1
|
Liu S, Xu Y, Yao X, Cao H, Zhou H, Luo J, Gao H, Chen B, Chen H, Xie T, Zhan X. Perillaldehyde ameliorates sepsis-associated acute kidney injury via inhibiting HSP90AA1-mediated ferroptosis and pyroptosis: Molecular structure and protein interaction of HSP90AA1. Int J Biol Macromol 2025; 304:140954. [PMID: 39947536 DOI: 10.1016/j.ijbiomac.2025.140954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
Heat shock protein 90α (HSP90AA1) is a molecular chaperone involved in a variety of cellular processes. Special attention is paid to how perillaldehyde ameliorates kidney injury by inhibiting HSP90AA1-mediated iron and pyrotoxicity, and in-depth analysis of the molecular structure and protein interactions of HSP90AA-1. The interaction between perillaldehyde and HSP90AA1 and the effect of perillaldehyde on the molecular structure of HSP90AA1 were analyzed by molecular docking and surface plasmon resonance technique. Western blot and immunohistochemical results showed that perillaldehyde could decrease the expression of HSP90AA1 and change its distribution in the kidney. Molecular docking and surface plasmonic resonance experiments revealed the high affinity binding between perillaldehyde and HSP90AA1, and further analysis showed that perillaldehyde could induce the conformational change of HSP90AA1, thereby inhibiting its function.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yunfei Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Heng Cao
- Department of Urology, The Third the People's Hospital of Bengbu, Bengbu Medical College, Bengbu 233000, China
| | - Hongmin Zhou
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jun Luo
- Department of Urology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Hanlu Gao
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Bowen Chen
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Hao Chen
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Tiancheng Xie
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xiangcheng Zhan
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
2
|
Hu M, Bai C, Zhao H, Wu J, Luan X. Research Progress on the Role of the Interleukin Family in the Pathogenesis of Cerebral Palsy in Children. J Integr Neurosci 2024; 23:213. [PMID: 39735959 DOI: 10.31083/j.jin2312213] [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: 03/20/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 12/31/2024] Open
Abstract
Cerebral palsy (CP), a common neurological disorder in children, remains a significant research focus. The interleukin (IL) family, pivotal mediators in inflammatory responses, shows increased expression in various neuroinflammatory diseases, markedly influencing their onset and progression. Elevated IL levels in the brains of children with CP, in contrast to healthy peers, reflect similar elevations in neurological conditions linked to CP, indicating a strong association between CP and the IL family. Anti-inflammatory therapies, particularly those targeting ILs, have shown effectiveness in animal models, diverging from traditional CP management methods. This shift suggests IL modulation as a promising therapeutic strategy in pediatric CP. This review consolidates recent findings on the IL family's role in CP, illuminating their evolving relationship.
Collapse
Affiliation(s)
- Mingbo Hu
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Chao Bai
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Hong Zhao
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Junjie Wu
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| | - Xinping Luan
- Cerebral Palsy Center in Neurosurgery, Second Affiliated Hospital of Xinjiang Medical University, 830063 Urumqi, Xinjiang, China
| |
Collapse
|
3
|
Prasad J, Van Steenwinckel J, Gunn AJ, Bennet L, Korzeniewski SJ, Gressens P, Dean JM. Chronic Inflammation Offers Hints About Viable Therapeutic Targets for Preeclampsia and Potentially Related Offspring Sequelae. Int J Mol Sci 2024; 25:12999. [PMID: 39684715 PMCID: PMC11640791 DOI: 10.3390/ijms252312999] [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: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The combination of hypertension with systemic inflammation during pregnancy is a hallmark of preeclampsia, but both processes also convey dynamic information about its antecedents and correlates (e.g., fetal growth restriction) and potentially related offspring sequelae. Causal inferences are further complicated by the increasingly frequent overlap of preeclampsia, fetal growth restriction, and multiple indicators of acute and chronic inflammation, with decreased gestational length and its correlates (e.g., social vulnerability). This complexity prompted our group to summarize information from mechanistic studies, integrated with key clinical evidence, to discuss the possibility that sustained or intermittent systemic inflammation-related phenomena offer hints about viable therapeutic targets, not only for the prevention of preeclampsia, but also the neurobehavioral and other developmental deficits that appear to be overrepresented in surviving offspring. Importantly, we feel that carefully designed hypothesis-driven observational studies are necessary if we are to translate the mechanistic evidence into child health benefits, namely because multiple pregnancy disorders might contribute to heightened risks of neuroinflammation, arrested brain development, or dysconnectivity in survivors who exhibit developmental problems later in life.
Collapse
Affiliation(s)
- Jaya Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | | | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Steven J. Korzeniewski
- C.S. Mott Center for Human Growth and Development, Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Pierre Gressens
- Inserm, Neurodiderot, Université de Paris, 75019 Paris, France;
- Centre for the Developing Brain, Division of Imaging Sciences and Department of Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Justin M. Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| |
Collapse
|
4
|
Shi Z, Sharif N, Luo K, Tan S. Development of a New Scoring System in Higher Animals for Testing Cognitive Function in the Newborn Period: Effect of Prenatal Hypoxia-Ischemia. Dev Neurosci 2024; 47:12-26. [PMID: 38547848 PMCID: PMC11436483 DOI: 10.1159/000538607] [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: 07/05/2023] [Accepted: 03/26/2024] [Indexed: 05/30/2024] Open
Abstract
INTRODUCTION Enhanced models for assessing cognitive function in the neonatal period are imperative in higher animals. Postnatal motor deficits, characteristic of cerebral palsy, emerge in newborn kits within our prenatal rabbit model of hypoxia-ischemia (HI). In humans, prenatal HI leads to intellectual disability and cerebral palsy. In a study examining cognitive function in newborn rabbits, we explored several questions. Is there a distinction between conditioned and unconditioned kits? Can the kits discern the human face or the laboratory coat? Do motorically normal kits, born after prenatal HI, exhibit cognitive deficits? METHODS The conditioning protocol was randomly assigned to kits from each litter. For conditioning, the same human, wearing a laboratory coat, fed the rabbit kits for 9 days before the cognitive test. The 6-arm radial maze was chosen for its simplicity and ease of use. Normally appearing kits, born after uterine ischemia at 79% or 92% term in New Zealand White rabbits, were compared to naïve kits. On postpartum day 22/23 or 29/30, the 6-arm maze helped determine if the kits recognized the original feeder from bystander (test 1) or the laboratory coat on bystander (test 2). The use of masks of feeder/bystander (test 3) assessed confounding cues. A weighted score was devised to address variability in entry to maze arms, time, and repeated-trial learning. RESULTS In conditioned kits, both naïve and HI kits exhibited a significant preference for the face of the feeder but not the laboratory coat. Cognitive deficits were minimal in normal-appearing HI kits. CONCLUSION The weighted score was amenable to statistical manipulation. INTRODUCTION Enhanced models for assessing cognitive function in the neonatal period are imperative in higher animals. Postnatal motor deficits, characteristic of cerebral palsy, emerge in newborn kits within our prenatal rabbit model of hypoxia-ischemia (HI). In humans, prenatal HI leads to intellectual disability and cerebral palsy. In a study examining cognitive function in newborn rabbits, we explored several questions. Is there a distinction between conditioned and unconditioned kits? Can the kits discern the human face or the laboratory coat? Do motorically normal kits, born after prenatal HI, exhibit cognitive deficits? METHODS The conditioning protocol was randomly assigned to kits from each litter. For conditioning, the same human, wearing a laboratory coat, fed the rabbit kits for 9 days before the cognitive test. The 6-arm radial maze was chosen for its simplicity and ease of use. Normally appearing kits, born after uterine ischemia at 79% or 92% term in New Zealand White rabbits, were compared to naïve kits. On postpartum day 22/23 or 29/30, the 6-arm maze helped determine if the kits recognized the original feeder from bystander (test 1) or the laboratory coat on bystander (test 2). The use of masks of feeder/bystander (test 3) assessed confounding cues. A weighted score was devised to address variability in entry to maze arms, time, and repeated-trial learning. RESULTS In conditioned kits, both naïve and HI kits exhibited a significant preference for the face of the feeder but not the laboratory coat. Cognitive deficits were minimal in normal-appearing HI kits. CONCLUSION The weighted score was amenable to statistical manipulation.
Collapse
Affiliation(s)
- Zhongjie Shi
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nadiya Sharif
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Kehuan Luo
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sidhartha Tan
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
5
|
Fleiss B, Gressens P. Role of Microglial Modulation in Therapies for Perinatal Brain Injuries Leading to Neurodevelopmental Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:591-606. [PMID: 39207715 DOI: 10.1007/978-3-031-55529-9_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neurodevelopmental disorders (NDDs) encompass various conditions stemming from changes during brain development, typically diagnosed early in life. Examples include autism spectrum disorder, intellectual disability, cerebral palsy, seizures, dyslexia, and attention deficit hyperactivity disorder. Many NDDs are linked to perinatal events like infections, oxygen disturbances, or insults in combination. This chapter outlines the causes and effects of perinatal brain injury as they relate to microglia, along with efforts to prevent or treat such damage. We primarily discuss therapies targeting microglia modulation, focusing on those either clinically used or in advanced development, often tested in large animal models such as sheep, non-human primates, and piglets-standard translational models in perinatal medicine. Additionally, it touches on experimental studies showcasing advancements in the field.
Collapse
Affiliation(s)
- Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Pierre Gressens
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
- Université de Paris, NeuroDiderot, Inserm, Paris, France.
| |
Collapse
|
6
|
She HQ, Sun YF, Chen L, Xiao QX, Luo BY, Zhou HS, Zhou D, Chang QY, Xiong LL. Current analysis of hypoxic-ischemic encephalopathy research issues and future treatment modalities. Front Neurosci 2023; 17:1136500. [PMID: 37360183 PMCID: PMC10288156 DOI: 10.3389/fnins.2023.1136500] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is the leading cause of long-term neurological disability in neonates and adults. Through bibliometric analysis, we analyzed the current research on HIE in various countries, institutions, and authors. At the same time, we extensively summarized the animal HIE models and modeling methods. There are various opinions on the neuroprotective treatment of HIE, and the main therapy in clinical is therapeutic hypothermia, although its efficacy remains to be investigated. Therefore, in this study, we discussed the progress of neural circuits, injured brain tissue, and neural circuits-related technologies, providing new ideas for the treatment and prognosis management of HIE with the combination of neuroendocrine and neuroprotection.
Collapse
Affiliation(s)
- Hong-Qing She
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Translational Neurology Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- WANG TINGHUA Translation Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi-Fei Sun
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Li Chen
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu-Xia Xiao
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Bo-Yan Luo
- WANG TINGHUA Translation Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hong-Su Zhou
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Translational Neurology Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- WANG TINGHUA Translation Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Di Zhou
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Quan-Yuan Chang
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Translational Neurology Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- WANG TINGHUA Translation Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
7
|
Kelly SB, Dean JM, Zahra VA, Dudink I, Thiel A, Polglase GR, Miller SL, Hooper SB, Bennet L, Gunn AJ, Galinsky R. Progressive inflammation reduces high-frequency EEG activity and cortical dendritic arborisation in late gestation fetal sheep. J Neuroinflammation 2023; 20:124. [PMID: 37226206 DOI: 10.1186/s12974-023-02805-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Antenatal infection/inflammation is associated with disturbances in neuronal connectivity, impaired cortical growth and poor neurodevelopmental outcomes. The pathophysiological substrate that underpins these changes is poorly understood. We tested the hypothesis that progressive inflammation in late gestation fetal sheep would alter cortical neuronal microstructure and neural function assessed using electroencephalogram band power analysis. METHODS Fetal sheep (0.85 of gestation) were surgically instrumented for continuous electroencephalogram (EEG) recording and randomly assigned to repeated saline (control; n = 9) or LPS (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng; n = 8) infusions to induce inflammation. Sheep were euthanised 4 days after the first LPS infusion for assessment of inflammatory gene expression, histopathology and neuronal dendritic morphology in the somatosensory cortex. RESULTS LPS infusions increased delta power between 8 and 50 h, with reduced beta power from 18 to 96 h (P < 0.05 vs. control). Basal dendritic length, numbers of dendritic terminals, dendritic arborisation and numbers of dendritic spines were reduced in LPS-exposed fetuses (P < 0.05 vs. control) within the somatosensory cortex. Numbers of microglia and interleukin (IL)-1β immunoreactivity were increased in LPS-exposed fetuses compared with controls (P < 0.05). There were no differences in total numbers of cortical NeuN + neurons or cortical area between the groups. CONCLUSIONS Exposure to antenatal infection/inflammation was associated with impaired dendritic arborisation, spine number and loss of high-frequency EEG activity, despite normal numbers of neurons, that may contribute to disturbed cortical development and connectivity.
Collapse
Affiliation(s)
- Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Justin M Dean
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
| | - Ingrid Dudink
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Alison Thiel
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - 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
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Melbourne, VIC, 3168, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
PSMB5 Alleviates Ulcerative Colitis by Inhibiting ROS-Dependent NLRP3 Inflammasome-Mediated Pyroptosis. DISEASE MARKERS 2022; 2022:2329904. [PMID: 36061354 PMCID: PMC9439905 DOI: 10.1155/2022/2329904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 12/04/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease. Intestinal mucosal injury is a significant factor in UC. Pyroptosis is a kind of programmed cell death induced by inflammatory caspases. Proteasome 20S subunit beta 5 (PSMB5) promotes cell viability. The purpose of this study was to determine the impact of PSMB5 on intestinal mucosal injury and to elucidate the underlying processes in dextran sulfate sodium- (DSS-) induced UC mice. Kunming (KM) mice received 3% DSS for 5 days to induce UC. We collected clinical symptoms, body weight, colon length, and histological changes. MDA (malondialdehyde) and SOD (superoxide dismutase) levels were determined using an ELISA assay. RT-PCR was used to assess the expression of IL-1β and IL-18. PSMB5 demonstrated a significant effect against UC by increasing body weight and colon length and decreasing DAI (disease activity index), colon macroscopic damage index (CMDI), histological injury scores, and reactive oxygen species (ROS), MDA, and SOD levels, thereby alleviating histopathological changes and inhibiting oxidative stress. HIEC-6 cells were exposed to lipopolysaccharide (LPS) condition with or without PSMB5, along with caspase-1 inhibitor (Z-VAD-FMK), NLRP3 inhibitor (MCC950), and ROS scavenger N-acetylcysteine (NAC). The viability of the cells, the release of lactate dehydrogenase (LDH), and intracellular ROS generation were determined using assay kits. Western blot analysis was used to determine the levels of NLRP3, ASC, cleaved caspase-1 (p20), pro-IL-1β, IL-1β, pro-IL-18, and IL-18. PSMB5 overexpression enhanced the inflammatory damage in LPS-treated HIEC-6 cells by activating the NLRP3 inflammasome and mediating pyroptosis, as demonstrated by increased LDH release and lower cell viability, as well as increased expression of NLRP3, ASC, cleaved caspase-1 (p20), IL-1, and IL-18. Meanwhile, NAC protected HIEC-6 cells from LPS-induced damage by reversing the activation of the NLRP3 inflammasome-mediated pyroptosis. In conclusion, PSMB5 may lower HIEC-6 cell susceptibility to LPS and ameliorate UC-induced HIEC-6 cell damage by decreasing ROS generation and hence inhibiting NLRP3-mediated pyroptosis.
Collapse
|
9
|
Gressens P. Extracellular vesicles at the rescue of the preterm brain. Brain Behav Immun 2022; 102:135-136. [PMID: 35150859 DOI: 10.1016/j.bbi.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022] Open
Affiliation(s)
- Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France.
| |
Collapse
|
10
|
Galinsky R, van de Looij Y, Mitchell N, Dean JM, Dhillon SK, Yamaguchi K, Lear CA, Wassink G, Davidson JO, Nott F, Zahra VA, Kelly SB, King VJ, Sizonenko SV, Bennet L, Gunn AJ. Magnetic Resonance Imaging Correlates of White Matter Gliosis and Injury in Preterm Fetal Sheep Exposed to Progressive Systemic Inflammation. Int J Mol Sci 2020; 21:ijms21238891. [PMID: 33255257 PMCID: PMC7727662 DOI: 10.3390/ijms21238891] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022] Open
Abstract
Progressive fetal infection/inflammation is strongly associated with neural injury after preterm birth. We aimed to test the hypotheses that progressively developing fetal inflammation leads to neuroinflammation and impaired white matter development and that the histopathological changes can be detected using high-field diffusion tensor magnetic resonance imaging (MRI). Chronically instrumented preterm fetal sheep at 0.7 of gestation were randomly assigned to receive intravenous saline (control; n = 6) or a progressive infusion of lipopolysaccharide (LPS, 200 ng intravenous over 24 h then doubled every 24 h for 5 days to induce fetal inflammation, n = 7). Sheep were killed 10 days after starting the infusions, for histology and high-field diffusion tensor MRI. Progressive LPS infusion was associated with increased circulating interleukin (IL)-6 concentrations and moderate increases in carotid artery perfusion and the frequency of electroencephalogram (EEG) activity (p < 0.05 vs. control). In the periventricular white matter, fractional anisotropy (FA) was increased, and orientation dispersion index (ODI) was reduced (p < 0.05 vs. control for both). Histologically, in the same brain region, LPS infusion increased microglial activation and astrocyte numbers and reduced the total number of oligodendrocytes with no change in myelination or numbers of immature/mature oligodendrocytes. Numbers of astrocytes in the periventricular white matter were correlated with increased FA and reduced ODI signal intensities. Astrocyte coherence was associated with increased FA. Moderate astrogliosis, but not loss of total oligodendrocytes, after progressive fetal inflammation can be detected with high-field diffusion tensor MRI.
Collapse
Affiliation(s)
- Robert Galinsky
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria 3800, Australia
| | - Yohan van de Looij
- Division of Child Development & Growth, Department of Pediatrics, Gynaecology & Obstetrics, School of Medicine, University of Geneva, 1015 Geneva, Switzerland; (Y.v.d.L.); (S.V.S.)
| | - Natasha Mitchell
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Justin M. Dean
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Simerdeep K. Dhillon
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Kyohei Yamaguchi
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Christopher A. Lear
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Guido Wassink
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Joanne O. Davidson
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Fraser Nott
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
| | - Valerie A. Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
| | - Sharmony B. Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria 3800, Australia
| | - Victoria J. King
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Stéphane V. Sizonenko
- Division of Child Development & Growth, Department of Pediatrics, Gynaecology & Obstetrics, School of Medicine, University of Geneva, 1015 Geneva, Switzerland; (Y.v.d.L.); (S.V.S.)
| | - Laura Bennet
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Alistair J. Gunn
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
- Correspondence:
| |
Collapse
|
11
|
Shi J, Wang W, Sang G, Xi H, Sun Y, Lu C, Ye H, Huang L. Short Term Usage of Omega-3 Polyunsaturated Fatty Acids Ameliorate Lipopolysaccharide-Induced Inflammatory Response and Oxidative Stress in the Neonatal Rat Hippocampal Tissue. Front Nutr 2020; 7:572363. [PMID: 33282898 PMCID: PMC7705230 DOI: 10.3389/fnut.2020.572363] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022] Open
Abstract
Objective: To investigate the effect of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) on lipopolysaccharide (LPS)-induced inflammatory response and oxidative stress in neonatal rat brain. Methods: Ninety-six 3-day-old Sprague Dawley rats were divided into four groups: control (saline/saline), LPS/ω-3, LPS/ω-6, and LPS/saline (n = 24/group). All rats, except those in the control group, were intraperitoneally challenged once with LPS (0.6 mg/kg) and were treated with ω-3 PUFAs, ω-6 PUFAs, or saline at 15 mL/kg for 1 or 5 consecutive days beginning on the day of LPS-challenge. Rats in the control group underwent the same procedures and received saline (vehicle). After 1 or 5 days of treatment, 12 rats from each group were sacrificed and their hippocampuses were collected. The expression of inflammation-related genes as well as the levels of oxidative stress markers in hippocampal tissues were determined. Results: After 1 or 5 days of treatment, the expression of toll-like receptor 4 and multiple proinflammatory cytokines were significantly decreased in the LPS/ω-3 group compared with those in the LPS/saline group. The activities of superoxide dismutase and glutathione (GSH) were significantly elevated, whereas amounts of malondialdehyde and oxidized glutathione (GSSG) and the ratio of GSSG/GSH were remarkably lowered in the LPS/ω-3 group compared with those in the LPS/saline group after 1 day of treatment. Opposite effects were observed in the LPS/ω-6 group. Conclusion: ω-3 PUFAs may protect rat brain tissue against LPS-induced inflammatory response and oxidative stress.
Collapse
Affiliation(s)
- Jipeng Shi
- Henan Key Laboratory of Neurorestoratology, Department of Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Weiwei Wang
- Henan Key Laboratory of Neurorestoratology, Department of Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Guimei Sang
- Henan Key Laboratory of Neurorestoratology, Department of Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Huifang Xi
- Henan Key Laboratory of Neurorestoratology, Department of Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yazhou Sun
- Henan Key Laboratory of Neurorestoratology, Department of Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Chaosheng Lu
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Hezhen Ye
- The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| | - Limi Huang
- The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, China
| |
Collapse
|
12
|
Fernandez A, Dumon C, Guimond D, Tyzio R, Bonifazi P, Lozovaya N, Burnashev N, Ferrari DC, Ben-Ari Y. The GABA Developmental Shift Is Abolished by Maternal Immune Activation Already at Birth. Cereb Cortex 2020; 29:3982-3992. [PMID: 30395185 DOI: 10.1093/cercor/bhy279] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/14/2018] [Accepted: 10/11/2018] [Indexed: 01/27/2023] Open
Abstract
Epidemiological and experimental studies suggest that maternal immune activation (MIA) leads to developmental brain disorders, but whether the pathogenic mechanism impacts neurons already at birth is not known. We now report that MIA abolishes in mice the oxytocin-mediated delivery γ-aminobutyric acid (GABA) shift from depolarizing to hyperpolarizing in CA3 pyramidal neurons, and this is restored by the NKCC1 chloride importer antagonist bumetanide. Furthermore, MIA hippocampal pyramidal neurons at birth have a more exuberant apical arbor organization and increased apical dendritic length than age-matched controls. The frequency of spontaneous glutamatergic postsynaptic currents is also increased in MIA offspring, as well as the pairwise correlation of the synchronized firing of active cells in CA3. These alterations produced by MIA persist, since at P14-15 GABA action remains depolarizing, produces excitatory action, and network activity remains elevated with a higher frequency of spontaneous glutamatergic postsynaptic currents. Therefore, the pathogenic actions of MIA lead to important morphophysiological and network alterations in the hippocampus already at birth.
Collapse
Affiliation(s)
- Amandine Fernandez
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Camille Dumon
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Damien Guimond
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France
| | - Roman Tyzio
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Paolo Bonifazi
- Biocruces Health Research Institute, Barakaldo, Spain.,IKERBASQUE: The Basque Foundation for Science, Bilbao, Spain
| | - Natalia Lozovaya
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France
| | - Nail Burnashev
- Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Diana C Ferrari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France
| | - Yehezkel Ben-Ari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France
| |
Collapse
|
13
|
Gilles FH, Leviton A. Neonatal white matter damage and the fetal inflammatory response. Semin Fetal Neonatal Med 2020; 25:101111. [PMID: 32299712 DOI: 10.1016/j.siny.2020.101111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In 1962 a long-recognized pathologic abnormality in neonatal brains characterized by multiple telencephalic focal white matter necroses was renamed periventricular leukomalacia (PVL) and the authors inappropriately asserted that their entity was caused by anoxia. They also failed to include three other white matter histologic abnormalities. In this essay, we identify the breadth of white matter pathology, especially in very preterm newborns, and show that none of the four histologic expressions of white matter damage, including focal necrosis, are associated with hypoxemia or correlates as hypotension, but are instead associated with markers of fetal or perinatal inflammation, particularly in preterm babies. We begin with the background needed to evaluate the evidence.
Collapse
Affiliation(s)
- F H Gilles
- Children's Hospital Los Angeles, The University of Southern California, USA.
| | - A Leviton
- Boston Children's Hospital, Harvard Medical School, USA.
| |
Collapse
|
14
|
Pires JM, Foresti ML, Silva CS, Rêgo DB, Calió ML, Mosini AC, Nakamura TKE, Leslie ATF, Mello LE. Lipopolysaccharide-Induced Systemic Inflammation in the Neonatal Period Increases Microglial Density and Oxidative Stress in the Cerebellum of Adult Rats. Front Cell Neurosci 2020; 14:142. [PMID: 32581717 PMCID: PMC7283979 DOI: 10.3389/fncel.2020.00142] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/28/2020] [Indexed: 01/09/2023] Open
Abstract
Inflammatory processes occurring in the perinatal period may affect different brain regions, resulting in neurologic sequelae. Injection of lipopolysaccharide (LPS) at different neurodevelopmental stages produces long-term consequences in several brain structures, but there is scarce evidence regarding alterations in the cerebellum. The aim of this study was to evaluate the long-term consequences on the cerebellum of a systemic inflammatory process induced by neonatal LPS injection. For this, neonatal rats were randomly assigned to three different groups: naïve, sham, and LPS. Saline (sham group) or LPS solution (1 mg/kg) was intraperitoneally injected on alternate postnatal days (PN) PN1, PN3, PN5, and PN7. Spontaneous activity was evaluated with the open field test in adulthood. The cerebellum was evaluated for different parameters: microglial and Purkinje cell densities, oxidative stress levels, and tumor necrosis factor alpha (TNF-α) mRNA expression. Our results show that administration of LPS did not result in altered spontaneous activity in adult animals. Our data also indicate increased oxidative stress in the cerebellum, as evidenced by an increase in superoxide fluorescence by dihydroethidium (DHE) indicator. Stereological analyses indicated increased microglial density in the cerebellum that was not accompanied by Purkinje cell loss or altered TNF-α expression in adult animals. Interestingly, Purkinje cells ectopically positioned in the granular and molecular layers of the cerebellum were observed in animals of the LPS group. Our data suggest that neonatal LPS exposure causes persistent cellular and molecular changes to the cerebellum, indicating the susceptibility of this region to systemic inflammatory insults in infancy. Further investigation of the consequences of these changes and the development of strategies to avoid those should be subject of future studies.
Collapse
Affiliation(s)
| | - Maira Licia Foresti
- Physiology Department, Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | | | | | | | - Amanda Cristina Mosini
- Physiology Department, Universidade Federal de São Paulo, São Paulo, Brazil.,Associação Brasileira de Epilepsia, São Paulo, Brazil
| | | | | | - Luiz Eugênio Mello
- Physiology Department, Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Truttmann AC, Ginet V, Puyal J. Current Evidence on Cell Death in Preterm Brain Injury in Human and Preclinical Models. Front Cell Dev Biol 2020; 8:27. [PMID: 32133356 PMCID: PMC7039819 DOI: 10.3389/fcell.2020.00027] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
Despite tremendous advances in neonatal intensive care over the past 20 years, prematurity carries a high burden of neurological morbidity lasting lifelong. The term encephalopathy of prematurity (EoP) coined by Volpe in 2009 encompasses all aspects of the now known effects of prematurity on the immature brain, including altered and disturbed development as well as specific lesional hallmarks. Understanding the way cells are damaged is crucial to design brain protective strategies, and in this purpose, preclinical models largely contribute to improve the comprehension of the cell death mechanisms. While neuronal cell death has been deeply investigated and characterized in (hypoxic–ischemic) encephalopathy of the newborn at term, little is known about the types of cell death occurring in preterm brain injury. Three main different morphological cell death types are observed in the immature brain, specifically in models of hypoxic–ischemic encephalopathy, namely, necrotic, apoptotic, and autophagic cell death. Features of all three types may be present in the same dying neuron. In preterm brain injury, description of cell death types is sparse, and cell loss primarily concerns immature oligodendrocytes and, infrequently, neurons. In the present review, we first shortly discuss the different main severe preterm brain injury conditions that have been reported to involve cell death, including periventricular leucomalacia (PVL), diffuse white matter injury (dWMI), and intraventricular hemorrhages, as well as potentially harmful iatrogenic conditions linked to premature birth (anesthesia and caffeine therapy). Then, we present an overview of current evidence concerning cell death in both clinical human tissue data and preclinical models by focusing on studies investigating the presence of cell death allowing discriminating between the types of cell death involved. We conclude that, to improve brain protective strategies, not only apoptosis but also other cell death (such as regulated necrotic and autophagic) pathways now need to be investigated together in order to consider all cell death mechanisms involved in the pathogenesis of preterm brain damage.
Collapse
Affiliation(s)
- Anita C Truttmann
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland
| | - Vanessa Ginet
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland.,Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
16
|
Shi JP, Fu W, Liu J. <Omega>-3 PUFA Attenuates LPS-Induced Neuro-Injury of Neonatal Rats through the PI3K/AKT Pathway. Neuroscience 2019; 414:112-127. [PMID: 31260648 DOI: 10.1016/j.neuroscience.2019.06.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 02/05/2023]
Abstract
Inflammation may result in periventricular leukomalacia, which is the leading cause of preterm brain encephalopathy. Moreover, -3 polyunsaturated fatty acids (-3 PUFAs) play a pivotal role against central nervous system injury, which is likely related to its anti-inflammatory effect. However, the mechanism regarding the remedial effects of -3 PUFA for LPS-induced neuro-injury has remained unclear. In this study, newborn SD rats were intraperitoneally injected with LPS or < omega>-3 PUFA, and the proliferation and apoptosis of neurocytes in the hippocampus were measured by TUNEL and BrdU. Quantitative real-time PCR (qPCR) and Western blot assay were used to analyze the mRNA and protein levels of PI3K, AKT and β-catenin in vitro and in vivo. We found that -3 PUFA promoted the proliferation and migration of neurocytes in vitro and in vivo and inhibited apoptosis. Furthermore, we confirmed that -3 PUFA through the PI3K/AKT signaling pathway positively regulated the expression of PI3K and further caused the phosphorylation of AKT activation, followed by the upregulation of β-catenin expression. Interestingly, this phenomenon became more noticeable with the combined application of -3 PUFA and a PI3K/AKT agonist. In conclusion, we confirm that -3 PUFA plays an important role in neuroprotection by activating the PI3K/AKT/β-catenin pathway. It may be a promising strategy against brain injury.
Collapse
Affiliation(s)
- Ji-Peng Shi
- Department of Paediatrics, The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; Department of Neonatology and NICU of Bayi Children's Hospital, the Army General Hospital of the Chinese PLA, Southern Medical University, Beijing 100700, China; Department of Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, PR China
| | - Wei Fu
- Department of Neonatology and NICU, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing 100101, China; Department of Neonatology and NICU of Bayi Children's Hospital, the Army General Hospital of the Chinese PLA, Southern Medical University, Beijing 100700, China
| | - Jing Liu
- Department of Paediatrics, The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; Department of Neonatology and NICU, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing 100101, China; Department of Neonatology and NICU of Bayi Children's Hospital, the Army General Hospital of the Chinese PLA, Southern Medical University, Beijing 100700, China.
| |
Collapse
|
17
|
Suppression of PDGF induces neuronal apoptosis after neonatal cerebral hypoxia and ischemia by inhibiting P-PI3K and P-AKT signaling pathways. Brain Res 2019; 1719:77-88. [PMID: 31082354 DOI: 10.1016/j.brainres.2019.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 02/05/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) always results in severe neurologic dysfunction, nevertheless effective treatments are limited and the underlying mechanism also remains unclear. In this study, we firstly established the neonatal HIE model in the postnatal day 7 SD rats, Zea-Longa score and TTC staining were employed to assess the neurological behavior and infarct volume of the brain after cerebral hypoxia-ischemia (HI). Afterwards, protein chip was adopted to detect the differential proteins in the right cortex, hippocampus and lung, ultimately, PDGF was noticed. Then, immunohistochemistry, immunofluorescence double staining of NeuN/PDGF, and western blot were used to validate the expression level of PDGF in the cortex and hippocampus at 6 hours (h), 12 h and 24 h after HI. To determine the role of PDGF, the primary cortical neurons were prepared and performed PDGF shRNA administration. The results showed that HIE induced a severe behavioral dysfunction and brain infarction in neonatal rats, and the expression of PDGF in right cortex and hippocampus was remarkably increased after HI. Whereas, suppressing PDGF resulted in a significant loss of neurons and inhibition of neurite growth. Moreover, the protein level of P-PI3K and P-AKT signaling pathways were largely decreased following PDGF-shRNA application in the cortical neurons. In conclusion, PDGF suppression aggravated neuronal dysfunction, and the underlying mechanism is associated with inhibiting the phosphorylation of P-PI3K and P-AKT. Together, PDGF regulating PI3K and AKT may be an important panel in HIE events and therefore may provide possible strategy for the treatment of HIE in future clinic trail.
Collapse
|
18
|
Abstract
Despite notable advances in the care and survival of preterm infants, a significant proportion of preterm neonates will have life-long cognitive, behavioral, and motor deficits, and robustly effective neuroprotective strategies are still missing. These therapies must target the pathophysiologic mechanisms observed in contemporaneous infants and rely on modern epidemiology, imaging, and experimental models and assessment techniques. Two drugs, magnesium sulfate and caffeine, are already in use in several units, and although their targets are apnea of prematurity and myometrial contractility (respectively), they do offer improved odds of positive outcomes. Nevertheless, these drugs have limited efficacy, and NICU-to-NICU administration varies greatly. As such, there is an obvious need for additional specific neurotherapeutic strategies to further enhance the outcome of this very fragile population of neonates. The chapter reviews these issues, highlights bottlenecks that need to be solved for meaningful progress in the field, and proposes future innovative avenues for intervention, including delayed interventions.
Collapse
Affiliation(s)
- Bobbi Fleiss
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, London, United Kingdom
| | - Pierre Gressens
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, London, United Kingdom.
| |
Collapse
|
19
|
Nguyen DN, Thymann T, Goericke-Pesch SK, Ren S, Wei W, Skovgaard K, Damborg P, Brunse A, van Gorp C, Kramer BW, Wolfs TG, Sangild PT. Prenatal Intra-Amniotic Endotoxin Induces Fetal Gut and Lung Immune Responses and Postnatal Systemic Inflammation in Preterm Pigs. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2629-2643. [PMID: 30314768 DOI: 10.1016/j.ajpath.2018.07.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/14/2018] [Accepted: 07/05/2018] [Indexed: 10/28/2022]
Abstract
Prenatal inflammation is a major risk for preterm birth and neonatal morbidity, but its effects on postnatal immunity and organ functions remain unclear. Using preterm pigs as a model for preterm infants, we investigated whether prenatal intra-amniotic (IA) inflammation modulates postnatal systemic immune status and organ functions. Preterm pigs exposed to IA lipopolysaccharide (LPS) for 3 days were compared with controls at birth and postnatal day 5 after formula feeding. IA LPS induced mild chorioamnionitis but extensive intra-amniotic inflammation. There were minor systemic effects at birth (increased blood neutrophil counts), but a few days later, prenatal LPS induced delayed neonatal arousal, systemic inflammation (increased blood leukocytes, plasma cytokines, and splenic bacterial counts), altered serum biochemistry (lower albumin and cholesterol and higher iron and glucose values), and increased urinary protein and sodium excretion. In the gut and lungs, IA LPS-induced inflammatory responses were observed mainly at birth (increased LPS, CXCL8, and IL-1β levels and myeloperoxidase-positive cell density, multiple increases in innate immune gene expressions, and reduced villus heights), but not on postnatal day 5 (except elevated lung CXCL8 and diarrhea symptoms). Finally, IA LPS did not affect postnatal gut brush-border enzymes, hexose absorption, permeability, or sensitivity to necrotizing enterocolitis on day 5. Short-term IA LPS exposure predisposes preterm pigs to postnatal systemic inflammation after acute fetal gut and lung inflammatory responses.
Collapse
Affiliation(s)
- Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Thomas Thymann
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Sandra K Goericke-Pesch
- Section for Veterinary Reproduction and Obstetrics, Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Shuqiang Ren
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Wei Wei
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Peter Damborg
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
| | - Anders Brunse
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Charlotte van Gorp
- Department of Pediatrics, School of Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Boris W Kramer
- Department of Pediatrics, School of Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Tim G Wolfs
- Department of Pediatrics, School of Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Per T Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark; Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
20
|
Snyder JM, Wood TR, Corry K, Moralejo DH, Parikh P, Juul SE. Ontogeny of white matter, toll-like receptor expression, and motor skills in the neonatal ferret. Int J Dev Neurosci 2018; 70:25-33. [PMID: 29791868 DOI: 10.1016/j.ijdevneu.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/09/2018] [Accepted: 05/15/2018] [Indexed: 10/16/2022] Open
Abstract
Inflammation caused by perinatal infection, superimposed with hypoxia and/or hyperoxia, appears to be important in the pathogenesis of preterm neonatal encephalopathy, with white matter particularly vulnerable during the third trimester. The associated inflammatory response is at least partly mediated through Toll-like receptor (TLR)-dependent mechanisms. Immunohistochemistry, gene expression, and behavioral studies were used to characterize white matter development and determine TLR3 and TLR4 expression and accumulation in the neonatal ferret brain. Expression of markers of white matter development increased significantly between postnatal day (P)1 and P10 (NG2, PDGFRα) or P15 (Olig2), and either remained elevated (NG2), or decreased again at P40 (PDGFRα, Olig2). Olig2 immunostaining within the internal capsule was also greatest at P15. Myelin basic protein (MBP) immunostaining and mRNA expression increased markedly from P15 to P40 and into adulthood, which correlated with increasing performance on behavioral tests (negative geotaxis, cliff aversion, righting reflex, and catwalk gait analysis). TLR4 and TLR3 positive staining was low at all ages, but TLR3 and TLR4 mRNA expression both increased significantly from P1 to P40. Following lipopolysaccharide (LPS) and hypoxia/hyperoxia exposure at P10, meningeal and parenchymal inflammation was seen, including an increase in TLR4 positive cells. These data suggest that the neuroinflammation associated with prematurity could be modeled in the newborn ferret.
Collapse
Affiliation(s)
- Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Thomas R Wood
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Kylie Corry
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Daniel H Moralejo
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Pratik Parikh
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Sandra E Juul
- Department of Pediatrics, University of Washington, Seattle, WA, United States.
| |
Collapse
|
21
|
Leviton A, Joseph RM, Allred EN, O’Shea TM, Taylor HG, Kuban KKC. Antenatal and Neonatal Antecedents of Executive Dysfunctions in Extremely Preterm Children. J Child Neurol 2018; 33:198-208. [PMID: 29322860 PMCID: PMC5807158 DOI: 10.1177/0883073817750499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
To find out why children born extremely preterm are at heightened risk of executive dysfunctions, the authors assessed 716 children who were 10 years old born extremely preterm whose IQ was ≥ 70. A working memory dysfunction (n = 169), an inhibition dysfunction (n = 360), a switching dysfunction (355), and all 3 (executive dysfunction; n = 107) were defined on the basis of Z-scores ≤ -1 on the Differential Ability Scales-II Working Memory composite, and/or on the NEPSY-II Inhibition-Inhibition and Inhibition-Switching subtests. All risk profiles include an indicator of socioeconomic disadvantage. The risk profile of each of the 3 individual dysfunctions includes an indicator of the newborn's immaturity, and the risk profiles of the inhibition dysfunction and switching dysfunction also include an indicator of inflammation. Only the switching dysfunction was associated with fetal growth restriction. The risk factors for executive dysfunction can be subsumed under the 4 themes of socioeconomic disadvantage, immaturity/vulnerability, inflammation, and fetal growth restriction.
Collapse
Affiliation(s)
- Alan Leviton
- Boston Children’s Hospital and Harvard Medical School,
Boston MA, USA
| | | | | | - T. Michael O’Shea
- University of North Carolina School of Medicine, Chapel Hill NC,
USA
| | - H. Gerry Taylor
- Nationwide Children’s Hospital and The Ohio State
University, Columbus, OH, USA
| | - Karl KC Kuban
- Boston Medical Center and Boston University School of Medicine,
Boston, MA, USA
| |
Collapse
|
22
|
Galinsky R, Lear CA, Dean JM, Wassink G, Dhillon SK, Fraser M, Davidson JO, Bennet L, Gunn AJ. Complex interactions between hypoxia-ischemia and inflammation in preterm brain injury. Dev Med Child Neurol 2018; 60:126-133. [PMID: 29194585 DOI: 10.1111/dmcn.13629] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 12/12/2022]
Abstract
UNLABELLED Children surviving preterm birth have a high risk of disability, particularly cognitive and learning problems. There is extensive clinical and experimental evidence that disability is now primarily related to dysmaturation of white and gray matter, defined by failure of oligodendrocyte maturation and neuronal dendritic arborization, rather than cell death alone. The etiology of this dysmaturation is multifactorial, with contributions from hypoxia-ischemia, infection/inflammation and barotrauma. Intriguingly, these factors can interact to both increase and decrease damage. In this review we summarize preclinical and clinical evidence that all of these factors trigger secondary or chronic inflammation and gliosis. Thus, we hypothesize that these shared pathological features play a key role in a final common pathway that leads to the impaired neural maturation and connectivity and cognitive/motor impairments that are commonly observed in infants born preterm. This raises the possibility that secondary or chronic inflammation may be a viable therapeutic target for delayed interventions to improve neurodevelopmental outcomes after preterm birth. WHAT THIS PAPER ADDS Hypoxia-ischemia, infection/inflammation, and barotrauma/volutrauma all contribute to preterm brain injury. Multiple different triggers of preterm brain injury are associated with central nervous system dysmaturation. Secondary brain inflammation may be a viable target to improve neurodevelopment after preterm birth.
Collapse
Affiliation(s)
- Robert Galinsky
- The Department of Physiology, University of Auckland, Auckland, New Zealand.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Christopher A Lear
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Justin M Dean
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Mhoyra Fraser
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
23
|
Shi Z, Vasquez-Vivar J, Luo K, Yan Y, Northington F, Mehrmohammadi M, Tan S. Ascending Lipopolysaccharide-Induced Intrauterine Inflammation in Near-Term Rabbits Leading to Newborn Neurobehavioral Deficits. Dev Neurosci 2018; 40:534-546. [PMID: 31163416 PMCID: PMC9873358 DOI: 10.1159/000499960] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/26/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Chorioamnionitis from ascending bacterial infection through the endocervix is a potential risk factor for cerebral palsy. Tetrahydrobiopterin, an essential cofactor for nitric oxide synthase (NOS) and amino acid hydroxylases, when augmented in the fetal brain, prevents some of the cerebral palsy-like deficits in a rabbit hypoxia-ischemia model. OBJECTIVES To study the effect of lipopolysaccharide (LPS)-induced intrauterine inflammation in preterm gestation on motor deficits in the newborn, and whether biosynthesis of tetrahydrobiopterin or inflammatory mediators is affected in the fetal brain. METHODS Pregnant rabbits at 28 days gestation (89% term) were administered either saline or LPS into both endocervical openings. One group underwent spontaneous delivery, and neurobehavioral tests were performed at postnatal day (P) 1 and P11, with some kits being sacrificed at P1 for histological analysis. Another group underwent Cesarean section 24 h after LPS administration. Gene sequences for rabbit biosynthetic enzymes of tetra-hydrobiopterin pathways were determined and analyzed in addition to cytokines, using quantitative real-time polymerase chain reaction. RESULTS Exposure to 200 μg/kg/mL LPS caused a locomotion deficit and mild hypertonia at P1. By P11, most animals turned into normal-appearing kits. There was no difference in neuronal cell death in the caudate between hypertonic and nonhypertonic kits at P1 (n = 3-5 in each group). Fetal brain GTP cyclohydrolase I was increased, whereas sepiapterin reductase and 6-pyruvoyltetrahydropterin synthase were decreased, 24 h after LPS administration. Neuronal NOS was also increased. Regardless of the position in the uterus or the brain region, expression of TNF-α and TGF-β was decreased, whereas that of IL-1β, IL-6, and IL-8 was increased (n = 3-4 in each group). CONCLUSIONS This is the first study using an ascending LPS-induced intrauterine inflammation model in rabbits, showing mostly transient hypertonia and mainly locomotor deficits in the kits. Not all proinflammatory cytokines are increased in the fetal brain following LPS administration. Changes in key tetrahydro-biopterin biosynthetic enzymes possibly indicate different effects of the inflammatory insult.
Collapse
Affiliation(s)
- Zhongjie Shi
- Department of Pediatrics, Children’s Hospital of Michigan, Wayne State University, Detroit, MI
| | | | - Kehuan Luo
- Department of Pediatrics, Children’s Hospital of Michigan, Wayne State University, Detroit, MI
| | - Yan Yan
- Department of Biomedical Engineering, Wayne State University, Detroit, MI
| | | | | | - Sidhartha Tan
- Department of Pediatrics, Children’s Hospital of Michigan, Wayne State University, Detroit, MI
| |
Collapse
|
24
|
Systemic inflammation combined with neonatal cerebellar haemorrhage aggravates long-term structural and functional outcomes in a mouse model. Brain Behav Immun 2017; 66:257-276. [PMID: 28755859 DOI: 10.1016/j.bbi.2017.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/11/2017] [Accepted: 07/19/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Despite the increased recognition of cerebellar injury in survivors of preterm birth, the neurodevelopmental consequences of isolated cerebellar injury have been largely unexplored and our current understanding of the functional deficits requires further attention in order to translate knowledge to best practices. Preterm infants are exposed to multiple stressors during their postnatal development including perinatal cerebellar haemorrhage (CBH) and postnatal infection, two major risk factors for neurodevelopmental impairments. METHODS We developed a translational mouse model of CBH and/or inflammation to measure the short- and long-term outcomes in cerebellar structure and function. RESULTS Mice exposed to early combined insults of CBH and early inflammatory state (EIS) have a delay in grasping acquisition, neonatal motor deficits and deficient long-term memory. CBH combined with late inflammatory state (LIS) does not induce neonatal motor problems but leads to poor fine motor function and long-term memory deficits at adulthood. Early combined insults result in poor cerebellar growth from postnatal day 15 until adulthood shown by MRI, which are reflected in diminished volumes of cerebellar structures. There are also decreases in volumes of gray matter and hippocampus. Cerebellar microgliosis appears 24h after the combined insults and persists until postnatal day 15 in the cerebellar molecular layer and cerebellar nuclei in association with a disrupted patterning of myelin deposition, a delay of oligodendrocyte maturation and reduced white matter cerebellar volume. CONCLUSIONS Together, these findings reveal poor outcomes in developing brains exposed to combined cerebellar perinatal insults in association with cerebellar hypoplasia, persistence of microgliosis and alterations of cerebellar white matter maturation and growth.
Collapse
|
25
|
Yanni D, Korzeniewski SJ, Allred EN, Fichorova RN, O'Shea TM, Kuban K, Dammann O, Leviton A. Both antenatal and postnatal inflammation contribute information about the risk of brain damage in extremely preterm newborns. Pediatr Res 2017; 82:691-696. [PMID: 28549057 PMCID: PMC5599336 DOI: 10.1038/pr.2017.128] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/15/2017] [Indexed: 12/02/2022]
Abstract
BackgroundPreterm newborns exposed to intrauterine inflammation are at an increased risk of neurodevelopmental disorders. We hypothesized that adverse outcomes are more strongly associated with a combination of antenatal and postnatal inflammation than with either of them alone.MethodsWe defined antenatal inflammation as histologic inflammation in the placenta. We measured the concentrations of seven inflammation-related proteins in blood obtained on postnatal days 1, 7, and 14 from 763 infants born before 28 weeks of gestation. We defined postnatal inflammation as a protein concentration in the highest quartile on at least 2 days. We used logistic regression models to evaluate the contribution of antenatal and postnatal inflammation to the risk of neurodevelopmental disorders.ResultsThe risk of white matter damage was increased when placental inflammation was followed by sustained elevation of C-reactive protein or ICAM-1. We found the same for spastic cerebral palsy when placental inflammation was followed by elevation of TNF-α or IL-8. The presence of both placental inflammation and elevated levels of IL-6, TNF-α, or ICAM-1 was associated with an increased risk for microcephaly.ConclusionCompared with a single hit, two inflammatory hits are associated with stronger risk for abnormal cranial ultrasound, spastic cerebral palsy, and microcephaly at 2 years.
Collapse
Affiliation(s)
- Diana Yanni
- Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts
| | - Steven J. Korzeniewski
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan
| | - Elizabeth N. Allred
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raina N. Fichorova
- Departments of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - T. Michael O'Shea
- Department of Pediatrics, University of North Carolina, Chapel Hill North Carolina
| | - Karl Kuban
- Departments of Pediatrics and Neurology, Boston Medical Center and Boston University School of Medicine, Boston, Massachusetts
| | - Olaf Dammann
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | - Alan Leviton
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
26
|
Abstract
Worldwide, children are often not meeting their developmental potential owing to malnutrition, infection, lack of stimulation, and toxic stress. Children with disabilities are more likely to experience poverty, neglect, and abuse, and are less likely to have adequate access to education and medical care. Early childhood developmental stimulation can improve language, learning, and future participation in communities. Therapeutic supports and endeavors to reduce stigma for people of all abilities strengthen communities and allow for human thriving.
Collapse
|
27
|
Pascual M, Montesinos J, Montagud-Romero S, Forteza J, Rodríguez-Arias M, Miñarro J, Guerri C. TLR4 response mediates ethanol-induced neurodevelopment alterations in a model of fetal alcohol spectrum disorders. J Neuroinflammation 2017; 14:145. [PMID: 28738878 PMCID: PMC5525270 DOI: 10.1186/s12974-017-0918-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/12/2017] [Indexed: 01/15/2023] Open
Abstract
Background Inflammation during brain development participates in the pathogenesis of early brain injury and cognitive dysfunctions. Prenatal ethanol exposure affects the developing brain and causes neural impairment, cognitive and behavioral effects, collectively known as fetal alcohol spectrum disorders (FASD). Our previous studies demonstrate that ethanol activates the innate immune response and TLR4 receptor and causes neuroinflammation, brain damage, and cognitive defects in the developmental brain stage of adolescents. We hypothesize that by activating the TLR4 response, maternal alcohol consumption during pregnancy triggers the release of cytokines and chemokines in both the maternal sera and brains of fetuses/offspring, which impairs brain ontogeny and causes cognitive dysfunction. Methods WT and TLR4-KO female mice treated with or without 10% ethanol in the drinking water during gestation and lactation were used. Cytokine/chemokine levels were determined by ELISA in the amniotic fluid, maternal serum, and cerebral cortex, as well as in the offspring cerebral cortex. Microglial and neuronal markers (evaluated by western blotting), myelin proteins (immunohistochemical and western blotting) and synaptic parameters (western blotting and electron microscopy) were assessed in the cortices of the WT and TLR4-KO pups on PND 0, 20, and 66. Behavioral tests (elevated plus maze and passive avoidance) were performed in the WT and TLR4-KO mice on PND 66 exposed or not to ethanol. Results We show that alcohol intake during gestation and lactation increases the levels of several cytokines/chemokines (IL-1β, IL-17, MIP-1α, and fractalkine) in the maternal sera, amniotic fluid, and brains of fetuses and offspring. The upregulation of cytokines/chemokines is associated with an increase in activated microglia markers (CD11b and MHC-II), and with a reduction in some synaptic (synaptotagmin, synapsin IIa) and myelin (MBP, PLP) proteins in the brains of offspring on days 0, 20, and 66 (long-term effects). These changes are associated with long-term behavioral impairments, in the 66-day-old alcohol-exposed pups. TLR4-deficient mice are protected against ethanol-induced cytokine/chemokine production in alcohol-treated dams and offspring, along with synaptic and myelin alterations, and the log-term behavioral dysfunction induced by ethanol in offspring. Conclusions These results suggest that the immune system activation, through the TLR4 response, might play an important role in the neurodevelopmental defects in FASD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0918-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.,Department of Physiology, School of Medicine, Universitat de Valencia, Valencia, Spain
| | - Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Sandra Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Jerónimo Forteza
- Instituto Valenciano de Patología, Unidad Mixta de Patología Molecular, Principe Felipe Research Center, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - José Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012, Valencia, Spain. .,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
28
|
Williams M, Zhang Z, Nance E, Drewes JL, Lesniak WG, Singh S, Chugani DC, Rangaramanujam K, Graham DR, Kannan S. Maternal Inflammation Results in Altered Tryptophan Metabolism in Rabbit Placenta and Fetal Brain. Dev Neurosci 2017; 39:399-412. [PMID: 28490020 DOI: 10.1159/000471509] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/08/2017] [Indexed: 12/13/2022] Open
Abstract
Maternal inflammation has been linked to neurodevelopmental and neuropsychiatric disorders such as cerebral palsy, schizophrenia, and autism. We had previously shown that intrauterine inflammation resulted in a decrease in serotonin, one of the tryptophan metabolites, and a decrease in serotonin fibers in the sensory cortex of newborns in a rabbit model of cerebral palsy. In this study, we hypothesized that maternal inflammation results in alterations in tryptophan pathway enzymes and metabolites in the placenta and fetal brain. We found that intrauterine endotoxin administration at gestational day 28 (G28) resulted in a significant upregulation of indoleamine 2,3-dioxygenase (IDO) in both the placenta and fetal brain at G29 (24 h after treatment). This endotoxin-mediated IDO induction was also associated with intense microglial activation, an increase in interferon gamma expression, and increases in kynurenine and the kynurenine pathway metabolites kynurenine acid and quinolinic acid, as well as a significant decrease in 5-hydroxyindole acetic acid (a precursor of serotonin) levels in the periventricular region of the fetal brain. These results indicate that maternal inflammation shunts tryptophan metabolism away from the serotonin to the kynurenine pathway, which may lead to excitotoxic injury along with impaired development of serotonin-mediated thalamocortical fibers in the newborn brain. These findings provide new targets for prevention and treatment of maternal inflammation-induced fetal and neonatal brain injury leading to neurodevelopmental disorders such as cerebral palsy and autism.
Collapse
Affiliation(s)
- Monica Williams
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University SOM, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
30
|
Rand KM, Austin NC, Inder TE, Bora S, Woodward LJ. Neonatal Infection and Later Neurodevelopmental Risk in the Very Preterm Infant. J Pediatr 2016; 170:97-104. [PMID: 26707582 DOI: 10.1016/j.jpeds.2015.11.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/01/2015] [Accepted: 11/06/2015] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To document associations between confirmed and suspected neonatal infection and motor, cognitive, educational, and mental health outcomes of very preterm (VPT)-born children at 9 years of age; to examine the potential intervening role of cerebral white matter abnormalities (WMAs) and structural development on term magnetic resonance imaging. STUDY DESIGN A regional cohort of 110 infants born VPT in Christchurch, New Zealand were studied from birth to age of 9 years. Confirmed infection was defined as positive blood, cerebrospinal fluid or urine culture, and/or necrotizing enterocolitis ≥ stage 2. Suspected infection was defined as ≥ 5 days of antibiotics with evidence of clinical correlates. At term gestational equivalence, infants underwent structural magnetic resonance imaging. At age 9 years, neuromotor function, IQ, educational achievement, and mental health were assessed. RESULTS During hospitalization, 25% of VPT infants had confirmed and 23% had suspected infection. Longer-term neurodevelopmental impairments were largely confined to infants with confirmed infection (relative risk 1.4-3.1, vs uninfected). After accounting for other neonatal factors, these infants were at increased risk of severe motor impairment (OR 3.3, 95% CI 1.3-8), attention deficit hyperactivity disorder (ADHD) (OR 3.6, 95% CI 1.6-8), and IQ delay (OR 2.0, 95% CI 1-3.9). Cerebral WMAs contributed to associations between confirmed infection and motor and IQ impairments but not to ADHD (P = .005). CONCLUSIONS Confirmed neonatal infection heightens VPT infants' risk for neurodevelopmental impairment. WMA appears to be an important intervening factor linking infection and severe motor and IQ impairments. Further analysis of the neurologic mechanism accounting for ADHD in infants with infection is needed.
Collapse
Affiliation(s)
- Katherine M Rand
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA
| | - Nicola C Austin
- Christchurch Women's Hospital, Christchurch, New Zealand; University of Otago, Dunedin, New Zealand
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Samudragupta Bora
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Lianne J Woodward
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| |
Collapse
|
31
|
Bernstein HG, Piontkewitz Y, Keilhoff G. Commentary: Maternal immune activation evoked by polyinosinic: polycytidylic acid does not evoke microglial cell activation in the embryo. Front Cell Neurosci 2016; 10:41. [PMID: 26941606 PMCID: PMC4763168 DOI: 10.3389/fncel.2016.00041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/05/2016] [Indexed: 12/31/2022] Open
Affiliation(s)
| | - Yael Piontkewitz
- George S. Wise Faculty of Life Sciences, Strauss Center for Computational Neuroimaging, Tel Aviv University Tel Aviv, Israel
| | - Gerburg Keilhoff
- Department of Biochemistry and Cell Biology, University of Magdeburg Magdeburg, Germany
| |
Collapse
|
32
|
Subclinical decelerations during developing hypotension in preterm fetal sheep after acute on chronic lipopolysaccharide exposure. Sci Rep 2015; 5:16201. [PMID: 26537688 PMCID: PMC4633652 DOI: 10.1038/srep16201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/12/2015] [Indexed: 12/16/2022] Open
Abstract
Subclinical (shallow) heart rate decelerations occur during neonatal sepsis, but there is limited information on their relationship with hypotension or whether they occur before birth. We examined whether subclinical decelerations, a fall in fetal heart rate (FHR) that remained above 100 bpm, were associated with hypotension in preterm fetal sheep exposed to lipopolysaccharide (LPS). Chronically-instrumented fetal sheep at 0.7 gestation received continuous low-dose LPS infusions (n = 15, 100 ng/kg over 24 h, followed by 250 ng/kg/24 h for 96 h) or saline (n = 8). Boluses of 1 μg LPS or saline were given at 48 and 72 h. FHR variability (FHRV) was calculated, and sample asymmetry was used to assess the severity and frequency of decelerations. Low-dose LPS infusion did not affect FHR. After the first LPS bolus, 7 fetuses remained normotensive, while 8 developed hypotension (a fall in mean arterial blood pressure of ≥5 mmHg). Developing hypotension was associated with subclinical decelerations, with a corresponding increase in sample asymmetry and FHRV (p < 0.05). The second LPS bolus was associated with similar but attenuated changes in FHR and blood pressure (p < 0.05). In conclusion, subclinical decelerations are not consistently seen during prenatal exposure to LPS, but may be a useful marker of developing inflammation-related hypotension before birth.
Collapse
|
33
|
Dhillon SK, Gunn AJ, Jung Y, Mathai S, Bennet L, Fraser M. Lipopolysaccharide-Induced Preconditioning Attenuates Apoptosis and Differentially Regulates TLR4 and TLR7 Gene Expression after Ischemia in the Preterm Ovine Fetal Brain. Dev Neurosci 2015; 37:497-514. [PMID: 26184807 DOI: 10.1159/000433422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/13/2015] [Indexed: 11/19/2022] Open
Abstract
Acute exposure to subclinical infection modulates subsequent hypoxia-ischemia (HI) injury in a time-dependent manner, likely by cross-talk through Toll-like receptors (TLRs), but the specific pathways are unclear in the preterm-equivalent brain. In the present study, we tested the hypothesis that repeated low-dose exposure to lipopolysaccharide (LPS) before acute ischemia would be associated with induction of specific TLRs that are potentially neuroprotective. Fetal sheep at 0.65 gestation (term is ∼145 days) received intravenous boluses of low-dose LPS for 5 days (day 1, 50 ng/kg; days 2-5, 100 ng/kg) or the same volume of saline. Either 4 or 24 h after the last bolus of LPS, complete carotid occlusion was induced for 22 min. Five days after LPS, brains were collected. Pretreatment with LPS for 5 days decreased cellular apoptosis, microglial activation and reactive astrogliosis in response to HI injury induced 24 but not 4 h after the last dose of LPS. This was associated with upregulation of TLR4, TLR7 and IFN-β mRNA, and increased fetal plasma IFN-β concentrations. The association of reduced white matter apoptosis and astrogliosis after repeated low-dose LPS finishing 24 h but not 4 h before cerebral ischemia, with central and peripheral induction of IFN-β, suggests the possibility that IFN-β may be an important mediator of endogenous neuroprotection in the developing brain.
Collapse
|
34
|
Empie K, Rangarajan V, Juul SE. Is the ferret a suitable species for studying perinatal brain injury? Int J Dev Neurosci 2015; 45:2-10. [PMID: 26102988 PMCID: PMC4793918 DOI: 10.1016/j.ijdevneu.2015.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/09/2015] [Accepted: 06/01/2015] [Indexed: 11/26/2022] Open
Abstract
Ferret brain architecture, composition, and development are similar to humans. Postnatal ferret brain development is comparable to that of premature infants. Ferrets have potential to model preterm and term neonatal brain injury. Ferrets may fulfill the need for an intermediate model species of neurodevelopment. Many opportunities exist to expand the use of ferrets as research subjects.
Complications of prematurity often disrupt normal brain development and/or cause direct damage to the developing brain, resulting in poor neurodevelopmental outcomes. Physiologically relevant animal models of perinatal brain injury can advance our understanding of these influences and thereby provide opportunities to develop therapies and improve long-term outcomes. While there are advantages to currently available small animal models, there are also significant drawbacks that have limited translation of research findings to humans. Large animal models such as newborn pig, sheep and nonhuman primates have complex brain development more similar to humans, but these animals are expensive, and developmental testing of sheep and piglets is limited. Ferrets (Mustela putorius furo) are born lissencephalic and undergo postnatal cortical folding to form complex gyrencephalic brains. This review examines whether ferrets might provide a novel intermediate animal model of neonatal brain disease that has the benefit of a gyrified, altricial brain in a small animal. It summarizes attributes of ferret brain growth and development that make it an appealing animal in which to model perinatal brain injury. We postulate that because of their innate characteristics, ferrets have great potential in neonatal neurodevelopmental studies.
Collapse
Affiliation(s)
- Kristen Empie
- Department of Neonatology, University of Washington, Seattle, USA
| | | | - Sandra E Juul
- Department of Neonatology, University of Washington, Seattle, USA.
| |
Collapse
|
35
|
Potential neuroprotective strategies for perinatal infection and inflammation. Int J Dev Neurosci 2015; 45:44-54. [DOI: 10.1016/j.ijdevneu.2015.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 02/16/2015] [Accepted: 02/16/2015] [Indexed: 01/17/2023] Open
|