1
|
Tran NT, Hale N, Maung AAW, Wiersma M, Walker DW, Polglase G, Castillo-Melendez M, Wong FY. Intrauterine inflammation and postnatal intravenous dopamine alter the neurovascular unit in preterm newborn lambs. J Neuroinflammation 2024; 21:142. [PMID: 38807204 PMCID: PMC11134744 DOI: 10.1186/s12974-024-03137-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Intrauterine inflammation is considered a major cause of brain injury in preterm infants, leading to long-term neurodevelopmental deficits. A potential contributor to this brain injury is dysregulation of neurovascular coupling. We have shown that intrauterine inflammation induced by intra-amniotic lipopolysaccharide (LPS) in preterm lambs, and postnatal dopamine administration, disrupts neurovascular coupling and the functional cerebral haemodynamic responses, potentially leading to impaired brain development. In this study, we aimed to characterise the structural changes of the neurovascular unit following intrauterine LPS exposure and postnatal dopamine administration in the brain of preterm lambs using cellular and molecular analyses. METHODS At 119-120 days of gestation (term = 147 days), LPS was administered into the amniotic sac in pregnant ewes. At 126-7 days of gestation, the LPS-exposed lambs were delivered, ventilated and given either a continuous intravenous infusion of dopamine at 10 µg/kg/min or isovolumetric vehicle solution for 90 min (LPS, n = 6; LPSDA, n = 6). Control preterm lambs not exposed to LPS were also administered vehicle or dopamine (CTL, n = 9; CTLDA, n = 7). Post-mortem brain tissue was collected 3-4 h after birth for immunohistochemistry and RT-qPCR analysis of components of the neurovascular unit. RESULTS LPS exposure increased vascular leakage in the presence of increased vascular density and remodelling with increased astrocyte "end feet" vessel coverage, together with downregulated mRNA levels of the tight junction proteins Claudin-1 and Occludin. Dopamine administration decreased vessel density and size, decreased endothelial glucose transporter, reduced neuronal dendritic coverage, increased cell proliferation within vessel walls, and increased pericyte vascular coverage particularly within the cortical and deep grey matter. Dopamine also downregulated VEGFA and Occludin tight junction mRNA, and upregulated dopamine receptor DRD1 and oxidative protein (NOX1, SOD3) mRNA levels. Dopamine administration following LPS exposure did not exacerbate any effects induced by LPS. CONCLUSION LPS exposure and dopamine administration independently alters the neurovascular unit in the preterm brain. Alterations to the neurovascular unit may predispose the developing brain to further injury.
Collapse
Affiliation(s)
- Nhi T Tran
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Nadia Hale
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | | | - Manon Wiersma
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - David W Walker
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
- Monash Newborn, Monash Medical Centre, Melbourne, Australia
| | - Graeme Polglase
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
- Department of Paediatrics, Monash University, Melbourne, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Flora Y Wong
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.
- Department of Paediatrics, Monash University, Melbourne, Australia.
- Monash Newborn, Monash Medical Centre, Melbourne, Australia.
- Monash Children's Hospital, Level 5, 246 Clayton Rd, Clayton, VIC, 3168, Australia.
| |
Collapse
|
2
|
Logsdon AF, Erickson MA, Herbert MJ, Noonan C, Foresi BD, Qiu J, Lim YP, Banks WA, Stonestreet BS. Inter-alpha inhibitor proteins attenuate lipopolysaccharide-induced blood-brain barrier disruption in neonatal mice. Exp Neurol 2023; 370:114563. [PMID: 37806514 DOI: 10.1016/j.expneurol.2023.114563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/21/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
There is a paucity of information regarding efficacious pharmacological neuroprotective strategies to attenuate or reduce brain injury in neonates. Lipopolysaccharide (LPS) disrupts blood-brain barrier (BBB) function in adult rodents and increases inflammation in adults and neonates. Human blood-derived Inter-alpha Inhibitor Proteins (IAIPs) are neuroprotective, improve neonatal survival after LPS, and attenuate LPS-induced disruption of the BBB in adult male mice. We hypothesized that LPS also disrupts the function of the BBB in neonatal mice and that IAIPs attenuate the LPS-induced BBB disruption in male and female neonatal mice. IAIPs were administered to neonatal mice after LPS and BBB permeability quantified with intravenous 14C-sucrose and 99mTc-albumin. Although repeated high doses (3 mg/kg) of LPS in neonates resulted in high mortality rates and a robust increase in BBB permeability, repeated lower doses (1 mg/kg) of LPS resulted in lower mortality rates and disruption of the BBB in both male and female neonates. IAIP treatment attenuated disruption of the BBB similarly to sucrose and albumin after exposure to low-dose LPS in neonatal mice. Exposure to low-dose LPS elevated IAIP concentrations in blood, but it did not appear to increase the systemic levels of Pre-alpha inhibitor (PaI), one of the family members of the IAIPs that contains heavy chain 3. We conclude that IAIPs attenuate LPS-related disruption of the BBB in both male and female neonatal mice.
Collapse
Affiliation(s)
- Aric F Logsdon
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Melanie J Herbert
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Cassidy Noonan
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Brian D Foresi
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Joseph Qiu
- ProThera Biologics, Inc., Providence, RI 02903, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02903, USA; Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Barbara S Stonestreet
- The Alpert Medical School of Brown University, Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI 02905, USA
| |
Collapse
|
3
|
Time Course of Changes in the Neurovascular Unit after Hypoxic-Ischemic Injury in Neonatal Rats. Int J Mol Sci 2022; 23:ijms23084180. [PMID: 35456999 PMCID: PMC9027443 DOI: 10.3390/ijms23084180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Exposure to hypoxic-ischemic (HI) insults in newborns can predispose them to severe neurological sequela. The mechanisms underlying HI-related brain injury have not been completely elucidated. The neurovascular unit (NVU) is a composite of structures that protect the brain from the influx of detrimental molecules. Changes in the NVU after HI are important because they could reveal endogenous neuroprotective pathways in the cerebral microvasculature. Furthermore, the time course of changes in the NVU after exposure to HI in the newborn remains to be determined. In this study, we examined the effects of severe HI on the time course of changes in the NVU in neonatal rats. Brains were collected from rats exposed to right carotid artery ligation and 2 h of hypoxia on postnatal day 7 with recovery for 6 or 48 h after exposure to sham treatment (Sham) or HI. The right HI and left hypoxic alone sides of the brains were examined by quantitative immunohistochemistry for vascular density (laminin), pericyte vascular coverage (PDGFRβ), astrocyte vascular coverage (GFAP), and claudin-5 expression in the microvasculature of the cerebral cortex, white matter, and hippocampus. HI-related brain injury in neonatal rats was associated with increases in vascular density in the cortex and hippocampus 48 h after HI as well as neurovascular remodeling, including loss of pericyte coverage in the cortex and increases in claudin-5 in the hippocampus 6 h after HI. Astrocyte coverage was not affected by HI injury. The time course of the responses in the different components of the NVU varied after exposure to HI. There were also differential regional responses in the elements of the NVU in response to HI and hypoxia alone.
Collapse
|
4
|
The cerebral haemodynamic response to somatosensory stimulation in preterm newborn lambs is reduced following intrauterine inflammation and dopamine infusion. Exp Neurol 2022; 352:114049. [DOI: 10.1016/j.expneurol.2022.114049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/01/2022] [Accepted: 03/13/2022] [Indexed: 11/18/2022]
|
5
|
Rasile M, Lauranzano E, Mirabella F, Matteoli M. Neurological consequences of neurovascular unit and brain vasculature damages: potential risks for pregnancy infections and COVID-19-babies. FEBS J 2021; 289:3374-3392. [PMID: 33998773 PMCID: PMC8237015 DOI: 10.1111/febs.16020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023]
Abstract
Intragravidic and perinatal infections, acting through either direct viral effect or immune-mediated responses, are recognized causes of liability for neurodevelopmental disorders in the progeny. The large amounts of epidemiological data and the wealth of information deriving from animal models of gestational infections have contributed to delineate, in the last years, possible underpinning mechanisms for this phenomenon, including defects in neuronal migration, impaired spine and synaptic development, and altered activation of microglia. Recently, dysfunctions of the neurovascular unit and anomalies of the brain vasculature have unexpectedly emerged as potential causes at the origin of behavioral abnormalities and psychiatric disorders consequent to prenatal and perinatal infections. This review aims to discuss the up-to-date literature evidence pointing to the neurovascular unit and brain vasculature damages as the etiological mechanisms in neurodevelopmental syndromes. We focus on the inflammatory events consequent to intragravidic viral infections as well as on the direct viral effects as the potential primary triggers. These authors hope that a timely review of the literature will help to envision promising research directions, also relevant for the present and future COVID-19 longitudinal studies.
Collapse
Affiliation(s)
- Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,IRCCS Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Michela Matteoli
- IRCCS Humanitas Clinical and Research Center, Rozzano, Italy.,CNR Institute of Neuroscience, Milano, Italy
| |
Collapse
|
6
|
Gussenhoven R, Ophelders DRMG, Dudink J, Pieterman K, Lammens M, Mays RW, Zimmermann LJ, Kramer BW, Wolfs TGAM, Jellema RK. Systemic multipotent adult progenitor cells protect the cerebellum after asphyxia in fetal sheep. Stem Cells Transl Med 2020; 10:57-67. [PMID: 32985793 PMCID: PMC7780812 DOI: 10.1002/sctm.19-0157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/29/2020] [Accepted: 08/09/2020] [Indexed: 12/30/2022] Open
Abstract
Involvement of the cerebellum in the pathophysiology of hypoxic‐ischemic encephalopathy (HIE) in preterm infants is increasingly recognized. We aimed to assess the neuroprotective potential of intravenously administered multipotent adult progenitor cells (MAPCs) in the preterm cerebellum. Instrumented preterm ovine fetuses were subjected to transient global hypoxia‐ischemia (HI) by 25 minutes of umbilical cord occlusion at 0.7 of gestation. After reperfusion, two doses of MAPCs were administered intravenously. MAPCs are a plastic adherent bone‐marrow‐derived population of adult progenitor cells with neuroprotective potency in experimental and clinical studies. Global HI caused marked cortical injury in the cerebellum, histologically indicated by disruption of cortical strata, impeded Purkinje cell development, and decreased dendritic arborization. Furthermore, global HI induced histopathological microgliosis, hypomyelination, and disruption of white matter organization. MAPC treatment significantly prevented cortical injury and region‐specifically attenuated white matter injury in the cerebellum following global HI. Diffusion tensor imaging (DTI) detected HI‐induced injury and MAPC neuroprotection in the preterm cerebellum. This study has demonstrated in a preclinical large animal model that early systemic MAPC therapy improved structural injury of the preterm cerebellum following global HI. Microstructural improvement was detectable with DTI. These findings support the potential of MAPC therapy for the treatment of HIE and the added clinical value of DTI for the detection of cerebellar injury and the evaluation of cell‐based therapy.
Collapse
Affiliation(s)
- Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital and Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Kay Pieterman
- Biomedical Imaging Group Rotterdam, Department of Radiology and Medical Informatics, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Martin Lammens
- Department of Pathology, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - Robert W Mays
- Regenerative Medicine, Athersys, Inc., Cleveland, Ohio, USA
| | - Luc J Zimmermann
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Reint K Jellema
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
7
|
Disdier C, Awa F, Chen X, Dhillon SK, Galinsky R, Davidson JO, Lear CA, Bennet L, Gunn AJ, Stonestreet BS. Lipopolysaccharide-induced changes in the neurovascular unit in the preterm fetal sheep brain. J Neuroinflammation 2020; 17:167. [PMID: 32466771 PMCID: PMC7257152 DOI: 10.1186/s12974-020-01852-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background Exposure to inflammation during pregnancy can predispose to brain injury in premature infants. In the present study, we investigated the effects of prolonged exposure to inflammation on the cerebrovasculature of preterm fetal sheep. Methods Chronically instrumented fetal sheep at 103–104 days of gestation (full term is ~ 147 days) received continuous low-dose lipopolysaccharide (LPS) infusions (100 ng/kg over 24 h, followed by 250 ng/kg/24 h for 96 h plus boluses of 1 μg LPS at 48, 72, and 96 h) or the same volume of normal saline (0.9%, w/v). Ten days after the start of LPS exposure at 113–114 days of gestation, the sheep were killed, and the fetal brain perfused with formalin in situ. Vessel density, pericyte and astrocyte coverage of the blood vessels, and astrogliosis in the cerebral cortex and white matter were determined using immunohistochemistry. Results LPS exposure reduced (P < 0.05) microvascular vessel density and pericyte vascular coverage in the cerebral cortex and white matter of preterm fetal sheep, and increased the activation of perivascular astrocytes, but decreased astrocytic vessel coverage in the white matter. Conclusions Prolonged exposure to LPS in preterm fetal sheep resulted in decreased vessel density and neurovascular remodeling, suggesting that chronic inflammation adversely affects the neurovascular unit and, therefore, could contribute to long-term impairment of brain development.
Collapse
Affiliation(s)
- Clémence Disdier
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | - Fares Awa
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | - Xiaodi Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | | | - Robert Galinsky
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA.
| |
Collapse
|
8
|
Bell AH, Miller SL, Castillo-Melendez M, Malhotra A. The Neurovascular Unit: Effects of Brain Insults During the Perinatal Period. Front Neurosci 2020; 13:1452. [PMID: 32038147 PMCID: PMC6987380 DOI: 10.3389/fnins.2019.01452] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
The neurovascular unit (NVU) is a relatively recent concept in neuroscience that broadly describes the relationship between brain cells and their blood vessels. The NVU incorporates cellular and extracellular components involved in regulating cerebral blood flow and blood-brain barrier function. The NVU within the adult brain has attracted strong research interest and its structure and function is well described, however, the NVU in the developing brain over the fetal and neonatal period remains much less well known. One area of particular interest in perinatal brain development is the impact of known neuropathological insults on the NVU. The aim of this review is to synthesize existing literature to describe structure and function of the NVU in the developing brain, with a particular emphasis on exploring the effects of perinatal insults. Accordingly, a brief overview of NVU components and function is provided, before discussion of NVU development and how this may be affected by perinatal pathologies. We have focused this discussion around three common perinatal insults: prematurity, acute hypoxia, and chronic hypoxia. A greater understanding of processes affecting the NVU in the perinatal period may enable application of targeted therapies, as well as providing a useful basis for research as it expands further into this area.
Collapse
Affiliation(s)
- Alexander H. Bell
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Ellery SJ, Goss MG, Brew N, Dickinson H, Hale N, LaRosa DA, Walker DW, Wong FY. Evaluation of 3K3A-Activated Protein C to Treat Neonatal Hypoxic Ischemic Brain Injury in the Spiny Mouse. Neurotherapeutics 2019; 16:231-243. [PMID: 30225791 PMCID: PMC6361063 DOI: 10.1007/s13311-018-0661-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) resulting from intrapartum asphyxia is a global problem that causes severe disabilities and up to 1 million deaths annually. A variant form of activated protein C, 3K3A-APC, has cytoprotective properties that attenuate brain injury in models of adult stroke. In this study, we compared the ability of 3K3A-APC and APC (wild-type (wt)) to attenuate neonatal brain injury, using the spiny mouse (Acomys cahirinus) model of intrapartum asphyxia. Pups were delivered at 38 days of gestation (term = 39 days), with an intrapartum hypoxic insult of 7.5 min (intrapartum asphyxia cohort), or immediate removal from the uterus (control cohort). After 1 h, pups received a subcutaneous injection of 3K3A-APC or wild-type APC (wtAPC) at 7 mg/kg, or vehicle (saline). At 24 h of age, pups were killed and brain tissue was collected for measurement of inflammation and cell death using RT-qPCR and histopathology. Intrapartum asphyxia increased weight loss, inflammation, and apoptosis/necrosis in the newborn brain. 3K3A-APC administration maintained body weight and ameliorated an asphyxia-induced increase of TGFβ1 messenger RNA expression in the cerebral cortex, immune cell aggregation in the corpus callosum, and cell death in the deep gray matter and hippocampus. In the cortex, 3K3A-APC appeared to exacerbate the immune response to the hypoxic ischemic insult. While wtAPC reduced cell death in the corpus callosum and hippocampus following intrapartum asphyxia, it increased markers of neuro-inflammation and cell death in control pups. These findings suggest 3K3A-APC administration may be a useful therapy to reduce cell death and neonatal brain injury associated with HIE.
Collapse
Affiliation(s)
- Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia.
| | - Madeleine G Goss
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
| | - Nadine Brew
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
| | - Hayley Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
| | - Domenic A LaRosa
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
- Women and Infants Hospital, Alpert Medical School, Brown University, Providence, RI, USA
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Flora Y Wong
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Paediatrics, Monash University, Clayton, Australia
- Monash Newborn, Monash Medical Centre, Clayton, Melbourne, Australia
| |
Collapse
|
10
|
Dobutamine treatment reduces inflammation in the preterm fetal sheep brain exposed to acute hypoxia. Pediatr Res 2018; 84:442-450. [PMID: 29976968 DOI: 10.1038/s41390-018-0045-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Impaired cerebral autoregulation in preterm infants makes circulatory management important to avoid cerebral hypoxic-ischemic injury. Dobutamine is frequently used as inotropic treatment in preterm neonates, but its effects on the brain exposed to cerebral hypoxia are unknown. We hypothesized that dobutamine would protect the immature brain from cerebral hypoxic injury. METHODS In preterm (0.6 gestation) fetal sheep, dobutamine (Dob, 10 μg/kg/min) or saline (Sal) was infused intravenously for 74 h. Two hours after the beginning of the infusion, umbilical cord occlusion (UCO) was performed to produce fetal asphyxia (Sal+UCO: n = 9, Dob+UCO: n = 7), or sham occlusion (Sal+sham: n = 7, Dob+sham: n = 6) was performed. Brains were collected 72 h later for neuropathology. RESULTS Dobutamine did not induce cerebral changes in the sham UCO group. UCO increased apoptosis and microglia density in white matter, hippocampus, and caudate nucleus, and astrocyte density in the caudate nucleus. Dobutamine commenced before UCO reduced microglia infiltration in the white matter, and microglial and astrocyte density in the caudate. CONCLUSION In preterm hypoxia-induced brain injury, dobutamine decreases neuroinflammation in the white matter and caudate, and reduces astrogliosis in the caudate. Early administration of dobutamine in preterm infants for cardiovascular stabilization appears safe and may be neuroprotective against unforeseeable cerebral hypoxic injury.
Collapse
|
11
|
Mallard C, Ek CJ, Vexler ZS. The myth of the immature barrier systems in the developing brain: role in perinatal brain injury. J Physiol 2018. [PMID: 29528501 DOI: 10.1113/jp274938] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Central nervous system homeostasis is maintained by cellular barriers that protect the brain from external environmental changes and protect the CNS from harmful molecules and pathogens in the blood. Historically, for many years these barriers were thought of as immature, with limited functions, during brain development. In this review, we will present advances in the understanding of the barrier systems during development and evidence to show that in fact the barriers serve many important neurodevelopmental functions and that fetal and newborn brains are well protected. We will also discuss how ischaemic injury or systemic inflammation may breach the integrity of the barriers in the developing brain.
Collapse
Affiliation(s)
- Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - C Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, USA
| |
Collapse
|
12
|
Abstract
Drug bioavailability to the developing brain is a major concern in the treatment of neonates and infants as well as pregnant and breast-feeding women. Central adverse drug reactions can have dramatic consequences for brain development, leading to major neurological impairment. Factors setting the cerebral bioavailability of drugs include protein-unbound drug concentration in plasma, local cerebral blood flow, permeability across blood-brain interfaces, binding to neural cells, volume of cerebral fluid compartments, and cerebrospinal fluid secretion rate. Most of these factors change during development, which will affect cerebral drug concentrations. Regarding the impact of blood-brain interfaces, the blood-brain barrier located at the cerebral endothelium and the blood-cerebrospinal fluid barrier located at the choroid plexus epithelium both display a tight phenotype early on in embryos. However, the developmental regulation of some multispecific efflux transporters that also limit the entry of numerous drugs into the brain through barrier cells is expected to favor drug penetration in the neonatal brain. Finally, drug cerebral bioavailability is likely to be affected following perinatal injuries that alter blood-brain interface properties. A thorough investigation of these mechanisms is mandatory for a better risk assessment of drug treatments in pregnant or breast-feeding women, and in neonate and pediatric patients.
Collapse
Affiliation(s)
- Jean-François Ghersi-Egea
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028 CRNS UMR 5292, Université Claude Bernard Lyon-1, 69008, Lyon, France.
- Blood-Brain Interfaces Exploratory Platform BIP, Lyon Neurosciences Research Center, 69008, Lyon, France.
| | - Elodie Saudrais
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028 CRNS UMR 5292, Université Claude Bernard Lyon-1, 69008, Lyon, France
- Blood-Brain Interfaces Exploratory Platform BIP, Lyon Neurosciences Research Center, 69008, Lyon, France
| | - Nathalie Strazielle
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028 CRNS UMR 5292, Université Claude Bernard Lyon-1, 69008, Lyon, France
- Blood-Brain Interfaces Exploratory Platform BIP, Lyon Neurosciences Research Center, 69008, Lyon, France
- Brain-I, 69008, Lyon, France
| |
Collapse
|
13
|
McDougall ARA, Hale N, Rees S, Harding R, De Matteo R, Hooper SB, Tolcos M. Erythropoietin Protects Against Lipopolysaccharide-Induced Microgliosis and Abnormal Granule Cell Development in the Ovine Fetal Cerebellum. Front Cell Neurosci 2017; 11:224. [PMID: 28804448 PMCID: PMC5532439 DOI: 10.3389/fncel.2017.00224] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 07/13/2017] [Indexed: 11/13/2022] Open
Abstract
Erythropoietin (EPO) ameliorates inflammation-induced injury in cerebral white matter (WM). However, effects of inflammation on the cerebellum and neuroprotective effects of EPO are unknown. Our aims were to determine: (i) whether lipopolysaccharide (LPS)-induced intrauterine inflammation causes injury to, and/or impairs development of the cerebellum; and (ii) whether recombinant human EPO (rhEPO) mitigates these changes. At 107 ± 1 days gestational age (DGA; ~0.7 of term), fetal sheep received LPS (~0.9 μg/kg; i.v.) or an equivalent volume of saline, followed 1 h later with 5000 IU/kg rhEPO (i.v.) or an equivalent volume of saline (i.v.). This generated the following experimental groups: control (saline + saline; n = 6), LPS (LPS + saline, n = 8) and LPS + rhEPO (n = 8). At necropsy (116 ± 1 DGA; ~0.8 of term) the brain was perfusion-fixed and stained histologically (H&E) and immunostained to identify granule cells (Neuronal Nuclei, NeuN), granule cell proliferation (Ki67), Bergmann glia (glial fibrillary acidic protein, GFAP), astrogliosis (GFAP) and microgliosis (Iba-1). In comparison to controls, LPS fetuses had an increased density of Iba-1-positive microglia (p < 0.005) in the lobular WM; rhEPO prevented this increase (p < 0.05). The thickness of both the proliferative (Ki67-positive) and post-mitotic zones (Ki67-negative) of the EGL were increased in LPS-exposed fetuses compared to controls (p < 0.05), but were not different between controls and LPS + rhEPO fetuses. LPS also increased (p < 0.001) the density of granule cells (NeuN-positive) in the internal granule layer (IGL); rhEPO prevented the increase (p < 0.01). There was no difference between groups in the areas of the vermis (total cross-section), molecular layer (ML), IGL or WM, the density of NeuN-positive granule cells in the ML, the linear density of Bergmann glial fibers, the areal density or somal area of the Purkinje cells, the areal coverage of GFAP-positive astrocytes in the lobular and deep WM, the density of Iba-1-positive microglia in the deep WM or the density of apopotic cells in the cerebellum. LPS-induced intrauterine inflammation caused microgliosis and abnormal development of granule cells. rhEPO ameliorated these changes, suggesting that it is neuroprotective against LPS-induced inflammatory effects in the cerebellum.
Collapse
Affiliation(s)
- Annie R A McDougall
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Sandra Rees
- Department of Anatomy and Neuroscience, University of MelbourneParkville, VIC, Australia
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash UniversityClayton, VIC, Australia
| | - Robert De Matteo
- Department of Anatomy and Developmental Biology, Monash UniversityClayton, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT UniversityMelbourne, VIC, Australia
| |
Collapse
|
14
|
Patra A, Chen X, Sadowska GB, Zhang J, Lim YP, Padbury JF, Banks WA, Stonestreet BS. Neutralizing anti-interleukin-1β antibodies reduce ischemia-related interleukin-1β transport across the blood-brain barrier in fetal sheep. Neuroscience 2017; 346:113-125. [PMID: 28089577 DOI: 10.1016/j.neuroscience.2016.12.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
Hypoxic ischemic insults predispose to perinatal brain injury. Pro-inflammatory cytokines are important in the evolution of this injury. Interleukin-1β (IL-1β) is a key mediator of inflammatory responses and elevated IL-1β levels in brain correlate with adverse neurodevelopmental outcomes after brain injury. Impaired blood-brain barrier (BBB) function represents an important component of hypoxic-ischemic brain injury in the fetus. In addition, ischemia-reperfusion increases cytokine transport across the BBB of the ovine fetus. Reducing pro-inflammatory cytokine entry into brain could represent a novel approach to attenuate ischemia-related brain injury. We hypothesized that infusions of neutralizing IL-1β monoclonal antibody (mAb) reduce IL-1β transport across the BBB after ischemia in the fetus. Fetal sheep were studied 24-h after 30-min of carotid artery occlusion. Fetuses were treated with placebo- or anti-IL-1β mAb intravenously 15-min and 4-h after ischemia. Ovine IL-1β protein expressed from IL-1β pGEX-2T vectors in Escherichia coli (E. coli) BL-21 cells was produced, purified, and radiolabeled with 125I. BBB permeability was quantified using the blood-to-brain transfer constant (Ki) with 125I-radiolabeled-IL-1β. Increases in anti-IL-1β mAb were observed in the brain of the mAb-treated group (P<0.001). Blood-to-brain transport of 125I-IL-1β was lower (P<0.04) across brain regions in the anti-IL-1β mAb-treated than placebo-treated ischemic fetuses. Plasma 125I-IL-1β counts were higher (P<0.001) in the anti-IL-1β mAb- than placebo-treated ischemic fetuses. Systemic infusions of anti-IL-1β mAb reduce IL-1β transport across the BBB after ischemia in the ovine fetus. Our findings suggest that conditions associated with increases in systemic pro-inflammatory cytokines and neurodevelopmental impairment could benefit from an anti-cytokine therapeutic strategy.
Collapse
Affiliation(s)
- Aparna Patra
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States.
| | - Xiaodi Chen
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - Grazyna B Sadowska
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - Jiyong Zhang
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - Yow-Pin Lim
- ProThera Biologics, Providence, RI 02903, United States
| | - James F Padbury
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, United States
| | - Barbara S Stonestreet
- Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, United States.
| |
Collapse
|
15
|
Spasova MS, Chen X, Sadowska GB, Horton ER, Lim YP, Stonestreet BS. Ischemia reduces inter-alpha inhibitor proteins in the brain of the ovine fetus. Dev Neurobiol 2016; 77:726-737. [PMID: 27618403 DOI: 10.1002/dneu.22451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 01/04/2023]
Abstract
Hypoxic-ischemic (HI) brain injury is a major cause of neurological abnormalities in the perinatal period. Inflammation contributes to the evolution of HI brain injury. Inter-alpha inhibitor proteins (IAIPs) are a family of proteins that are part of the innate immune system. We have reported that endogenous IAIPs exhibit developmental changes in ovine brain and that exogenous IAIP treatment reduces neuronal death in HI neonatal rats. However, the effects of HI on endogenous IAIPs in brain have not been previously examined. In this study, we examined the effects of ischemia-reperfusion on endogenous IAIPs levels in fetal sheep brain. Cerebral cortex, cerebellum, cervical spinal cord, choroid plexus, and CSF were snap frozen from sham control fetuses at 127 days gestation and after 30-min of carotid occlusion and 4-, 24-, and 48-h of reperfusion. IAIP levels were determined by Western immunoblot. IAIP expressions of the 250 kDa Inter-alpha inhibitor (IaI) and 125 kDa Pre-alpha inhibitor (PaI) in cerebral cortex and PaI in cerebellum were reduced (p < 0.05) 4-h after ischemia compared with controls and returned toward control levels 24- and 48-h after ischemia. CSF PaI and IaI were reduced 48 h after ischemia. We conclude that IAIPs in cerebral cortex and cerebellum are reduced by brain ischemia, and return toward control levels between 24 and 48 h after ischemia. However, changes in CSF IAIPs were delayed, exhibiting decreases 48 h after ischemia. We speculate that the decreases in endogenous IAIPs reflect increased utilization, potentially suggesting that they have endogenous neuroprotective properties. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 726-737, 2017.
Collapse
Affiliation(s)
- Mariya S Spasova
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Xiaodi Chen
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Grazyna B Sadowska
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Edward R Horton
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| | - Yow-Pin Lim
- ProThera Biologics, Inc, Providence, RI, 02903
| | - Barbara S Stonestreet
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, 02905
| |
Collapse
|
16
|
Brew N, Azhan A, den Heijer I, Boomgardt M, Davies G, Nitsos I, Miller S, Walker A, Walker D, Wong F. Dopamine treatment during acute hypoxia is neuroprotective in the developing sheep brain. Neuroscience 2016; 316:82-93. [DOI: 10.1016/j.neuroscience.2015.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 12/04/2015] [Accepted: 12/14/2015] [Indexed: 11/15/2022]
|
17
|
Rizzo GS, Sen S. Maternal obesity and immune dysregulation in mother and infant: A review of the evidence. Paediatr Respir Rev 2015; 16:251-7. [PMID: 25454382 DOI: 10.1016/j.prrv.2014.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/17/2014] [Indexed: 01/06/2023]
Abstract
Obesity is a worldwide public health epidemic. Increasing numbers of reproductive-age women enter pregnancy overweight or obese and there is now convincing data that this adverse in utero environment impacts both fetal and lifelong development. Epidemiologic evidence has shown a simultaneous increase in obesity and asthma rates in developed countries and maternal obesity is a risk factor for infant asthma and wheeze. Here we review the state of research linking maternal obesity and immunomodulation in both mother and infant, with specific attention to the relationship between maternal obesity and offspring asthma. We will also propose several different mechanisms by which maternal obesity may predispose offspring to this chronic condition and briefly summarize interventions that have been trialed to limit this association.
Collapse
Affiliation(s)
- Giulia S Rizzo
- Tufts University School of Medicine, 145 Harrison Ave, Boston, MA 02111, USA.
| | - Sarbattama Sen
- Brigham and Women's Hospital, Department of Pediatric Newborn Medicine, 75 Francis St, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Interleukin-1β transfer across the blood-brain barrier in the ovine fetus. J Cereb Blood Flow Metab 2015; 35:1388-95. [PMID: 26082012 PMCID: PMC4640327 DOI: 10.1038/jcbfm.2015.134] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 11/09/2022]
Abstract
Pro-inflammatory cytokines contribute to hypoxic-ischemic brain injury. Blood-brain barrier (BBB) dysfunction represents an important component of hypoxic-ischemic brain injury in the fetus. Hypoxic-ischemic injury could accentuate systemic cytokine transfer across the fetal BBB. There has been considerable conjecture suggesting that systemic cytokines could cross the BBB during the perinatal period. Nonetheless, evidence to support this contention is sparse. We hypothesized that ischemia-reperfusion increases the transfer of systemic interleukin-1β (IL-1β) across the BBB in the fetus. Ovine fetuses at 127 days of gestation were studied 4 hours after 30 minutes of bilateral carotid artery occlusion and compared with a nonischemic group. Recombinant ovine IL-1β protein was expressed from an IL-1β pGEX-2 T vector in E. coli BL-21 cells and purified. The BBB function was quantified in 12 brain regions using a blood-to-brain transfer constant with intravenous (125)I-radiolabeled IL-1β ((125)I-IL-1β). Interleukin-1β crossed the intact BBB in nonischemic fetuses. Blood-to-brain transport of (125)I-IL-1β was higher (P<0.05) across brain regions in fetuses exposed to ischemia-reperfusion than nonischemic fetuses. We conclude that systemic IL-1β crosses the intact fetal BBB, and that ischemia-reperfusion increases transfer of this cytokine across the fetal BBB. Therefore, altered BBB function after hypoxia-ischemia facilitates entry of systemic cytokines into the brain of the fetus.
Collapse
|
19
|
Cao M, Cortes M, Moore CS, Leong SY, Durosier LD, Burns P, Fecteau G, Desrochers A, Auer RN, Barreiro LB, Antel JP, Frasch MG. Fetal microglial phenotype in vitro carries memory of prior in vivo exposure to inflammation. Front Cell Neurosci 2015; 9:294. [PMID: 26300730 PMCID: PMC4524165 DOI: 10.3389/fncel.2015.00294] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 07/16/2015] [Indexed: 01/01/2023] Open
Abstract
Objective: Neuroinflammation in utero may result in life-long neurological disabilities. The molecular mechanisms whereby microglia contribute to this response remain incompletely understood. Methods: Lipopolysaccharide (LPS) or saline were administered intravenously to non-anesthetized chronically instrumented near-term fetal sheep to model fetal inflammation in vivo. Microglia were then isolated from in vivo LPS and saline (naïve) exposed animals. To mimic the second hit of neuroinflammation, these microglia were then re-exposed to LPS in vitro. Cytokine responses were measured in vivo and subsequently in vitro in the primary microglia cultures derived from these animals. We sequenced the whole transcriptome of naïve and second hit microglia and profiled their genetic expression to define molecular pathways disrupted during neuroinflammation. Results:In vivo LPS exposure resulted in IL-6 increase in fetal plasma 3 h post LPS exposure. Even though not histologically apparent, microglia acquired a pro-inflammatory phenotype in vivo that was sustained and amplified in vitro upon second hit LPS exposure as measured by IL-1β response in vitro and RNAseq analyses. While NFKB and Jak-Stat inflammatory pathways were up regulated in naïve microglia, heme oxygenase 1 (HMOX1) and Fructose-1,6-bisphosphatase (FBP) genes were uniquely differentially expressed in the second hit microglia. Compared to the microglia exposed to LPS in vitro only, the transcriptome of the in vivo LPS pre-exposed microglia showed a diminished differential gene expression in inflammatory and metabolic pathways prior and upon re-exposure to LPS in vitro. Notably, this desensitization response was also observed in histone deacetylases (HDAC) 1, 2, 4, and 6. Microglial calreticulin/LRP genes implicated in microglia-neuronal communication relevant for the neuronal development were up regulated in second hit microglia. Discussion: We identified a unique HMOX1down and FBPup phenotype of microglia exposed to the double-hit suggesting interplay of inflammatory and metabolic pathways. Our findings suggest that epigenetic mechanisms mediate this immunological and metabolic memory of the prior inflammatory insult relevant to neuronal development and provide new therapeutic targets for early postnatal intervention to prevent brain injury.
Collapse
Affiliation(s)
- Mingju Cao
- Department of Obstetrics and Gynaecology, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada ; Department of Neurosciences, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada
| | - Marina Cortes
- Faculty of Veterinary Medicine, Animal Reproduction Research Centre, Université de Montréal Montréal, QC, Canada
| | - Craig S Moore
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University Montréal, QC, Canada
| | - Soo Yuen Leong
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University Montréal, QC, Canada
| | - Lucien D Durosier
- Department of Obstetrics and Gynaecology, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada ; Department of Neurosciences, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada
| | - Patrick Burns
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal QC, Canada
| | - Gilles Fecteau
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal QC, Canada
| | - Andre Desrochers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal QC, Canada
| | - Roland N Auer
- Département de Pathologie, University Hospital Ste-Justine, Université de Montréal QC, Canada
| | - Luis B Barreiro
- Department of Pediatrics, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University Montréal, QC, Canada
| | - Martin G Frasch
- Department of Obstetrics and Gynaecology, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada ; Department of Neurosciences, Faculty of Medicine, CHU Ste-Justine Research Centre, Université de Montréal Montréal, QC, Canada ; Faculty of Veterinary Medicine, Animal Reproduction Research Centre, Université de Montréal Montréal, QC, Canada
| |
Collapse
|
20
|
Cerebrovascular adaptations to chronic hypoxia in the growth restricted lamb. Int J Dev Neurosci 2015; 45:55-65. [PMID: 25639519 DOI: 10.1016/j.ijdevneu.2015.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/27/2015] [Accepted: 01/27/2015] [Indexed: 12/11/2022] Open
Abstract
Chronic moderate hypoxia induces angiogenic adaptation in the brain, reflecting a modulatory role for oxygen in determining cerebrovascular development. Chronic intrauterine fetal hypoxia, such as occurs in intrauterine growth restriction (IUGR) is likely to lead to a reduction in oxygen delivery to the brain and long-term neurological abnormalities. Thus we investigated whether vascular remodeling and vascular abnormalities were evident in the brain of IUGR newborn lambs that were chronically hypoxic in utero. Single uterine artery ligation (SUAL) surgery was performed in fetuses at ∼ 105 days gestation (term ∼ 145 days) to induce placental insufficiency and IUGR. Ewes delivered naturally at term and lambs were euthanased 24h later. IUGR brains (n = 9) demonstrated a significant reduction in positive staining for the number of blood vessels (laminin immunohistochemistry) compared with control (n = 8): from 1650 ± 284 to 416 ± 47 cells/mm(2) in subcortical white matter (SCWM) 1793 ± 298 to 385 ± 20 cells/mm(2) in periventricular white matter (PVWM), and 1717 ± 161 to 405 ± 84 cells/mm(2) in the subventricular zone (SVZ). The decrease in vascular density was associated with a significant decrease in VEGF immunoreactivity. The percentage of blood vessels exhibiting endothelial cell proliferation (Ki67 positive) varied regionally between 14 to 22% in white matter of control lambs, while only 1-3% of blood vessels in IUGR brains showed proliferation. A 66% reduction in pericyte coverage (α-SMA and desmin) of blood vessels was observed in SCWM, 71% in PVWM, and 73% in SVZ of IUGR lambs, compared to controls. A reduction in peri-vascular astrocytes (GFAP and laminin) was also observed throughout the white matter of IUGR lambs, and extravasation of albumin into the brain parenchyma was present, indicative of increased permeability of the blood brain barrier. Chronic hypoxia associated with IUGR results in a reduction in vascular density in the white matter of IUGR newborn brains. Vascular pericyte coverage and peri-vascular astrocytes, both of which are essential for stabilisation of blood vessels and the maintenance of vascular permeability, were also decreased in the white matter of IUGR lambs. In turn, these vascular changes could lead to inadequate oxygen supply and contribute to under-perfusion and increased vulnerability of white matter in IUGR infants.
Collapse
|
21
|
Abstract
BACKGROUND Fetal hypoxia contributes significantly to the pathogenesis of permanent perinatal brain injury. We hypothesized that hypoxia-induced cerebral angiogenesis and microvascular changes would occur in fetal sheep subjected to a severe hypoxic insult produced by umbilical cord occlusion (UCO) for 10 min. METHODS At 124-126 d of gestation, singleton fetal sheep underwent surgery for implantation of catheters and placement of an inflatable cuff around the umbilical cord. A 10-min UCO or sham UCO (n = 5) was induced at 130 d gestation. The fetal brain was collected at 24 h (n = 5) or 48 h (n = 4) after UCO for immunohistochemical analysis of vascular endothelial growth factor (VEGF), Ki67, and serum albumin. RESULTS By 48 h after UCO, the percentage of blood vessels expressing VEGF had increased in the subventricular zone, periventricular and subcortical white matter, corpus callosum, and cortex. Alterations in vascular permeability (albumin extravasation) were observed only in the periventricular and subcortical white matter and the subventricular zone following UCO. CONCLUSION The upregulation of VEGF expression and increased leakage of plasma protein in the fetal sheep brain show that the microvasculature in white matter is sensitive to hypoxia in the near-term brain.
Collapse
|
22
|
Strackx E, Gantert M, Moers V, van Kooten IAJ, Rieke R, Hürter H, Lemmens MAM, Steinbusch HWM, Zimmermann LJI, Vles JSH, Garnier Y, Gavilanes AWD, Kramer BW. Increased number of cerebellar granule cells and astrocytes in the internal granule layer in sheep following prenatal intra-amniotic injection of lipopolysaccharide. THE CEREBELLUM 2012; 11:132-44. [PMID: 21773814 PMCID: PMC3311858 DOI: 10.1007/s12311-011-0297-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chorioamnionitis is an important problem in perinatology today, leading to brain injury and neurological handicaps. However, there are almost no data available regarding chorioamnionitis and a specific damage of the cerebellum. Therefore, this study aimed at determining if chorioamnionitis causes cerebellar morphological alterations. Chorioamnionitis was induced in sheep by the intra-amniotic injection of lipopolysaccharide (LPS) at a gestational age (GA) of 110 days. At a GA of 140 days, we assessed the mean total and layer-specific volume and the mean total granule cell (GCs) and Purkinje cell (PC) number in the cerebelli of LPS-exposed and control animals using high-precision design-based stereology. Astrogliosis was assessed in the gray and white matter (WM) using a glial fibrillary acidic protein staining combined with gray value image analysis. The present study showed an unchanged volume of the total cerebellum as well as the molecular layer, outer and inner granular cell layers (OGL and IGL, respectively), and WM. Interestingly, compared with controls, the LPS-exposed brains showed a statistically significant increase (+20.4%) in the mean total number of GCs, whereas the number of PCs did not show any difference between the two groups. In addition, LPS-exposed animals showed signs of astrogliosis specifically affecting the IGL. Intra-amniotic injection of LPS causes morphological changes in the cerebellum of fetal sheep still detectable at full-term birth. In this study, changes were restricted to the inner granule layer. These cerebellar changes might correspond to some of the motor or non-motor deficits seen in neonates from compromised pregnancies.
Collapse
Affiliation(s)
- Eveline Strackx
- Department of Neuroscience and European Graduate School of Neuroscience, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hsiao EY, Patterson PH. Placental regulation of maternal-fetal interactions and brain development. Dev Neurobiol 2012; 72:1317-26. [PMID: 22753006 DOI: 10.1002/dneu.22045] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 06/09/2012] [Accepted: 06/19/2012] [Indexed: 01/16/2023]
Abstract
A variety prenatal insults are associated with the incidence of neurodevelopmental disorders such as schizophrenia, autism and cerebral palsy. While the precise mechanisms underlying how transient gestational challenges can lead to later life dysfunctions are largely unknown, the placenta is likely to play a key role. The literal interface between maternal and fetal cells resides in the placenta, and disruptions to the maternal or intrauterine environment are necessarily conveyed to the developing embryo via the placenta. Placental cells bear the responsibility of promoting maternal tolerance of the semiallogeneic fetus and regulating selective permeability of nutrients, gases, and antibodies, while still providing physiological protection of the embryo from adversity. The placenta's critical role in modulating immune protection and the availability of nutrients and endocrine factors to the offspring implicates its involvement in autoimmunity, growth restriction and hypoxia, all factors associated with the development of neurological complications. In this review, we summarize primary maternal-fetal interactions that occur in the placenta and describe pathways by which maternal insults can impair these processes and disrupt fetal brain development. We also review emerging evidence for placental dysfunction in the prenatal programming of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elaine Y Hsiao
- Biology Division, California Institute of Technology, Pasadena, California, USA.
| | | |
Collapse
|
24
|
Miller SL, Wallace EM, Walker DW. Antioxidant therapies: a potential role in perinatal medicine. Neuroendocrinology 2012; 96:13-23. [PMID: 22377769 DOI: 10.1159/000336378] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/30/2011] [Indexed: 11/19/2022]
Abstract
Pregnancies complicated by impaired placentation, acute severe reductions in oxygen supply to the fetus, or intrauterine infection are associated with oxidative stress to the mother and developing baby. Such oxidative stress is characterized as an upregulation in the production of oxidative or nitrative free radicals and a concomitant decrease in the availability of antioxidant species, thereby creating a state of fetoplacental oxidative imbalance. Recently, there has been a good deal of interest in the potential for the use of antioxidant therapies in the perinatal period to protect the fetus, particularly the developing brain, against oxidative stress in complications of pregnancy and birth. This review will examine why the immature brain is particularly susceptible to oxidative imbalance and will provide discussion on antioxidant treatments currently receiving attention in the adult and perinatal literature - allopurinol, melatonin, α-lipoic acid, and vitamins C and E. In addition, we aim to address the interaction between oxidative stress and the fetal inflammatory response, an interaction that may be vital when proposing antioxidant or other neuroprotective strategies.
Collapse
Affiliation(s)
- S L Miller
- The Ritchie Centre, Monash Institute of Medical Research, Clayton, Vic., Australia
| | | | | |
Collapse
|
25
|
Dean JM, van de Looij Y, Sizonenko SV, Lodygensky GA, Lazeyras F, Bolouri H, Kjellmer I, Huppi PS, Hagberg H, Mallard C. Delayed cortical impairment following lipopolysaccharide exposure in preterm fetal sheep. Ann Neurol 2011; 70:846-56. [PMID: 22002627 DOI: 10.1002/ana.22480] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 04/28/2011] [Accepted: 05/06/2011] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Preterm infants exhibit chronic deficits in white matter (WM) and cortical maturation. Although fetal infection/inflammation may contribute to WM pathology, the factors contributing to cortical changes are largely unknown. We examined the effect of fetal lipopolysaccharide (LPS) exposure on WM and cortical development as assessed by magnetic resonance imaging (MRI), electroencephalography (EEG), and histopathology in fetal sheep at preterm human equivalent age. METHODS LPS was administered to fetal sheep at 102.5 ± 0.5 days of gestation. Continuous biophysical recordings were analyzed for 10 days after LPS. At postmortem, measurement of cerebral WM and cortical tissue volumes was achieved by stereological techniques. Specific effects of LPS on MRI-assessed T(1)-weighted and T(2)-weighted images, and immunohistochemical expression of oligodendrocytes, proliferating cells, cortical NeuN-positive and Nurr1-positive neurons (subplate marker), and cell death mechanisms were examined. RESULTS We observed reductions in WM (~21%; LPS, 1.19 ± 0.04 vs control, 1.51 ± 0.07 cm(3); p < 0.001) and cortical (~18%; LPS, 2.34 ± 0.10 vs control, 2.85 ± 0.07 cm(3); p < 0.001) volumes, associated with overt and diffuse WM injury, T(1)-/T(2) -weighted signal alterations, and reduced numbers of WM oligodendrocytes (LPS, 485 ± 31 vs control, 699 ± 69 cells/mm(2); p = 0.0189) and NeuN-positive (LPS, 421 ± 71 vs control 718 ± 92 cells/mm(2); p = 0.04) and Nurr1-positive (control, 2.5 ± 0.6 vs LPS, 0.6 ± 0.1 cells/mm(2); p = 0.007) cortical neurons after LPS. Moreover, there was loss of the normal maturational increase in cortical EEG amplitude, which correlated with reduced cortical volumes. INTERPRETATION Fetal exposure to LPS prior to myelination onset can impair both white matter and cortical development in a preclinical large animal model, supporting a role for maternal/fetal infection in the pathogenesis of preterm brain injury.
Collapse
Affiliation(s)
- Justin M Dean
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Silbereis JC, Huang EJ, Back SA, Rowitch DH. Towards improved animal models of neonatal white matter injury associated with cerebral palsy. Dis Model Mech 2011; 3:678-88. [PMID: 21030421 DOI: 10.1242/dmm.002915] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Newborn neurological injuries are the leading cause of intellectual and motor disabilities that are associated with cerebral palsy. Cerebral white matter injury is a common feature in hypoxic-ischemic encephalopathy (HIE), which affects full-term infants, and in periventricular leukomalacia (PVL), which affects preterm infants. This article discusses recent efforts to model neonatal white matter injury using mammalian systems. We emphasize that a comprehensive understanding of oligodendrocyte development and physiology is crucial for obtaining new insights into the pathobiology of HIE and PVL as well as for the generation of more sophisticated and faithful animal models.
Collapse
Affiliation(s)
- John C Silbereis
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
27
|
Wang W, Li S, Dong HP, Lv S, Tang YY. Differential impairment of spatial and nonspatial cognition in a mouse model of brain aging. Life Sci 2009; 85:127-35. [DOI: 10.1016/j.lfs.2009.05.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 04/24/2009] [Accepted: 05/03/2009] [Indexed: 02/04/2023]
|
28
|
Dean J, Farrag D, Zahkouk S, El Zawahry E, Hagberg H, Kjellmer I, Mallard C. Cerebellar white matter injury following systemic endotoxemia in preterm fetal sheep. Neuroscience 2009; 160:606-15. [DOI: 10.1016/j.neuroscience.2009.02.071] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 02/24/2009] [Accepted: 02/26/2009] [Indexed: 10/21/2022]
|
29
|
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: 3.0] [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
|
30
|
Microglial activation, macrophage infiltration, and evidence of cell death in the fetal brain after uteroplacental administration of lipopolysaccharide in sheep in late gestation. Am J Obstet Gynecol 2008; 198:117.e1-11. [PMID: 18166323 DOI: 10.1016/j.ajog.2007.06.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 05/07/2007] [Accepted: 06/08/2007] [Indexed: 11/20/2022]
Abstract
OBJECTIVE The purpose of this study was to determine whether uteroplacental delivery of endotoxin produces fetal systemic and central nervous system reactions that are suggestive of inflammation. STUDY DESIGN Lipopolysaccharide (30 or 60 microg) was administered into the uterine artery of late gestation (135 +/- 0.3 days) pregnant sheep. Fetal blood was assayed to determine changes in levels of quinolinic acid, which is a metabolite of tryptophan that is produced by monocytes (macrophages, microglia). Fetal brains were collected after 72 hours and examined for the presence of activated microglia and parenchymal macrophages. RESULTS The brains of treated fetuses showed microglial activation and macrophage infiltration, which varied between brain region and lipopolysaccharide dose. Cell death that had been determined by cresyl violet/acid fuchsin staining was observed in the external capsule. There was significant increase of quinolinic acid in the fetal circulation, but no lipopolysaccharide was detected. CONCLUSION Uteroplacental inflammation results in significant microglial activation and macrophage infiltration without direct fetal exposure to endotoxin, which suggests that placental responses contribute to perinatal brain damage that is associated with infection during pregnancy.
Collapse
|