101
|
Chew LJ, Fusar-Poli P, Schmitz T. Oligodendroglial alterations and the role of microglia in white matter injury: relevance to schizophrenia. Dev Neurosci 2013; 35:102-29. [PMID: 23446060 PMCID: PMC4531048 DOI: 10.1159/000346157] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/07/2012] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a chronic and debilitating mental illness characterized by a broad range of abnormal behaviors, including delusions and hallucinations, impaired cognitive function, as well as mood disturbances and social withdrawal. Due to the heterogeneous nature of the disease, the causes of schizophrenia are very complex; its etiology is believed to involve multiple brain regions and the connections between them, and includes alterations in both gray and white matter regions. The onset of symptoms varies with age and severity, and there is some debate over a degenerative or developmental etiology. Longitudinal magnetic resonance imaging studies have detected progressive gray matter loss in the first years of disease, suggesting neurodegeneration; but there is also increasing recognition of a temporal association between clinical complications at birth and disease onset that supports a neurodevelopmental origin. Presently, neuronal abnormalities in schizophrenia are better understood than alterations in myelin-producing cells of the brain, the oligodendrocytes, which are the predominant constituents of white matter structures. Proper white matter development and its structural integrity critically impacts brain connectivity, which affects sensorimotor coordination and cognitive ability. Evidence of defective white matter growth and compromised white matter integrity has been found in individuals at high risk of psychosis, and decreased numbers of mature oligodendrocytes are detected in schizophrenia patients. Inflammatory markers, including proinflammatory cytokines and chemokines, are also associated with psychosis. A relationship between risk of psychosis, white matter defects and prenatal inflammation is being established. Animal models of perinatal brain injury are successful in producing white matter damage in the brain, typified by hypomyelination and/or dysmyelination, impaired motor coordination and prepulse inhibition of the acoustic startle reflex, recapitulating structural and functional characteristics observed in schizophrenia. In addition, elevated expression of inflammation-related genes in brain tissue and increased production of cytokines by blood cells from patients with schizophrenia indicate immunological dysfunction and abnormal inflammatory responses, which are also important underlying features in experimental models. Microglia, resident immune defenders of the central nervous system, play important roles in the development and protection of neural cells, but can contribute to injury under pathological conditions. This article discusses oligodendroglial changes in schizophrenia and focuses on microglial activity in the context of the disease, in neonatal brain injury and in various experimental models of white matter damage. These include disorders associated with premature birth, and animal models of perinatal bacterial and viral infection, oxygen deprivation (hypoxia) and excess (hyperoxia), and elevated systemic proinflammatory cytokine levels. We briefly review the effects of treatment with antipsychotic and anti-inflammatory agents in models of perinatal brain injury, and comment on the therapeutic potential of these strategies. By understanding the neurobiological basis of oligodendroglial abnormalities in schizophrenia, it is hoped that patients will benefit from the availability of targeted and more efficacious treatment options.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.
| | | | | |
Collapse
|
102
|
Fan LW, Kaizaki A, Tien LT, Pang Y, Tanaka S, Numazawa S, Bhatt AJ, Cai Z. Celecoxib attenuates systemic lipopolysaccharide-induced brain inflammation and white matter injury in the neonatal rats. Neuroscience 2013; 240:27-38. [PMID: 23485816 DOI: 10.1016/j.neuroscience.2013.02.041] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 11/25/2022]
Abstract
Lipopolysaccharide (LPS)-induced white matter injury in the neonatal rat brain is associated with inflammatory processes. Cyclooxygenase-2 (COX-2) can be induced by inflammatory stimuli, such as cytokines and pro-inflammatory molecules, suggesting that COX-2 may be considered as the target for anti-inflammation. The objective of the present study was to examine whether celecoxib, a selective COX-2 inhibitor, can reduce systemic LPS-induced brain inflammation and brain damage. Intraperitoneal (i.p.) injection of LPS (2mg/kg) was performed in postnatal day 5 (P5) of Sprague-Dawley rat pups and celecoxib (20mg/kg) or vehicle was administered i.p. 5 min after LPS injection. The body weight and wire-hanging maneuver test was performed 24h after the LPS exposure, and brain injury was examined after these tests. Systemic LPS exposure resulted in an impairment of behavioral performance and acute brain injury, as indicated by apoptotic death of oligodendrocytes (OLs) and loss of OL immunoreactivity in the neonatal rat brain. Treatments with celecoxib significantly reduced systemic LPS-induced neurobehavioral disturbance and brain damage. Celecoxib administration significantly attenuated systemic LPS-induced increments in the number of activated microglia and astrocytes, concentrations of IL-1β and TNFα, and protein levels of phosphorylated-p38 MAPK in the neonatal rat brain. The protection of celecoxib was also associated with a reduction of systemic LPS-induced COX-2+ cells which were double labeled with GFAP+ (astrocyte) cells. The overall results suggest that celecoxib was capable of attenuating the brain injury and neurobehavioral disturbance induced by systemic LPS exposure, and the protective effects are associated with its anti-inflammatory properties.
Collapse
Affiliation(s)
- L-W Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216-4505, USA.
| | | | | | | | | | | | | | | |
Collapse
|
103
|
Wang KC, Fan LW, Kaizaki A, Pang Y, Cai Z, Tien LT. Neonatal lipopolysaccharide exposure induces long-lasting learning impairment, less anxiety-like response and hippocampal injury in adult rats. Neuroscience 2013; 234:146-57. [PMID: 23298854 DOI: 10.1016/j.neuroscience.2012.12.049] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/27/2012] [Accepted: 12/28/2012] [Indexed: 12/20/2022]
Abstract
Infection during early neonatal period has been shown to cause lasting neurological disabilities and is associated with the subsequent impairment in development of learning and memory ability and anxiety-related behavior in adults. We have previously reported that neonatal lipopolysaccharide (LPS) exposure resulted in cognitive deficits in juvenile rats (P21); thus, the goal of the present study was to determine whether neonatal LPS exposure has long-lasting effects in adult rats. After an LPS (1mg/kg) intracerebral (i.c.) injection in postnatal day 5 (P5) Sprague-Dawley female rat pups, neurobehavioral tests were carried out on P21 and P22, P49 and P50 or P70 and P71 and brain injury was examined at 66days after LPS injection (P71). Our data indicate that neonatal LPS exposure resulted in learning deficits in the passive avoidance task, less anxiety-like (anxiolytic-like) responses in the elevated plus-maze task, reductions in the hippocampal volume and the number of neuron-specific nuclear protein (NeuN)+ cells, as well as axonal injury in the CA1 region of the middle dorsal hippocampus in P71 rats. Neonatal LPS exposure also resulted in sustained inflammatory responses in the P71 rat hippocampus, as indicated by an increased number of activated microglia and elevation of interleukin-1β content in the rat hippocampus. This study reveals that neonatal LPS exposure causes persistent injuries to the hippocampus and results in long-lasting learning disabilities, and these effects are related to the chronic inflammation in the rat hippocampus.
Collapse
Affiliation(s)
- K-C Wang
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
104
|
Brunssen SH, Moy SS, Toews AD, McPherson CA, Harry GJ. Interleukin-6 (IL-6) receptor/IL-6 fusion protein (Hyper IL-6) effects on the neonatal mouse brain: possible role for IL-6 trans-signaling in brain development and functional neurobehavioral outcomes. Brain Behav Immun 2013; 27:42-53. [PMID: 22985907 PMCID: PMC3565379 DOI: 10.1016/j.bbi.2012.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 08/17/2012] [Accepted: 08/30/2012] [Indexed: 12/30/2022] Open
Abstract
Adverse neurodevelopmental outcomes are linked to perinatal production of inflammatory mediators, including interleukin 6 (IL-6). While a pivotal role for maternal elevation in IL-6 has been established in determining neurobehavioral outcomes in the offspring and considered the primary target mediating the fetal inflammatory response, questions remain as to the specific actions of IL-6 on the developing brain. CD-1 male mice received a subdural injection of the bioactive fusion protein, hyper IL-6 (HIL-6) on postnatal-day (PND)4 and assessed from preweaning until adulthood. Immunohistochemical evaluation of astrocytes and microglia and mRNA levels for pro-inflammatory cytokines and host response genes indicated no evidence of an acute neuroinflammatory injury response. HIL-6 accelerated motor development and increased reactivity to stimulation and number of entries in a light/dark chamber, decreased ability to learn to withhold a response in passive avoidance, and effected deficits in social novelty behavior. No changes were observed in motor activity, pre-pulse startle inhibition, or learning and memory in the Morris water maze or radial arm maze, as have been reported for models of more severe developmental neuroinflammation. In young animals, mRNA levels for MBP and PLP/DM20 decreased and less complexity of MBP processes in the cortex was evident by immunohistochemistry. The non-hydroxy cerebroside fraction of cerebral lipids was increased. These results provide evidence for selective effects of IL-6 signaling, particularly trans-signaling, in the developing brain in the absence of a general neuroinflammatory response. These data contribute to our further understanding of the multiple aspects of IL-6 signaling in the developing brain.
Collapse
Affiliation(s)
- Susan H. Brunssen
- School of Nursing, University of North Carolina, North Carolina
- Carolina Institute for Developmental Disabilities, University of North Carolina, North Carolina
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Sheryl S. Moy
- Carolina Institute for Developmental Disabilities, University of North Carolina, North Carolina
| | - Arrel D. Toews
- Department of Cellular and Molecular Biology, University of North Carolina, North Carolina
| | - Christopher A. McPherson
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - G. Jean Harry
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| |
Collapse
|
105
|
Wang Y, Yin P, Huang S, Wang J, Sun R. Ethyl pyruvate protects against lipopolysaccharide-induced white matter injury in the developing rat brain. Int J Dev Neurosci 2012; 31:181-8. [PMID: 23280059 DOI: 10.1016/j.ijdevneu.2012.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 12/17/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022] Open
Abstract
The neuroprotective effects of ethyl pyruvate (EP) have been proved in several brain injury models, yet very little is known about its action on neonatal white matter injury. To investigate the effect of EP on white matte damage, a stereotactic intracerebral injection of lipopolysaccharide (LPS, 1mg/kg) was performed on postnatal day 5 Sprague-Dawley rat pups, and EP was administrated intraperitoneally at a dose of 40mg/kg immediately, 1h and 12h after LPS exposure. Significantly, treatment with EP reduced LPS-induced ventricle dilation, loss of O4+ and O1+ oligodendrocytes, apoptosis of oligodendrocytes, and hypomyelination. The protective effect of EP was associated with suppressed inflammatory responses, indicated by the inhibition of activation of microglia and astrocytes, as well as the decreased expression of tumor necrosis factor-alpha (TNF-α) and interleukin-1beta (IL-1β) in rat brains. Also, EP prevented the elevation of cleaved caspase-3 in periventricular white matter tissue after LPS insult. Taken together, these results suggest that EP confers potent protection against LPS-induced white matter injury via its anti-inflammatory and anti-apoptotic properties.
Collapse
Affiliation(s)
- Yingyan Wang
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | | | | | | | | |
Collapse
|
106
|
He YF, Chen HJ, Qian LH, He LF, Buzby JS. Diphenyleneiodonium protects preoligodendrocytes against endotoxin-activated microglial NADPH oxidase-generated peroxynitrite in a neonatal rat model of periventricular leukomalacia. Brain Res 2012; 1492:108-21. [PMID: 23174417 DOI: 10.1016/j.brainres.2012.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 10/06/2012] [Accepted: 11/12/2012] [Indexed: 01/17/2023]
Abstract
The contribution of microglial activation to preoligodendroglial (preOL) damage in the central nervous system (CNS) is considered to be one of the principal causes of periventricular leukomalacia (PVL) pathogenesis. The present study explores the effect of diphenyleneiodonium (DPI), a NADPH oxidase (NOX) inhibitor, on protection of preOLs from bacterial lipopolysaccharide (LPS)-induced microglial toxicity in vivo and in vitro. In vitro, preOLs co-cultured with microglia exhibited increased preOL apoptosis, accompanied by overproduction of superoxide anion (O(2)(-)) and the formation of peroxynitrite (ONOO(-)) after LPS exposure. LPS also significantly up-regulated accumulation of activated microglial NOX subunits p67-phox and gp91-phox in the plasma membrane. Diphenyleneiodonium (DPI) (10μm) was found to significantly attenuate up-regulation of this NOX activity. In vivo, DPI was administered (1mg/kg/day) by subcutaneous injection for 3 days to two-day-old neonatal Sprague-Dawley rats subjected to intracerebral injection of LPS. Treatment with DPI within 24h of LPS injection significantly ameliorated white matter injury, decreasing preOL loss, O(2)(-) generation, and ONOO(-) formation, and inhibiting p67-phox, gp91-phox synthesis and p67phox membrane translocation in microglia. These results indicated that LPS-induced preOL apoptosis may have been mediated by microglia-derived ONOO(-). DPI prevented this LPS-induced brain injury, most likely by inhibiting ONOO(-) formation via NOX, thereby preventing preOL loss and immature white matter injury.
Collapse
Affiliation(s)
- Ya-Fang He
- Shanghai Institute for Pediatric Research, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai 200092, China.
| | | | | | | | | |
Collapse
|
107
|
Brehmer F, Bendix I, Prager S, van de Looij Y, Reinboth BS, Zimmermanns J, Schlager GW, Brait D, Sifringer M, Endesfelder S, Sizonenko S, Mallard C, Bührer C, Felderhoff-Mueser U, Gerstner B. Interaction of inflammation and hyperoxia in a rat model of neonatal white matter damage. PLoS One 2012; 7:e49023. [PMID: 23155446 PMCID: PMC3498343 DOI: 10.1371/journal.pone.0049023] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 10/03/2012] [Indexed: 12/27/2022] Open
Abstract
Intrauterine infection and inflammation are major reasons for preterm birth. The switch from placenta-mediated to lung-mediated oxygen supply during birth is associated with a sudden rise of tissue oxygen tension that amounts to relative hyperoxia in preterm infants. Both infection/inflammation and hyperoxia have been shown to be involved in brain injury of preterm infants. Hypothesizing that they might be additive or synergistic, we investigated the influence of a systemic lipopolysaccharide (LPS) application on hyperoxia-induced white matter damage (WMD) in newborn rats. Three-day-old Wistar rat pups received 0.25 mg/kg LPS i.p. and were subjected to 80% oxygen on P6 for 24 h. The extent of WMD was assessed by immunohistochemistry, western blots, and diffusion tensor (DT) magnetic resonance imaging (MRI). In addition, the effects of LPS and hyperoxia were studied in an in vitro co-culture system of primary rat oligodendrocytes and microglia cells. Both noxious stimuli, hyperoxia, and LPS caused hypomyelination as revealed by western blot, immunohistochemistry, and altered WM microstructure on DT-MRI. Even so, cellular changes resulting in hypomyelination seem to be different. While hyperoxia induces cell death, LPS induces oligodendrocyte maturity arrest without cell death as revealed by TUNEL-staining and immunohistological maturation analysis. In the two-hit scenario cell death is reduced compared with hyperoxia treated animals, nevertheless white matter alterations persist. Concordantly with these in vivo findings we demonstrate that LPS pre-incubation reduced premyelinating-oligodendrocyte susceptibility towards hyperoxia in vitro. This protective effect might be caused by upregulation of interleukin-10 and superoxide dismutase expression after LPS stimulation. Reduced expression of transcription factors controlling oligodendrocyte development and maturation further indicates oligodendrocyte maturity arrest. The knowledge about mechanisms that triggered hypomyelination contributes to a better understanding of WMD in premature born infants.
Collapse
Affiliation(s)
- Felix Brehmer
- Department of Neonatology, Charité University Medical Center, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Doehner J, Genoud C, Imhof C, Krstic D, Knuesel I. Extrusion of misfolded and aggregated proteins--a protective strategy of aging neurons? Eur J Neurosci 2012; 35:1938-50. [PMID: 22708604 DOI: 10.1111/j.1460-9568.2012.08154.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cellular senescence is the consequence of repetitive exposures to oxidative stress, perturbed energy homeostasis, accumulation of damaged proteins and lesions in their nucleic acids. Whereas mitotic cells are equipped with efficient cell replacement strategies; postmitotic neurons have--with a few exceptions--no mechanism to substitute dysfunctional cells within a complex neuronal network. Here we propose a potential strategy by which aging neurons contend against abnormal accumulation of damaged/misfolded proteins. The suggested mechanism involves the formation of 'budding-like' extrusions and their subsequent clearance by glia. This hypothesis emerged from our previous investigations of the aged hippocampus revealing layer-specific accumulations of Reelin, a glycoprotein with fundamental roles during brain development and adult synaptic plasticity. We showed that Reelin deposits constitute a conserved neuropathological feature of aging, which is significantly accelerated in adult wild-type mice prenatally exposed to a viral-like infection. Here, we employed two- and three-dimensional immunoelectron microscopy to elucidate their morphological properties, localization and origin in immune challenged vs. control mice. In controls, Reelin-positive deposits were dispersed in the neuropil, some being engulfed by glia. In immune challenged mice, however, significantly more Reelin-immunoreactive deposits were associated with neuritic swellings containing mitochondria, vacuoles and cellular debris, pointing to their intracellular origin and suggesting that 'budding-like' neuronal extrusions of misfolded proteins and glial clearance may represent a protective strategy to counteract aging-associated impairments in proteosomal/lysosomal degradation. Neurons exposed to chronic neuroinflammation with increased levels of misfolded/damaged proteins, however, may fail to combat intraneuronal protein accumulations, a process probably underlying neuronal dysfunction and degeneration during aging.
Collapse
Affiliation(s)
- Jana Doehner
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, Zurich, Switzerland
| | | | | | | | | |
Collapse
|
109
|
Barateiro A, Miron VE, Santos SD, Relvas JB, Fernandes A, ffrench-Constant C, Brites D. Unconjugated Bilirubin Restricts Oligodendrocyte Differentiation and Axonal Myelination. Mol Neurobiol 2012; 47:632-44. [DOI: 10.1007/s12035-012-8364-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 10/08/2012] [Indexed: 01/05/2023]
|
110
|
Cao C, Yu X, Liao Z, Zhu N, Huo H, Wang M, Ji G, She H, Luo Z, Yue S. Hypertonic saline reduces lipopolysaccharide-induced mouse brain edema through inhibiting aquaporin 4 expression. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:R186. [PMID: 23036239 PMCID: PMC3682288 DOI: 10.1186/cc11670] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 10/04/2012] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Three percent sodium chloride (NaCl) treatment has been shown to reduce brain edema and inhibited brain aquaporin 4 (AQP4) expression in bacterial meningitis induced by Escherichia coli. Lipopolysaccharide (LPS) is the main pathogenic component of E. coli. We aimed to explore the effect of 3% NaCl in mouse brain edema induced by LPS, as well as to elucidate the potential mechanisms of action. METHODS Three percent NaCl was used to treat cerebral edema induced by LPS in mice in vivo. Brain water content, IL-1β, TNFα, immunoglobulin G (IgG), AQP4 mRNA and protein were measured in brain tissues. IL-1β, 3% NaCl and calphostin C (a specific inhibitor of protein kinase C) were used to treat the primary astrocytes in vitro. AQP4 mRNA and protein were measured in astrocytes. Differences in various groups were determined by one-way analysis of variance. RESULTS Three percent NaCl attenuated the increase of brain water content, IL-1β, TNFα, IgG, AQP4 mRNA and protein in brain tissues induced by LPS. Three percent NaCl inhibited the increase of AQP4 mRNA and protein in astrocytes induced by IL-1β in vitro. Calphostin C blocked the decrease of AQP4 mRNA and protein in astrocytes induced by 3% NaCl in vitro. CONCLUSIONS Osmotherapy with 3% NaCl ameliorated LPS-induced cerebral edema in vivo. In addition to its osmotic force, 3% NaCl exerted anti-edema effects possibly through down-regulating the expression of proinflammatory cytokines (IL-1β and TNFα) and inhibiting the expression of AQP4 induced by proinflammatory cytokines. Three percent NaCl attenuated the expression of AQP4 through activation of protein kinase C in astrocytes.
Collapse
|
111
|
Harvey L, Boksa P. Prenatal and postnatal animal models of immune activation: Relevance to a range of neurodevelopmental disorders. Dev Neurobiol 2012; 72:1335-48. [DOI: 10.1002/dneu.22043] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 06/18/2012] [Indexed: 11/11/2022]
|
112
|
Chhor V, Schang AL, Favrais G, Fleiss B, Gressens P. [Long-term cerebral effects of perinatal inflammation]. Arch Pediatr 2012; 19:946-52. [PMID: 22885003 DOI: 10.1016/j.arcped.2012.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 06/21/2012] [Indexed: 12/12/2022]
Abstract
Perinatal inflammation can lead to fetal/neonatal inflammatory syndrome, a risk factor for brain lesions, especially in the white matter. Perinatal inflammation is associated with increased incidence of cerebral palsy in humans and animal models and there is a strong relationship with increased incidence of autism and schizophrenia in humans. Perinatal inflammation causes acute microglial and astroglial activation, blood-brain barrier dysfunction, and disrupts oligodendrocyte maturation leading to hypomyelination. Inflammation also sensitizes the brain to additional perinatal insults, including hypoxia-ischemia. Furthermore, long after the primary cause of inflammation has resolved, gliosis may also persist and predispose to neurodegenerative diseases including Alzheimer's and Parkinson's disease, but this relation is still hypothetical. Finding of acute and chronic changes in brain structure and function due to perinatal inflammation highlights the need for treatments. As gliosis appears to be involved in the acute and chronic effects of perinatal inflammation, modulating the glial phenotype may be an effective strategy to prevent damage to the brain.
Collapse
Affiliation(s)
- V Chhor
- Inserm U676, hôpital Robert-Debré, 48, boulevard Sérurier, 75019 Paris, France
| | | | | | | | | |
Collapse
|
113
|
Bilbo SD, Schwarz JM. The immune system and developmental programming of brain and behavior. Front Neuroendocrinol 2012; 33:267-86. [PMID: 22982535 PMCID: PMC3484177 DOI: 10.1016/j.yfrne.2012.08.006] [Citation(s) in RCA: 402] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/28/2012] [Accepted: 08/29/2012] [Indexed: 12/16/2022]
Abstract
The brain, endocrine, and immune systems are inextricably linked. Immune molecules have a powerful impact on neuroendocrine function, including hormone-behavior interactions, during health as well as sickness. Similarly, alterations in hormones, such as during stress, can powerfully impact immune function or reactivity. These functional shifts are evolved, adaptive responses that organize changes in behavior and mobilize immune resources, but can also lead to pathology or exacerbate disease if prolonged or exaggerated. The developing brain in particular is exquisitely sensitive to both endogenous and exogenous signals, and increasing evidence suggests the immune system has a critical role in brain development and associated behavioral outcomes for the life of the individual. Indeed, there are associations between many neuropsychiatric disorders and immune dysfunction, with a distinct etiology in neurodevelopment. The goal of this review is to describe the important role of the immune system during brain development, and to discuss some of the many ways in which immune activation during early brain development can affect the later-life outcomes of neural function, immune function, mood and cognition.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, 572 Research Drive, Box 91050, Durham, NC 27708, USA.
| | | |
Collapse
|
114
|
Intracerebral lipopolysaccharide induces neuroinflammatory change and augmented brain injury in growth-restricted neonatal rats. Pediatr Res 2012; 71:645-52. [PMID: 22337231 PMCID: PMC3601589 DOI: 10.1038/pr.2012.26] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Intrauterine growth restriction (IUGR) alters fetal development and is associated with neurodevelopmental abnormalities. We hypothesized that growth restriction from reduced intrauterine perfusion would predispose neonatal rats to subsequent inflammatory brain injury. METHODS In this study, IUGR was achieved by induced placental insufficiency in pregnant rats at 14 days of gestation. IUGR offspring and sham-operated control pups were subsequently injected with intracerebral lipopolysaccharide (LPS) as a model of periventricular leukomalacia (PVL). RESULTS LPS similarly elevates proinflammatory cytokines in the brains of both IUGR and control rat pups. However, the chemokines cytokine-induced neutrophil chemoattractant-1 (CINC-1) and macrophage chemoattractant protein-1 (MCP-1), as well as microglia activation, were significantly higher in LPS-treated IUGR rat pups as compared with LPS-treated controls. In addition to the unique brain inflammatory response, IUGR rat pups demonstrated increased brain damage with an increased number of apoptotic cells, larger lateral ventricular size, and more severe impairment of myelination. DISCUSSION This study provides evidence that placental insufficiency may sensitize the innate immune system in the immature brain and reveals a possible link between brain inflammation and injury.
Collapse
|
115
|
Weaver-Mikaere L, Gibbons HM, De Silva D, Fraser M. Primary mixed glial cultures from fetal ovine forebrain are a valid model of inflammation-mediated white matter injury. Dev Neurosci 2012; 34:30-42. [PMID: 22627272 DOI: 10.1159/000338039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Astrocytes, microglial cells and oligodendrocytes (OLs) have been employed separately in vitro to assess cellular pathways following a variety of stimuli. Mixed glial cell cultures, however, have not been utilized to the same extent, despite the observed discrepancy in outcomes resulting from cell-to-cell contact of different glia in culture. Our objective was to standardize and morphologically characterize a primary culture of preterm ovine glial cells in order to attain a relevant in vitro model to assess the intracellular effects of infection and inflammation. This would provide a high-throughput model necessary for in-depth studies on the various pathophysiological mechanisms of white matter injury (WMI), which may occur in the preterm infant as a consequence of maternal infection or the fetal inflammatory response. Glial cells from the forebrains of 0.65-gestation ovine fetuses (comparable to 24- to 26-week human fetal brain development) were mechanically and enzymatically isolated and plated at a final density of 250,000 cells per well. When reaching confluence at 5 days after plating, the cultures contained astrocytes, microglial cells, as well as progenitor, precursor and immature OLs. Glial cell morphology and phenotypic immunoreactivity were characteristic of and consistent with previous observations of separately cultured cell types. To determine the effects of infection or inflammation in our in vitro model, we then treated mixed glial cultures with tumour necrosis factor-α (TNF-α; 50 or 100 ng/ml) or lipopolysaccharide (LPS; 1 µg/ml) for a period of 48 h. Cytokine levels were measured by ELISA and cell numbers for specific glial cell types were determined along with OL proliferation and apoptosis by Ki67 and caspase-3 immunocytochemistry, respectively. Our results showed that exposure to TNF-α or LPS resulted in a characteristic inflammatory response entailed by up-regulation of pro-inflammatory cytokines, a lack of astrogliosis and a marked reduction in OLs attributable to increased apoptosis. In LPS-treated cultures, there was a marked increase in the pro-inflammatory cytokine TNF-α at both 24 and 48 h. In conclusion, this is the first report of the immunocytochemical description and characterization of fetal ovine-derived mixed glial cell primary cultures. This in vitro model provides a novel and efficient system to explore the mechanisms of infection/inflammation-mediated WMI at the cellular level and for screening candidate therapeutic strategies.
Collapse
Affiliation(s)
- Luke Weaver-Mikaere
- The Liggins Institute, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
116
|
Grin’kina NM, Karnabi EE, Damania D, Wadgaonkar S, Muslimov IA, Wadgaonkar R. Sphingosine kinase 1 deficiency exacerbates LPS-induced neuroinflammation. PLoS One 2012; 7:e36475. [PMID: 22615770 PMCID: PMC3355156 DOI: 10.1371/journal.pone.0036475] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 04/09/2012] [Indexed: 11/24/2022] Open
Abstract
The pathogenesis of inflammation in the central nervous system (CNS), which contributes to numerous neurodegenerative diseases and results in encephalopathy and neuroinflammation, is poorly understood. Sphingolipid metabolism plays a crucial role in maintaining cellular processes in the CNS, and thus mediates the various pathological consequences of inflammation. For a better understanding of the role of sphingosine kinase activation during neuroinflammation, we developed a bacterial lipopolysaccharide (LPS)-induced brain injury model. The onset of the inflammatory response was observed beginning 4 hours after intracerebral injection of LPS into the lateral ventricles of the brain. A comparison of established neuroinflammatory parameters such as white matter rarefactions, development of cytotoxic edema, astrogliosis, loss of oligodendrocytes, and major cytokines levels in wild type and knockout mice suggested that the neuroinflammatory response in SphK1-/- mice was significantly upregulated. At 6 hours after intracerebroventricular injection of LPS in SphK1-/- mice, the immunoreactivity of the microglia markers and astrocyte marker glial fibrillary acidic protein (GFAP) were significantly increased, while the oligodendrocyte marker O4 was decreased compared to WT mice. Furthermore, western blotting data showed increased levels of GFAP. These results suggest that SphK1 activation is involved in the regulation of LPS induced brain injury. RESEARCH HIGHLIGHTS: • Lipopolysaccharide (LPS) intracerebral injection induces severe neuroinflammation. • Sphingosine kinase 1 deletion worsens the effect of the LPS. • Overexpression of SphK1 might be a potential new treatment approach to neuroinflammation.
Collapse
Affiliation(s)
- Natalia M. Grin’kina
- SUNY Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Research and Development VA Medical Center, Brooklyn, New York, United States of America
| | - Eddy E. Karnabi
- SUNY Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Research and Development VA Medical Center, Brooklyn, New York, United States of America
| | - Dushyant Damania
- SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Sunil Wadgaonkar
- Department of Research and Development VA Medical Center, Brooklyn, New York, United States of America
| | - Ilham A. Muslimov
- SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Raj Wadgaonkar
- SUNY Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Research and Development VA Medical Center, Brooklyn, New York, United States of America
| |
Collapse
|
117
|
Pang Y, Fan LW, Zheng B, Campbell LR, Cai Z, Rhodes PG. Dexamethasone and betamethasone protect against lipopolysaccharide-induced brain damage in neonatal rats. Pediatr Res 2012; 71:552-8. [PMID: 22314662 PMCID: PMC3609027 DOI: 10.1038/pr.2012.9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION The aim of this study was to test whether dexamethasone (Dex) and betamethasone (Beta), two of the most commonly used corticosteroids, protect against lipopolysaccharide (LPS)-induced white matter damage and neurobehavioral dysfunction. METHODS LPS or sterile saline was injected into the brain white matter of rat pups at postnatal day 5 (P5), and Dex or Beta was given intraperitoneally to the rat pups 1 h before the LPS microinjection. Brain inflammatory response, brain damage, and myelination were examined at P6, P8, and P14. Neurobehavioral tests were performed from P3 through P22. RESULTS Our results demonstrate that Dex and Beta markedly diminish the LPS-induced brain inflammatory response, restore myelin basic protein (MBP) expression, and alleviate lateral ventricle dilation. Both corticosteroids demonstrate significant protection against most LPS-induced behavioral deficits, including those in rearing, vibrissa-elicited forelimb-placing, beam walking, learning, and elevated plus-maze test. Of note, only Beta improved the locomotion and stereotype dysfunction. In contrast to their beneficial effects, neither drug prevented LPS-induced delay in body weight gain from P6 through P21. DISCUSSION Our study suggests that if their adverse effects are minimized, corticosteroids may be the potential candidate drugs to prevent brain damage in premature infants.
Collapse
Affiliation(s)
- Yi Pang
- Department of Pediatrics (Y.P., LW.F., B.Z., L.R.C, Z.C., and P.G.R.), University of Mississippi Medical Center, Jackson, MS 39216
| | - Lir-Wan Fan
- Department of Pediatrics (Y.P., LW.F., B.Z., L.R.C, Z.C., and P.G.R.), University of Mississippi Medical Center, Jackson, MS 39216
| | - Baoying Zheng
- Department of Pediatrics (Y.P., LW.F., B.Z., L.R.C, Z.C., and P.G.R.), University of Mississippi Medical Center, Jackson, MS 39216
| | - Leigh R. Campbell
- Department of Pediatrics (Y.P., LW.F., B.Z., L.R.C, Z.C., and P.G.R.), University of Mississippi Medical Center, Jackson, MS 39216
| | - Zhengwei Cai
- Department of Pediatrics (Y.P., LW.F., B.Z., L.R.C, Z.C., and P.G.R.), University of Mississippi Medical Center, Jackson, MS 39216
| | - Philip G. Rhodes
- Department of Pediatrics (Y.P., LW.F., B.Z., L.R.C, Z.C., and P.G.R.), University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
118
|
The role of cytokines and inflammatory cells in perinatal brain injury. Neurol Res Int 2012; 2012:561494. [PMID: 22530124 PMCID: PMC3317045 DOI: 10.1155/2012/561494] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 11/25/2011] [Accepted: 12/13/2011] [Indexed: 12/25/2022] Open
Abstract
Perinatal brain injury frequently complicates preterm birth and leads to significant long-term morbidity. Cytokines and inflammatory cells are mediators in the common pathways associated with perinatal brain injury induced by a variety of insults, such as hypoxic-ischemic injury, reperfusion injury, toxin-mediated injury, and infection. This paper examines our current knowledge regarding cytokine-related perinatal brain injury and specifically discusses strategies for attenuating cytokine-mediated brain damage.
Collapse
|
119
|
Bilbo SD, Smith SH, Schwarz JM. A lifespan approach to neuroinflammatory and cognitive disorders: a critical role for glia. J Neuroimmune Pharmacol 2012; 7:24-41. [PMID: 21822589 PMCID: PMC3267003 DOI: 10.1007/s11481-011-9299-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/14/2011] [Indexed: 12/15/2022]
Abstract
Cognitive decline is a common problem of aging. Whereas multiple neural and glial mechanisms may account for these declines, microglial sensitization and/or dystrophy has emerged as a leading culprit in brain aging and dysfunction. However, glial activation is consistently observed in normal brain aging as well, independent of frank neuroinflammation or functional impairment. Such variability suggests the existence of additional vulnerability factors that can impact neuronal-glial interactions and thus overall brain and cognitive health. The goal of this review is to elucidate our working hypothesis that an individual's risk or resilience to neuroinflammatory disorders and poor cognitive aging may critically depend on their early life experience, which can change immune reactivity within the brain for the remainder of the lifespan. For instance, early-life infection in rats can profoundly disrupt memory function in young adulthood, as well as accelerate age-related cognitive decline, both of which are linked to enduring changes in glial function that occur in response to the initial infection. We discuss these findings within the context of the growing literature on the role of immune molecules and neuroimmune crosstalk in normal brain development. We highlight the intrinsic factors (e.g., chemokines, hormones) that regulate microglial development and their colonization of the embryonic and postnatal brain, and the capacity for disruption or "re-programming" of this crucial process by external events (e.g., stress, infection). An impact on glia, which in turn alters neural development, has the capacity to profoundly impact cognitive and mental health function at all stages of life.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology & Neuroscience, Duke University, Durham, NC 27708, USA.
| | | | | |
Collapse
|
120
|
Titomanlio L, Kavelaars A, Dalous J, Mani S, El Ghouzzi V, Heijnen C, Baud O, Gressens P. Stem cell therapy for neonatal brain injury: perspectives and challenges. Ann Neurol 2012; 70:698-712. [PMID: 22162055 DOI: 10.1002/ana.22518] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebral palsy is a major health problem caused by brain damage during pregnancy, delivery, or the immediate postnatal period. Perinatal stroke, intraventricular hemorrhage, and asphyxia are the most common causes of neonatal brain damage. Periventricular white matter damage (periventricular leukomalacia) is the predominant form in premature infants and the most common antecedent of cerebral palsy. Stem cell treatment has proven effective in restoring injured organs and tissues in animal models. The potential of stem cells for self-renewal and differentiation translates into substantial neuroprotection and neuroregeneration in the animal brain, with minimal risks of rejection and side effects. Stem cell treatments described to date have used neural stem cells, embryonic stem cells, mesenchymal stem cells, umbilical cord stem cells, and induced pluripotent stem cells. Most of these treatments are still experimental. In this review, we focus on the efficacy of stem cell therapy in animal models of cerebral palsy, and discuss potential implications for current and future clinical trials.
Collapse
|
121
|
Pang Y, Zheng B, Kimberly SL, Cai Z, Rhodes PG, Lin RCS. Neuron-oligodendrocyte myelination co-culture derived from embryonic rat spinal cord and cerebral cortex. Brain Behav 2012; 2:53-67. [PMID: 22574274 PMCID: PMC3343299 DOI: 10.1002/brb3.33] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 12/13/2011] [Accepted: 12/18/2011] [Indexed: 02/07/2023] Open
Abstract
An in vitro myelination model derived from rat central nervous system (CNS) remains to be established. Here, we describe a simple and reproducible myelination culture method using dissociated neuron-oligodendrocyte (OL) co-cultures from either the embryonic day 16 (E16) rat spinal cord or cerebral cortex. The dissociated cells are plated directly on poly-L-lysine-coated cover slips and maintained in a modified myelination medium that supports both OL and neuron differentiation. The spinal cord derived OL progenitor cells develop quickly into myelin basic protein (MBP)+ mature OLs and start to myelinate axons around 17 days in vitro (DIV17). Myelination reaches its peak around six weeks (DIV40) and the typical nodes of Ranvier are revealed by paranodal proteins Caspr and juxaparanodal protein Kv1.2 immunoreactivity. Electron microscopy (EM) shows typical myelination cytoarchitecture and synaptic organization. In contrast, the cortical-derived co-culture requires triiodothyronine (T3) in the culture medium for myelination. Finally, either hypomyelination and/or demyelination can be induced by exposing proinflammatory cytokines or demyelinating agents to the co-culture, suggesting the feasibility of this modified in vitro myelination model for myelin-deficit investigation.
Collapse
Affiliation(s)
- Yi Pang
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Baoying Zheng
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Simpson L. Kimberly
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Zhengwei Cai
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Philip G. Rhodes
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Rick C. S. Lin
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216
| |
Collapse
|
122
|
Bsibsi M, Nomden A, van Noort JM, Baron W. Toll-like receptors 2 and 3 agonists differentially affect oligodendrocyte survival, differentiation, and myelin membrane formation. J Neurosci Res 2011; 90:388-98. [PMID: 21971760 DOI: 10.1002/jnr.22767] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 07/08/2011] [Accepted: 07/17/2011] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) play a key role in controlling innate immune responses to a wide variety of pathogen-associated molecules as well as endogenous signals. In addition, TLR expression within nonimmune cells has been recognized as as modulator of cell behavior. In this study we have addressed the question of whether functional TLRs are expressed on oligodendrocytes, the myelinating cells of the central nervous system. Primary cultures of rat oligodendrocytes at different maturation stages were found to express TLR2 and, to lesser extent, TLR3. Immunocytochemical analysis revealed that both TLRs were localized at the cell body and primary processes and were excluded from myelin-like membranes. Interestingly, innate immune receptor ligands were able to modulate oligodendrocyte survival, differentiation, and myelin-like membrane formation, indicating that TLRs on oligodendrocytes are functional. In highly purified oligodendrocytes cultures, the TLR2 agonist zymosan promoted survival, differentiation, and myelin-like membrane formation, whereas poly-I:C, a TLR3 ligand, was a potent inducer of apoptosis. Together, these data indicate that, in addition to other neural cell types, also oligodendrocytes express functional TLRs, which play a role in regulating various aspects of oligodendrocyte behavior.
Collapse
|
123
|
Cai Z, Fan LW, Lin S, Pang Y, Rhodes PG. Intranasal administration of insulin-like growth factor-1 protects against lipopolysaccharide-induced injury in the developing rat brain. Neuroscience 2011; 194:195-207. [PMID: 21840378 DOI: 10.1016/j.neuroscience.2011.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 08/01/2011] [Accepted: 08/01/2011] [Indexed: 11/27/2022]
Abstract
Our previous studies show that insulin-like growth factor-1 (IGF-1) can either protect against or increase lipopolysaccharide (LPS)-induced damage in the developing brain, depending on the dose, when it is co-administered with LPS through intracerebral injection. To further explore effects of IGF-1 on central inflammation associated brain injury, IGF-1 was administered through intranasal infusion in the current study. Postnatal day 5 (P5) rats were exposed to LPS at a dose of 1 μg/g body weight or sterile saline through intracerebral injection. Recombinant human insulin-like growth factor-1 (rhIGF-1) at a dose of 50 μg/pup or vehicle was administered intranasally 1 or 2 h after the LPS injection. Neonatal LPS exposure resulted in oligodendrocyte (OL) and white matter injury in the P6 or P21 rat brain. The damages include dilatation of lateral ventricles, pyknotic cell death, loss of OL progenitor cells and mature OLs in the cingulum area, and impairment of myelination at the corpus callosum area. Neurological dysfunctions were observed in juvenile rats with neonatal LPS exposure. Intranasal IGF-1 treatment at either 1 or 2 h after LPS exposure significantly attenuated LPS-induced brain injury and improved some behavioral deficits. Intranasal IGF-1 treatment also reduced infiltration of polymorphonuclear (PMN) leukocytes and activation of microglia in the rat brain 24 h after LPS exposure, but it did not prevent the elevation in concentrations of interleukin-1β (IL-1β) and tumor necrosis factor alpha (TNFα) in the LPS-exposed rat brain during the first 24 h. This is an indication that direct anti-inflammation might not be the primary mechanism for the protection of IGF-1, and other mechanisms, such as anti-apoptotic effects, are likely involved in its protective effects.
Collapse
Affiliation(s)
- Z Cai
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | | | | | | | | |
Collapse
|
124
|
Fan LW, Tien LT, Lin RCS, Simpson KL, Rhodes PG, Cai Z. Neonatal exposure to lipopolysaccharide enhances vulnerability of nigrostriatal dopaminergic neurons to rotenone neurotoxicity in later life. Neurobiol Dis 2011; 44:304-16. [PMID: 21798348 DOI: 10.1016/j.nbd.2011.07.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/29/2011] [Accepted: 07/11/2011] [Indexed: 12/21/2022] Open
Abstract
Brain inflammation in early life has been proposed to play important roles in the development of neurodegenerative disorders in adult life. To test this hypothesis, we used a neonatal rat model of lipopolysaccharide (LPS) exposure (1000 EU/g body weight, intracerebral injection on P5) to produce brain inflammation. By P70, when LPS-induced behavioral deficits were spontaneously recovered, animals were challenged with rotenone, a commonly used pesticide, through subcutaneous mini-pump infusion at a dose of 1.25 mg/kg per day for 14 days. This rotenone treatment regimen ordinarily does not produce toxic effects on behaviors in normal adult rats. Our results show that neonatal LPS exposure enhanced the vulnerability of nigrostriatal dopaminergic neurons to rotenone neurotoxicity in later life. Rotenone treatment resulted in motor neurobehavioral impairments in rats with the neonatal LPS exposure, but not in those without the neonatal LPS exposure. Rotenone induced losses of tyrosine hydroxylase immunoreactive neurons in the substantia nigra and decreased mitochondrial complex I activity in the striatum of rats with neonatal LPS exposure, but not in those without this exposure. Neonatal LPS exposure with later exposure to rotenone decreased retrogradely labeled nigrostriatal dopaminergic projecting neurons. The current study suggests that perinatal brain inflammation may enhance adult susceptibility to the development of neurodegenerative disorders triggered later on by environmental toxins at an ordinarily non-toxic or sub-toxic dose. Our model may be useful for studying mechanisms involved in the pathogenesis of nonfamilial Parkinson's disease and the development of potential therapeutic treatments.
Collapse
Affiliation(s)
- Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | | | | | | | | |
Collapse
|
125
|
Tien LT, Cai Z, Rhodes PG, Fan LW. Neonatal exposure to lipopolysaccharide enhances methamphetamine-induced reinstated behavioral sensitization in adult rats. Behav Brain Res 2011; 224:166-73. [PMID: 21669234 DOI: 10.1016/j.bbr.2011.05.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 05/27/2011] [Accepted: 05/29/2011] [Indexed: 01/28/2023]
Abstract
Our previous studies have shown that neonatal exposure to lipopolysaccharide (LPS) resulted in long-lasting dopaminergic injury and enhanced methamphetamine (METH)-induced increase of locomotion in the adult male rat. To further investigate the effect of neonatal LPS exposure-induced dopaminergic injury, we used our neonatal rat model of LPS exposure (1mg/kg, intracerebral injection in postnatal day 5, P5, rats) to examine the METH sensitization as an indicator of drug addiction in the adult rats. On P70, animals began a treatment schedule of 5 daily subcutaneous (s.c.) administration of METH (0.5mg/kg) or saline (P70-P74) to induce behavioral sensitization. Ninety-six hours after the 5th treatment with METH or saline (P78), animals received a single dose of 0.5mg/kg METH (s.c.) or saline. Neonatal LPS exposure enhanced the level of development of behavioral sensitization including distance traveled, rearing events and stereotypy to METH administration in both male and female rats. Neonatal LPS exposure also enhanced the reinstated behavioral sensitization in both male and female rats after the administration had ceased for 96h. However, neonatal LPS exposure induced alteration in the reinstated behaviors sensitization of distance traveled and rearing events to METH administration appears to be greater in male than in female rats. These results indicate that neonatal brain LPS exposure produces a persistent lesion in the dopaminergic system, as indicated by enhanced METH-induced locomotor and stereotyped behavioral sensitization in later life. These findings show that early-life brain inflammation may enhance susceptibility to the development of drug addiction in later life.
Collapse
Affiliation(s)
- Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan
| | | | | | | |
Collapse
|
126
|
Souvenir R, Fathali N, Ostrowski RP, Lekic T, Zhang JH, Tang J. Tissue inhibitor of matrix metalloproteinase-1 mediates erythropoietin-induced neuroprotection in hypoxia ischemia. Neurobiol Dis 2011; 44:28-37. [PMID: 21689752 DOI: 10.1016/j.nbd.2011.05.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/17/2011] [Accepted: 05/28/2011] [Indexed: 02/06/2023] Open
Abstract
Previous studies have shown that erythropoietin (EPO) is neuroprotective in both in vivo and in vitro models of hypoxia ischemia. However these studies hold limited clinical translations because the underlying mechanism remains unclear and the key molecules involved in EPO-induced neuroprotection are still to be determined. This study investigated if tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) and its upstream regulator signaling molecule Janus kinase-2 (JAK-2) are critical in EPO-induced neuroprotection. Hypoxia ischemia (HI) was modeled in-vitro by oxygen and glucose deprivation (OGD) and in-vivo by a modified version of Rice-Vannucci model of HI in 10-day-old rat pups. EPO treated cells were exposed to AG490, an inhibitor of JAK-2 or TIMP-1 neutralizing antibody for 2h with OGD. Cell death, phosphorylation of JAK-2 and signal transducers and activators of transcription protein-3 (STAT-3), TIMP-1 expression, and matrix metalloproteinase-9 (MMP-9) activity were measured and compared with normoxic group. Hypoxic ischemic animals were treated one hour following HI and evaluated 48 h after. Our data showed that EPO significantly increased cell survival, associated with increased TIMP-1 activity, phosphorylation of JAK-2 and STAT-3, and decreased MMP-9 activity in vivo and in vitro. EPO's protective effects were reversed by inhibition of JAK-2 or TIMP-1 in both models. We concluded that JAK-2, STAT-3 and TIMP-1 are key mediators of EPO-induced neuroprotection during hypoxia ischemia injury.
Collapse
Affiliation(s)
- Rhonda Souvenir
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | | | |
Collapse
|
127
|
Effects of neonatal systemic inflammation on blood-brain barrier permeability and behaviour in juvenile and adult rats. Cardiovasc Psychiatry Neurol 2011; 2011:469046. [PMID: 21547250 PMCID: PMC3085330 DOI: 10.1155/2011/469046] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 01/09/2011] [Accepted: 01/18/2011] [Indexed: 12/03/2022] Open
Abstract
Several neurological disorders have been linked to inflammatory insults suffered during development. We investigated the effects of neonatal systemic inflammation, induced by LPS injections, on blood-brain barrier permeability, endothelial tight junctions and behaviour of juvenile (P20) and adult rats. LPS-treatment resulted in altered cellular localisation of claudin-5 and changes in ultrastructural morphology of a few cerebral blood vessels. Barrier permeability to sucrose was significantly increased in LPS treated animals when adult but not at P20 or earlier. Behavioural tests showed that LPS treated animals at P20 exhibited altered behaviour using prepulse inhibition (PPI) analysis, whereas adults demonstrated altered behaviour in the dark/light test. These data indicate that an inflammatory insult during brain development can change blood-brain barrier permeability and behaviour in later life. It also suggests that the impact of inflammation can occur in several phases (short- and long-term) and that each phase might lead to different behavioural modifications.
Collapse
|
128
|
Saunders N, Habgood M. Understanding barrier mechanisms in the developing brain to aid therapy for the dysfunctional brain. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.10.84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The brain, both in the adult and during development, is protected by morphological barriers and functional mechanisms that provide a stable internal environment. Understanding these processes in the developing brain may lead to novel therapies for brain disorders, as some transport mechanisms, particularly those in the choroid plexus, may prove more amenable to devising novel delivery systems. Based on results from studies of the transfer of specific proteins across the blood–cerebrospinal fluid interface in the developing brain, the steps required to develop such a delivery system are outlined. Knowledge of barrier mechanisms in the developing brain may be relevant to treating neuropsychiatric conditions in children and adults for whom barrier dysfunction in the fetus, precipitated by adverse factors such as maternal infection, may contribute to the neuropathology underlying disorders such as autism and schizophrenia.
Collapse
Affiliation(s)
- Norman Saunders
- University of Melbourne, Department of Pharmacology, Parkville, Victoria 3010, Australia
| | - Mark Habgood
- University of Melbourne, Department of Pharmacology, Parkville, Victoria 3010, Australia
| |
Collapse
|
129
|
Sato Y, Ishida-Nakajima W, Kawamura M, Miura S, Oguma R, Arai H, Takahashi T. Hypoxia-ischemia induces hypo-phosphorylation of collapsin response mediator protein 2 in a neonatal rat model of periventricular leukomalacia. Brain Res 2011; 1386:165-74. [PMID: 21333637 DOI: 10.1016/j.brainres.2011.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 02/10/2011] [Accepted: 02/10/2011] [Indexed: 01/14/2023]
Abstract
Collapsin response mediator protein2 (CRMP2) is a brain-specific protein involved in neuronal polarity and axonal guidance, and phosphorylation of CRMP2 regulates the function and the activity. CRMP2 has shown to be implicated in several neurodegenerative diseases (Alzheimer's disease, epilepsy and ischemia) and this study was designed to assess the role of CRMP2 in periventricular leukomalacia (PVL). We developed a PVL model using 3-day-old rats to investigate the expression and phosphorylation of CRMP2 in the newborn brain. Hypoxia-ischemia was applied by unilateral carotid ligation followed by exposure to 5% oxygen for 30min. Pathological changes were evaluated from 0h to 21d post-HI, and white matter damage including severe necrosis, white matter rarefaction and lateral ventricle dilatation were found. In the PVL model astrogliosis and axonal damage were detected in the injured white matter by immunohistochemistry at 48-168h post-HI, and delayed myelination was verified by Western blotting after 21-day post-HI. We confirmed that this model showed neuropathological features of PVL. Next, significant changes of CRMP2 were observed in the brain of the PVL model. Western blotting and immunohistochemistry showed that cleavage and hypo-phosphorylation of CRMP2 occurred after 48h post-HI in the PVL brain. Our results suggest that cleaved CRMP2 could represent hypo-phosphorylated-CRMP2 and HI could induce activation of CRMP2 in the PVL brain. The activated CRMP2 may play an important role in neuronal plasticity in PVL. Our findings suggest that future treatment strategies of PVL should target the phosphorylation mechanism of CRMP2.
Collapse
Affiliation(s)
- Yoko Sato
- Department of Reproductive and Developmental Medicine, Akita University School of Medicine, Akita, Japan.
| | | | | | | | | | | | | |
Collapse
|
130
|
Fan LW, Tien LT, Zheng B, Pang Y, Lin RCS, Simpson KL, Ma T, Rhodes PG, Cai Z. Dopaminergic neuronal injury in the adult rat brain following neonatal exposure to lipopolysaccharide and the silent neurotoxicity. Brain Behav Immun 2011; 25:286-97. [PMID: 20875849 PMCID: PMC3025048 DOI: 10.1016/j.bbi.2010.09.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 09/07/2010] [Accepted: 09/22/2010] [Indexed: 01/17/2023] Open
Abstract
Our previous studies have shown that neonatal exposure to lipopolysaccharide (LPS) resulted in motor dysfunction and dopaminergic neuronal injury in the juvenile rat brain. To further examine whether neonatal LPS exposure has persisting effects in adult rats, motor behaviors were examined from postnatal day 7 (P7) to P70 and brain injury was determined in P70 rats following an intracerebral injection of LPS (1 mg/kg) in P5 Sprague-Dawley male rats. Although neonatal LPS exposure resulted in hyperactivity in locomotion and stereotyped tasks, and other disturbances of motor behaviors, the impaired motor functions were spontaneously recovered by P70. On the other hand, neonatal LPS-induced injury to the dopaminergic system such as the loss of dendrites and reduced tyrosine hydroxylase immunoreactivity in the substantia nigra persisted in P70 rats. Neonatal LPS exposure also resulted in sustained inflammatory responses in the P70 rat brain, as indicated by an increased number of activated microglia and elevation of interleukin-1β and interleukin-6 content in the rat brain. In addition, when challenged with methamphetamine (METH, 0.5 mg/kg) subcutaneously, rats with neonatal LPS exposure had significantly increased responses in METH-induced locomotion and stereotypy behaviors as compared to those without LPS exposure. These results indicate that although neonatal LPS-induced neurobehavioral impairment is spontaneously recoverable, the LPS exposure-induced persistent injury to the dopaminergic system and the chronic inflammation may represent the existence of silent neurotoxicity. Our data further suggest that the compromised dendritic mitochondrial function might contribute, at least partially, to the silent neurotoxicity.
Collapse
Affiliation(s)
- Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lu-Tai Tien
- School of Medicine, Fu-Jen Catholic University, Hsin-Chuang, Taipei County 24205, Taiwan
| | - Baoying Zheng
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Rick C. S. Lin
- Departments of Anatomy, Psychiatry & Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Kimberly L. Simpson
- Departments of Anatomy, Psychiatry & Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Tangeng Ma
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Philip G. Rhodes
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Zhengwei Cai
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA,Corresponding author: Dr. Zhengwei Cai, Ph.D., Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216-4504, USA, Tel.: +1-601-984-2786; Fax: +1-601-815-3666, (Z. Cai)
| |
Collapse
|
131
|
Interleukin-1 receptor antagonist ameliorates neonatal lipopolysaccharide-induced long-lasting hyperalgesia in the adult rats. Toxicology 2010; 279:123-9. [PMID: 20937348 DOI: 10.1016/j.tox.2010.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/02/2010] [Accepted: 10/04/2010] [Indexed: 11/23/2022]
Abstract
An increasing amount of data show that central inflammation contributes to many debilitating diseases and produces spontaneous pain and hyperalgesia (an increased sensitivity to painful stimuli), and these processes may be associated with the production of proinflammatory cytokines by activated microglia. In the present study, we demonstrate that neonatal intracerebral injection of lipopolysaccharide (LPS) (1mg/kg) in postnatal day 5 (P5) rats produced hyperalgesia that lasted into adulthood as indicated by decreased latency in the tail-flick test. Neonatal LPS administration resulted in a long-lasting increase in the number of activated microglial in the P70 rat brain. The effects of interleukin-1beta (IL-1β) and IL-1 receptor antagonists on hyperalgesia were determined to examine the possible role of inflammatory cytokines in LPS-induced hyperalgesia. Our data show that neonatal intracerebral injection of IL-1β (1 μg/kg) produced a hyperalgesic tendency similar to that induced by LPS. Neonatal administration of an IL-1 receptor antagonist (0.1mg/kg) significantly attenuated long-lasting hyperalgesia induced by LPS and reduced the number of activated microglia in the adult rat brain. These data reveal that neonatal intracerebral LPS exposure results in long-lasting hyperalgesia and an elevated number of activated microglia in later life. This effect is similar to that induced by IL-1β and can be prevented by an IL-1 receptor antagonist. The present study suggests that an IL-1 receptor antagonist effectively attenuates or blocks long-lasting hyperalgesia and microglia activation produced by LPS exposure in the neonatal period of rats.
Collapse
|
132
|
Rico JLR, Ferraz DB, Ramalho-Pinto FJ, Morato S. Neonatal exposure to LPS leads to heightened exploratory activity in adolescent rats. Behav Brain Res 2010; 215:102-9. [PMID: 20620170 DOI: 10.1016/j.bbr.2010.07.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 06/23/2010] [Accepted: 07/02/2010] [Indexed: 01/30/2023]
Abstract
Although several reports have demonstrated physiological and behavioral changes in adult rats due to neonatal immune challenges, little is known about their effects in adolescence. Since neonatal exposure to lipopolysaccharide (LPS) alters the neural substrates involved in cognitive disorders, we tested the hypothesis that it may also alter the response to novel environments in adolescent rats. At 3 and 5 days of age, male Wistar rats received intraperitoneal injections of either vehicle solution or E. coli LPS (0.05mg/kg) or were left undisturbed. In the mid-adolescent period, between 40 and 46 days of age, the rats were exposed to the following behavioral tests: elevated plus-maze, open-field, novel-object exploration task, hole-board and the modified Porsolt forced swim test. The results showed that, in comparison with control animals, LPS-treated rats exhibited (1) less anxiety-related behaviors and enhanced patterns of locomotion and rearing in the plus-maze and the open-field tests, (2) high levels of exploration of both objects in the novel-object task and of corner and central holes in hole-board test, and (3) more time spent diving, an active behavior in the forced swim test. The present findings suggest that neonatal LPS exposure has long-lasting effects on the behavior profile adolescent rats exhibit in response to novelty. This behavioral pattern, characterized by heightened exploratory activity in novel environments, also suggests that early immune stimulation may contribute to the development of impulsive behavior in adolescent rats.
Collapse
|
133
|
Lodygensky GA, West T, Stump M, Holtzman DM, Inder TE, Neil JJ. In vivo MRI analysis of an inflammatory injury in the developing brain. Brain Behav Immun 2010; 24:759-67. [PMID: 19945527 PMCID: PMC2885544 DOI: 10.1016/j.bbi.2009.11.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 11/13/2009] [Accepted: 11/15/2009] [Indexed: 11/30/2022] Open
Abstract
Cerebral periventricular white matter injury stands as a leading cause of cognitive, behavioral and motor impairment in preterm infants. There is epidemiological and histopathological evidence demonstrating the role of prenatal or neonatal inflammation in brain injury in preterm infants. In order to define the effect of an inflammatory insult in the developing brain on magnetic resonance (MR) imaging, we obtained high resolution conventional and diffusion MR images of the brain of rat pups after an inflammatory injury. Rat pups were subjected on postnatal day 5 (P5) to a stereotaxic injection of lipopolysaccharide in the corpus callosum and then imaged at 11.7 T on days 0, 2 and 4 following the injury. They were subsequently sacrificed for immunohistochemistry. Diffusion tensor imaging (DTI) acquired at high spatial resolution showed an initial reduction of the apparent diffusion coefficient (ADC) in the white matter. This was followed by an increase in ADC value and in T2 relaxation time constant in the white matter, with an associated increase of radial diffusivity of the corpus callosum, and a 10-fold increase in ventricular size. On histology, these MR changes corresponded to widespread astrogliosis, and decreased proportion of the section areas containing cresyl violet positive stain. The increase in radial diffusivity, typically attributed to myelin loss, occurred in this case despite the absence of myelin at this developmental stage.
Collapse
Affiliation(s)
- GA Lodygensky
- Department of the Child and Adolescent, Pediatric and Neonatal ICU, University of Geneva, Switzerland
,Departments of Pediatrics, Washington University, St Louis, Missouri, United States
| | - T West
- Department of Neurology and Developmental Biology, Washington University, St Louis, Missouri, United States
| | - M Stump
- Department of Neurology and Developmental Biology, Washington University, St Louis, Missouri, United States
| | - DM Holtzman
- Department of Neurology and Developmental Biology, Washington University, St Louis, Missouri, United States
,Hope Center for Neurological Disorders, Washington University, St Louis, Missouri, United States
| | - TE Inder
- Department of Neurology and Developmental Biology, Washington University, St Louis, Missouri, United States
,Departments of Radiology, Washington University, St Louis, Missouri, United States
,Departments of Pediatrics, Washington University, St Louis, Missouri, United States
| | - JJ Neil
- Department of Neurology and Developmental Biology, Washington University, St Louis, Missouri, United States
,Departments of Radiology, Washington University, St Louis, Missouri, United States
,Departments of Pediatrics, Washington University, St Louis, Missouri, United States
| |
Collapse
|
134
|
FAN LW, TIEN LT, ZHENG B, PANG Y, RHODES P, CAI Z. Interleukin-1beta-induced brain injury and neurobehavioral dysfunctions in juvenile rats can be attenuated by alpha-phenyl-n-tert-butyl-nitrone. Neuroscience 2010; 168:240-52. [PMID: 20346393 PMCID: PMC2873102 DOI: 10.1016/j.neuroscience.2010.03.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 02/23/2010] [Accepted: 03/12/2010] [Indexed: 02/02/2023]
Abstract
Our previous study showed that perinatal exposure to interleukin-1beta (IL-1beta), an inflammatory cytokine, induces acute injury to developing white matter in the neonatal rat brain, and alpha-phenyl-n-tert-butyl-nitrone (PBN), a free radical scavenger and antioxidant, protects against IL-1beta-induced acute brain injury. The objective of the present study was to further examine whether perinatal exposure to IL-1beta resulted in persistent brain damage and neurological disabilities, and whether PBN offers lasting protection. Intracerebral injection of IL-1beta (1 microg/kg) was performed in postnatal day 5 (P5) Sprague-Dawley rat pups and PBN (100 mg/kg) or saline was administered intraperitoneally 5 min after IL-1beta injection. Perinatal IL-1beta exposure significantly affected neurobehavioral functions in juvenile rats. Although some neurobehavioral deficits such as performance in negative geotaxis, cliff avoidance, beam walking, and locomotion were spontaneously reversible, sustained deficits such as poor performance in the vibrissa-elicited forelimb-placing test, the pole test, the passive avoidance task, and the elevated plus-maze task were still observable at P21. Perinatal IL-1beta exposure resulted in persistent brain damage including enlargement of ventricles, loss of mature oligodendrocytes, impaired myelination as indicated by the decrease in myelin basic protein immunostaining, axonal and dendritic injury, and loss of hippocampal CA1 neurons and tyrosine hydroxylase positive neurons in the substantia nigra and ventral tegmental areas of the rat brain. Treatments with PBN provided lasting protection against the IL-1beta-induced brain injury and improved the associated neurological dysfunctions in juvenile rats, suggesting that prompt treatments for brain injury induced by perinatal infection/inflammation might have important long-term consequences.
Collapse
Affiliation(s)
- L.-W. FAN
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - L.-T. TIEN
- School of Medicine, Fu-Jen Catholic University, Hsin-Chuang, Taipei County, Taiwan
| | - B. ZHENG
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Y. PANG
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - P.G. RHODES
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Z. CAI
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
135
|
Pang Y, Zheng B, Campbell LR, Fan LW, Cai Z, Rhodes PG. IGF-1 can either protect against or increase LPS-induced damage in the developing rat brain. Pediatr Res 2010; 67:579-84. [PMID: 20220546 PMCID: PMC3076081 DOI: 10.1203/pdr.0b013e3181dc240f] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Periventricular leukomalacia (PVL) is a major form of brain damage in premature infants. This study was to test whether IGF-1 can prevent PVL-like brain damage induced by lipopolysaccharide (LPS) in the neonatal rat. Intraventricular delivery of LPS resulted in an acute brain inflammatory response, i.e., rapid recruitment of polymorphonuclear leukocytes (PMNs), activation of microglia and astrocytes, and induction of IL-1beta (IL1beta) expression. Brain inflammation was associated with the loss of O4+ preoligodendrocytes (preOLs), a decrease of myelin basic protein (MBP) in the white matter and an increase of pyknotic cells in the cortex. IGF-1 at a low dose significantly prevented LPS-induced deleterious effects without alteration of IL-1beta expression and microglia/astrocytes activation. On the other hand, the low dose of IGF-1 enhanced LPS-induced PMNs recruitment and blood-brain barrier (BBB) permeability, and caused intracerebral hemorrhage. At higher doses, co-application of IGF-1 with LPS resulted in a high mortality rate. Brains from the surviving rats showed massive PMN infiltration and intracerebral hemorrhage. However, these adverse effects were not found in rats treated with IGF-1 alone. This study provides the alarming evidence that in an acute inflammatory condition, IGF-1 may have severe, harmful effects on the developing brain.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Astrocytes/drug effects
- Astrocytes/pathology
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/metabolism
- Brain/drug effects
- Brain/growth & development
- Brain/metabolism
- Brain/pathology
- Capillary Permeability/drug effects
- Cell Death
- Cerebral Hemorrhage/chemically induced
- Cerebral Hemorrhage/pathology
- Cerebral Hemorrhage/physiopathology
- Chemotaxis, Leukocyte/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalitis/chemically induced
- Encephalitis/metabolism
- Encephalitis/pathology
- Encephalitis/physiopathology
- Encephalitis/prevention & control
- Female
- Humans
- Infant, Newborn
- Inflammation Mediators/metabolism
- Injections, Intraventricular
- Insulin-Like Growth Factor I/administration & dosage
- Insulin-Like Growth Factor I/toxicity
- Interleukin-1beta/metabolism
- Leukomalacia, Periventricular/chemically induced
- Leukomalacia, Periventricular/metabolism
- Leukomalacia, Periventricular/pathology
- Leukomalacia, Periventricular/physiopathology
- Leukomalacia, Periventricular/prevention & control
- Lipopolysaccharides
- Male
- Microglia/drug effects
- Microglia/pathology
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/toxicity
- Rats
- Rats, Sprague-Dawley
- Recombinant Proteins/administration & dosage
Collapse
Affiliation(s)
- Yi Pang
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | | | | | | | | | | |
Collapse
|
136
|
He LF, Chen HJ, Qian LH, Chen GY, Buzby JS. Curcumin protects pre-oligodendrocytes from activated microglia in vitro and in vivo. Brain Res 2010; 1339:60-9. [DOI: 10.1016/j.brainres.2010.04.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 04/07/2010] [Accepted: 04/08/2010] [Indexed: 12/11/2022]
|
137
|
|
138
|
Yuan TM, Yu HM. Notch signaling: key role in intrauterine infection/inflammation, embryonic development, and white matter damage? J Neurosci Res 2010; 88:461-8. [PMID: 19768798 DOI: 10.1002/jnr.22229] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The mechanisms or pathophysiologies that lead to cerebral white matter damage during development are complex and not fully understood. It is postulated that exposure of the preterm brain to inflammatory cytokines during intrauterine infection/inflammation contributes to brain white matter damage, and this damage may affect the function and differentiation of progenitor oligodendrocyte cells under physiological conditions. The Notch pathway, an important signaling pathway controlling various cells' differentiation, functions in the timing of oligodendrocyte differentiation, and Notch signaling may contribute to white matter damage and may mediate neurogenesis in a pathophysiological phase. Recent studies have led to recognition of the role of the Notch pathway in neurogenesis in cerebral ischemic damage and in myelination and axonal damage of neurodegenerative diseases. Moreover, Notch plays a critical role in steering an immune response toward inflammation by regulating expression of various cytokines and proinflammatory cytokines resulting in the activation of Notch signaling. Thus, the Notch signaling pathway likely plays a key role in intrauterine infection/inflammation, brain development, and white matter damage, and future research directed toward understanding its role will be important. Insofar as Notch signaling could have an important effect on neurogenesis, mobilization of progenitor cells is one strategy for compensating for the neuronal losses seen in white matter damage after intrauterine infection/inflammation.
Collapse
Affiliation(s)
- Tian-Ming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University, School of Medicine, Hangzhou, People's Republic of China
| | | |
Collapse
|
139
|
Willette AA, Bendlin BB, McLaren DG, Canu E, Kastman EK, Kosmatka KJ, Xu G, Field AS, Alexander AL, Colman RJ, Weindruch RH, Coe CL, Johnson SC. Age-related changes in neural volume and microstructure associated with interleukin-6 are ameliorated by a calorie-restricted diet in old rhesus monkeys. Neuroimage 2010; 51:987-94. [PMID: 20298794 DOI: 10.1016/j.neuroimage.2010.03.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 03/02/2010] [Accepted: 03/04/2010] [Indexed: 11/24/2022] Open
Abstract
Systemic levels of proinflammatory cytokines such as interleukin-6 (IL-6) increase in old age and may contribute to neural atrophy in humans. We investigated IL-6 associations with age in T1-weighted segments and microstructural diffusion indices using MRI in aged rhesus monkeys (Macaca mulatta). Further, we determined if long-term 30% calorie restriction (CR) reduced IL-6 and attenuated its association with lower tissue volume and density. Voxel-based morphometry (VBM) and diffusion-weighted voxelwise analyses were conducted. IL-6 was associated with less global gray and white matter (GM and WM), as well as smaller parietal and temporal GM volumes. Lower fractional anisotropy (FA) was associated with higher IL-6 levels along the corpus callosum and various cortical and subcortical tracts. Higher IL-6 concentrations across subjects were also associated with increased mean diffusivity (MD) throughout many brain regions, particularly in corpus callosum, cingulum, and parietal, frontal, and prefrontal areas. CR monkeys had significantly lower IL-6 and less associated atrophy. An IL-6xCR interaction across modalities also indicated that CR mitigated IL-6 related changes in several brain regions compared to controls. Peripheral IL-6 levels were correlated with atrophy in regions sensitive to aging, and this relationship was decreased by CR.
Collapse
Affiliation(s)
- A A Willette
- Harlow Primate Laboratory, Department of Psychology, Madison, WI 53715, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Bland ST, Beckley JT, Young S, Tsang V, Watkins LR, Maier SF, Bilbo SD. Enduring consequences of early-life infection on glial and neural cell genesis within cognitive regions of the brain. Brain Behav Immun 2010; 24:329-38. [PMID: 19782746 PMCID: PMC2826544 DOI: 10.1016/j.bbi.2009.09.012] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 09/07/2009] [Accepted: 09/19/2009] [Indexed: 01/14/2023] Open
Abstract
Systemic infection with Escherichia coli on postnatal day (P) 4 in rats results in significantly altered brain cytokine responses and behavioral changes in adulthood, but only in response to a subsequent immune challenge with lipopolysaccharide [LPS]. The basis for these changes may be long-term changes in glial cell function. We assessed glial and neural cell genesis in the hippocampus, parietal cortex (PAR), and prefrontal cortex (PFC), in neonates just after the infection, as well as in adulthood in response to LPS. E. coli increased the number of newborn microglia within the hippocampus and PAR compared to controls. The total number of microglia was also significantly increased in E. coli-treated pups, with a concomitant decrease in total proliferation. On P33, there were large decreases in numbers of cells coexpressing BrdU and NeuN in all brain regions of E. coli rats compared to controls. In adulthood, basal neurogenesis within the dentate gyrus (DG) did not differ between groups; however, in response to LPS, there was a decrease in neurogenesis in early-infected rats, but an increase in controls to the same challenge. There were also significantly more microglia in the adult DG of early-infected rats, although microglial proliferation in response to LPS was increased in controls. Taken together, we have provided evidence that systemic infection with E. coli early in life has significant, enduring consequences for brain development and subsequent adult function. These changes include marked alterations in glia, as well as influences on neurogenesis in brain regions important for cognition.
Collapse
Affiliation(s)
- Sondra T. Bland
- Department of Psychology, University of Colorado, Denver, CO, 80204
| | - Jacob T. Beckley
- Department of Psychology, University of Colorado, Boulder, CO, 80309
| | - Sarah Young
- Department of Psychology, University of Colorado, Boulder, CO, 80309
| | - Verne Tsang
- Department of Psychology & Neuroscience, Duke University, Durham, NC, 27708
| | - Linda R. Watkins
- Department of Psychology, University of Colorado, Boulder, CO, 80309
| | - Steven F. Maier
- Department of Psychology, University of Colorado, Boulder, CO, 80309
| | - Staci D. Bilbo
- Department of Psychology & Neuroscience, Duke University, Durham, NC, 27708, To whom correspondence should be addressed: Department of Psychology and Neuroscience, Duke University, Durham, NC, 27708; Phone: 919-681-7005; Fax: 919-660-5798;
| |
Collapse
|
141
|
Pang Y, Campbell L, Zheng B, Fan L, Cai Z, Rhodes P. Lipopolysaccharide-activated microglia induce death of oligodendrocyte progenitor cells and impede their development. Neuroscience 2009; 166:464-75. [PMID: 20035837 DOI: 10.1016/j.neuroscience.2009.12.040] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 12/14/2009] [Accepted: 12/16/2009] [Indexed: 12/30/2022]
Abstract
Damage to oligodendrocyte (OL) progenitor cells (OPCs) and hypomyelination are two hallmark features of periventricular leukomalacia (PVL), the most common form of brain damage in premature infants. Clinical and animal studies have linked the incidence of PVL to maternal infection/inflammation, and activated microglia have been proposed to play a central role. However, the precise mechanism of how activated microglia adversely affects the survival and development of OPCs is still not clear. Here we demonstrate that lipopolysaccharide (LPS)-activated microglia are deleterious to OPCs, that is, impeding OL lineage progression, reducing the production of myelin basic protein (MBP), and mediating OPC death. We further demonstrate that LPS-activated microglia mediate OPC death by two distinct mechanisms in a time-dependent manner. The early phase of cell damage occurs within 24 h after LPS treatment, which is mediated by nitric oxide (NO)-dependent oxidative damage and is prevented by N(G)-nitro-l-arginine methyl ester (l-NAME), a general inhibitor of nitric oxide synthase. The delayed cell death is evident at 48 h after LPS treatment, is mediated by cytokines, and is prevented by blocking the activity of tumor necrosis factor-alpha (TNF-alpha) and pro-nerve growth factor (proNGF), but not by l-NAME. Furthermore, microglia-derived insulin-like growth factor-1 (IGF-1) and ciliary neurotrophic factor (CNTF) were significantly suppressed by LPS, and exogenous IGF-1 and CNTF synergistically protected OLs from death induced by LPS-treated microglia conditioned medium, indicating that a deficiency in trophic support may also be involved in OL death. Our finding that LPS-activated microglia not only induce two waves of cell death but also greatly impair OL development may shed some light on the mechanisms underlying selective white matter damage and hypomyelination in PVL.
Collapse
Affiliation(s)
- Y Pang
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | | | | | | | | | | |
Collapse
|
142
|
Mallard C, Wang X, Hagberg H. The role of Toll-like receptors in perinatal brain injury. Clin Perinatol 2009; 36:763-72, v-vi. [PMID: 19944834 DOI: 10.1016/j.clp.2009.07.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The toll-like receptors (TLRs) are a family of microbe-sensing receptors on peripheral immune cells. TLRs have also been discovered to be present in the brain, particularly in circumventricular organs, microglia, and astrocytes. Some TLRs are strongly expressed in the embryonic brain and TLR3 and TLR8 have been implicated in neurogenesis and neurite outgrowth in the developing brain, whereas TLR2 and TLR4 have been shown to regulate adult neurogenesis. TLR2 and TLR4 also play a role in acute ischemic brain injury in the adult, although no neuroprotection was observed following perinatal hypoxic-ischemic injury. These findings suggest that different TLRs have specific roles in the immature and adult brain following brain damage.
Collapse
Affiliation(s)
- Carina Mallard
- Department of Neuroscience and Physiology, Perinatal Center, Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 11, 41390 Gothenburg, Sweden
| | | | | |
Collapse
|
143
|
Scantlebury MH, Galanopoulou AS, Chudomelova L, Raffo E, Betancourth D, Moshé SL. A model of symptomatic infantile spasms syndrome. Neurobiol Dis 2009; 37:604-12. [PMID: 19945533 DOI: 10.1016/j.nbd.2009.11.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 10/27/2009] [Accepted: 11/13/2009] [Indexed: 11/16/2022] Open
Abstract
Infantile spasms are characterized by age-specific expression of epileptic spasms and hypsarrhythmia and often result in significant cognitive impairment. Other epilepsies or autism often ensue especially in symptomatic IS (SIS). Cortical or subcortical damage, including white matter, have been implicated in the pathogenesis of SIS. To generate a model of SIS, we recreated this pathology by injecting rats with lipopolysaccharide and doxorubicin intracerebrally at postnatal day (P) 3 and with p-chlorophenylalanine intraperitoneally at P5. Spasms occurred between P4 and 13 and were associated with ictal EEG correlates, interictal EEG abnormalities and neurodevelopmental decline. After P9 other seizures, deficits in learning and memory, and autistic-like behaviors (indifference to other rats, increased grooming) were observed. Adrenocorticotropic hormone (ACTH) did not affect spasms. Vigabatrin transiently suppressed spasms at P5. This new model of SIS will be useful to study the neurobiology and treatment of SIS, including those that are refractory to ACTH.
Collapse
Affiliation(s)
- Morris H Scantlebury
- Saul R. Korey Department of Neurology, Laboratory of Developmental Neurology, and Montefiore/Einstein Epilepsy Management Center, USA.
| | | | | | | | | | | |
Collapse
|
144
|
Fan LW, Mitchell HJ, Tien LT, Rhodes PG, Cai Z. Interleukin-1beta-induced brain injury in the neonatal rat can be ameliorated by alpha-phenyl-n-tert-butyl-nitrone. Exp Neurol 2009; 220:143-53. [PMID: 19682987 PMCID: PMC2761495 DOI: 10.1016/j.expneurol.2009.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 06/24/2009] [Accepted: 08/06/2009] [Indexed: 11/19/2022]
Abstract
To examine the possible role of inflammatory cytokines in mediating perinatal brain injury, we investigated effects of intracerebral injection of interleukin-1beta (IL-1beta) on brain injury in the neonatal rat and the mechanisms involved. Intracerebral administration of IL-1beta (1 microg/kg) resulted in acute brain injury, as indicated by enlargement of ventricles bilaterally, apoptotic death of oligodendrocytes (OLs) and loss of OL immunoreactivity in the neonatal rat brain. IL-1beta also induced axonal and neuronal injury in the cerebral cortex as indicated by elevated expression of beta-amyloid precursor protein, short beaded axons and dendrites, and loss of tyrosine hydroxylase-positive neurons in the substantia nigra and the ventral tegmental areas. Administration of alpha-phenyl-n-tert-butyl-nitrone (PBN, 100 mg/kg i.p.) immediately after the IL-1beta injection protected the brain from IL-1beta-induced injury. Protection of PBN was linked with the attenuated oxidative stress induced by IL-1beta, as indicated by decreased elevation of 8-isoprostane content and by the reduced number of 4-hydroxynonenal or malondialdehyde or nitrotyrosine-positive cells following IL-1beta exposure. PBN also attenuated IL-1beta-stimulated inflammatory responses as indicated by the reduced activation of microglia. The finding that IL-1beta induced perinatal brain injury was very similar to that induced by lipopolysaccharide (LPS), as we previously reported and that PBN was capable to attenuate the injury induced by either LPS or IL-1beta suggests that IL-1beta may play a critical role in mediating brain injury associated with perinatal infection/inflammation.
Collapse
Affiliation(s)
- Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Helen J. Mitchell
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lu-Tai Tien
- School of Medicine, Fu-Jen Catholic University, Hsin-Chuang, Taipei County, Taiwan
| | - Philip G. Rhodes
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Zhengwei Cai
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
145
|
Webber DJ, van Blitterswijk M, Chandran S. Neuroprotective effect of oligodendrocyte precursor cell transplantation in a long-term model of periventricular leukomalacia. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2332-42. [PMID: 19850891 DOI: 10.2353/ajpath.2009.090051] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Perinatal white matter injury, or periventricular leukomalacia (PVL), is the most common cause of brain injury in premature infants and is the leading cause of cerebral palsy. Despite increasing numbers of surviving extreme premature infants and associated long-term neurological morbidity, our understanding and treatment of PVL remains incomplete. Inflammation- or ischemia/hypoxia-based rodent models, although immensely valuable, are largely restricted to reproducing short-term features of up to 3 weeks after injury. Given the long-term sequelae of PVL, there is a need for subchronic models that will enable testing of putative neuroprotective therapies. Here, we report long term characterization of a neonatal inflammation-induced rat model of PVL. We show bilateral ventriculomegaly, inflammation, reactive astrogliosis, injury to pre-oligodendrocytes, and neuronal loss 8 weeks after injury. We demonstrate neuroprotective effects of oligodendrocyte precursor cell transplantation. Our findings present a subchronic model of PVL and highlight the tissue protective effects of oligodendrocyte precursor cell transplants that demonstrate the potential of cell-based therapy for PVL.
Collapse
Affiliation(s)
- Daniel J Webber
- Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
146
|
Wang X, Hellgren G, Löfqvist C, Li W, Hellström A, Hagberg H, Mallard C. White matter damage after chronic subclinical inflammation in newborn mice. J Child Neurol 2009; 24:1171-8. [PMID: 19745089 PMCID: PMC3674559 DOI: 10.1177/0883073809338068] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preterm infants exposed to inflammation are at increased risk of white matter injury and/or cerebral palsy. To investigate the effect of chronic inflammation on the developing white matter, we administered low-dose lipopolysaccharide once a day from postnatal days 3 to 11, examined white matter changes at postnatal day 12, and monitored serum levels of insulin-like growth factor 1 and insulin-like factor binding protein-3. A single injection of lipopolysaccharide decreased the serum insulin-like growth factor 1 level but not the insulin-like factor binding protein-3 level. At postnatal day 12, quantification of immunohistochemical staining for axonal, myelin, and oligodendrocyte markers revealed impaired myelination in subcortical white matter. In addition, brain gray matter volume decreased and spleen and liver weight increased at postnatal day 12. These data suggest chronic subclinical inflammation hampers development of white and gray matter in early life, which may be associated with insulin-like growth factor 1 deficiency.
Collapse
Affiliation(s)
- Xiaoyang Wang
- Perinatal Center, Department of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, S-405 30 Gothenburg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
147
|
Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci 2009; 3:14. [PMID: 19738918 PMCID: PMC2737431 DOI: 10.3389/neuro.08.014.2009] [Citation(s) in RCA: 467] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 07/22/2009] [Indexed: 12/12/2022] Open
Abstract
The immune system is well characterized for its critical role in host defense. Far beyond this limited role however, there is mounting evidence for the vital role the immune system plays within the brain, in both normal, “homeostatic” processes (e.g., sleep, metabolism, memory), as well as in pathology, when the dysregulation of immune molecules may occur. This recognition is especially critical in the area of brain development. Microglia and astrocytes, the primary immunocompetent cells of the CNS, are involved in every major aspect of brain development and function, including synaptogenesis, apoptosis, and angiogenesis. Cytokines such as tumor necrosis factor (TNF)α, interleukin [IL]-1β, and IL-6 are produced by glia within the CNS, and are implicated in synaptic formation and scaling, long-term potentiation, and neurogenesis. Importantly, cytokines are involved in both injury and repair, and the conditions underlying these distinct outcomes are under intense investigation and debate. Evidence from both animal and human studies implicates the immune system in a number of disorders with known or suspected developmental origins, including schizophrenia, anxiety/depression, and cognitive dysfunction. We review the evidence that infection during the perinatal period of life acts as a vulnerability factor for later-life alterations in cytokine production, and marked changes in cognitive and affective behaviors throughout the remainder of the lifespan. We also discuss the hypothesis that long-term changes in brain glial cell function underlie this vulnerability.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology & Neuroscience, Duke University Durham, NC, USA.
| | | |
Collapse
|
148
|
Gavilanes AD, Strackx E, Kramer BW, Gantert M, Van den Hove D, Steinbusch H, Garnier Y, Cornips E, Steinbusch H, Zimmermann L, Vles J. Chorioamnionitis induced by intraamniotic lipopolysaccharide resulted in an interval-dependent increase in central nervous system injury in the fetal sheep. Am J Obstet Gynecol 2009; 200:437.e1-8. [PMID: 19217590 DOI: 10.1016/j.ajog.2008.12.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 07/20/2008] [Accepted: 12/04/2008] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We quantified the impact of chorioamnionitis on both the white and gray matter structures of the preterm ovine central nervous system (CNS). STUDY DESIGN The CNS was studied at 125 days of gestation, either 2 or 14 days after the intraamniotic administration of 10 mg of lipopolysaccharide (LPS) (Escherichia coli) or saline. Apoptotic cells and cell types were analyzed in the brain, cerebellum, and spinal cord using flow cytometry. RESULTS Apoptosis and microglial activation increased in all regions with prolonged exposure to LPS-induced chorioamnionitis. Astrocytes were increased in the brain and cerebellum of LPS-exposed fetuses but not in the spinal cord. Mature oligodendrocytes decreased in the cerebral and cerebellar white matter, the cerebral cortex, caudate putamen, and hippocampus 14 days after LPS. Neurons in the cerebral cortex, hippocampus, and substantia nigra were reduced 14 days after LPS. CONCLUSION Fetal inflammation globally but differentially affected the CNS depending on the maturational stage of the brain region.
Collapse
|
149
|
Stolp HB, Dziegielewska KM. Review: Role of developmental inflammation and blood-brain barrier dysfunction in neurodevelopmental and neurodegenerative diseases. Neuropathol Appl Neurobiol 2009; 35:132-46. [DOI: 10.1111/j.1365-2990.2008.01005.x] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
150
|
Stolp HB, Ek CJ, Johansson PA, Dziegielewska KM, Bethge N, Wheaton BJ, Potter AM, Saunders NR. Factors involved in inflammation-induced developmental white matter damage. Neurosci Lett 2009; 451:232-6. [PMID: 19152829 DOI: 10.1016/j.neulet.2009.01.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 11/28/2008] [Accepted: 01/08/2009] [Indexed: 01/14/2023]
Abstract
Developmental white matter damage is a brain pathology associated with several long-term neurological disorders. An inflammatory insult has been suggested as the major instigating event. This study investigated the relative influence of inflammation, blood-brain barrier permeability and glial ontogeny in white matter damage. Systemic inflammation was induced in Monodelphis domestica (opossum) by serial intraperitoneal injections of lipopolysaccharide at different stages of brain development. Volume of white matter was estimated for the external capsule. Blood-brain barrier permeability was assessed immunocytochemically. Quantitative RT-PCR was used to measure relative levels of mRNA for IL-1beta, IL-6 and COX-2. Developmental changes in numbers and appearance of microglia and astrocytes were estimated. Results showed that in response to systemic inflammation, white matter was reduced in the external capsule during a circumscribed period only. At the same developmental stage blood-brain barrier permeability was altered, cerebral inflammatory response was present and numbers of microglia increased. However, the periods of altered blood-brain barrier permeability and the cerebral inflammatory response were longer than the period of the external capsule's susceptibility to white matter damage, which coincided with the developmental increase in the number of astrocytes in this tract. Thus, the mechanism of white matter damage following systemic inflammation is multifactorial, including cerebral inflammation and breakdown of brain barriers occurring simultaneously at specific stages of glial cell development.
Collapse
Affiliation(s)
- Helen B Stolp
- Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia.
| | | | | | | | | | | | | | | |
Collapse
|