1
|
Valderrama-Mantilla AI, Martín-Cuevas C, Gómez-Garrido A, Morente-Montilla C, Crespo-Facorro B, García-Cerro S. Shared molecular signature in Alzheimer's disease and schizophrenia: A systematic review of the reelin signaling pathway. Neurosci Biobehav Rev 2025; 169:106032. [PMID: 39894421 DOI: 10.1016/j.neubiorev.2025.106032] [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: 12/23/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
The Reelin signaling pathway, particularly the RELN-APOER2-DAB1 complex, has emerged as a key contributor to the neuropathology of Alzheimer's disease (AD) and Schizophrenia (SZ). Despite being distinct clinical conditions, these disorders exhibit similar patterns of cognitive decline, including early disruptions in synaptic function and memory impairments. Notably, individuals with SZ have a 2-4 fold increased risk of developing AD or other dementias, highlighting potential shared molecular mechanisms, and positioning Reelin as a pivotal link between them. This systematic review explores the role of Reelin and its signaling components across these disorders. In AD, Reelin disruption correlates with hallmark features such as Tau hyperphosphorylation, amyloid-beta accumulation, and cognitive deficits. In SZ, alterations in Reelin signaling, including epigenetic modifications affecting RELN expression, are linked to disruptions in neuronal development and synaptic plasticity, particularly in the parietal and prefrontal cortices. Additionally, genomic studies reveal specific RELN variants and allelic imbalances that may influence disease severity and treatment response in SZ, suggesting RELN's role as a potential biomarker for therapeutic outcomes. Region-specific Reelin alterations in both AD and SZ suggest differing impacts yet underscore a potential common molecular origin. Our findings highlight the Reelin pathway as a molecular convergence point, warranting further investigation as a therapeutic and diagnostic target for AD, SZ, and potentially other neuropsychiatric disorders. The interplay between genetic and epigenetic regulation of RELN may provide novel insights into neurodegeneration, with implications for personalized intervention strategies in AD and SZ.
Collapse
Affiliation(s)
| | - Celia Martín-Cuevas
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, Seville 41013, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, Madrid 28029, Spain.
| | - Ana Gómez-Garrido
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, Seville 41013, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, Madrid 28029, Spain.
| | - Cristina Morente-Montilla
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, Seville 41013, Spain.
| | - Benedicto Crespo-Facorro
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, Seville 41013, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, Madrid 28029, Spain; Department of Psychiatry, School of Medicine, University of Seville, Manuel Siurot AV, Seville 41013, Spain.
| | - Susana García-Cerro
- Instituto de Biomedicina de Sevilla (IBiS)/University Hospital Virgen del Rocío/CSIC/University of Sevilla, Manuel Siurot AV, Seville 41013, Spain; Spanish Network for Research in Mental Health (CIBERSAM, ISCIII), Monforte de Lemos AV, 3-5, Madrid 28029, Spain.
| |
Collapse
|
2
|
Garcia IJP, Kinoshita PF, Valadares JMDM, de Carvalho LED, Cortes VF, Barbosa LA, Scavone C, Santos HDL. Effect of Ouabain on Glutamate Transport in the Hippocampus of Rats with LPS-Induced Neuroinflammation. Biomedicines 2023; 11:biomedicines11030920. [PMID: 36979899 PMCID: PMC10045517 DOI: 10.3390/biomedicines11030920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
A lipopolysaccharide (LPS)-induced neuroinflammation rat model was used to study the effects of ouabain (OUA) at low concentrations, which can interact with the Na,K-ATPase, causing the modulation of intracellular signalling pathways in the Central Nervous System. Our study aimed to analyse the effects of OUA on glutamate transport in the hippocampus of rats with LPS-induced neuroinflammation. Adult male Wistar rats were divided into four groups: OUA (1.8 µg/kg), saline (CTR), LPS (200 µg/kg), and OUA + LPS (OUA 20 min before LPS). The animals were sacrificed after 2 h, and the hippocampus was collected for analysis. After treatment, we determined the activities of Na,K-ATPase and glutamine synthetase (GS). In addition, expression of the α1, α2, and α3 isoforms of Na,K-ATPase and the glutamate transporters, EAAT1 and EAAT2, were also analysed. Treatment with OUA caused a specific increase in the α2 isoform expression (~20%), whereas LPS decreased its expression (~22%), and treatment with OUA before LPS prevented the effects of LPS. Moreover, LPS caused a decrease of approximately 50% in GS activity compared with that in the CTR group; however, OUA pre-treatment attenuated this effect of LPS. Notably, it was found that treatment with OUA caused an increase in the expression of EAAT1 (~30%) and EAAT2 (~25%), whereas LPS caused a decrease in the expression of EAAT1 (~23%) and EAAT2 (~25%) compared with that in the CTR group. When treated with OUA, the effects of LPS were abrogated. In conclusion, the OUA pre-treatment abolished the effect caused by LPS, suggesting that this finding may be related to the restoration of the interaction between FXYD2 and the studied membrane proteins.
Collapse
Affiliation(s)
- Israel José Pereira Garcia
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Paula Fernanda Kinoshita
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jéssica Martins de Moura Valadares
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Luciana Estefani Drumond de Carvalho
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Vanessa Faria Cortes
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Leandro Augusto Barbosa
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Cristoforo Scavone
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil
- Correspondence: (C.S.); (H.d.L.S.)
| | - Hérica de Lima Santos
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Correspondence: (C.S.); (H.d.L.S.)
| |
Collapse
|
3
|
Preeclampsia-induced alterations in brain and liver gene expression and DNA methylation patterns in fetal mice. J Dev Orig Health Dis 2023; 14:146-151. [PMID: 35748176 DOI: 10.1017/s2040174422000344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Exposure to pregnancy complications, including preeclampsia (PE), has lifelong influences on offspring's health. We have previously reported that experimental PE, induced in mice by administration of adenoviral sFlt1 at gestational day 8.5 combined with LPS at day 10.5, results in symmetrical growth restriction in female and asymmetrical growth restriction in male offspring. Here, we characterize the molecular phenotype of the fetal brain and liver with respect to gene transcription and DNA methylation at the end of gestation.In fetal brain and liver, expression and DNA methylation of several key regulatory genes is altered by PE exposure, mostly independent of fetal sex. These alterations point toward a decreased gluconeogenesis in the liver and stimulated neurogenesis in the brain, potentially affecting long-term brain and liver function. The observed sex-specific growth restriction pattern is not reflected in the molecular data, showing that PE, rather than tissue growth, drives the molecular phenotype of PE-exposed offspring.
Collapse
|
4
|
Lin K, Liu M, Sun L, Qiao H, Wang S, Pan S, Fu H, Wang J, Wei Q, Gao H. The effects of inactivated SARS-CoV-2 vaccination and subsequent infection of pregnant mice on the behaviors of offspring. Animal Model Exp Med 2022; 5:430-435. [PMID: 35909330 PMCID: PMC9353298 DOI: 10.1002/ame2.12261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/30/2022] [Indexed: 11/07/2022] Open
Abstract
The mass inoculation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines to induce herd immunity is one of the most effective measures we can deploy in the fight against coronavirus disease 2019 (COVID-19). Pregnant women are prone to a higher risk of COVID-19, and maternal infection is a risk factor for a range of neurological disorders leading to abnormal behavior in adulthood. However, there are limited clinical data to support whether vaccination or infection post-immunization in pregnant women can affect the behavioral cognition of fetuses in adulthood. In this study, human angiotensin-converting enzyme 2 pregnant mice (F0 generation) were immunized with CoronaVac and then infected with SARS-CoV-2. Subsequently, we analyzed the behavioral cognition of their adult offspring (F1 generation) using the open-field test and Morris water maze test. The adult F1 generation did not exhibit any impairments in spontaneous locomotor activity or spatial reference memory.
Collapse
Affiliation(s)
- Kaili Lin
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Meixuan Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Lu Sun
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Hongwei Qiao
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Shunyi Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Sidan Pan
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Hanjun Fu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Jingzhu Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Qiang Wei
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| | - Hong Gao
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina
| |
Collapse
|
5
|
Maternal Inflammation Exaggerates Offspring Susceptibility to Cerebral Ischemia–Reperfusion Injury via the COX-2/PGD2/DP2 Pathway Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1571705. [PMID: 35437456 PMCID: PMC9013311 DOI: 10.1155/2022/1571705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 12/04/2022]
Abstract
The pathogenesis of cerebral ischemia–reperfusion (I/R) injury is complex and does not exhibit an effective strategy. Maternal inflammation represents one of the most important factors involved in the etiology of brain injury in newborns. We aimed to investigate the effect of maternal inflammation on offspring susceptibility to cerebral I/R injury and the mechanisms by which it exerts its effects. Pregnant SD rats were intraperitoneally injected with LPS (300 μg/kg/day) at gestational days 11, 14, and 18. Pups were subjected to MCAO/R on postnatal day 60. Primary neurons were obtained from postnatal day 0 SD rats and subjected to OGD/R. Neurological deficits, brain injury, neuronal viability, neuronal damage, and neuronal apoptosis were assessed. Oxidative stress and inflammation were evaluated, and the expression levels of COX-2/PGD2/DP pathway-related proteins and apoptotic proteins were detected. Maternal LPS exposure significantly increased the levels of oxidative stress and inflammation, significantly activated the COX-2/PGD2/DP2 pathway, and increased proapoptotic protein expression. However, maternal LPS exposure significantly decreased the antiapoptotic protein expression, which subsequently increased neurological deficits and cerebral I/R injury in offspring rats. The corresponding results were observed in primary neurons. Moreover, these effects of maternal LPS exposure were reversed by a COX-2 inhibitor and DP1 agonist but exacerbated by a DP2 agonist. In conclusion, maternal inflammatory exposure may increase offspring susceptibility to cerebral I/R injury. Moreover, the underlying mechanism might be related to the activation of the COX-2/PGD2/DP2 pathway. These findings provide a theoretical foundation for the development of therapeutic drugs for cerebral I/R injury.
Collapse
|
6
|
Sánchez-Hidalgo AC, Martín-Cuevas C, Crespo-Facorro B, Garrido-Torres N. Reelin Alterations, Behavioral Phenotypes, and Brain Anomalies in Schizophrenia: A Systematic Review of Insights From Rodent Models. Front Neuroanat 2022; 16:844737. [PMID: 35401125 PMCID: PMC8986979 DOI: 10.3389/fnana.2022.844737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/14/2022] [Indexed: 12/09/2022] Open
Abstract
Reelin is an extracellular matrix glycoprotein reduced in brain regions (the prefrontal cortex and the hippocampus) of patients with schizophrenia. There are diverse rodent models of schizophrenia that mimic patient symptoms based on various causal theories; however, likely shared reelin alterations have not yet been systematically assessed in those models. A systematic review of the literature was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) model. Articles focused on psychotic disorders or schizophrenia and their relationship with reelin in rodent models were selected. Data (first author, publication year, results, both open field and prepulse inhibition test results, and type of reelin alteration) were extracted in duplicate by two independent reviewers. The 37 reviewed articles reported about various schizophrenia models and their reelin alterations, brain morphology, and behavioral defects. We conclude that reelin is an altered preclinical biomarker common to all models included, mainly prenatal or genetic models, and a key protein in schizophrenia disease, making the reelin signaling pathway in prenatal stages a target of special interest for future preclinical and clinical studies. All models presented at least one of the four described reelin alteration types. Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021210568], identifier [CRD42021210568].
Collapse
Affiliation(s)
- Ana C. Sánchez-Hidalgo
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
| | - Celia Martín-Cuevas
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
| | - Benedicto Crespo-Facorro
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocío-IBiS, Seville, Spain
- *Correspondence: Benedicto Crespo-Facorro,
| | - Nathalia Garrido-Torres
- Spanish Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Seville Biomedical Research Centre (IBiS), Seville, Spain
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocío-IBiS, Seville, Spain
| |
Collapse
|
7
|
Park S, Luk SHC, Bains RS, Whittaker DS, Chiem E, Jordan MC, Roos KP, Ghiani CA, Colwell CS. Targeted Genetic Reduction of Mutant Huntingtin Lessens Cardiac Pathology in the BACHD Mouse Model of Huntington's Disease. Front Cardiovasc Med 2022; 8:810810. [PMID: 35004919 PMCID: PMC8739867 DOI: 10.3389/fcvm.2021.810810] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/07/2021] [Indexed: 01/16/2023] Open
Abstract
Individuals affected by Huntington's disease (HD) present with progressive degeneration that results in a wide range of symptoms, including cardiovascular (CV) dysfunction. The huntingtin gene (HTT) and its product are ubiquitously expressed, hence, the cardiomyopathy could also be driven by defects caused by its mutated form (mHTT) in the cardiomyocytes themselves. In the present study, we sought to determine the contribution of the mHTT expressed in the cardiomyocytes to CV symptoms. We utilized the BACHD mouse model, which exhibits many of the HD core symptoms, including CV dysfunction. This model allows the targeted genetic reduction of mHTT expression in the cardiomyocytes while maintaining the expression of the mHTT in the rest of the body. The BACHD line was crossed with a line of mice in which the expression of Cre recombinase is driven by the cardiac-specific alpha myosin-heavy chain (Myh6) promoter. The offspring of this cross (BMYO mice) exhibited a dramatic reduction in mHTT in the heart but not in the striatum. The BMYO mice were evaluated at 6 months old, as at this age, the BACHD line displays a strong CV phenotype. Echocardiogram measurements found improvement in the ejection fraction in the BMYO line compared to the BACHD, while hypertrophy was observed in both mutant lines. Next, we examined the expression of genes known to be upregulated during pathological cardiac hypertrophy. As measured by qPCR, the BMYO hearts exhibited significantly less expression of collagen1a as well as Gata4, and brain natriuretic peptide compared to the BACHD. Fibrosis in the hearts assessed by Masson's trichrome stain and the protein levels of fibronectin were reduced in the BMYO hearts compared to BACHD. Finally, we examined the performance of the mice on CV-sensitive motor tasks. Both the overall activity levels and grip strength were improved in the BMYO mice. Therefore, we conclude that the reduction of mHtt expression in the heart benefits CV function in the BACHD model, and suggest that cardiomyopathy should be considered in the treatment strategies for HD.
Collapse
Affiliation(s)
- Saemi Park
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shu Hon Christopher Luk
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Raj S Bains
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel S Whittaker
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Emily Chiem
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maria C Jordan
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kenneth P Roos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cristina A Ghiani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
8
|
Eve M, Gandawijaya J, Yang L, Oguro-Ando A. Neuronal Cell Adhesion Molecules May Mediate Neuroinflammation in Autism Spectrum Disorder. Front Psychiatry 2022; 13:842755. [PMID: 35492721 PMCID: PMC9051034 DOI: 10.3389/fpsyt.2022.842755] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by restrictive and repetitive behaviors, alongside deficits in social interaction and communication. The etiology of ASD is largely unknown but is strongly linked to genetic variants in neuronal cell adhesion molecules (CAMs), cell-surface proteins that have important roles in neurodevelopment. A combination of environmental and genetic factors are believed to contribute to ASD pathogenesis. Inflammation in ASD has been identified as one of these factors, demonstrated through the presence of proinflammatory cytokines, maternal immune activation, and activation of glial cells in ASD brains. Glial cells are the main source of cytokines within the brain and, therefore, their activity is vital in mediating inflammation in the central nervous system. However, it is unclear whether the aforementioned neuronal CAMs are involved in modulating neuroimmune signaling or glial behavior. This review aims to address the largely unexplored role that neuronal CAMs may play in mediating inflammatory cascades that underpin neuroinflammation in ASD, primarily focusing on the Notch, nuclear factor-κB (NF-κB), and mitogen-activated protein kinase (MAPK) cascades. We will also evaluate the available evidence on how neuronal CAMs may influence glial activity associated with inflammation. This is important when considering the impact of environmental factors and inflammatory responses on ASD development. In particular, neural CAM1 (NCAM1) can regulate NF-κB transcription in neurons, directly altering proinflammatory signaling. Additionally, NCAM1 and contactin-1 appear to mediate astrocyte and oligodendrocyte precursor proliferation which can alter the neuroimmune response. Importantly, although this review highlights the limited information available, there is evidence of a neuronal CAM regulatory role in inflammatory signaling. This warrants further investigation into the role other neuronal CAM family members may have in mediating inflammatory cascades and would advance our understanding of how neuroinflammation can contribute to ASD pathology.
Collapse
Affiliation(s)
- Madeline Eve
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Liming Yang
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
9
|
Shende P, Gupta S. Role of lipopolysaccharides in potential applications of nanocarrier systems. Curr Pharm Des 2021; 28:1000-1010. [PMID: 34818999 DOI: 10.2174/1381612827666211124094302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lipopolysaccharides (LPS) are considered the main molecular component in the outer membrane of gram-negative bacteria. The LPS molecule in the bacterial cell wall acts as a primary physical barrier and protects gram-negative bacteria from the surrounding environment. LPS (endotoxins) show immunomodulatory therapeutic properties as well as toxicity to the host cell, whereas potential applications encompass. OBJECTIVE This review article aims to describe the recent developments of lipopolysaccharides in nanocarrier systems for various applications such as vaccination, cancer chemotherapy and immune stimulants action. Different nanocarriers like cubosomes, niosomes, dendrimers and metal nanoparticles used in the delivery of actives are employed to decorate lipopolysaccharide molecules superficially. METHODS A narrative review of all the relevant papers known to the author was conducted. CONCLUSION Commercially available lipid nanoparticles contribute to many advances as promising nanocarriers in cancer therapy and are used as a vaccine adjuvant by improving the immune response due to their properties such as size, shape, biocompatibility, and biodegradability. Whereas lipopolysaccharide-decorated nanoparticles change the host's tolerability and increase the effectiveness of molecule in cancer immunotherapy. These nanoconjugate systems enhance overall immunogenic response and effectiveness in vaccine immunotherapy and targeted therapy, not only limited to humans application but also for poultry and aquaculture. Newer opportunities using lipopolysaccharides for the treatment and management of diseases with unique characteristics like the presence of lipoprotein that act as an alternative for bacterial infections over conventional dosage forms.
Collapse
Affiliation(s)
- Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai. India
| | - Shubham Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai. India
| |
Collapse
|
10
|
Prevention of Teratogenesis in Pregnancies of Obese Rats by Vitamin E Supplementation. Antioxidants (Basel) 2021; 10:antiox10081173. [PMID: 34439421 PMCID: PMC8389020 DOI: 10.3390/antiox10081173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 01/15/2023] Open
Abstract
Congenital malformations are a common adverse outcome in pregnancies complicated by pregestational obesity, although the underlying mechanisms are still unrevealed. Our aim was to study the effect of oxidative stress in obesity-induced teratogenesis. Wistar rats were fed a high-fat diet for 13 weeks, with (OE group) or without (O group) vitamin E supplementation. Then, rats were mated and sacrificed at day 11.5 of gestation. Embryos from O dams presented a 25.9 ± 3.5% rate of malformations (vs. 8.7 ± 3.4% in C rats), which was reduced in the OE group (11.5 ± 2.3%). Pregestational obesity induced hepatic protein and DNA oxidation and a decline in antioxidant enzymes. Importantly, glutathione content was also decreased, limiting the availability of this antioxidant in the embryos. Vitamin E supplementation efficiently maintained glutathione levels in the obese mothers, which could be used in their embryos to prevent oxidation-induced malformations. To test the effect of decreasing glutathione levels alone in a cell culture model of neuroepithelium, murine embryonic stem cells (ESC) were induced to form neuronal precursors and glutathione synthesis was inhibited with the gamma–glutamylcysteine synthesis inhibitor, buthionine sulfoximine (BSO). BSO inhibited the expression of Pax3, a gene required for neural tube closure that is also inhibited by oxidative stress. Taken together, our data indicate that obesity causes malformations through the depletion of maternal glutathione, thereby decreasing glutathione-dependent free radical scavenging in embryos, which can be prevented by vitamin E supplementation.
Collapse
|
11
|
Influence of Cerebral Vasodilation on Blood Reelin Levels in Growth Restricted Fetuses. Diagnostics (Basel) 2021; 11:diagnostics11061036. [PMID: 34199942 PMCID: PMC8228107 DOI: 10.3390/diagnostics11061036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 11/23/2022] Open
Abstract
Fetal growth restriction (FGR) is one of the most important obstetric pathologies. It is frequently caused by placental insufficiency. Previous studies have shown a relationship between FGR and impaired new-born neurodevelopment, although the molecular mechanisms involved in this association have not yet been completely clarified. Reelin is an extracellular matrix glycoprotein involved in development of neocortex, hippocampus, cerebellum and spinal cord. Reelin has been demonstrated to play a key role in regulating perinatal neurodevelopment and to contribute to the emergence and development of various psychiatric pathologies, and its levels are highly influenced by pathological conditions of hypoxia. The purpose of this article is to study whether reelin levels in new-borns vary as a function of severity of fetal growth restriction by gestational age and sex. We sub-grouped fetuses in: normal weight group (Group 1, n = 17), FGR group with normal umbilical artery Doppler and cerebral redistribution at middle cerebral artery Doppler (Group 2, n = 9), and FGR with abnormal umbilical artery Doppler (Group 3, n = 8). Our results show a significant association of elevated Reelin levels in FGR fetuses with cerebral blood redistribution compared to the normal weight group and the FGR with abnormal umbilical artery group. Future research should focus on further expanding the knowledge of the relationship of reelin and its regulated products with neurodevelopment impairment in new-borns with FGR and should include larger and more homogeneous samples and the combined use of different in vivo techniques in neonates with impaired growth during their different adaptive phases.
Collapse
|
12
|
Chen C, Lu D, Xue L, Ren P, Zhang H, Zhang J. Association between Placental Inflammatory Pathology and Offspring Neurodevelopment at 8 Months and 4 and 7 Years of Age. J Pediatr 2020; 225:132-137.e2. [PMID: 32479832 DOI: 10.1016/j.jpeds.2020.05.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To examine whether placental inflammatory pathology is associated with subsequent child neurodevelopment. STUDY DESIGN We used the data of US Collaborative Perinatal Project cohort study. Placentas were examined by pathologists and child neurodevelopment was evaluated at 8 months and 4 and 7 years of age. Multivariable logistic regression modelling was used to assess the association. A mediation analysis was used to evaluate whether the association was mediated through shorter gestational age. RESULTS Multivariable analysis after adjusting for confounders showed that placental inflammatory pathology was significantly associated with low Bayley motor (adjusted OR (aOR), 2.15; 95% CI, 1.50-3.06) and mental scales (aOR, 1.51; 95% CI, 1.05-2.17) at 8 months and an IQ of 70-84 (aOR, 1.13; 95% CI, 1.01-1.26) at 4 years of age. The association diminished at 7 years of age (IQ of <70, aOR 1.20 [95% CI, 0.97-1.48]; IQ of 70-79, aOR 1.03 [95% CI, 0.89-1.18]). The mediation analysis demonstrated that associations between placental inflammatory pathology and development were primarily due to direct effects of placental inflammatory pathology rather than indirect effects of shorter gestational age. CONCLUSIONS Placental inflammation was associated with adverse offspring neurodevelopment up to 4 years of age.
Collapse
Affiliation(s)
- Chang Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danni Lu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Xue
- Department of Obstetrics, Jiaxing Maternity and Child Health Care Hospital, Zhejiang, China
| | - Peng Ren
- Department of Pathology, Jiaxing Maternity and Child Health Care Hospital, Zhejiang, China
| | - Huijuan Zhang
- Department of Pathology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Costa LG, Cole TB, Dao K, Chang YC, Coburn J, Garrick JM. Effects of air pollution on the nervous system and its possible role in neurodevelopmental and neurodegenerative disorders. Pharmacol Ther 2020; 210:107523. [PMID: 32165138 PMCID: PMC7245732 DOI: 10.1016/j.pharmthera.2020.107523] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
Recent extensive evidence indicates that air pollution, in addition to causing respiratory and cardiovascular diseases, may also negatively affect the brain and contribute to central nervous system diseases. Air pollution is comprised of ambient particulate matter (PM) of different sizes, gases, organic compounds, and metals. An important contributor to PM is represented by traffic-related air pollution, mostly ascribed to diesel exhaust (DE). Epidemiological and animal studies have shown that exposure to air pollution may be associated with multiple adverse effects on the central nervous system. In addition to a variety of behavioral abnormalities, the most prominent effects caused by air pollution are oxidative stress and neuro-inflammation, which are seen in both humans and animals, and are supported by in vitro studies. Among factors which can affect neurotoxic outcomes, age is considered most relevant. Human and animal studies suggest that air pollution may cause developmental neurotoxicity, and may contribute to the etiology of neurodevelopmental disorders, including autism spectrum disorder. In addition, air pollution exposure has been associated with increased expression of markers of neurodegenerative disease pathologies, such as alpha-synuclein or beta-amyloid, and may thus contribute to the etiopathogenesis of neurodegenerative diseases, particularly Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Lucio G Costa
- Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Dept. of Medicine & Surgery, University of Parma, Italy.
| | - Toby B Cole
- Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Center on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Khoi Dao
- Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Yu-Chi Chang
- Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Jacki Coburn
- Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Jacqueline M Garrick
- Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Microglia along sex lines: From brain colonization, maturation and function, to implication in neurodevelopmental disorders. Semin Cell Dev Biol 2019; 94:152-163. [PMID: 31201858 DOI: 10.1016/j.semcdb.2019.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022]
Abstract
In addition to their traditional role as immune sentinels, recent discoveries over the last decade have shown that microglial functions now include regulation of neuronal/glial cell migration, differentiation and maturation, as well as neuronal network formation. It was thus proposed that disruption of these microglial roles, during critical periods of brain development, could lead to the pathological onset of several neurodevelopmental disorders, including autism spectrum disorder, attention deficit hyperactivity disorder, epilepsy, schizophrenia, and major depressive disorder. The prevalence of these disorders exhibits a clear distinction along sex lines with very little known about the mechanisms underlying this difference. One of the fundamental discoveries that arose from recent research into the physiological roles of microglia in neurodevelopment is their sexual dimorphism, raising the intriguing possibility that sex differences in microglial colonization, maturation and/or function in the developing brain could underlie the emergence of various neurodevelopmental disorders. This review discusses the physiological roles of microglia across neurodevelopment, these roles in the two sexes, and the recent evidence that microglial sexually dimorphic nature may contribute, at least partially, to neurodevelopmental disorders.
Collapse
|
15
|
Kuljis D, Kudo T, Tahara Y, Ghiani CA, Colwell CS. Pathophysiology in the suprachiasmatic nucleus in mouse models of Huntington's disease. J Neurosci Res 2018; 96:1862-1875. [PMID: 30168855 DOI: 10.1002/jnr.24320] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/04/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022]
Abstract
Disturbances in sleep/wake cycle are a common complaint of individuals with Huntington's disease (HD) and are displayed by HD mouse models. The underlying mechanisms, including the possible role of the circadian timing system, are not well established. The BACHD mouse model of HD exhibits disrupted behavioral and physiological rhythms, including decreased electrical activity in the central circadian clock (suprachiasmatic nucleus, SCN). In this study, electrophysiological techniques were used to explore the ionic underpinning of the reduced spontaneous neural activity in male mice. We found that SCN neural activity rhythms were lost early in the disease progression and was accompanied by loss of the normal daily variation in resting membrane potential in the mutant SCN neurons. The low neural activity could be transiently reversed by direct current injection or application of exogenous N-methyl-d-aspartate (NMDA) thus demonstrating that the neurons have the capacity to discharge at WT levels. Exploring the potassium currents known to regulate the electrical activity of SCN neurons, our most striking finding was that these cells in the mutants exhibited an enhancement in the large-conductance calcium activated K+ (BK) currents. The expression of the pore forming subunit (Kcnma1) of the BK channel was higher in the mutant SCN. We found a similar decrease in daytime electrical activity and enhancement in the magnitude of the BK currents early in disease in another HD mouse model (Q175). These findings suggest that SCN neurons of both HD models exhibit early pathophysiology and that dysregulation of BK current may be responsible.
Collapse
Affiliation(s)
- Dika Kuljis
- Department of Neurobiology, University of California Los Angeles, Los Angeles, California.,Department of Biological Sciences, Mellon Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Takashi Kudo
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California.,Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Yu Tahara
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Cristina A Ghiani
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California.,Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California
| | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
16
|
Wu Y, Qi F, Song D, He Z, Zuo Z, Yang Y, Liu Q, Hu S, Wang X, Zheng X, Yang J, Yuan Q, Zou J, Guo K, Yao Z. Prenatal influenza vaccination rescues impairments of social behavior and lamination in a mouse model of autism. J Neuroinflammation 2018; 15:228. [PMID: 30103815 PMCID: PMC6090662 DOI: 10.1186/s12974-018-1252-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022] Open
Abstract
Background Prenatal infection is a substantial risk factor for neurodevelopmental disorders such as autism in offspring. We have previously reported that influenza vaccination (VAC) during early pregnancy contributes to neurogenesis and behavioral function in offspring. Results Here, we probe the efficacy of VAC pretreatment on autism-like behaviors in a lipopolysaccharide (LPS)-induced maternal immune activation (MIA) mouse model. We show that VAC improves abnormal fetal brain cytoarchitecture and lamination, an effect associated with promotion of intermediate progenitor cell differentiation in MIA fetal brain. These beneficial effects are sufficient to prevent social deficits in adult MIA offspring. Furthermore, whole-genome analysis suggests a strong interaction between Ikzf1 (IKAROS family zinc-finger 1) and neuronal differentiation. Intriguingly, VAC rescues excessive microglial Ikzf1 expression and attenuates microglial inflammatory responses in the MIA fetal brain. Conclusions Our study implies that a preprocessed influenza vaccination prevents maternal bacterial infection from causing neocortical lamination impairments and autism-related behaviors in offspring. Electronic supplementary material The online version of this article (10.1186/s12974-018-1252-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingying Wu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Dan Song
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zitian He
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Yunjie Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Qiongliang Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Saisai Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Qunfang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China.
| |
Collapse
|
17
|
Hester MS, Tulina N, Brown A, Barila G, Elovitz MA. Intrauterine inflammation reduces postnatal neurogenesis in the hippocampal subgranular zone and leads to accumulation of hilar ectopic granule cells. Brain Res 2018; 1685:51-59. [PMID: 29448014 PMCID: PMC5880291 DOI: 10.1016/j.brainres.2018.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/04/2018] [Accepted: 02/02/2018] [Indexed: 01/09/2023]
Abstract
Prenatal inflammation is associated with poor neurobehavioral outcomes in exposed offspring. A common route of exposure for the fetus is intrauterine infection, which is often associated with preterm birth. Hippocampal development may be particularly vulnerable to an inflammatory insult during pregnancy as this region remains highly neurogenic both prenatally and postnatally. These studies sought to determine if intrauterine inflammation specifically altered hippocampal neurogenesis and migration of newly produced granule neurons during the early postnatal period. Microglial and astroglial cell populations known to play a role in the regulation of postnatal neurogenesis were also examined. We show that intrauterine inflammation significantly reduced hippocampal neurogenesis between postnatal days 7 (P7) and P14 as well as decreased granule cell density at P28. Ectopic migration of granule cells was observed in LPS-exposed mice at P14, but not at P28. Intrauterine inflammation had no effect on hippocampal astrocyte or microglia density or on apoptosis rate at the postnatal time points examined. Thus, exposure to intrauterine inflammation disrupts early postnatal neurogenesis and leads to aberrant migration of newly born granule cells.
Collapse
Affiliation(s)
- Michael S Hester
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Natalia Tulina
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Amy Brown
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guillermo Barila
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Acute in utero exposure to lipopolysaccharide induces inflammation in the pre- and postnatal brain and alters the glial cytoarchitecture in the developing amygdala. J Neuroinflammation 2017; 14:212. [PMID: 29096641 PMCID: PMC5667487 DOI: 10.1186/s12974-017-0981-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022] Open
Abstract
Background Maternal immune activation (MIA) is a risk factor for neurodevelopmental disorders such as autism and schizophrenia, as well as seizure development. The amygdala is a brain region involved in the regulation of emotions, and amygdalar maldevelopment due to infection-induced MIA may lead to amygdala-related disorders. MIA priming of glial cells during development has been linked to abnormalities seen in later life; however, little is known about its effects on amygdalar biochemical and cytoarchitecture integrity. Methods Time-mated C57BL6J mice were administered a single intraperitoneal injection of 50 μg/kg lipopolysaccharide (LPS) on embryonic day 12 (E12), and the effects of MIA were examined at prenatal, neonatal, and postnatal developmental stages using immunohistochemistry, real-time quantitative PCR, and stereological quantification of cytoarchitecture changes. Results Fetal brain expression of pro-inflammatory cytokines (IL-1β, TNFα, and IL-6) was significantly upregulated at 4 h postinjection (E12) and remained elevated until the day of birth (P0). In offspring from LPS-treated dams, amygdalar expression of pro-inflammatory cytokines was also increased on day 7 (P7) and expression was sustained on day 40 (P40). Toll-like receptor (TLR-2, TLR-4) expression was also upregulated in fetal brains and in the postnatal amygdala in LPS-injected animals. Morphological examination of cells expressing ionized calcium-binding adaptor molecule 1 (Iba-1) and glial fibrillary acidic protein (GFAP) suggested long-term microglial activation and astrogliosis in postnatal amygdalar regions. Conclusions Our results showed that LPS-induced MIA at E12 induces a pro-inflammatory cytokine profile in the developing fetal brain that continues up to early adulthood in the amygdala. Inflammation elicited by MIA may activate cells in the fetal brain and lead to alterations in glial (microglia and astrocyte) cells observed in the postnatal amygdala. Moreover, increased pro-inflammatory cytokines and their effects on glial subpopulations may in turn have deleterious consequences for neuronal viability. These MIA-induced changes may predispose offspring to amygdala-related disorders such as heightened anxiety and depression and also neurodevelopmental disorders, such as autism spectrum disorders.
Collapse
|
19
|
Radtke FA, Chapman G, Hall J, Syed YA. Modulating Neuroinflammation to Treat Neuropsychiatric Disorders. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5071786. [PMID: 29181395 PMCID: PMC5664241 DOI: 10.1155/2017/5071786] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/13/2017] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is recognised as one of the potential mechanisms mediating the onset of a broad range of psychiatric disorders and may contribute to nonresponsiveness to current therapies. Both preclinical and clinical studies have indicated that aberrant inflammatory responses can result in altered behavioral responses and cognitive deficits. In this review, we discuss the role of inflammation in the pathogenesis of neuropsychiatric disorders and ask the question if certain genetic copy-number variants (CNVs) associated with psychiatric disorders might play a role in modulating inflammation. Furthermore, we detail some of the potential treatment strategies for psychiatric disorders that may operate by altering inflammatory responses.
Collapse
Affiliation(s)
- Franziska A. Radtke
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Gareth Chapman
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Yasir A. Syed
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| |
Collapse
|
20
|
Child neurodevelopmental outcomes following preterm and term birth: What can the placenta tell us? Placenta 2017; 57:79-86. [DOI: 10.1016/j.placenta.2017.06.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 11/21/2022]
|
21
|
Costa LG, Chang YC, Cole TB. Developmental Neurotoxicity of Traffic-Related Air Pollution: Focus on Autism. Curr Environ Health Rep 2017; 4:156-165. [PMID: 28417440 PMCID: PMC5952375 DOI: 10.1007/s40572-017-0135-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Epidemiological and animal studies suggest that air pollution may negatively affect the central nervous system (CNS) and contribute to CNS diseases. Traffic-related air pollution is a major contributor to global air pollution, and diesel exhaust (DE) is its most important component. RECENT FINDINGS Several studies suggest that young individuals may be particularly susceptible to air pollution-induced neurotoxicity and that perinatal exposure may cause or contribute to developmental disabilities and behavioral abnormalities. In particular, a number of recent studies have found associations between exposures to traffic-related air pollution and autism spectrum disorders (ASD), which are characterized by impairment in socialization and in communication and by the presence of repetitive and unusual behaviors. The cause(s) of ASD are unknown, and while it may have a hereditary component, environmental factors are increasingly suspected as playing a pivotal role in its etiology, particularly in genetically susceptible individuals. Autistic children present higher levels of neuroinflammation and systemic inflammation, which are also hallmarks of exposure to traffic-related air pollution. Gene-environment interactions may play a relevant role in determining individual susceptibility to air pollution developmental neurotoxicity. Given the worldwide presence of elevated air pollution, studies on its effects and mechanisms on the developing brain, genetic susceptibility, role in neurodevelopmental disorders, and possible therapeutic interventions are certainly warranted.
Collapse
Affiliation(s)
- Lucio G Costa
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt, Suite No. 100, Seattle, WA, 98105, USA.
- Department of Neuroscience, University of Parma, Parma, Italy.
| | - Yu-Chi Chang
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt, Suite No. 100, Seattle, WA, 98105, USA
| | - Toby B Cole
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, 4225 Roosevelt, Suite No. 100, Seattle, WA, 98105, USA
- Center on Human Development and Disability, University of Washington, Seattle, WA, USA
| |
Collapse
|
22
|
Ronovsky M, Berger S, Molz B, Berger A, Pollak DD. Animal Models of Maternal Immune Activation in Depression Research. Curr Neuropharmacol 2017; 14:688-704. [PMID: 26666733 PMCID: PMC5050397 DOI: 10.2174/1570159x14666151215095359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/24/2015] [Accepted: 11/09/2015] [Indexed: 01/17/2023] Open
Abstract
Abstract: Background Depression and schizophrenia are debilitating mental illnesses with significant socio-economic impact. The high degree of comorbidity between the two disorders, and shared symptoms and risk factors, suggest partly common pathogenic mechanisms. Supported by human and animal studies, maternal immune activation (MIA) has been intimately associated with the development of schizophrenia. However, the link between MIA and depression has remained less clear, in part due to the lack of appropriate animal models. Objective Here we aim to summarize findings obtained from studies using MIA animal models and discuss their relevance for preclinical depression research. Methods Results on molecular, cellular and behavioral phenotypes in MIA animal models were collected by literature search (PubMed) and evaluated for their significance for depression. Results Several reports on offspring depression-related behavioral alterations indicate an involvement of MIA in the development of depression later in life. Depression-related behavioral phenotypes were frequently paralleled by neurogenic and neurotrophic deficits and modulated by several genetic and environmental factors. Conclusion Literature evidence analyzed in this review supports a relevance of MIA as animal model for a specific early life adversity, which may prime an individual for the development of distinct psychopathologies later life. MIA animal models may present a unique tool for the identification of additional exogenous and endogenous factors, which are required for the manifestation of a specific neuropsychiatric disorder, such as depression, later in life. Hereby, novel insights into the molecular mechanisms involved in the pathophysiology of depression may be obtained, supporting the identification of alternative therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| |
Collapse
|
23
|
Traffic-Related Air Pollution and Neurodegenerative Diseases: Epidemiological and Experimental Evidence, and Potential Underlying Mechanisms. ADVANCES IN NEUROTOXICOLOGY 2017. [DOI: 10.1016/bs.ant.2017.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Wang HB, Whittaker DS, Truong D, Mulji AK, Ghiani CA, Loh DH, Colwell CS. Blue light therapy improves circadian dysfunction as well as motor symptoms in two mouse models of Huntington's disease. Neurobiol Sleep Circadian Rhythms 2017; 2:39-52. [PMID: 31236494 PMCID: PMC6575206 DOI: 10.1016/j.nbscr.2016.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 01/23/2023] Open
Abstract
Patients with Huntington's disease (HD) exhibit movement disorders, psychiatric disturbance and cognitive impairments as the disease progresses. Abnormal sleep/wake cycles are common among HD patients with reports of delayed sleep onset, fatigue during the day, and a delayed pattern of melatonin secretion all of which suggest circadian dysfunction. Mouse models of HD confirm disrupted circadian rhythms with pathophysiology found in the central circadian clock (suprachiasmatic nucleus). Importantly, circadian dysfunction manifests early in disease, even before the classic motor symptoms, in both patients and mouse models. Therefore, we hypothesize that the circadian dysfunction may interact with the disease pathology and exacerbate the HD symptoms. If correct, early intervention may benefit patients and delay disease progression. One test of this hypothesis is to determine whether light therapy designed to strengthen this intrinsic timing system can delay the disease progression in mouse models. Therefore, we determined the impact of blue wavelength-enriched light on two HD models: the BACHD and Q175 mice. Both models received 6 h of blue-light at the beginning of their daily light cycle for 3 months. After treatment, both genotypes showed improvements in their locomotor activity rhythm without significant change to their sleep behavior. Critically, treated mice of both lines exhibited improved motor performance compared to untreated controls. Focusing on the Q175 genotype, we sought to determine whether the treatment altered signaling pathways in brain regions known to be impacted by HD using NanoString gene expression assays. We found that the expression of several HD relevant markers was altered in the striatum and cortex of the treated mice. Our study demonstrates that strengthening the circadian system can delay the progression of HD in pre-clinical models. This work suggests that lighting conditions should be considered when managing treatment of HD and other neurodegenerative disorders.
Collapse
Key Words
- BACHD
- BACHD, bacterial artificial chromosome mouse model of HD
- Blue light therapy
- Circadian rhythms
- HD, Huntington's disease
- HTT, Huntingtin protein
- Htt, huntingtin gene
- Huntington's disease
- KI, knock in
- Photic therapy
- Q175
- SCN, suprachiasmatic nucleus
- UCLA, University of California, Los Angeles
- ZT, Zeitgeber time
- ipRGCs, intrinsically photoreceptive retinal ganglion cells
Collapse
Affiliation(s)
- Huei-Bin Wang
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | - Daniel S. Whittaker
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | - Danny Truong
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | - Aly K. Mulji
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
- Integrative Biology and Physiology, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | - Cristina A. Ghiani
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
- Department of Pathology, Laboratory of Circadian and Sleep Medicine, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | - Dawn H. Loh
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| | - Christopher S. Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California - Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90024-1759, USA
| |
Collapse
|
25
|
Chao MW, Chen CP, Yang YH, Chuang YC, Chu TY, Tseng CY. N-acetylcysteine attenuates lipopolysaccharide-induced impairment in lamination of Ctip2-and Tbr1- expressing cortical neurons in the developing rat fetal brain. Sci Rep 2016; 6:32373. [PMID: 27577752 PMCID: PMC5006028 DOI: 10.1038/srep32373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/03/2016] [Indexed: 02/02/2023] Open
Abstract
Oxidative stress and inflammatory insults are the major instigating events of bacterial intrauterine infection that lead to fetal brain injury. The purpose of this study is to investigate the remedial effects of N-acetyl-cysteine (NAC) for inflammation-caused deficits in brain development. We found that lipopolysaccharide (LPS) induced reactive oxygen species (ROS) production by RAW264.7 cells. Macrophage-conditioned medium caused noticeable cortical cell damage, specifically in cortical neurons. LPS at 25 μg/kg caused more than 75% fetal loss in rats. An increase in fetal cortical thickness was noted in the LPS-treated group. In the enlarged fetal cortex, laminar positioning of the early born cortical cells expressing Tbr1 and Ctip2 was disrupted, with a scattered distribution. The effect was similar, but minor, in later born Satb2-expressing cortical cells. NAC protected against LPS-induced neuron toxicity in vitro and counteracted pregnancy loss and alterations in thickness and lamination of the neocortex in vivo. Fetal loss and abnormal fetal brain development were due to LPS-induced ROS production. NAC is an effective protective agent against LPS-induced damage. This finding highlights the key therapeutic impact of NAC in LPS-caused abnormal neuronal laminar distribution during brain development.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Chie-Pein Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Hsiu Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Tzu-Yun Chu
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- International Master Program of Biomedical Material and Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- Center for Nano-Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| |
Collapse
|
26
|
Lee JH, Espinera AR, Chen D, Choi KE, Caslin AY, Won S, Pecoraro V, Xu GY, Wei L, Yu SP. Neonatal inflammatory pain and systemic inflammatory responses as possible environmental factors in the development of autism spectrum disorder of juvenile rats. J Neuroinflammation 2016; 13:109. [PMID: 27184741 PMCID: PMC4867541 DOI: 10.1186/s12974-016-0575-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/06/2016] [Indexed: 02/28/2023] Open
Abstract
Background Autism spectrum disorder (ASD) affects many children and juveniles. The pathogenesis of ASD is not well understood. Environmental factors may play important roles in the development of ASD. We examined a possible relationship of inflammatory pain in neonates and the development of ASD in juveniles. Methods Acute inflammation pain was induced by 5 % formalin (5 μl/day) subcutaneous injection into two hindpaws of postnatal day 3 to 5 (P3–P5) rat pups. Western blot, immunohistochemical, and behavioral examinations were performed at different time points after the insult. Results Formalin injection caused acute and chronic inflammatory responses including transient local edema, increased levels of inflammatory cytokines, TNF-α, and IL-1β in the blood as well as in the brain, and increased microglia in the brain. One day after the pain insult, there was significant cell death in the cortex and hippocampus. Two weeks later, although the hindpaw local reaction subsided, impaired axonal growth and demyelization were seen in the brain of P21 juvenile rats. The number of bromodeoxyuridine (BrdU) and doublecortin (DCX) double-positive cells in the hippocampal dentate gyrus of P21 rats was significantly lower than that in controls, indicating reduced neurogenesis. In the P21 rat’s brain of the formalin group, the expression of autism-related gene neurexin 1 (NRXN1), fragile X mental retardation 1 (FMR1), and oxytocin was significantly downregulated, consistent with the gene alteration in ASD. Juvenile rats in the formalin group showed hyperalgesia, repetitive behaviors, abnormal locomotion, sleep disorder, and distinct deficits in social memory and social activities. These alterations in neuroinflammatory reactions, gene expression, and behaviors were more evident in male than in female rats. Importantly, an anti-inflammation treatment using indomethacin (10 mg/kg, i.p.) at the time of formalin injections suppressed inflammatory responses and neuronal cell death and prevented alterations in ASD-related genes and the development of abnormal behaviors. Conclusions These novel observations indicate that severe inflammatory pain in neonates and persistent inflammatory reactions may predispose premature infants to development delays and psychiatric disorders including ASD. The prevention of pain stimuli and prompt treatments of inflammation during development appear vitally important in disrupting possible evolution of ASD syndromes. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0575-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alyssa R Espinera
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dongdong Chen
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,The Laboratory of Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ko-Eun Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Asha Yoshiko Caslin
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Soonmi Won
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Valentina Pecoraro
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Guang-Yin Xu
- The Laboratory of Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Atlanta, GA, 30033, USA. .,Emory University School of Medicine, 101 Woodruff Circle, WMB Suite 620, Atlanta, GA, 30322, USA.
| |
Collapse
|
27
|
Toll-like receptor 4-mediated immune stress in pregnant rats activates STAT3 in the fetal brain: role of interleukin-6. Pediatr Res 2016; 79:781-7. [PMID: 25938734 DOI: 10.1038/pr.2015.86] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/28/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prenatal exposure to pathogens induces long lasting effect on brain function and plasticity. It is unclear how maternal immune stress impacts fetal brain development. Immune challenged pregnant rats induce the production of inflammatory cytokines including tumor necrosis factor (TNF)α, interleukin (IL)1β, and IL-6. IL-6 crosses the placenta but its mechanism of action on fetal brain is unclear. METHODS Gestation day 15 (GD15) rats were given a single injection of lipopolysaccharide (LPS) (100 µg/kg) in the presence or the absence of an IL-6 neutralizing antibody (IL-6Ab, 10 µg/kg). The activation of the intracellular signal of IL-6; signal transducer and activator of transcription (STAT3) and levels of glucocorticoids (GCs) were monitored in fetal brains. RESULTS LPS administration to GD15 rats significantly increased the phosphorylation levels of STAT3 in fetal brains. Such activation was blunted by IL-6Ab. LPS induced a significant rise in GCs in the plasma of dams but not in fetal brains. IL-6Ab significantly reduced LPS-induced GCs in maternal plasma. CONCLUSION Toll-like receptor 4 (TLR4)-induced activation of the maternal innate immune system affects fetal brains likely via the mobilization of IL-6/STAT3 pathway. In contrast, TLR4-stimulated maternal GCs release is less likely to play a significant role in fetal brain development.
Collapse
|
28
|
Imai K, Kotani T, Tsuda H, Mano Y, Nakano T, Ushida T, Li H, Miki R, Sumigama S, Iwase A, Hirakawa A, Ohno K, Toyokuni S, Takeuchi H, Mizuno T, Suzumura A, Kikkawa F. Neuroprotective potential of molecular hydrogen against perinatal brain injury via suppression of activated microglia. Free Radic Biol Med 2016; 91:154-63. [PMID: 26709014 DOI: 10.1016/j.freeradbiomed.2015.12.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 12/15/2022]
Abstract
Exposure to inflammation in utero is related to perinatal brain injury, which is itself associated with high rates of long-term morbidity and mortality in children. Novel therapeutic interventions during the perinatal period are required to prevent inflammation, but its pathogenesis is incompletely understood. Activated microglia are known to play a central role in brain injury by producing a variety of pro-inflammatory cytokines and releasing oxidative products. The study is aimed to investigate the preventative potential of molecular hydrogen (H2), which is an antioxidant and anti-inflammatory agent without mutagenicity. Pregnant ICR mice were injected with lipopolysaccharide (LPS) intraperitoneally on embryonic day 17 to create a model of perinatal brain injury caused by prenatal inflammation. In this model, the effect of maternal administration of hydrogen water (HW) on pups was also evaluated. The levels of pro-inflammatory cytokines, oxidative damage and activation of microglia were determined in the fetal brains. H2 reduced the LPS-induced expression of pro-inflammatory cytokines, oxidative damage and microglial activation in the fetal brains. Next, we investigated how H2 contributes to neuroprotection, focusing on microglia, using primary cultured microglia and neurons. H2 prevented LPS- or cytokine-induced generation of reactive oxidative species by microglia and reduced LPS-induced microglial neurotoxicity. Finally, we identified several molecules influenced by H2, involved in the process of activating microglia. These results suggested that H2 holds promise for the prevention of inflammation related to perinatal brain injury.
Collapse
Affiliation(s)
- Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Hiroyuki Tsuda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yukio Mano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomoko Nakano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hua Li
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Rika Miki
- Laboratory of Bell Research Centre-Department of Obstetrics and Gynecology Collaborative Research, Bell Research Centre for Reproductive Health and Cancer, Department of Reproduction, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Seiji Sumigama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akihiro Hirakawa
- Biostatistics Section, Center for Advanced Medicine and Clinical Research Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
29
|
Smit AL, Lambermont VA, Stokroos RJ, Anteunis LJC, Chenault MN, Schaefer SM, Schoenmakers LWG, Kremer B, Kramer BW. Intrauterine Lipopolysaccharide-Induced Chorioamnionitis in a Sheep: Does It Affect the Auditory System? Reprod Sci 2015; 23:257-63. [PMID: 26702124 DOI: 10.1177/1933719115602759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Fetal exposure to in utero inflammation such as chorioamnionitis is related to central nervous system injury. We hypothesized that chorioamnionitis can provoke inflammatory changes in the perilymph and alter hearing outcome. METHODS Pregnant ewes were randomized into 2 groups: intrauterine injection with lipopolysaccharide (LPS; n = 19) or saline (n = 21). In the first experiment, fetal perilymph samples were taken for cytokine analysis. In the second experiment, consecutive bone-conducted auditory brain stem responses were obtained from 1 to 7 months after birth. RESULTS Perilymph samples showed a significant elevation in interleukin 8 in the LPS group. Auditory brain stem response analysis demonstrated higher response thresholds and a prolongation of absolute peak V and interpeak intervals I to V and III to V in the LPS group compared to sham treatment. CONCLUSION Our study confirms the hypothesis that an intrauterine inflammation by LPS can result in a fetal perilymphatic inflammatory response and functional impaired hearing outcomes after birth in a sheep model.
Collapse
Affiliation(s)
- Adriana L Smit
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Verena A Lambermont
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Robert J Stokroos
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Lucien J C Anteunis
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Michelene N Chenault
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands Department of Methodology and Statistics, Maastricht University, Maastricht, the Netherlands
| | - Simone M Schaefer
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Luke W G Schoenmakers
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Bernd Kremer
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands School of Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Boris W Kramer
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands Department of Pediatrics, Maastricht University Medical Centre, Maastricht, the Netherlands School of Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
30
|
Loh DH, Jami SA, Flores RE, Truong D, Ghiani CA, O'Dell TJ, Colwell CS. Misaligned feeding impairs memories. eLife 2015; 4. [PMID: 26652002 PMCID: PMC4729691 DOI: 10.7554/elife.09460] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/19/2015] [Indexed: 01/23/2023] Open
Abstract
Robust sleep/wake rhythms are important for health and cognitive function. Unfortunately, many people are living in an environment where their circadian system is challenged by inappropriate meal- or work-times. Here we scheduled food access to the sleep time and examined the impact on learning and memory in mice. Under these conditions, we demonstrate that the molecular clock in the master pacemaker, the suprachiasmatic nucleus (SCN), is unaltered while the molecular clock in the hippocampus is synchronized by the timing of food availability. This chronic circadian misalignment causes reduced hippocampal long term potentiation and total CREB expression. Importantly this mis-timed feeding resulted in dramatic deficits in hippocampal-dependent learning and memory. Our findings suggest that the timing of meals have far-reaching effects on hippocampal physiology and learned behaviour. DOI:http://dx.doi.org/10.7554/eLife.09460.001 Many processes within the body follow an approximately 24-hour cycle. In addition to patterns of sleep and wakefulness, such circadian rhythms help to regulate body temperature, blood pressure and hormone levels. They also affect when we feel hungry, when our muscles work most efficiently, and when we are mentally at our sharpest. A region of the brain called the suprachiasmatic nucleus (SCN) generates and maintains circadian rhythms, and thus acts as the body’s master clock. Daily exposure to light keeps the SCN synchronized with the 24-hour day/night cycle. However, most organs, from the heart to the pancreas, also possess their own clocks, which help to regulate organ-specific processes. These secondary clocks normally operate in synchrony with the SCN. Exposure to light has long been known to influence circadian rhythms. However, more recent evidence suggests that the timing of meals may also affect circadian clocks, particularly those within the digestive system. Loh et al. therefore decided to investigate whether eating outside normal waking hours would also affect other key physiological processes, specifically the cognitive processes of learning and memory. Mice normally consume most of their food after sunset. Loh et al. showed that rodents that were instead fed during the day performed less well on cognitive tests than other mice who received the same food at night. The daytime-fed mice showed changes in a region of the brain called the hippocampus, which supports learning and memory. In particular, daytime feeding changed the timing of the secondary circadian clock within the hippocampus, although it had no effect on the master clock in the SCN. Loh et al. therefore suggest that the misalignment of these circadian clocks impairs cognition. Further experiments are needed to determine whether a similar relationship exists between the timing of meals and cognitive performance in humans. If so, these findings will have implications for the many individuals whose mealtimes, for work or social reasons, are out of synchrony with their body clocks. DOI:http://dx.doi.org/10.7554/eLife.09460.002
Collapse
Affiliation(s)
- Dawn H Loh
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,UCLA Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, United States
| | - Shekib A Jami
- UCLA Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, United States.,Molecular, Cellular, and Integrative Physiology PhD Program, University of California, Los Angeles, Los Angeles, United States
| | - Richard E Flores
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Danny Truong
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Cristina A Ghiani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Thomas J O'Dell
- UCLA Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, United States.,Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,UCLA Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
31
|
Yin P, Zhang XT, Li J, Yu L, Wang JW, Lei GF, Sun RP, Li BM. Maternal immune activation increases seizure susceptibility in juvenile rat offspring. Epilepsy Behav 2015; 47:93-7. [PMID: 25982885 DOI: 10.1016/j.yebeh.2015.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 11/15/2022]
Abstract
Epidemiological data suggest a relationship between maternal infection and a high incidence of childhood epilepsy in offspring. However, there is little experimental evidence that links maternal infection with later seizure susceptibility in juvenile offspring. Here, we asked whether maternal immune challenge during pregnancy can alter seizure susceptibility and seizure-associated brain damage in adolescence. Pregnant Sprague-Dawley rats were treated with lipopolysaccharide (LPS) or normal saline (NS) on gestational days 15 and 16. At postnatal day 21, seizure susceptibility to kainic acid (KA) was evaluated in male offspring. Four groups were studied, including normal control (NS-NS), prenatal infection (LPS-NS), juvenile seizure (NS-KA), and "two-hit" (LPS-KA) groups. Our results demonstrated that maternal LPS exposure caused long-term reactive astrogliosis and increased seizure susceptibility in juvenile rat offspring. Compared to the juvenile seizure group, animals in the "two-hit" group showed exaggerated astrogliosis, followed by worsened spatial learning ability in adulthood. In addition, prenatal immune challenge alone led to spatial learning impairment in offspring but had no effect on anxiety. These data suggest that prenatal immune challenge causes a long-term increase in juvenile seizure susceptibility and exacerbates seizure-induced brain injury, possibly by priming astroglia.
Collapse
Affiliation(s)
- Ping Yin
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China
| | - Xin-Ting Zhang
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China
| | - Jun Li
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China
| | - Lin Yu
- Women's Hospital, School of Medicine, Zhejiang University, China
| | - Ji-Wen Wang
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China
| | - Ge-Fei Lei
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China
| | - Ruo-Peng Sun
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China
| | - Bao-Min Li
- Pediatric Department of Qilu Hospital, Shandong University, Jinan, China; Brain Science Research Institute, Shandong University, Jinan, China.
| |
Collapse
|
32
|
Missault S, Van den Eynde K, Vanden Berghe W, Fransen E, Weeren A, Timmermans JP, Kumar-Singh S, Dedeurwaerdere S. The risk for behavioural deficits is determined by the maternal immune response to prenatal immune challenge in a neurodevelopmental model. Brain Behav Immun 2014; 42:138-46. [PMID: 24973728 DOI: 10.1016/j.bbi.2014.06.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 06/06/2014] [Accepted: 06/16/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Schizophrenia is a highly disabling psychiatric disorder with a proposed neurodevelopmental basis. One mechanism through which genetic and environmental risk factors might act is by triggering persistent brain inflammation, as evidenced by long-lasting neuro-immunological disturbances in patients. Our goal was to investigate whether microglia activation is a neurobiological correlate to the altered behaviour in the maternal immune activation (MIA) model, a well-validated animal model with relevance to schizophrenia. A recent observation in the MIA model is the differential maternal body weight response to the immune stimulus, correlated with a different behavioural outcome in the offspring. Although it is generally assumed that the differences in maternal weight response reflect differences in cytokine response, this has not been investigated so far. Our aim was to investigate whether (i) the maternal weight response to MIA reflects differences in the maternal cytokine response, (ii) the differential behavioural phenotype of the offspring extends to depressive symptoms such as anhedonia and (iii) there are changes in chronic microglia activation dependent on the behavioural phenotype. METHODS Based on a dose-response study, MIA was induced in pregnant rats by injecting 4mg/kg Poly I:C at gestational day 15. Serum samples were collected to assess the amount of TNF-α in the maternal blood following MIA. MIA offspring were divided into weight loss (WL; n=14) and weight gain (WG; n=10) groups, depending on the maternal body weight response to Poly I:C. Adult offspring were behaviourally phenotyped for prepulse inhibition, locomotor activity with and without amphetamine and MK-801 challenge, and sucrose preference. Finally, microglia activation was scored on CD11b- and Iba1-immunohistochemically stained sections. RESULTS Pregnant dams that lost weight following MIA showed increased levels of TNF-α compared to controls, unlike dams that gained weight following MIA. Poly I:C WL offspring showed the most severe behavioural outcome. Poly I:C WG offspring, on the other hand, did not show clear behavioural deficits. Most interestingly a reduced sucrose preference indicative of anhedonia was found in Poly I:C WL but not Poly I:C WG offspring compared to controls. Finally, there were no significant differences in microglia activation scores between any of the investigated groups. CONCLUSIONS The individual maternal immune response to MIA is an important determinant of the behavioural outcome in offspring, including negative symptoms such as anhedonia. We failed to find any significant difference in the level of microglia activation between Poly I:C WL, Poly I:C WG and control offspring.
Collapse
Affiliation(s)
- S Missault
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - K Van den Eynde
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - W Vanden Berghe
- Laboratory of Protein Science, Proteomics & Epigenetic Signaling, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - E Fransen
- StatUA, University of Antwerp, Campus Drie Eiken, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
| | - A Weeren
- StatUA, University of Antwerp, City Campus, Prinsstraat 13, 2000 Antwerpen, Belgium
| | - J P Timmermans
- Laboratory of Cell Biology & Histology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - S Kumar-Singh
- Laboratory of Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - S Dedeurwaerdere
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| |
Collapse
|
33
|
Prenatal lipopolysaccharide disrupts maternal behavior, reduces nest odor preference in pups, and induces anxiety: Studies of F1 and F2 generations. Eur J Pharmacol 2014; 738:342-51. [DOI: 10.1016/j.ejphar.2014.05.058] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 05/11/2014] [Accepted: 05/23/2014] [Indexed: 11/19/2022]
|
34
|
Rose KM, Parmar MS, Cavanaugh JE. Dietary supplementation with resveratrol protects against striatal dopaminergic deficits produced by in utero LPS exposure. Brain Res 2014; 1573:37-43. [DOI: 10.1016/j.brainres.2014.05.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 05/14/2014] [Accepted: 05/16/2014] [Indexed: 01/08/2023]
|
35
|
Green HF, Nolan YM. Inflammation and the developing brain: Consequences for hippocampal neurogenesis and behavior. Neurosci Biobehav Rev 2014; 40:20-34. [DOI: 10.1016/j.neubiorev.2014.01.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 01/12/2014] [Accepted: 01/13/2014] [Indexed: 02/06/2023]
|
36
|
Prenatal immune challenge in rats increases susceptibility to seizure-induced brain injury in adulthood. Brain Res 2013; 1519:78-86. [DOI: 10.1016/j.brainres.2013.04.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/23/2013] [Accepted: 04/23/2013] [Indexed: 01/22/2023]
|
37
|
Folsom TD, Fatemi SH. The involvement of Reelin in neurodevelopmental disorders. Neuropharmacology 2013; 68:122-35. [PMID: 22981949 PMCID: PMC3632377 DOI: 10.1016/j.neuropharm.2012.08.015] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 08/14/2012] [Accepted: 08/16/2012] [Indexed: 12/21/2022]
Abstract
Reelin is a glycoprotein that serves important roles both during development (regulation of neuronal migration and brain lamination) and in adulthood (maintenance of synaptic function). A number of neuropsychiatric disorders including autism, schizophrenia, bipolar disorder, major depression, Alzheimer's disease and lissencephaly share a common feature of abnormal Reelin expression in the brain. Altered Reelin expression has been hypothesized to impair neuronal connectivity and synaptic plasticity, leading ultimately to the cognitive deficits present in these disorders. The mechanisms for abnormal Reelin expression in some of these disorders are currently unknown although possible explanations include early developmental insults, mutations, hypermethylation of the promoter for the Reelin gene (RELN), miRNA silencing of Reelin mRNA, FMRP underexpression and Reelin processing abnormalities. Increasing Reelin expression through pharmacological therapies may help ameliorate symptoms resulting from Reelin deficits. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Timothy D. Folsom
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
| | - S. Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
- Department of Pharmacology, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA
| |
Collapse
|
38
|
Soto AM, Kirsten TB, Reis-Silva TM, Martins MF, Teodorov E, Flório JC, Palermo-Neto J, Bernardi MM, Bondan EF. Single early prenatal lipopolysaccharide exposure impairs striatal monoamines and maternal care in female rats. Life Sci 2013; 92:852-8. [DOI: 10.1016/j.lfs.2013.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/19/2013] [Accepted: 03/02/2013] [Indexed: 11/30/2022]
|
39
|
Xu M, Sulkowski ZL, Parekh P, Khan A, Chen T, Midha S, Iwasaki T, Shimokawa N, Koibuchi N, Zavacki AM, Sajdel-Sulkowska EM. Effects of Perinatal Lipopolysaccharide (LPS) Exposure on the Developing Rat Brain; Modeling the Effect of Maternal Infection on the Developing Human CNS. THE CEREBELLUM 2013; 12:572-86. [DOI: 10.1007/s12311-013-0465-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
40
|
Cai Z, Fan LW, Kaizaki A, Tien LT, Ma T, Pang Y, Lin S, Lin RCS, Simpson KL. Neonatal systemic exposure to lipopolysaccharide enhances susceptibility of nigrostriatal dopaminergic neurons to rotenone neurotoxicity in later life. Dev Neurosci 2013; 35:155-71. [PMID: 23446007 PMCID: PMC3777222 DOI: 10.1159/000346156] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/26/2012] [Indexed: 12/21/2022] Open
Abstract
Brain inflammation via intracerebral injection with lipopolysaccharide (LPS) in early life has been shown to increase risks for the development of neurodegenerative disorders in adult rats. To determine if neonatal systemic LPS exposure has the same effects on enhancement of adult dopaminergic neuron susceptibility to rotenone neurotoxicity as centrally injected LPS does, LPS (2 μg/g body weight) was administered intraperitoneally into postnatal day 5 (P5) rats and when grown to P70, rats were challenged with rotenone, a commonly used pesticide, through subcutaneous minipump infusion at a dose of 1.25 mg/kg/day for 14 days. Systemically administered LPS can penetrate into the neonatal rat brain and cause acute and chronic brain inflammation, as evidenced by persistent increases in IL-1β levels, cyclooxygenase-2 expression and microglial activation in the substantia nigra (SN) of P70 rats. Neonatal LPS exposure resulted in suppression of tyrosine hydroxylase (TH) expression, but not actual death of dopaminergic neurons in the SN, as indicated by the reduced number of TH+ cells and unchanged total number of neurons (NeuN+) in the SN. Neonatal LPS exposure also caused motor function deficits, which were spontaneously recoverable by P70. A small dose of rotenone at P70 induced loss of dopaminergic neurons, as indicated by reduced numbers of both TH+ and NeuN+ cells in the SN, and Parkinson's disease (PD)-like motor impairment in P98 rats that had experienced neonatal LPS exposure, but not in those without the LPS exposure. These results indicate that although neonatal systemic LPS exposure may not necessarily lead to death of dopaminergic neurons in the SN, such an exposure could cause persistent functional alterations in the dopaminergic system and indirectly predispose the nigrostriatal system in the adult brain to be damaged by environmental toxins at an ordinarily nontoxic or subtoxic dose and develop PD-like pathological features and motor dysfunction.
Collapse
Affiliation(s)
- Zhengwei Cai
- Division of Newborn Medicine, Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216-4504, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Stolp HB. Neuropoietic cytokines in normal brain development and neurodevelopmental disorders. Mol Cell Neurosci 2012; 53:63-8. [PMID: 22926235 DOI: 10.1016/j.mcn.2012.08.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/17/2022] Open
Abstract
Inflammation has been implicated in a wide variety of neurological disorders and there is increasing evidence for long-term consequences of inflammation during early brain development. A number of immune mediators, termed neuropoietic cytokines, have a role in normal brain development. Neuropoietic cytokines contribute to proliferation of neural precursors; fate determination and differentiation; migration of neurons and glia; as well as cell survival and activity dependent alteration of synaptic function. Inflammation during development, therefore, may cause widespread injury to the brain by interfering with the normal balance of cytokine signalling and therefore developmental processes. This review will examine the normal role of neuropoietic cytokines and the potential contribution of inflammatory insults to a number of neurodevelopmental disorders. It will also discuss the potential for developmental inflammation to sensitise the brain to later insult, possibly contributing to neurodegenerative disorders later in life. This article is part of a Special Issue entitled 'Neuroinflammation in neurodegeneration and neurodysfunction'.
Collapse
Affiliation(s)
- H B Stolp
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QT, UK.
| |
Collapse
|
42
|
Oskvig DB, Elkahloun AG, Johnson KR, Phillips TM, Herkenham M. Maternal immune activation by LPS selectively alters specific gene expression profiles of interneuron migration and oxidative stress in the fetus without triggering a fetal immune response. Brain Behav Immun 2012; 26:623-34. [PMID: 22310921 PMCID: PMC3285385 DOI: 10.1016/j.bbi.2012.01.015] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/12/2012] [Accepted: 01/20/2012] [Indexed: 01/07/2023] Open
Abstract
Maternal immune activation (MIA) is a risk factor for the development of schizophrenia and autism. Infections during pregnancy activate the mother's immune system and alter the fetal environment, with consequential effects on CNS function and behavior in the offspring, but the cellular and molecular links between infection-induced altered fetal development and risk for neuropsychiatric disorders are unknown. We investigated the immunological, molecular, and behavioral effects of MIA in the offspring of pregnant Sprague-Dawley rats given an intraperitoneal (0.25 mg/kg) injection of lipopolysaccharide (LPS) on gestational day 15. LPS significantly elevated pro-inflammatory cytokine levels in maternal serum, amniotic fluid, and fetal brain at 4 h, and levels decreased but remained elevated at 24 h. Offspring born to LPS-treated dams exhibited reduced social preference and exploration behaviors as juveniles and young adults. Whole genome microarray analysis of the fetal brain at 4 h post maternal LPS was performed to elucidate the possible molecular mechanisms by which MIA affects the fetal brain. We observed dysregulation of 3285 genes in restricted functional categories, with increased mRNA expression of cellular stress and cell death genes and reduced expression of developmentally-regulated and brain-specific genes, specifically those that regulate neuronal migration of GABAergic interneurons, including the Distal-less (Dlx) family of transcription factors required for tangential migration from progenitor pools within the ganglionic eminences into the cerebral cortex. Our results provide a novel mechanism by which MIA induces the widespread down-regulation of critical neurodevelopmental genes, including those previously associated with autism.
Collapse
Affiliation(s)
- Devon B. Oskvig
- Section on Functional Neuroanatomy, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Abdel G. Elkahloun
- Division of Intramural Research Programs Microarray Core Facility, NIH, Bethesda, MD, 20892 USA
| | - Kory R. Johnson
- Bioinformatics Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892 USA
| | - Terry M. Phillips
- Ultramicro Immunodiagnostics Section, Laboratory of Bioengineering and Physical Science, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, MD, 20892, USA
| | - Miles Herkenham
- Section on Functional Neuroanatomy, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA,Corresponding Author: Address: Bldg. 35, Rm. 1C913, Bethesda, MD 20892-3724, USA. (M. Herkenham)
| |
Collapse
|