dong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yeon Mee Kim
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan Korea
| | | |
Collapse
|
154
|
Jin SJ, Liu Y, Deng SH, Lin TL, Rashid A, Liao LH, Ning Q, Luo XP. Protective effects of activated protein C on neurovascular unit in a rat model of intrauterine infection-induced neonatal white matter injury. ACTA ACUST UNITED AC 2015; 35:904-909. [PMID: 26670444 DOI: 10.1007/s11596-015-1526-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 10/14/2015] [Indexed: 02/08/2023]
Abstract
Activated protein C (APC), a natural anticoagulant, has been reported to exert direct vasculoprotective, neural protective, anti-inflammatory, and proneurogenic activities in the central nervous system. This study was aimed to explore the neuroprotective effects and potential mechanisms of APC on the neurovascular unit of neonatal rats with intrauterine infection-induced white matter injury. Intraperitoneal injection of 300 μg/kg lipopolysaccharide (LPS) was administered consecutively to pregnant Sprague-Dawley rats at embryonic days 19 and 20 to establish the rat model of intrauterine infection- induced white matter injury. Control rats were injected with an equivalent amount of sterile saline on the same time. APC at the dosage of 0.2 mg/kg was intraperitoneally injected to neonatal rats immediately after birth. Brain tissues were collected at postnatal day 7 and stained with hematoxylin and eosin (H&E). Immunohistochemistry was used to evaluate myelin basic protein (MBP) expression in the periventricular white matter region. Blood-brain barrier (BBB) permeability and brain water content were measured using Evens Blue dye and wet/dry weight method. Double immunofluorescence staining and real-time quantitative PCR were performed to detect microglial activation and the expression of protease activated receptor 1 (PAR1). Typical pathological changes of white matter injury were observed in rat brains exposed to LPS, and MBP expression in the periventricular region was significantly decreased. BBB was disrupted and the brain water content was increased. Microglia were largely activated and the mRNA and protein levels of PAR1 were elevated. APC administration ameliorated the pathological lesions of the white matter and increased MBP expression. BBB permeability and brain water content were reduced. Microglia activation was inhibited and the PAR1 mRNA and protein expression levels were both down-regulated. Our results suggested that APC exerted neuroprotective effects on multiple components of the neurovascular unit in neonatal rats with intrauterine infection- induced white matter injury, and the underlying mechanisms might involve decreased expression of PAR1.
Collapse
Affiliation(s)
- Sheng-Juan Jin
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Liu
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shi-Hua Deng
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tu-Lian Lin
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Abid Rashid
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Hong Liao
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Ping Luo
- Department of Pediatrics, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
155
|
Nadeau-Vallée M, Obari D, Quiniou C, Lubell WD, Olson DM, Girard S, Chemtob S. A critical role of interleukin-1 in preterm labor. Cytokine Growth Factor Rev 2015; 28:37-51. [PMID: 26684042 DOI: 10.1016/j.cytogfr.2015.11.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/24/2015] [Accepted: 11/03/2015] [Indexed: 12/16/2022]
Abstract
Preterm birth (PTB) is a leading cause of neonatal mortality and morbidity worldwide, and represents a heavy economic and social burden. Despite its broad etiology, PTB has been firmly linked to inflammatory processes. Pro-inflammatory cytokines are produced in gestational tissues in response to stressors and can prematurely induce uterine activation, which precedes the onset of preterm labor. Of all cytokines implicated, interleukin (IL)-1 has been largely studied, revealing a central role in preterm labor. However, currently approved IL-1-targeting therapies have failed to show expected efficacy in pre-clinical studies of preterm labor. Herein, we (a) summarize animal and human studies in which IL-1 or IL-1-targeting therapeutics are implicated with preterm labor, (b) focus on novel IL-1-targeting therapies and diagnostic tests, and (c) develop the case for commercialization and translation means to hasten their development.
Collapse
Affiliation(s)
- Mathieu Nadeau-Vallée
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montréal H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montréal H3C 3J7, Canada
| | - Dima Obari
- Department of Pharmacology, Université de Montréal, Montréal H3C 3J7, Canada
| | - Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montréal H3T 1C5, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton AB TG6 2S2, Canada
| | - Sylvie Girard
- Departments of Obstetrics and Gynecology, CHU Sainte-Justine Research Centre, Montréal H3T 1C5, Canada.
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montréal H3T 1C5, Canada.
| |
Collapse
|
156
|
Association Between Prenatal Exposure to Maternal Infection and Offspring Mood Disorders: A Review of the Literature. Curr Probl Pediatr Adolesc Health Care 2015; 45:325-64. [PMID: 26476880 DOI: 10.1016/j.cppeds.2015.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/14/2015] [Indexed: 01/19/2023]
Abstract
The purpose of this article is to provide a systematic review of studies that have examined the association between prenatal exposure to maternal infection and development of mood disorders across the life course. Drawing from both human- and animal-based studies, we give an overview of hypothesized biological mechanisms by which exposure to maternal infection during critical periods of gestation may contribute to fetal programming of mood disorders in offspring. We discuss studies examining the association between prenatal exposure to maternal infection with pathogens including influenza as well as other respiratory viruses, herpesviruses, hepatitis viruses, and Toxoplasma gondii and mood disorders in human populations. Moreover, we outline strengths and limitations of the current body of evidence and make recommendations for future research. We also discuss findings in the context of well-documented gender and socioeconomic disparities in the prevalence and severity of mood disorders, particularly major depression, and the role that early exposure to infection may play in explaining the perpetuation of such disparities across generations. Overall, this review of the current knowledge on this topic has important implications for determining future research directions, designing interventions as well as prenatal care guidelines targeted at prevention or treatment of infection during pregnancy, and clinical practice for the identification of individuals that may be at increased risk for mood disorders beginning early in life. Importantly, such efforts may not only lower the overall burden of mood disorders but also serve to address social disparities in these adverse mental health conditions in the U.S.
Collapse
|
157
|
Comim CM, Bussmann RM, Simão SR, Ventura L, Freiberger V, Patrício JJ, Palmas D, Mendonça BP, Cassol-Jr OJ, Quevedo J. Experimental Neonatal Sepsis Causes Long-Term Cognitive Impairment. Mol Neurobiol 2015; 53:5928-5934. [DOI: 10.1007/s12035-015-9495-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 10/16/2015] [Indexed: 10/22/2022]
|
158
|
Maxwell JR, Denson JL, Joste NE, Robinson S, Jantzie LL. Combined in utero hypoxia-ischemia and lipopolysaccharide administration in rats induces chorioamnionitis and a fetal inflammatory response syndrome. Placenta 2015; 36:1378-84. [PMID: 26601766 DOI: 10.1016/j.placenta.2015.10.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/16/2015] [Accepted: 10/12/2015] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Preterm birth is a major cause of infant morbidity and long-term disability, and is associated with numerous central nervous system (CNS) deficits. Infants exposed to intrauterine inflammation, specifically chorioamnionitis, are at risk for very early preterm birth and neurological complications including cerebral palsy, epilepsy, and behavioral and cognitive deficits. However, placenta-brain axis abnormalities and their relationship to subsequent permanent CNS injury remain poorly defined. METHODS Intrauterine injury was induced in rats on embryonic day 18 (E18) by transient systemic hypoxia-ischemia (TSHI) and intra-amniotic lipopolysaccharide (LPS) injection. Placenta, brain and serum were collected from E19 to postnatal day 0 (P0). Histology, TUNEL staining, western blot and multiplex immunoassays were used to quantify placental and brain abnormalities, and fetal serum cytokine levels. RESULTS Prenatal TSHI + LPS caused acute and subacute placental injury hallmarked by inflammatory infiltrate, edema, hemorrhage and cell death along with placental increases in IL-1β and TNFα. TSHI + LPS increased a diverse array of circulating inflammatory proteins including IL-1β, TNFα, IL-6, IL-10, IL-4, IFNγ and CXCL1, both immediately after TSHI + LPS and in live born pups. CNS inflammation was characterized by increased CXCL1. DISCUSSION Prenatal TSHI + LPS in rats induces placental injury and inflammation histologically consistent with chorioamnionitis, concomitant with elevated serum and CNS pro-inflammatory cytokines. This model accurately recapitulates key pathophysiological processes observed in extremely preterm infants including placental, fetal, and CNS inflammation. Further investigation into the mechanism of CNS injury following chorioamnionitis and the placental-brain axis will guide the use of future interventions.
Collapse
Affiliation(s)
- Jessie R Maxwell
- Departments of Pediatrics and Neurosciences, University of New Mexico, Albuquerque, NM, USA
| | - Jesse L Denson
- Departments of Pediatrics and Neurosciences, University of New Mexico, Albuquerque, NM, USA
| | - Nancy E Joste
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Shenandoah Robinson
- Departments of Neurosurgery and Neurology, Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston MA, USA
| | - Lauren L Jantzie
- Departments of Pediatrics and Neurosciences, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
159
|
Ferraz MM, Sab IM, Silva MA, Santos DA, Ferraz MR. Prenatal Hypoxia Ischemia Increases Male Rat Sexual Behavior. J Sex Med 2015; 12:2013-21. [DOI: 10.1111/jsm.13006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
160
|
Cho GJ, Hong HR, Hong SC, Oh MJ, Kim HJ. The neuroprotective effect of magnesium sulfate in preterm fetal mice. J Perinat Med 2015; 43:537-43. [PMID: 25503462 DOI: 10.1515/jpm-2014-0176] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 11/10/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To investigate the use of magnesium sulfate (MgSO4) as a neuroprotective agent in a mouse model of inflammation-associated and noninflammation-associated preterm birth. METHODS On embryonic day 15 of gestation, lipopolysaccharide (LPS) and mifepristone (RU486) were used, respectively, to create mouse models of inflammation and noninflammation-associated preterm birth. After intraperitoneal injection of LPS, RU486, or normal saline solution (NS), dams were randomized to intraperitoneal MgSO4 or NS injection. From the 6 treatment groups (NS+NS, LPS+NS, NS+MgSO4, LPS+MgSO4, RU486+NS and RU486+MgSO4), fetal brains were collected for Western blot analysis and neuronal cultures. Protein expression of S100B was assessed, and immunohistochemistry was performed to detect NeuN. The numbers of NeuN-labeled cells were counted using confocal laser scanning microscopy. RESULTS The expression of S100B significantly differed among the groups and was decreased in the LPS+MgSO4 group compared to the LPS+NS group. The expression of S100B did not differ between the RU486+NS and RU486+MgSO4 groups. NeuN-labeled cells were increased in the LPS+MgSO4 group compared with the LPS+NS group. NeuN-labeled cells were decreased in the RU486+MgSO4 group compared with the RU486+NS group. CONCLUSIONS We observed that prenatal treatment with MgSO4 was associated with decreased expression of S100B and increased numbers of NeuN-labeled cells in an inflammation-associated preterm mouse model but not in a noninflammation-associated preterm mouse model. Our results suggest that prenatal treatment of MgSO4 reduces inflammation-associated brain injury in fetal mice.
Collapse
|
161
|
Interleukin-1β transfer across the blood-brain barrier in the ovine fetus. J Cereb Blood Flow Metab 2015; 35:1388-95. [PMID: 26082012 PMCID: PMC4640327 DOI: 10.1038/jcbfm.2015.134] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 11/09/2022]
Abstract
Pro-inflammatory cytokines contribute to hypoxic-ischemic brain injury. Blood-brain barrier (BBB) dysfunction represents an important component of hypoxic-ischemic brain injury in the fetus. Hypoxic-ischemic injury could accentuate systemic cytokine transfer across the fetal BBB. There has been considerable conjecture suggesting that systemic cytokines could cross the BBB during the perinatal period. Nonetheless, evidence to support this contention is sparse. We hypothesized that ischemia-reperfusion increases the transfer of systemic interleukin-1β (IL-1β) across the BBB in the fetus. Ovine fetuses at 127 days of gestation were studied 4 hours after 30 minutes of bilateral carotid artery occlusion and compared with a nonischemic group. Recombinant ovine IL-1β protein was expressed from an IL-1β pGEX-2 T vector in E. coli BL-21 cells and purified. The BBB function was quantified in 12 brain regions using a blood-to-brain transfer constant with intravenous (125)I-radiolabeled IL-1β ((125)I-IL-1β). Interleukin-1β crossed the intact BBB in nonischemic fetuses. Blood-to-brain transport of (125)I-IL-1β was higher (P<0.05) across brain regions in fetuses exposed to ischemia-reperfusion than nonischemic fetuses. We conclude that systemic IL-1β crosses the intact fetal BBB, and that ischemia-reperfusion increases transfer of this cytokine across the fetal BBB. Therefore, altered BBB function after hypoxia-ischemia facilitates entry of systemic cytokines into the brain of the fetus.
Collapse
|
162
|
A question of cytokine signaling in the fetus. J Cereb Blood Flow Metab 2015; 35:1387. [PMID: 26082011 PMCID: PMC4640328 DOI: 10.1038/jcbfm.2015.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/18/2015] [Indexed: 11/09/2022]
Abstract
The study by Sadowska and colleagues, featured in this edition of Journal of Cerebral Blood Flow and Metabolism, provides compelling insight into a critical mechanism of signaling that links somatic and brain inflammatory pathways. Brain inflammation is often treated as an isolated event, but the work by Sadowska et al clearly demonstrate that interleukin 1β crosses the fetal blood brain barrier, and that the rate of transfer increases after cerebral ischemia. It is, perhaps, time to link blood-borne cytokine signaling with alterations in the course of fetal brain development, especially after transient fetal hypoxia.
Collapse
|
163
|
Ahmad AS, Satriotomo I, Fazal J, Nadeau SE, Doré S. Considerations for the Optimization of Induced White Matter Injury Preclinical Models. Front Neurol 2015; 6:172. [PMID: 26322013 PMCID: PMC4532913 DOI: 10.3389/fneur.2015.00172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
White matter (WM) injury in relation to acute neurologic conditions, especially stroke, has remained obscure until recently. Current advances in imaging technologies in the field of stroke have confirmed that WM injury plays an important role in the prognosis of stroke and suggest that WM protection is essential for functional recovery and post-stroke rehabilitation. However, due to the lack of a reproducible animal model of WM injury, the pathophysiology and mechanisms of this injury are not well studied. Moreover, producing selective WM injury in animals, especially in rodents, has proven to be challenging. Problems associated with inducing selective WM ischemic injury in the rodent derive from differences in the architecture of the brain, most particularly, the ratio of WM to gray matter in rodents compared to humans, the agents used to induce the injury, and the location of the injury. Aging, gender differences, and comorbidities further add to this complexity. This review provides a brief account of the techniques commonly used to induce general WM injury in animal models (stroke and non-stroke related) and highlights relevance, optimization issues, and translational potentials associated with this particular form of injury.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Irawan Satriotomo
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Jawad Fazal
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Stephen E Nadeau
- Research Service, Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA ; Research Service, Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA ; Department of Neuroscience, University of Florida , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA ; Department of Pharmaceutics, University of Florida , Gainesville, FL , USA ; Department of Psychology, University of Florida , Gainesville, FL , USA ; Department of Psychiatry, University of Florida , Gainesville, FL , USA
| |
Collapse
|
164
|
Migale R, Herbert BR, Lee YS, Sykes L, Waddington SN, Peebles D, Hagberg H, Johnson MR, Bennett PR, MacIntyre DA. Specific Lipopolysaccharide Serotypes Induce Differential Maternal and Neonatal Inflammatory Responses in a Murine Model of Preterm Labor. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [PMID: 26212908 DOI: 10.1016/j.ajpath.2015.05.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Intrauterine inflammation is recognized as a key mediator of both normal and preterm birth but is also associated with neonatal neurological injury. Lipopolysaccharide (LPS) is often used to stimulate inflammatory pathways in animal models of infection/inflammation-induced preterm labor; however, inconsistencies in maternal and neonatal responses to LPS are frequently reported. We hypothesized that LPS serotype-specific responses may account for a portion of these inconsistencies. Four different Escherichia coli LPS serotypes (O111:B4, O55:B5, O127:B8, and O128:B12) were administered to CD1 mice via intrauterine injection at gestational day 16. Although control animals delivered at term 60 ± 15 hours postinjection (p.i.), those administered with O111:B4 delivered 7 ± 2 hours p.i., O55:B5 delivered 10 ± 3 hours p.i., O127:B8 delivered 16 ± 10 hours p.i., and O128:B12 delivered 17 ± 2 hours p.i. (means ± SD). A correlation between the onset of preterm labor and myometrial activation of the inflammatory transcription factor, activator protein 1, but not NF-κB was observed. Specific LPS serotypes induced differential activation of downstream contractile and inflammatory pathways in myometrium and neonatal pup brain. Our findings demonstrate functional disparity in inflammatory pathway activation in response to differing LPS serotypes. Selective use of LPS serotypes may represent a useful tool for targeting specific inflammatory response mechanisms in these models.
Collapse
Affiliation(s)
- Roberta Migale
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Bronwen R Herbert
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom; Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Yun S Lee
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Lynne Sykes
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, United Kingdom; Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witswatersrand, Johannesburg, South Africa
| | - Donald Peebles
- UCL Centre for Perinatal Brain Protection & Repair, Institute for Women's Health, University College London, London, United Kingdom
| | - Henrik Hagberg
- Department of Clinical Sciences, Perinatal Center, University of Gothenburg, Gothenburg, Sweden; Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Mark R Johnson
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom; Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Phillip R Bennett
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - David A MacIntyre
- Imperial College Parturition Research Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom.
| |
Collapse
|
165
|
Peri‐intraventricular hemorrhage and oxidative and inflammatory stress markers in very‐low birth weight newborns. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2015. [DOI: 10.1016/j.jpedp.2015.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
166
|
Labouesse MA, Langhans W, Meyer U. Long-term pathological consequences of prenatal infection: beyond brain disorders. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1-R12. [DOI: 10.1152/ajpregu.00087.2015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/21/2015] [Indexed: 12/24/2022]
Abstract
Prenatal immunological adversities such as maternal infection have been widely acknowledged to contribute to an increased risk of neurodevelopmental brain disorders. In recent years, epidemiological and experimental evidence has accumulated to suggest that prenatal exposure to immune challenges can also negatively affect various physiological and metabolic functions beyond those typically associated with primary defects in CNS development. These peripheral changes include excessive accumulation of adipose tissue and increased body weight, impaired glycemic regulation and insulin resistance, altered myeloid lineage development, increased gut permeability, hyperpurinergia, and changes in microbiota composition. Experimental work in animal models further suggests that at least some of these peripheral abnormalities could directly contribute to CNS dysfunctions, so that normalization of peripheral pathologies could lead to an amelioration of behavioral deficits. Hence, seemingly unrelated central and peripheral effects of prenatal infection could represent interrelated pathological entities that emerge in response to a common developmental stressor. Targeting peripheral abnormalities may thus represent a valuable strategy to improve the wide spectrum of behavioral abnormalities that can emerge in subjects with prenatal infection histories.
Collapse
Affiliation(s)
| | | | - Urs Meyer
- Physiology and Behavior Laboratory, ETH Zurich, Switzerland
| |
Collapse
|
167
|
Caldas JPS, Braghini CA, Mazzola TN, Vilela MMS, Marba STM. Peri-intraventricular hemorrhage and oxidative and inflammatory stress markers in very-low birth weight newborns. J Pediatr (Rio J) 2015; 91:373-9. [PMID: 25913045 DOI: 10.1016/j.jped.2014.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/21/2014] [Accepted: 09/22/2014] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES To evaluate the association between oxidative and inflammatory stress markers with peri-intraventricular hemorrhage (PIVH) in very-low birth weight newborns. METHODS This was a prospective study conducted in a level III neonatal unit. Basal and stimulated reactive oxygen intermediates (ROIs), reduced glutathione (GSH), and interleukin-6 (IL-6) levels were measured in umbilical cord blood. Newborns underwent serial ultrasound at the bedside, at 6, 12, 24, and 72hours of life and at seven days for the diagnosis of PIVH, classified as grades I to IV. Two groups were assessed, those with and without PIVH; maternal and neonatal control variables were used for comparison. Univariate and multiple regression analyses were applied. RESULTS A total of 125 newborns were assessed. PIVH incidence rate was 12.0%. In the univariate analysis, basal ROI, the use of two or more doses of corticosteroids, birth weight<1,000g, ventilatory support use, and SNAPPE II value ≥ 22 were significantly associated with PIVH. However, in the multivariate analysis, only antenatal steroid use was independently associated with the disease (OR 0,194; 95% CI: 0,048 to 0,773; p=0,02). CONCLUSION ROI, GSH, and IL-6 levels were not associated with the occurrence of PIVH in very-low birth weight infants.
Collapse
Affiliation(s)
- Jamil P S Caldas
- Neonatology Division, Hospital da Mulher Prof. Dr. José Aristodemo Pinotti, Centro de Atenção Integral à Saúde da Mulher (CAISM), Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Carolina A Braghini
- Centro de Investigação em Pediatria (CIPED), Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Taís N Mazzola
- Centro de Investigação em Pediatria (CIPED), Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Maria M S Vilela
- Department of Pediatrics, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Sérgio T M Marba
- Department of Pediatrics, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
168
|
Stoner TD, Weston TA, Trejo J, Doran KS. Group B streptococcal infection and activation of human astrocytes. PLoS One 2015; 10:e0128431. [PMID: 26030618 PMCID: PMC4452173 DOI: 10.1371/journal.pone.0128431] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/27/2015] [Indexed: 01/08/2023] Open
Abstract
Background Streptococcus agalactiae (Group B Streptococcus, GBS) is the leading cause of life-threatening meningitis in human newborns in industrialized countries. Meningitis results from neonatal infection that occurs when GBS leaves the bloodstream (bacteremia), crosses the blood-brain barrier (BBB), and enters the central nervous system (CNS), where the bacteria contact the meninges. Although GBS is known to invade the BBB, subsequent interaction with astrocytes that physically associate with brain endothelium has not been well studied. Methodology/Principal Findings We hypothesize that human astrocytes play a unique role in GBS infection and contribute to the development of meningitis. To address this, we used a well- characterized human fetal astrocyte cell line, SVG-A, and examined GBS infection in vitro. We observed that all GBS strains of representative clinically dominant serotypes (Ia, Ib, III, and V) were able to adhere to and invade astrocytes. Cellular invasion was dependent on host actin cytoskeleton rearrangements, and was specific to GBS as Streptococcus gordonii failed to enter astrocytes. Analysis of isogenic mutant GBS strains deficient in various cell surface organelles showed that anchored LTA, serine-rich repeat protein (Srr1) and fibronectin binding (SfbA) proteins all contribute to host cell internalization. Wild-type GBS also displayed an ability to persist and survive within an intracellular compartment for at least 12 h following invasion. Moreover, GBS infection resulted in increased astrocyte transcription of interleukin (IL)-1β, IL-6 and VEGF. Conclusions/Significance This study has further characterized the interaction of GBS with human astrocytes, and has identified the importance of specific virulence factors in these interactions. Understanding the role of astrocytes during GBS infection will provide important information regarding BBB disruption and the development of neonatal meningitis.
Collapse
Affiliation(s)
- Terri D. Stoner
- Department of Biology, Center for Microbial Sciences, San Diego State University, San Diego, CA, United States of America
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Thomas A. Weston
- Department of Biology, Center for Microbial Sciences, San Diego State University, San Diego, CA, United States of America
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Kelly S. Doran
- Department of Biology, Center for Microbial Sciences, San Diego State University, San Diego, CA, United States of America
- Departmant of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
- * E-mail:
| |
Collapse
|
169
|
Silva DD, Lopez P, Mantovani JC. Auditory brainstem response in term and preterm infants with neonatal complications: the importance of the sequential evaluation. Int Arch Otorhinolaryngol 2015; 19:161-5. [PMID: 25992173 PMCID: PMC4399179 DOI: 10.1055/s-0034-1378137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/26/2014] [Indexed: 11/24/2022] Open
Abstract
Introduction Literature data are not conclusive as to the influence of neonatal complications in the maturational process of the auditory system observed by auditory brainstem response (ABR) in infants at term and preterm. Objectives Check the real influence of the neonatal complications in infants by the sequential auditory evaluation. Methods Historical cohort study in a tertiary referral center. A total of 114 neonates met inclusion criteria: treatment at the Universal Neonatal Hearing Screening Program of the local hospital; at least one risk indicator for hearing loss; presence in both evaluations (the first one after hospital discharge from the neonatal unit and the second one at 6 months old); all latencies in ABR and transient otoacoustic emissions present in both ears. Results The complications that most influenced the ABR findings were Apgar scores less than 6 at 5 minutes, gestational age, intensive care unit stay, peri-intraventricular hemorrhage, and mechanical ventilation. Conclusion Sequential auditory evaluation is necessary in premature and term newborns with risk indicators for hearing loss to correctly identify injuries in the auditory pathway.
Collapse
Affiliation(s)
- Daniela da Silva
- Ophthalmology/ENT and Head and Neck Surgery Department, Faculdade de Medicina de Botucatu, Botucatu, São Paulo, Brazil
| | - Priscila Lopez
- Ophthalmology/ENT and Head and Neck Surgery Department, Faculdade de Medicina de Botucatu, Botucatu, São Paulo, Brazil
| | - Jair Cortez Mantovani
- Ophthalmology/ENT and Head and Neck Surgery Department, Faculdade de Medicina de Botucatu, UNESP, Botucatu, Brazil
| |
Collapse
|
170
|
Abstract
Molecular profiling studies have helped increase the understanding of the immune processes thought to be involved in the etiology and pathophysiology of psychiatric disorders such as schizophrenia. Current therapeutic interventions with first- and second-generation antipsychotics are suboptimal. Poor response rates and debilitating side effects often lead to poor treatment compliance. This highlights the pressing need to identify more effective treatments as well as objective biomarker based tests, which can help predict treatment response and identify diagnostic subpopulations. Such tests could enable early detection of patients who will benefit from particular therapeutic interventions. In this review, we discuss studies relating to dysfunctions of the immune system in patients with schizophrenia and the effects of antipsychotic medication on the molecular components of these systems. Immune system dysfunction may in part be related to genetic risk factors for schizophrenia, but there is substantial evidence that a wide range of environmental factors ranging from exposure to infectious agents such as influenza and Toxoplasma gondii to HPA axis dysfunction play an important role in the etiopathogenesis of schizophrenia. Ongoing research efforts, testing therapeutic efficacy of anti-inflammatory agents used as add-on medications are also discussed. From a therapeutic perspective, these represent the initial steps toward novel treatment approaches and more effective patient care in the field of mental health.
Collapse
|
171
|
Hagberg H, Mallard C, Ferriero DM, Vannucci SJ, Levison SW, Vexler ZS, Gressens P. The role of inflammation in perinatal brain injury. Nat Rev Neurol 2015; 11:192-208. [PMID: 25686754 PMCID: PMC4664161 DOI: 10.1038/nrneurol.2015.13] [Citation(s) in RCA: 597] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Inflammation is increasingly recognized as being a critical contributor to both normal development and injury outcome in the immature brain. The focus of this Review is to highlight important differences in innate and adaptive immunity in immature versus adult brain, which support the notion that the consequences of inflammation will be entirely different depending on context and stage of CNS development. Perinatal brain injury can result from neonatal encephalopathy and perinatal arterial ischaemic stroke, usually at term, but also in preterm infants. Inflammation occurs before, during and after brain injury at term, and modulates vulnerability to and development of brain injury. Preterm birth, on the other hand, is often a result of exposure to inflammation at a very early developmental phase, which affects the brain not only during fetal life, but also over a protracted period of postnatal life in a neonatal intensive care setting, influencing critical phases of myelination and cortical plasticity. Neuroinflammation during the perinatal period can increase the risk of neurological and neuropsychiatric disease throughout childhood and adulthood, and is, therefore, of concern to the broader group of physicians who care for these individuals.
Collapse
Affiliation(s)
- Henrik Hagberg
- 1] Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St Thomas' Hospital, London SE1 7EH, UK. [2] Perinatal Center, Institute of Physiology and Neurosciences and Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 435 43 Gothenburg, Sweden
| | - Carina Mallard
- Perinatal Center, Institute of Physiology and Neurosciences and Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 435 43 Gothenburg, Sweden
| | - Donna M Ferriero
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Susan J Vannucci
- Department of Pediatrics/Newborn Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Steven W Levison
- Department of Neurology and Neuroscience, Rutgers University, RBHS-New Jersey Medical School, Cancer Center, H-1226 205 South Orange Avenue, Newark, NJ 07103, USA
| | - Zinaida S Vexler
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
172
|
Fleiss B, Tann CJ, Degos V, Sigaut S, Van Steenwinckel J, Schang AL, Kichev A, Robertson NJ, Mallard C, Hagberg H, Gressens P. Inflammation-induced sensitization of the brain in term infants. Dev Med Child Neurol 2015; 57 Suppl 3:17-28. [PMID: 25800488 DOI: 10.1111/dmcn.12723] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 12/12/2022]
Abstract
Perinatal insults are a leading cause of infant mortality and amongst survivors are frequently associated with neurocognitive impairment, cerebral palsy (CP), and seizure disorders. The events leading to perinatal brain injury are multifactorial. This review describes how one subinjurious factor affecting the brain sensitizes it to a second injurious factor, causing an exacerbated injurious cascade. We will review the clinical and experimental evidence, including observations of high rates of maternal and fetal infections in term-born infants with neonatal encephalopathy and cerebral palsy. In addition, we will discuss preclinical evidence for the sensitizing effects of inflammation on injuries, such as hypoxia-ischaemia, our current understanding of the mechanisms underpinning the sensitization process, and the possibility for neuroprotection.
Collapse
Affiliation(s)
- Bobbi Fleiss
- Inserm, U1141, Paris, France; University Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; Department of Child Neurology, APHP, Robert Debré Hospital, Paris, France; PremUP, Paris, France; Division of Imaging Sciences, Department of Perinatal Imaging and Health, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Barton SK, Melville JM, Tolcos M, Polglase GR, McDougall ARA, Azhan A, Crossley KJ, Jenkin G, Moss TJM. Human Amnion Epithelial Cells Modulate Ventilation-Induced White Matter Pathology in Preterm Lambs. Dev Neurosci 2015; 37:338-48. [PMID: 25720586 DOI: 10.1159/000371415] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Preterm infants can be inadvertently exposed to high tidal volumes (VT) during resuscitation in the delivery room due to limitations of available equipment. High VT ventilation of preterm lambs produces cerebral white matter (WM) pathology similar to that observed in preterm infants who develop cerebral palsy. We hypothesized that human amnion epithelial cells (hAECs), which have anti-inflammatory and regenerative properties, would reduce ventilation-induced WM pathology in neonatal late preterm lamb brains. METHODS Two groups of lambs (0.85 gestation) were used, as follows: (1) ventilated lambs (Vent; n = 8) were ventilated using a protocol that induces injury (VT targeting 15 ml/kg for 15 min, with no positive end-expiratory pressure) and were then maintained for another 105 min, and (2) ventilated + hAECs lambs (Vent+hAECs; n = 7) were similarly ventilated but received intravenous and intratracheal administration of 9 × 10(7) hAECs (18 × 10(7) hAECs total) at birth. Oxygenation and ventilation parameters were monitored in real time; cerebral oxygenation was measured using near-infrared spectroscopy. qPCR (quantitative real-time PCR) and immunohistochemistry were used to assess inflammation, vascular leakage and astrogliosis in both the periventricular and subcortical WM of the frontal and parietal lobes. An unventilated control group (UVC; n = 5) was also used for qPCR analysis of gene expression. Two-way repeated measures ANOVA was used to compare physiological data. Student's t test and one-way ANOVA were used for immunohistological and qPCR data comparisons, respectively. RESULTS Respiratory parameters were not different between groups. Interleukin (IL)-6 mRNA levels in subcortical WM were lower in the Vent+hAECs group than the Vent group (p = 0.028). IL-1β and IL-6 mRNA levels in periventricular WM were higher in the Vent+hAECs group than the Vent group (p = 0.007 and p = 0.001, respectively). The density of Iba-1-positive microglia was lower in the subcortical WM of the parietal lobes (p = 0.010) in the Vent+hAECs group but not in the periventricular WM. The number of vessels in the WM of the parietal lobe exhibiting protein extravasation was lower (p = 0.046) in the Vent+hAECs group. Claudin-1 mRNA levels were higher in the periventricular WM (p = 0.005). The density of GFAP-positive astrocytes was not different between groups. CONCLUSIONS Administration of hAECs at the time of birth alters the effects of injurious ventilation on the preterm neonatal brain. Further studies are required to understand the regional differences in the effects of hAECs on ventilation-induced WM pathology and their net effect on the developing brain.
Collapse
Affiliation(s)
- Samantha K Barton
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, Vic., Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Bell JE, Becher JC, Keeling JW, McIntosh N. The neuropathology of stillbirth - correlation with apolipoprotein genotype in a Scottish population based study. Early Hum Dev 2015; 91:139-48. [PMID: 25617864 DOI: 10.1016/j.earlhumdev.2014.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/11/2014] [Accepted: 12/28/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND The neuropathology of stillbirths has been widely studied but rarely on a population basis. Whether foetal apolipoprotein E (APOE) genotype exerts any influence has been little investigated, despite well known effects in adult brains. AIMS To establish the neuropathology of a population cohort of stillbirths and compare with the APOE genotype. STUDY DESIGN AND SUBJECTS The brains of 191 stillbirths (≥24weeks of gestation) were recruited from a Scottish population cohort and grouped by clinical history. APOE genotype was available for 97%. RESULTS AND CONCLUSIONS One or more neuropathological features, most appearing relatively recent, were found in 54% of 157 antepartum singletons, 44% of 9 abruption-associated stillbirths, 85% of 13 in multiple pregnancies but in only 19% of 12 intrapartum stillbirths. White matter injury (WMI) occurred in 36% of preterm and 21% mature stillbirths. Fresh petechial haemorrhages were common in all groups (29%) but germinal matrix haemorrhage (GMH) (7%) and periventricular leucomalacia (1%) were confined to preterm. GMH was significantly associated with WMI (p=0.003). Placental inflammation was common in intrapartum stillbirths (50%), compared with antepartum (15%), multiple pregnancy (23%) and abruption (0%). β-Amyloid precursor protein (βAPP) positive axons (36% stillbirths overall) correlated closely with WMI (p<0.0001), justifying future routine inclusion in foetal neuropathological investigation. This study highlights the paucity of brain damage in intrapartum stillbirths. While APOE2 was significantly overrepresented in stillbirths, there was no correlation between APOE genotype and neuropathological findings. We conclude that APOE does not influence neuropathological outcomes in stillbirths.
Collapse
Affiliation(s)
- Jeanne Elisabeth Bell
- Neuropathology, University of Edinburgh Division of Pathology, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | - Julie-Clare Becher
- Child Life and Health, University of Edinburgh, 20 Sylvan Place, Edinburgh EH9 1UW, UK.
| | - Jean Winifred Keeling
- Paediatric Pathology, Royal Hospital for Sick Children, 9 Sciennes Rd, Edinburgh, Midlothian EH9 1LF, UK.
| | - Neil McIntosh
- Child Life and Health, University of Edinburgh, 20 Sylvan Place, Edinburgh EH9 1UW, UK.
| |
Collapse
|
175
|
Boldenow E, Hassan I, Chames MC, Xi C, Loch-Caruso R. The trichloroethylene metabolite S-(1,2-dichlorovinyl)-l-cysteine but not trichloroacetate inhibits pathogen-stimulated TNF-α in human extraplacental membranes in vitro. Reprod Toxicol 2015; 52:1-6. [PMID: 25653212 DOI: 10.1016/j.reprotox.2015.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/20/2014] [Accepted: 01/22/2015] [Indexed: 11/29/2022]
Abstract
Extraplacental membranes define the gestational compartment and provide a barrier to infectious microorganisms ascending the gravid female reproductive tract. We tested the hypothesis that bioactive metabolites of trichloroethylene (TCE) decrease pathogen-stimulated innate immune response of extraplacental membranes. Extraplacental membranes were cultured for 4, 8, and 24h with the TCE metabolites trichloroacetate (TCA) or S-(1,2-dichlorovinyl)-l-cysteine (DCVC) in the absence or presence of lipoteichoic acid (LTA) or lipopolysaccharide (LPS) to simulate infection. In addition, membranes were cocultured with DCVC and Group B Streptococcus (GBS). DCVC (5-50μM) significantly inhibited LTA-, LPS-, and GBS-stimulated cytokine release from tissue cultures as early as 4h (P≤0.05). In contrast, TCA (up to 500μM) did not inhibit LTA-stimulated cytokine release from tissue punches. Because cytokines are important mediators for host response to infectious microorganisms these findings suggest that TCE exposure could potentially modify susceptibility to infection during pregnancy.
Collapse
Affiliation(s)
- Erica Boldenow
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, USA.
| | - Iman Hassan
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, USA.
| | - Mark C Chames
- Departments of Pathology and of Obstetrics and Gynecology, Medical School, University of Michigan, 4215 Med Sci I SPC 5602, Ann Arbor, MI 48109-5602, USA.
| | - Chuanwu Xi
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, USA.
| | - Rita Loch-Caruso
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, USA.
| |
Collapse
|
176
|
Correlation of serum KL-6 and CC16 levels with neurodevelopmental outcome in premature infants at 12 months corrected age. Sci Rep 2015; 5:8121. [PMID: 25631862 PMCID: PMC4309974 DOI: 10.1038/srep08121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/06/2015] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to evaluate KL-6 and CC16 levels and their correlation with neurodevelopmental outcome among very low birth weight pre-term infants at 12 months corrected age. This prospective cohort study was performed from 2011 to 2013 by enrolling pre-term neonates of gestational age ≤ 32 weeks and birth weight ≤ 1500 g. Serum KL-6 and CC16 levels were determined 7 days after birth and their correlation with neurodevelopment was evaluated using Gesell Mental Developmental Scales. Of the 86 eligible pre-term infants, 63 completed follow-up, of which 15 had bronchopulmonary dysplasia. At 12 months corrected age, 49 infants had favorable outcomes and 14 infants had poor neurodevelopmental outcome. KL-6 levels were higher and CC16 levels were lower in infants with poor neurodevelopmental outcome compared with those infants who had favourable neurodevelopmental outcome. Serum KL-6 levels less than 90.0 ng/ml and CC16 levels greater than 320.0 pg/ml at 7 days of life were found to be predictive of a favourable outcome at 12 months corrected age. These biological markers could predict neurodevelopmental outcome at 12 months corrected age in very low birth weight premature infants, and help the clinician plan early therapeutic interventions to minimize or avoid poor neurodevelopmental outcome.
Collapse
|
177
|
Zhang J, Sadowska GB, Chen X, Park SY, Kim JE, Bodge CA, Cummings E, Lim YP, Makeyev O, Besio WG, Gaitanis J, Banks WA, Stonestreet BS. Anti-IL-6 neutralizing antibody modulates blood-brain barrier function in the ovine fetus. FASEB J 2015; 29:1739-53. [PMID: 25609424 DOI: 10.1096/fj.14-258822] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
Impaired blood-brain barrier function represents an important component of hypoxic-ischemic brain injury in the perinatal period. Proinflammatory cytokines could contribute to ischemia-related blood-brain barrier dysfunction. IL-6 increases vascular endothelial cell monolayer permeability in vitro. However, contributions of IL-6 to blood-brain barrier abnormalities have not been examined in the immature brain in vivo. We generated pharmacologic quantities of ovine-specific neutralizing anti-IL-6 mAbs and systemically infused mAbs into fetal sheep at 126 days of gestation after exposure to brain ischemia. Anti-IL-6 mAbs were measured by ELISA in fetal plasma, cerebral cortex, and cerebrospinal fluid, blood-brain barrier permeability was quantified using the blood-to-brain transfer constant in brain regions, and IL-6, tight junction proteins, and plasmalemma vesicle protein (PLVAP) were detected by Western immunoblot. Anti-IL-6 mAb infusions resulted in increases in mAb (P < 0.05) in plasma, brain parenchyma, and cerebrospinal fluid and decreases in brain IL-6 protein. Twenty-four hours after ischemia, anti-IL-6 mAb infusions attenuated ischemia-related increases in blood-brain barrier permeability and modulated tight junction and PLVAP protein expression in fetal brain. We conclude that inhibiting the effects of IL-6 protein with systemic infusions of neutralizing antibodies attenuates ischemia-related increases in blood-brain barrier permeability by inhibiting IL-6 and modulates tight junction proteins after ischemia.
Collapse
Affiliation(s)
- Jiyong Zhang
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Grazyna B Sadowska
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Xiaodi Chen
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Seon Yeong Park
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jeong-Eun Kim
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Courtney A Bodge
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Erin Cummings
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Yow-Pin Lim
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Oleksandr Makeyev
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Walter G Besio
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - John Gaitanis
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - William A Banks
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Barbara S Stonestreet
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
178
|
Zhang Y, Inder TE, Neil JJ, Dierker DL, Alexopoulos D, Anderson PJ, Van Essen DC. Cortical structural abnormalities in very preterm children at 7 years of age. Neuroimage 2015; 109:469-79. [PMID: 25614973 DOI: 10.1016/j.neuroimage.2015.01.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 12/15/2014] [Accepted: 01/05/2015] [Indexed: 10/24/2022] Open
Abstract
We analyzed long-lasting alterations in brain morphometry associated with preterm birth using volumetric and surface-based analyses applied to children at age 7 years. Comparison of 24 children born very preterm (VPT) to 24 healthy term-born children revealed reductions in total cortical gray matter volume, white matter volume, cortical surface area and gyrification index. Regional cortical shape abnormalities in VPT children included the following: shallower anterior superior temporal sulci, smaller relative surface area in the inferior sensori-motor cortex and posterior superior temporal cortex, larger relative surface area and a cingulate sulcus that was shorter or more interrupted in medial frontoparietal cortex. These findings indicate a complex pattern of regional vulnerabilities in brain development that may contribute to the diverse and long-lasting neurobehavioral consequences that can occur after very premature birth.
Collapse
Affiliation(s)
- Yuning Zhang
- Division of Biomedical and Biological Science, Washington University School of Medicine, St Louis, MO, USA
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey J Neil
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna L Dierker
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO, USA
| | - Dimitrios Alexopoulos
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Peter J Anderson
- Clinical Sciences, Murdoch Children's Research Institute, Victoria, Australia
| | - David C Van Essen
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
179
|
Neuroprotection in preterm infants. BIOMED RESEARCH INTERNATIONAL 2015; 2015:257139. [PMID: 25650134 PMCID: PMC4306255 DOI: 10.1155/2015/257139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/22/2014] [Indexed: 01/05/2023]
Abstract
Preterm infants born before the 30th week of pregnancy are especially at risk of perinatal brain damage which is usually a result of cerebral ischemia or an ascending intrauterine infection. Prevention of preterm birth and early intervention given signs of imminent intrauterine infection can reduce the incidence of perinatal cerebral injury. It has been shown that administering magnesium intravenously to women at imminent risk of a preterm birth leads to a significant reduction in the likelihood of the infant developing cerebral palsy and motor skill dysfunction. It has also been demonstrated that delayed clamping of the umbilical cord after birth reduces the rate of brain hemorrhage among preterm infants by up to 50%. In addition, mesenchymal stem cells seem to have significant neuroprotective potential in animal experiments, as they increase the rate of regeneration of the damaged cerebral area. Clinical tests of these types of therapeutic intervention measures appear to be imminent. In the last trimester of pregnancy, the serum concentrations of estradiol and progesterone increase significantly. Preterm infants are removed abruptly from this estradiol and progesterone rich environment. It has been demonstrated in animal experiments that estradiol and progesterone protect the immature brain from hypoxic-ischemic lesions. However, this neuroprotective strategy has unfortunately not yet been subject to sufficient clinical investigation.
Collapse
|
180
|
Immune System Related Markers: Changes in childhood Neuropsychiatry Disorders Cause and Consequence. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/978-3-319-13602-8_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
181
|
Gano D, Andersen SK, Partridge JC, Bonifacio SL, Xu D, Glidden DV, Ferriero DM, Barkovich AJ, Glass HC. Diminished white matter injury over time in a cohort of premature newborns. J Pediatr 2015; 166:39-43. [PMID: 25311709 PMCID: PMC4274204 DOI: 10.1016/j.jpeds.2014.09.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/30/2014] [Accepted: 09/05/2014] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To determine the rate of magnetic resonance imaging (MRI)-detected noncystic white matter injury (WMI) in a prospective cohort of premature newborns, and to evaluate its associations with changes in clinical predictors of WMI over the study period. STUDY DESIGN A prospective cohort of premature newborns (<33 weeks gestational age) was studied with MRI within 4 weeks of birth and near term-equivalent age. A pediatric neuroradiologist scored the severity of WMI on T1-weighted MRI according to published criteria. WMI was classified as none/mild or moderate/severe. Subjects with severe cystic WMI, periventricular hemorrhagic infarction, or motion artifact on MRI were excluded. Changes in clinical characteristics and predictors of WMI over the study period (1998-2011) were evaluated. Predictors of moderate/severe WMI, including birth year, were evaluated using multivariate logistic regression. RESULTS Among 267 newborns, 45 (17%) had moderate/severe WMI. The rate of moderate/severe WMI decreased over the study period (P = .002, χ(2) test for trends). On multivariate logistic regression, the odds of moderate/severe WMI decreased by 11% for each birth year of the cohort (OR, 0.89; 95% CI, 0.81-0.98; P = .02). Prolonged exposure to indomethacin also was independently associated with reduced odds of moderate/severe WMI. CONCLUSION The decreasing burden of MRI-detected moderate/severe noncystic WMI in our cohort of premature newborns is independent over time of changes in the known clinical predictors of WMI. Prolonged exposure to indomethacin is associated with reduced WMI.
Collapse
Affiliation(s)
- Dawn Gano
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Sarah K Andersen
- Department of Medicine, Queens University, Kingston, Ontario, Canada
| | - J Colin Partridge
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Sonia L Bonifacio
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Duan Xu
- Department of Radiology, University of California San Francisco, San Francisco, CA
| | - David V Glidden
- Department of Biostatistics, University of California San Francisco, San Francisco, CA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Neurology, University of California San Francisco, San Francisco, CA
| | - A James Barkovich
- Department of Radiology, University of California San Francisco, San Francisco, CA
| | - Hannah C Glass
- Department of Pediatrics, University of California San Francisco, San Francisco, CA; Department of Neurology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
182
|
Goldstein JM, Cherkerzian S, Seidman LJ, Donatelli JAL, Remington AG, Tsuang MT, Hornig M, Buka SL. Prenatal maternal immune disruption and sex-dependent risk for psychoses. Psychol Med 2014; 44:3249-3261. [PMID: 25065485 PMCID: PMC4477534 DOI: 10.1017/s0033291714000683] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Previous studies suggest that abnormalities in maternal immune activity during pregnancy alter the offspring's brain development and are associated with increased risk for schizophrenia (SCZ) dependent on sex. METHOD Using a nested case-control design and prospectively collected prenatal maternal sera from which interleukin (IL)-1β, IL-8, IL-6, tumor necrosis factor (TNF)-α and IL-10 were assayed, we investigated sex-dependent associations between these cytokines and 88 psychotic cases [SCZ = 44; affective psychoses (AP) = 44] and 100 healthy controls from a pregnancy cohort followed for > 40 years. Analyses included sex-stratified non-parametric tests adjusted for multiple comparisons to screen cytokines associated with SCZ risk, followed by deviant subgroup analyses using generalized estimating equation (GEE) models. RESULTS There were higher prenatal IL-6 levels among male SCZ than male controls, and lower TNF-α levels among female SCZ than female controls. The results were supported by deviant subgroup analyses with significantly more SCZ males with high IL-6 levels (>highest quartile) compared with controls [odd ratio (OR)75 = 3.33, 95% confidence interval (CI) 1.13-9.82], and greater prevalence of low TNF-α levels (<lowest quartile) among SCZ females compared with their controls (OR25 = 6.30, 95% CI 1.20-33.04) and SCZ males. Higher levels of IL-6 were only found among SCZ compared with AP cases. Lower TNF-α levels (non-significant) also characterized female AP cases versus controls, although the prevalence of the lowest levels was higher in SCZ than AP females (70% v. 40%), with no effect in SCZ or AP males. CONCLUSIONS The results underscore the importance of immunologic processes affecting fetal brain development and differential risk for psychoses depending on psychosis subtype and offspring sex.
Collapse
Affiliation(s)
- J. M Goldstein
- Connors Center for Women’s Health and Gender Biology, Brigham and Women’s Hospital, Boston, MA, USA
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA
- Division of Psychiatric Neuroscience, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - S. Cherkerzian
- Connors Center for Women’s Health and Gender Biology, Brigham and Women’s Hospital, Boston, MA, USA
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA
| | - L. J. Seidman
- Division of Psychiatric Neuroscience, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Department of Psychiatry, Massachusetts Mental Health Center; Division of Public Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - A. G. Remington
- Connors Center for Women’s Health and Gender Biology, Brigham and Women’s Hospital, Boston, MA, USA
| | - M. T. Tsuang
- Beth Israel Deaconess Hospital, Department of Psychiatry, Division of Public Psychiatry, Massachusetts Mental Health Center and Harvard Medical School, Boston, MA, USA
- Center for Behavioral Genomics, Department of Psychiatry; Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
- Harvard Institute of Psychiatric Epidemiology and Genetics, Harvard School of Public Heath, Boston, MA, USA
| | - M. Hornig
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - S. L. Buka
- Department of Epidemiology, Brown University, Providence, RI, USA
| |
Collapse
|
183
|
Kim SM, Romero R, Park JW, Oh KJ, Jun JK, Yoon BH. The relationship between the intensity of intra-amniotic inflammation and the presence and severity of acute histologic chorioamnionitis in preterm gestation. J Matern Fetal Neonatal Med 2014; 28:1500-9. [PMID: 25184305 DOI: 10.3109/14767058.2014.961009] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Acute histologic chorioamnionitis (HCA) is associated with an increased risk of perinatal mortality and morbidity. The purpose of this study was to determine the relationship between the intensity of intra-amniotic inflammation (IAI) and the severity of acute HCA in preterm gestation. METHODS The relationship between the intensity of IAI and the presence and severity of acute HCA was examined in 412 patients with singleton gestations who delivered within 120 h of transabdominal amniocentesis. The concentration of amniotic fluid (AF) matrix metalloproteinase (MMP)-8 was assayed to determine the presence and intensity of IAI. Acute HCA was defined as the presence of inflammatory change in any tissue samples according to the criteria previously reported. The total grade of acute HCA was used to determine the severity of HCA. RESULTS (1) Patients with IAI had a significantly higher rate of acute HCA than those without IAI [76.9% (133/173)] versus 20.9% (50/239), p < 0.001]. The AF MMP-8 concentration was significantly higher in patients with acute HCA than in those without acute HCA (median [range]; 188.3 ng/ml [0.3-6142.6] versus 1.8 ng/ml [0.3-2845.5], p < 0.001); (2) Of 183 patients with acute HCA, the AF MMP-8 concentration was positively correlated with the severity of acute HCA (p < 0.001). CONCLUSIONS AF MMP-8 concentration was not only a predictor of the presence of acute HCA, but its concentration also correlated with the severity of acute HCA. The higher the intensity of IAI, the worse the degree of acute HCA in preterm gestation.
Collapse
Affiliation(s)
- Sun Min Kim
- a Department of Obstetrics and Gynecology , Seoul National University College of Medicine , Seoul , Republic of Korea
| | | | | | | | | | | |
Collapse
|
184
|
Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol 2014; 10:643-60. [PMID: 25311587 DOI: 10.1038/nrneurol.2014.187] [Citation(s) in RCA: 636] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have shown a clear association between maternal infection and schizophrenia or autism in the progeny. Animal models have revealed maternal immune activation (mIA) to be a profound risk factor for neurochemical and behavioural abnormalities in the offspring. Microglial priming has been proposed as a major consequence of mIA, and represents a critical link in a causal chain that leads to the wide spectrum of neuronal dysfunctions and behavioural phenotypes observed in the juvenile, adult or aged offspring. Such diversity of phenotypic outcomes in the mIA model are mirrored by recent clinical evidence suggesting that infectious exposure during pregnancy is also associated with epilepsy and, to a lesser extent, cerebral palsy in children. Preclinical research also suggests that mIA might precipitate the development of Alzheimer and Parkinson diseases. Here, we summarize and critically review the emerging evidence that mIA is a shared environmental risk factor across CNS disorders that varies as a function of interactions between genetic and additional environmental factors. We also review ongoing clinical trials targeting immune pathways affected by mIA that may play a part in disease manifestation. In addition, future directions and outstanding questions are discussed, including potential symptomatic, disease-modifying and preventive treatment strategies.
Collapse
|
185
|
Li J, McDonald CA, Fahey MC, Jenkin G, Miller SL. Could cord blood cell therapy reduce preterm brain injury? Front Neurol 2014; 5:200. [PMID: 25346720 PMCID: PMC4191167 DOI: 10.3389/fneur.2014.00200] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/19/2014] [Indexed: 12/25/2022] Open
Abstract
Major advances in neonatal care have led to significant improvements in survival rates for preterm infants, but this occurs at a cost, with a strong causal link between preterm birth and neurological deficits, including cerebral palsy (CP). Indeed, in high-income countries, up to 50% of children with CP were born preterm. The pathways that link preterm birth and brain injury are complex and multifactorial, but it is clear that preterm birth is strongly associated with damage to the white matter of the developing brain. Nearly 90% of preterm infants who later develop spastic CP have evidence of periventricular white matter injury. There are currently no treatments targeted at protecting the immature preterm brain. Umbilical cord blood (UCB) contains a diverse mix of stem and progenitor cells, and is a particularly promising source of cells for clinical applications, due to ethical and practical advantages over other potential therapeutic cell types. Recent studies have documented the potential benefits of UCB cells in reducing brain injury, particularly in rodent models of term neonatal hypoxia–ischemia. These studies indicate that UCB cells act via anti-inflammatory and immuno-modulatory effects, and release neurotrophic growth factors to support the damaged and surrounding brain tissue. The etiology of brain injury in preterm-born infants is less well understood than in term infants, but likely results from episodes of hypoperfusion, hypoxia–ischemia, and/or inflammation over a developmental period of white matter vulnerability. This review will explore current knowledge about the neuroprotective actions of UCB cells and their potential to ameliorate preterm brain injury through neonatal cell administration. We will also discuss the characteristics of UCB-derived from preterm and term infants for use in clinical applications.
Collapse
Affiliation(s)
- Jingang Li
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia
| | | | - Michael C Fahey
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Paediatrics, Monash University , Clayton, VIC , Australia
| | - Graham Jenkin
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Obstetrics and Gynaecology, Monash University , Clayton, VIC , Australia
| | - Suzanne L Miller
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Obstetrics and Gynaecology, Monash University , Clayton, VIC , Australia
| |
Collapse
|
186
|
Albertsson AM, Bi D, Duan L, Zhang X, Leavenworth JW, Qiao L, Zhu C, Cardell S, Cantor H, Hagberg H, Mallard C, Wang X. The immune response after hypoxia-ischemia in a mouse model of preterm brain injury. J Neuroinflammation 2014; 11:153. [PMID: 25187205 PMCID: PMC4172879 DOI: 10.1186/s12974-014-0153-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/14/2014] [Indexed: 01/04/2023] Open
Abstract
Background Preterm brain injury consists primarily of periventricular leukomalacia accompanied by elements of gray-matter injury, and these injuries are associated with cerebral palsy and cognitive impairments. Inflammation is believed to be an important contributing factor to these injuries. The aim of this study was to examine the immune response in a postnatal day (PND) 5 mouse model of preterm brain injury induced by hypoxia-ischemia (HI) that is characterized by focal white and gray-matter injury. Methods C57Bl/6 mice at PND 5 were subjected to unilateral HI induced by left carotid artery ligation and subsequent exposure to 10% O2 for 50 minutes, 70 minutes, or 80 minutes. At seven days post-HI, the white/gray-matter injury was examined. The immune responses in the brain after HI were examined at different time points after HI using RT-PCR and immunohistochemical staining. Results HI for 70 minutes in PND 5 mice induced local white-matter injury with focal cortical injury and hippocampal atrophy, features that are similar to those seen in preterm brain injury in human infants. HI for 50 minutes resulted in a small percentage of animals being injured, and HI for 80 minutes produced extensive infarction in multiple brain areas. Various immune responses, including changes in transcription factors and cytokines that are associated with a T-helper (Th)1/Th17-type response, an increased number of CD4+ T-cells, and elevated levels of triggering receptor expressed on myeloid cells 2 (TREM-2) and its adaptor protein DNAX activation protein of 12 kDa (DAP12) were observed using the HI 70 minute preterm brain injury model. Conclusions We have established a reproducible model of HI in PND 5 mice that produces consistent local white/gray-matter brain damage that is relevant to preterm brain injury in human infants. This model provides a useful tool for studying preterm brain injury. Both innate and adaptive immune responses are observed after HI, and these show a strong pro-inflammatory Th1/Th17-type bias. Such findings provide a critical foundation for future studies on the mechanism of preterm brain injury and suggest that blocking the Th1/Th17-type immune response might provide neuroprotection after preterm brain injury.
Collapse
|
187
|
Sesso MLT, Borges MCL, Ferriani VPL, Geraldo-Martins VR, Rodrigues DBR, Nogueira RD. Prospective evaluation of cytokine in saliva of preterm and fullterm neonates. Immunobiology 2014; 219:830-5. [PMID: 25155058 DOI: 10.1016/j.imbio.2014.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 06/20/2014] [Accepted: 07/15/2014] [Indexed: 11/28/2022]
Abstract
Little is known about the ontogeny of the cytokines in saliva of newborn. Previous studies showed that levels of immunoglobulin A (IgA) in saliva could be influenced by prematurity. So, the aim of this study was to analyze the levels of interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin 12 (IL-12), and interferon gamma (IFN-γ) in sample saliva of fullterm (FT) and preterm (PT) neonates at birth (T0) and after 3 months of age (T3). Saliva from 50 infants (25 FT and 25 PT) were collected at T0 and T3 and analyzed by Luminex Corporation (Austin, Texas, United States) multiplex assay. Clinical characteristics and social-economic data were assessed through questionnaires. All cytokines could be detected at birth in levels higher than found in T3. The mean levels and frequency of detection of cytokines were significantly higher in PT than FT at T0 (P<0.05). There were a positive association between IL-10 and infection (P<0.05) and IL-6 and stress (P<0.005). Salivary cytokines were detected within the first hours after birth and their levels decreased after 3 months. The cytokine levels were different between PT and FT children and appear to be influenced by stress situation and/or antigenic microbial challenge. The results confirm the necessity for further studies about the mucosal immune system by using of saliva as a source of diagnostic by identification of biomarkers of the status of the immune.
Collapse
|
188
|
Differences between periventricular hemorrhagic infarction and periventricular leukomalacia. Brain Dev 2014; 36:555-62. [PMID: 23978489 DOI: 10.1016/j.braindev.2013.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 07/28/2013] [Accepted: 07/30/2013] [Indexed: 11/20/2022]
Abstract
PURPOSE To clarify the differences between infants with periventricular hemorrhagic infarction (PVHI) and those with periventricular leukomalacia (PVL). METHODS We retrospectively evaluated the clinical features, ultrasonography, and electroencephalogram (EEG) findings in 22 preterm infants with PVHI and 49 with PVL. EEG and cranial ultrasonography were serially performed in all participants starting immediately after birth. Acute and chronic stage EEG abnormalities were evaluated separately. RESULTS Gestational age and birth weight were significantly lower in infants with PVHI than those with PVL. EEGs were normal in the majority of infants with PVHI on days 1-2. However, EEG abnormalities appeared after ultrasonography abnormalities. The majority of infants with PVL showed acute-stage EEG abnormalities on days 1-2. The rate of infants with acute-stage EEG abnormalities decreased with age, whereas the rate of infants with chronic-stage EEG abnormalities increased with age. Normal EEG before ultrasonography abnormalities was more common in infants with PVHI than in those with PVL. However, deterioration of acute-stage EEG abnormalities was more frequent in infants with PVHI than in those with PVL. CONCLUSIONS PVHI was presumed to cause mostly postnatal injury, whereas PVL was presumed to cause mostly pre-or perinatal injury.
Collapse
|
189
|
Basu S, Dewangan S, Barman S, Anupurba S, Shukla RC, Kumar A. Cerebral blood flow velocity in asymptomatic premature neonates exposed to clinical chorioamnionitis. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2014. [DOI: 10.1016/j.cegh.2013.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
190
|
Biran V, Phan Duy A, Decobert F, Bednarek N, Alberti C, Baud O. Is melatonin ready to be used in preterm infants as a neuroprotectant? Dev Med Child Neurol 2014; 56:717-23. [PMID: 24575840 DOI: 10.1111/dmcn.12415] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2014] [Indexed: 01/08/2023]
Abstract
The prevention of neurological disabilities following preterm birth remains a major public health challenge and efforts are still needed to test the neuroprotective properties of candidate molecules. Melatonin serves as a neuroprotectant in adult models of cerebral ischemia through its potent antioxidant and anti-inflammatory effects. An increasing number of preclinical studies have consistently demonstrated that melatonin protects the damaged developing brain by preventing abnormal myelination and an inflammatory glial reaction, a major cause of white matter injury. The main questions asked in this review are whether preclinical data on the neuroprotective properties of melatonin are sufficient to translate this concept into the clinical setting, and whether melatonin can reduce white matter damage in preterm infants. This review provides support for our view that melatonin is now ready to be tested in human preterm neonates, and discusses ongoing and planned clinical trials.
Collapse
Affiliation(s)
- Valérie Biran
- Neonatal Intensive Care Unit, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Université Paris Diderot, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, INSERM, Paris, France; PremUP Foundation, Paris, France
| | | | | | | | | | | |
Collapse
|
191
|
Gagliardi L, Rusconi F, Bellù R, Zanini R. Association of maternal hypertension and chorioamnionitis with preterm outcomes. Pediatrics 2014; 134:e154-e161. [PMID: 24913788 DOI: 10.1542/peds.2013-3898] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES We compared the relative effect of hypertensive disorders of pregnancy and chorioamnionitis on adverse neonatal outcomes in very preterm neonates, and studied whether gestational age (GA) modulates these effects. METHODS A cohort of neonates 23 to 30 weeks' GA, born in 2008 to 2011 in 82 hospitals adhering to the Italian Neonatal Network, was analyzed. Infants born from mothers who had hypertensive disorders (N = 2096) were compared with those born after chorioamnionitis (N = 1510). Statistical analysis employed logistic models, adjusting for GA, hospital, and potential confounders. RESULTS Overall mortality was higher after hypertension than after chorioamnionitis (odds ratio [OR], 1.39; 95% confidence interval [CI], 1.08-1.80), but this relationship changed across GA weeks; the OR for hypertension was highest at low GA, whereas from 28 weeks' GA onward, mortality was higher for chorioamnionitis. For other outcomes, the relative risks were constant across GA; infants born after hypertension had an increased risk for bronchopulmonary dysplasia (OR, 2.20; 95% CI, 1.68-2.88) and severe retinopathy of prematurity (OR, 1.48; 95% CI, 1.02-2.15), whereas there was a lower risk for early-onset sepsis (OR, 0.25; 95% CI, 0.19-0.34), severe intraventricular hemorrhage (OR, 0.65; 95% CI, 0.48-0.88), periventricular leukomalacia (OR, 0.70; 95% CI, 0.48-1.01), and surgical necrotizing enterocolitis or gastrointestinal perforation (OR, 0.47; 95% CI, 0.31-0.72). CONCLUSIONS Mortality and other adverse outcomes in very preterm infants depend on antecedents of preterm birth. Hypertension and chorioamnionitis are associated with different patterns of outcomes; for mortality, the effect changes across GA weeks.
Collapse
Affiliation(s)
- Luigi Gagliardi
- Department of Woman and Child Health, Ospedale Versilia, Viareggio, Italy;
| | - Franca Rusconi
- Unit of Epidemiology, A Meyer Children's University Hospital, Florence, Italy; and
| | - Roberto Bellù
- Neonatal ICU, Ospedale Alessandro Manzoni, Lecco, Italy
| | | |
Collapse
|
192
|
Davis J, Moylan S, Harvey BH, Maes M, Berk M. Neuroprogression in schizophrenia: Pathways underpinning clinical staging and therapeutic corollaries. Aust N Z J Psychiatry 2014; 48:512-29. [PMID: 24803587 DOI: 10.1177/0004867414533012] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Whilst dopaminergic dysfunction remains a necessary component involved in the pathogenesis of schizophrenia, our current pharmacological armoury of dopamine antagonists does little to control the negative symptoms of schizophrenia. This suggests other pathological processes must be implicated. This paper aims to elaborate on such theories. METHODS Data for this review were sourced from the electronic database PUBMED, and was not limited by language or date of publication. RESULTS It has been suggested that multiple 'hits' may be required to unveil the clinical syndrome in susceptible individuals. Such hits potentially first occur in utero, leading to neuronal disruption, epigenetic changes and the establishment of an abnormal inflammatory response. The development of schizophrenia may therefore potentially be viewed as a neuroprogressive response to these early stressors, driven on by changes in tryptophan catabolite (TRYCAT) metabolism, reactive oxygen species handling and N-methyl d-aspartate (NMDA) circuitry. Given the potential for such progression over time, it is prudent to explore the new treatment strategies which may be implemented before such changes become established. CONCLUSIONS Outside of the dopaminergic model, the potential pathogenesis of schizophrenia has yet to be fully elucidated, but common themes include neuropil shrinkage, the development of abnormal neuronal circuitry, and a chronic inflammatory state which further disrupts neuronal function. Whilst some early non-dopaminergic treatments show promise, none have yet to be fully studied in appropriately structured randomized controlled trials and they remain little more than potential attractive targets.
Collapse
Affiliation(s)
- Justin Davis
- IMPACT Strategic Research Centre, Deakin University, School of Medicine, Barwon Health, Geelong, Australia
| | - Steven Moylan
- IMPACT Strategic Research Centre, Deakin University, School of Medicine, Barwon Health, Geelong, Australia
| | - Brian H Harvey
- Division of Pharmacology, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North West University, Potchefstroom, South Africa
| | - Michael Maes
- IMPACT Strategic Research Centre, Deakin University, School of Medicine, Barwon Health, Geelong, Australia Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, Deakin University, School of Medicine, Barwon Health, Geelong, Australia Orygen Youth Health Research Centre, Parkville, Australia Centre of Youth Mental Health, University of Melbourne, Parkville, Australia Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Australia University of Melbourne, Department of Psychiatry, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
193
|
Romero R, Kadar N, Miranda J, Korzeniewski SJ, Schwartz AG, Chaemsaithong P, Rogers W, Soto E, Gotsch F, Yeo L, Hassan SS, Chaiworapongsa T. The diagnostic performance of the Mass Restricted (MR) score in the identification of microbial invasion of the amniotic cavity or intra-amniotic inflammation is not superior to amniotic fluid interleukin-6. J Matern Fetal Neonatal Med 2014; 27:757-69. [PMID: 24028673 PMCID: PMC5881917 DOI: 10.3109/14767058.2013.844123] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Intra-amniotic infection/inflammation are major causes of spontaneous preterm labor and delivery. However, diagnosis of intra-amniotic infection is challenging because most are subclinical and amniotic fluid (AF) cultures take several days before results are available. Several tests have been proposed for the rapid diagnosis of microbial invasion of the amniotic cavity (MIAC) or intra-amniotic inflammation. The aim of this study was to examine the diagnostic performance of the AF Mass Restricted (MR) score in comparison with interleukin-6 (IL-6) and matrix metalloproteinase-8 (MMP-8) for the identification of MIAC or inflammation. METHODS AF samples were collected from patients with singleton gestations and symptoms of preterm labor (n = 100). Intra-amniotic inflammation was defined as >100 white blood cells/mm(3) (WBCs) in AF; MIAC was defined as a positive AF culture. AF IL-6 and MMP-8 were determined using ELISA. The MR score was obtained using the Surface-Enhanced Laser Desorption Ionization Time of Flight (SELDI-TOF) mass spectrometry. Sensitivity and specificity were calculated and logistic regression models were fit to construct receiver-operating characteristic (ROC) curves for the identification of each outcome. The McNemar's test and paired sample non-parametric statistical techniques were used to test for differences in diagnostic performance metrics. RESULTS (1) The prevalence of MIAC and intra-amniotic inflammation was 34% (34/100) and 40% (40/100), respectively; (2) there were no significant differences in sensitivity of the three tests under study (MR score, IL-6 or MMP-8) in the identification of either MIAC or intra-amniotic inflammation (using the following cutoffs: MR score >2, IL-6 >11.4 ng/mL, and MMP-8 >23 ng/mL); (3) there was no significant difference in the sensitivity among the three tests for the same outcomes when the false positive rate was fixed at 15%; (4) the specificity for IL-6 was not significantly different from that of the MR score in identifying either MIAC or intra-amniotic inflammation when using previously reported thresholds; and (5) there were no significant differences in the area under the ROC curve when comparing the MR score, IL-6 or MMP-8 in the identification of these outcomes. CONCLUSIONS IL-6 and the MR score have equivalent diagnostic performance in the identification of MIAC or intra-amniotic inflammation. Selection from among these three tests (MR score, IL-6 and MMP-8) for diagnostic purposes should be based on factors such as availability, reproducibility, and cost. The MR score requires a protein chip and a SELDI-TOF instrument which are not widely available or considered "state of the art". In contrast, immunoassays for IL-6 can be performed in the majority of clinical laboratories.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Nicholas Kadar
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
| | - Jezid Miranda
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Alyse G. Schwartz
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Wade Rogers
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eleazar Soto
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Francesca Gotsch
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Integrata Verona, Ostetricia Ginecologia, Azienda Ospedaliera Universitaria, Verona, Italy
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
194
|
Bolouri H, Sävman K, Wang W, Thomas A, Maurer N, Dullaghan E, Fjell CD, Ek CJ, Hagberg H, Hancock REW, Brown KL, Mallard C. Innate defense regulator peptide 1018 protects against perinatal brain injury. Ann Neurol 2014; 75:395-410. [DOI: 10.1002/ana.24087] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 09/20/2013] [Accepted: 12/03/2013] [Indexed: 12/24/2022]
Affiliation(s)
- Hayde Bolouri
- Institute of Neuroscience and Physiology; Department of Physiology, Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
| | - Karin Sävman
- Institute of Neuroscience and Physiology; Department of Physiology, Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
- Department of Pediatrics, Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| | - Wei Wang
- Institute of Neuroscience and Physiology; Department of Physiology, Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
| | - Anitha Thomas
- Centre for Drug Research and Development; Vancouver, British Columbia; Canada
| | - Norbert Maurer
- Centre for Drug Research and Development; Vancouver, British Columbia; Canada
| | - Edie Dullaghan
- Centre for Drug Research and Development; Vancouver, British Columbia; Canada
| | - Christopher D. Fjell
- James Hogg Research Centre; University of British Columbia at St Paul's Hospital; Vancouver British Columbia Canada
| | - C. Joakim Ek
- Institute of Neuroscience and Physiology; Department of Physiology, Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
| | - Henrik Hagberg
- Perinatal Center, Department of Clinical Sciences; Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
- Centre for the Developing Brain, King's College; Perinatal Imaging and Health, St Thomas' Hospital; London United Kingdom
| | - Robert E. W. Hancock
- James Hogg Research Centre; University of British Columbia at St Paul's Hospital; Vancouver British Columbia Canada
| | - Kelly L. Brown
- Department of Rheumatology and Inflammation Research; Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology; Department of Physiology, Sahlgrenska Academy, University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
195
|
Mitchell RHB, Goldstein BI. Inflammation in children and adolescents with neuropsychiatric disorders: a systematic review. J Am Acad Child Adolesc Psychiatry 2014; 53:274-96. [PMID: 24565356 DOI: 10.1016/j.jaac.2013.11.013] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/21/2013] [Accepted: 12/13/2013] [Indexed: 11/29/2022]
Abstract
OBJECTIVE There has been rapid growth in research regarding inflammation in neuropsychiatric disorders as it relates to youth. We therefore set out to systematically review the literature on inflammation and neuropsychiatric disorders in children and adolescents. METHOD A systematic review of the literature was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Studies were included if proinflammatory markers (PIMs) in children and/or adolescents with neuropsychiatric disorders were measured. RESULTS Sixty-seven studies were included, involving 3,952 youth. Evidence for a proinflammatory state is strongest for autism spectrum disorders (ASD). PIMs are elevated in children and adolescents with major depressive disorder (MDD), bipolar disorder (BD), post-traumatic stress disorder (PTSD), obsessive-compulsive disorder (OCD), Tourette's disorder (TD), attention-deficit/hyperactivity disorder (ADHD), and schizophrenia (SZ). However, the data are inconsistent. Evidence for specific PIMs is equivocal at this stage, although the findings in youth with MDD, BD, and PTSD converge with the extant adult literature in these areas. Definitive conclusions are limited by methodologic factors including cross-sectional and retrospective study design, between-study differences in specific markers and methods of analysis, small sample size, and other sources of heterogeneity. CONCLUSION The literature regarding inflammation among children and adolescents with neuropsychiatric disorders represents nearly 4,000 youth. There is preliminary evidence for elevated markers of inflammation in this population. Larger, prospective studies are needed to realize the goal of inflammatory markers informing clinical practice. In the interim, present findings suggest that further examination of this topic is warranted.
Collapse
|
196
|
Intermittent or sustained systemic inflammation and the preterm brain. Pediatr Res 2014; 75:376-80. [PMID: 24429547 PMCID: PMC3943674 DOI: 10.1038/pr.2013.238] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 09/13/2013] [Indexed: 12/22/2022]
Abstract
Exposure to perinatal infection and inflammation is associated with an increased risk for neonatal brain damage and developmental disabilities. In this integrated mechanism review, we discuss evidence in support of the contention that the preterm newborn is capable of intermittent or sustained systemic inflammation (ISSI), which appears to contribute more to adverse neurodevelopmental outcomes in preterm infants than does shorter duration inflammation.
Collapse
|
197
|
Chau V, McFadden DE, Poskitt KJ, Miller SP. Chorioamnionitis in the pathogenesis of brain injury in preterm infants. Clin Perinatol 2014; 41:83-103. [PMID: 24524448 DOI: 10.1016/j.clp.2013.10.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chorioamnionitis (or placental infection) is suspected to be a risk factor for brain injury in premature infants. The suggested association between chorioamnionitis and cystic periventricular leukomalacia and cerebral palsy is uncertain because of the variability of study designs and definitions of chorioamnionitis. Improvements in neonatal intensive care may have attenuated the impact of chorioamnionitis on brain health outcomes. Large multicenter studies using rigorous definitions of chorioamnionitis on placental pathologies and quantitative magnetic resonance techniques may offer the optimal way to clarify the complex role of chorioamnionitis in modifying brain health and long-term outcomes.
Collapse
Affiliation(s)
- Vann Chau
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada; University of Toronto, Department of Pediatrics, 563 Spadina Crescent, Toronto, Ontario, M5S 2J7, Canada; Child & Family Research Institute, 950 28th Avenue, Vancouver, British Columbia, V5Z 4H4, Canada.
| | - Deborah E McFadden
- Child & Family Research Institute, 950 28th Avenue, Vancouver, British Columbia, V5Z 4H4, Canada; Department of Pathology, BC Children's & Women's Health Center, 4480 Oak Street, Vancouver, British Columbia, V6H 3V4, Canada; University of British Columbia, Departments of Pediatrics, Pathology and Radiology, 2329 West Mall, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Kenneth J Poskitt
- Child & Family Research Institute, 950 28th Avenue, Vancouver, British Columbia, V5Z 4H4, Canada; University of British Columbia, Departments of Pediatrics, Pathology and Radiology, 2329 West Mall, Vancouver, British Columbia, V6T 1Z4, Canada; Departments of Pediatrics and Radiology, BC Children's & Women's Health Center, 4480 Oak Street, Vancouver, British Columbia, V6H 3V4, Canada
| | - Steven P Miller
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada; Neurosciences and Mental Health Program, Research Institute, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada; University of Toronto, Department of Pediatrics, 563 Spadina Crescent, Toronto, Ontario, M5S 2J7, Canada; Child & Family Research Institute, 950 28th Avenue, Vancouver, British Columbia, V5Z 4H4, Canada
| |
Collapse
|
198
|
Meyer U. Prenatal poly(i:C) exposure and other developmental immune activation models in rodent systems. Biol Psychiatry 2014; 75:307-15. [PMID: 23938317 DOI: 10.1016/j.biopsych.2013.07.011] [Citation(s) in RCA: 458] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/18/2013] [Accepted: 07/04/2013] [Indexed: 02/08/2023]
Abstract
It is increasingly appreciated that altered neuroimmune mechanisms might play a role in the development of schizophrenia and related psychotic illnesses. On the basis of human epidemiological findings, a number of translational rodent models have been established to explore the consequences of prenatal immune activation on brain and behavioral development. The currently existing models are based on maternal gestational exposure to human influenza virus, the viral mimic polyriboinosinic-polyribocytidilic acid [Poly(I:C)], the bacterial endotoxin lipopolysaccharide, the locally acting inflammatory agent turpentine, or selected inflammatory cytokines. These models are pivotal for establishing causal relationships and for identifying cellular and molecular mechanisms that affect normal brain development in the event of early-life immune exposures. An important aspect of developmental immune activation models is that they allow a multi-faceted, longitudinal monitoring of the disease process as it unfolds during the course of neurodevelopment from prenatal to adult stages of life. An important recent refinement of these models is the incorporation of multiple etiologically relevant risk factors by combining prenatal immune challenges with specific genetic manipulations or additional environmental adversities. Converging findings from such recent experimental attempts suggest that prenatal infection can act as a "neurodevelopmental disease primer" that is likely relevant for a number of chronic mental illnesses. Hence, the adverse effects induced by prenatal infection might reflect an early entry into the neuropsychiatric route, but the specificity of subsequent disease or symptoms is likely to be strongly influenced by the genetic and environmental context in which the prenatal infectious process occurs.
Collapse
Affiliation(s)
- Urs Meyer
- Physiology and Behavior Laboratory, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
199
|
Kichev A, Rousset CI, Baburamani AA, Levison SW, Wood TL, Gressens P, Thornton C, Hagberg H. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling and cell death in the immature central nervous system after hypoxia-ischemia and inflammation. J Biol Chem 2014; 289:9430-9. [PMID: 24509861 PMCID: PMC3979382 DOI: 10.1074/jbc.m113.512350] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family. The interaction of TRAIL with death receptor 4 (DR4) and DR5 can trigger apoptotic cell death. The aim of this study was to investigate the role of TRAIL signaling in neonatal hypoxia-ischemia (HI). Using a neonatal mouse model of HI, mRNA, and protein expression of TRAIL, DR5 and the TRAIL decoy receptors osteoprotegerin (OPG), mDcTRAILR1, and mDcTRAILR2 were determined. In vitro, mRNA expression of these genes was measured in primary neurons and oligodendrocyte progenitor cells (OPCs) after inflammatory cytokine (TNF-α/IFN-γ) treatment and/or oxygen and glucose deprivation (OGD). The toxicity of these various paradigms was also measured. The expression of TRAIL, DR5, OPG, and mDcTRAILR2 was significantly increased after HI. In vitro, inflammatory cytokines and OGD treatment significantly induced mRNAs for TRAIL, DR5, OPG, and mDcTRAILR2 in primary neurons and of TRAIL and OPG in OPCs. TRAIL protein was expressed primarily in microglia and astroglia, whereas DR5 co-localized with neurons and OPCs in vivo. OGD enhanced TNF-α/IFN-γ toxicity in both neuronal and OPC cultures. Recombinant TRAIL exerted toxicity alone or in combination with OGD and TNF-α/IFN-γ in primary neurons but not in OPC cultures. The marked increases in the expression of TRAIL and its receptors after cytokine exposure and OGD in primary neurons and OPCs were similar to those found in our animal model of neonatal HI. The toxicity of TRAIL in primary neurons suggests that TRAIL signaling participates in neonatal brain injury after inflammation and HI.
Collapse
Affiliation(s)
- Anton Kichev
- From the Centre for the Developing Brain, Perinatal Brain Injury Group, Kings College London, London SE1 7EH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Chicha L, Smith T, Guzman R. Stem cells for brain repair in neonatal hypoxia-ischemia. Childs Nerv Syst 2014; 30:37-46. [PMID: 24178233 DOI: 10.1007/s00381-013-2304-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/15/2022]
Abstract
Neonatal hypoxic-ischemic insults are a significant cause of pediatric encephalopathy, developmental delays, and spastic cerebral palsy. Although the developing brain's plasticity allows for remarkable self-repair, severe disruption of normal myelination and cortical development upon neonatal brain injury are likely to generate life-persisting sensory-motor and cognitive deficits in the growing child. Currently, no treatments are available that can address the long-term consequences. Thus, regenerative medicine appears as a promising avenue to help restore normal developmental processes in affected infants. Stem cell therapy has proven effective in promoting functional recovery in animal models of neonatal hypoxic-ischemic injury and therefore represents a hopeful therapy for this unmet medical condition. Neural stem cells derived from pluripotent stem cells or fetal tissues as well as umbilical cord blood and mesenchymal stem cells have all shown initial success in improving functional outcomes. However, much still remains to be understood about how those stem cells can safely be administered to infants and what their repair mechanisms in the brain are. In this review, we discuss updated research into pathophysiological mechanisms of neonatal brain injury, the types of stem cell therapies currently being tested in this context, and the potential mechanisms through which exogenous stem cells might interact with and influence the developing brain.
Collapse
Affiliation(s)
- L Chicha
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | |
Collapse
|