101
|
Inflammation in pediatric epilepsies: Update on clinical features and treatment options. Epilepsy Behav 2022; 131:107959. [PMID: 33867302 DOI: 10.1016/j.yebeh.2021.107959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023]
Abstract
The role of inflammation is increasingly recognized in triggering or sustaining epileptic activity. In the last decades, increasing research has provided definite evidence to support the link between immunity, inflammatory process, and epilepsy. Neuro- and systemic inflammation play a pivotal role in driving epileptogenesis through different pathogenetic mechanisms: the activation of innate immunity in glia, neurons, and microvasculature, the brain mediated by blood-brain barrier (BBB) impairment, and the imbalance of pro- and anti-inflammatory molecules produced by both arms of immunity. More recently, research has focused on the adverse effects of maternal or early-life immune activation and cytokine imbalance on fetal neurodevelopment and postnatal epilepsy. A complex crosstalk between the immune and nervous system, and a crucial interplay of genetic, epigenetic, and environmental factors may influence structures and functions of the developing brain. A better understanding of the inflammatory process in promoting epilepsy implies that targeting specific pathways may be effective in seizure control. Multiple targets have been identified so far, and several antiseizure interventions are obtained by inhibiting inflammatory signaling or protecting/restoring BBB. All this evidence has changed the field of epilepsy research and neuropharmacology. Further developments and new treatments will rapidly emerge to improve seizure management in inflammation-related epilepsies. This article is part of the Special Issue "Severe Infantile Epilepsies".
Collapse
|
102
|
Brynge M, Gardner RM, Sjöqvist H, Lee BK, Dalman C, Karlsson H. Maternal Levels of Cytokines in Early Pregnancy and Risk of Autism Spectrum Disorders in Offspring. Front Public Health 2022; 10:917563. [PMID: 35712277 PMCID: PMC9197505 DOI: 10.3389/fpubh.2022.917563] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/04/2022] [Indexed: 11/24/2022] Open
Abstract
Previous studies indicate a role of immune disturbances during early development in the etiology of autism spectrum disorders (ASD). Any potential disturbances during fetal development are best addressed by prospective evaluation of maternal markers of inflammation. Previous studies have investigated maternal cytokines, a group of powerful effectors of the immune system, with inconsistent results. In this study, we aimed to clarify the relationship between maternal cytokines and ASD by evaluating levels of 17 cytokines in first trimester maternal serum samples, from 318 mothers to ASD-cases and 429 mothers to ASD-unaffected controls, nested within the register-based Stockholm Youth Cohort. Overall, we observed no consistent associations between levels of maternal cytokines and ASD. While we observed a number of individual associations, the patterns varied across the diagnostic sub-groups. Levels above the 90th percentile of IL-1β (OR = 2.31, 95% CI 1.16-4.60), IL-7 (OR = 2.28, 95% CI 1.20-4.33), IL-13 (OR = 2.42, 95% CI 1.29-4.55), and MCP-1 (OR = 2.09, 95% CI 1.03-4.24) were associated with increased odds of ASD with co-occurring intellectual disability (ID), whereas GMCSF (OR = 2.06, 95% CI 1.03-4.11) and TNF-α (OR = 2.31, 95% CI 1.18-4.50) were associated with increased odds of ASD with ADHD but none survived correction for multiple comparisons. Also, none of the measured maternal cytokines were associated with ASD without co-occurring ID or ADHD. Implementing a data-driven approach using machine learning (Random Forest's Variable Importance measurement), we found no evidence to suggest that adding these cytokines and other markers of maternal immunity, to register-based maternal factors (e.g., psychiatric history) improves prediction of ASD. In summary, we found no robust evidence of an association between maternal immune markers during early pregnancy and ASD.
Collapse
Affiliation(s)
- Martin Brynge
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Renee M. Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Hugo Sjöqvist
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Brian K. Lee
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Department of Epidemiology and Biostatistics, Drexel University School of Public Health, Philadelphia, PA, United States
- A.J. Drexel Autism Institute, Philadelphia, PA, United States
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
103
|
Maternal Prenatal Inflammation Increases Brain Damage Susceptibility of Lipopolysaccharide in Adult Rat Offspring via COX-2/PGD-2/DPs Pathway Activation. Int J Mol Sci 2022; 23:ijms23116142. [PMID: 35682823 PMCID: PMC9181626 DOI: 10.3390/ijms23116142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
A growing body of research suggests that inflammatory insult contributes to the etiology of central nervous system diseases, such as depression, Alzheimer’s disease, and so forth. However, the effect of prenatal systemic inflammation exposure on offspring brain development and cerebral susceptibility to inflammatory insult remains unknown. In this study, we utilized the prenatal inflammatory insult model in vivo and the neuronal damage model in vitro. The results obtained show that prenatal maternal inflammation exacerbates LPS-induced memory impairment, neuronal necrosis, brain inflammatory response, and significantly increases protein expressions of COX-2, DP2, APP, and Aβ, while obviously decreasing that of DP1 and the exploratory behaviors of offspring rats. Meloxicam significantly inhibited memory impairment, neuronal necrosis, oxidative stress, and inflammatory response, and down-regulated the expressions of APP, Aβ, COX-2, and DP2, whereas significantly increased exploring behaviors and the expression of DP1 in vivo. Collectively, these findings suggested that maternal inflammation could cause offspring suffering from inflammatory and behavioral disorders and increase the susceptibility of offspring to cerebral pathological factors, accompanied by COX-2/PGD-2/DPs pathway activation, which could be ameliorated significantly by COX-2 inhibitor meloxicam treatment.
Collapse
|
104
|
Ciliary neurotrophic factor is increased in the plasma of patients with obesity and its levels correlate with diabetes and inflammation indices. Sci Rep 2022; 12:8331. [PMID: 35585213 PMCID: PMC9117681 DOI: 10.1038/s41598-022-11942-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/22/2022] [Indexed: 11/09/2022] Open
Abstract
To establish whether obesity involves activation of endogenous ciliary neurotrophic factor (CNTF) signalling, we evaluated its plasma levels in patients with obesity and correlated its values with the major clinical and haematological indices of obesity, insulin resistance and systemic inflammation. This study involved 118 subjects: 39 healthy controls (19 men), 39 subjects with obesity (19 men) and 40 subjects with obesity and diabetes (20 men). Plasma CNTF and CNTF receptor α (CNTFRα) were measured using commercial ELISA kits. The results showed that plasma CNTF was significantly higher in males and females with obesity with and without diabetes than in healthy subjects. Women consistently exhibited higher levels of circulating CNTF. In both genders, CNTF levels correlated significantly and positively with obesity (BMI, WHR, leptin), diabetes (fasting insulin, HOMA index and HbA1c) and inflammation (IL-6 and hsCRP) indices. Circulating CNTFRα and the CNTF/CNTFRα molar ratio tended to be higher in the patient groups than in controls. In conclusion, endogenous CNTF signalling is activated in human obesity and may help counteract some adverse effects of obesity. Studies involving a higher number of selected patients may reveal circulating CNTF and/or CNTFRα as potential novel diagnostic and/or prognostic markers of obesity, diabetes and associated diseases.
Collapse
|
105
|
I SP, I GD, L B, M S, J GR, A M EO, I M AB, C LM, E M PV, J A A, E B, J L V, R M DP, R R. The Absence of Caspase-8 in the Dopaminergic System Leads to Mild Autism-like Behavior. Front Cell Dev Biol 2022; 10:839715. [PMID: 35493109 PMCID: PMC9045412 DOI: 10.3389/fcell.2022.839715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
In the last decade, new non-apoptotic roles have been ascribed to apoptotic caspases. This family of proteins plays an important role in the sculpting of the brain in the early stages of development by eliminating excessive and nonfunctional synapses and extra cells. Consequently, impairments in this process can underlie many neurological and mental illnesses. This view is particularly relevant to dopamine because it plays a pleiotropic role in motor control, motivation, and reward processing. In this study, we analyze the effects of the elimination of caspase-8 (CASP8) on the development of catecholaminergic neurons using neurochemical, ultrastructural, and behavioral tests. To do this, we selectively delete the CASP8 gene in cells that express tyrosine hydroxylase with the help of recombination through the Cre-loxP system. Our results show that the number of dopaminergic neurons increases in the substantia nigra. In the striatum, the basal extracellular level of dopamine and potassium-evoked dopamine release decreased significantly in mice lacking CASP8, clearly showing the low dopamine functioning in tissues innervated by this neurotransmitter. This view is supported by electron microscopy analysis of striatal synapses. Interestingly, behavioral analysis demonstrates that mice lacking CASP8 show changes reminiscent of autism spectrum disorders (ASD). Our research reactivates the possible role of dopamine transmission in the pathogenesis of ASD and provides a mild model of autism.
Collapse
Affiliation(s)
- Suárez-Pereira I
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Sevilla, Spain.,Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - García-Domínguez I
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Bravo L
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Sevilla, Spain.,Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - Santiago M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - García-Revilla J
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Espinosa-Oliva A M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Alonso-Bellido I M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - López-Martín C
- Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - Pérez-Villegas E M
- Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain
| | - Armengol J A
- Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain
| | - Berrocoso E
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Sevilla, Spain.,Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - Venero J L
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - de Pablos R M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ruiz R
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
106
|
Ryan AM, Bauman MD. Primate Models as a Translational Tool for Understanding Prenatal Origins of Neurodevelopmental Disorders Associated With Maternal Infection. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:510-523. [PMID: 35276404 PMCID: PMC8902899 DOI: 10.1016/j.bpsc.2022.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/13/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023]
Abstract
Pregnant women represent a uniquely vulnerable population during an infectious disease outbreak, such as the COVID-19 pandemic. Although we are at the early stages of understanding the specific impact of SARS-CoV-2 exposure during pregnancy, mounting epidemiological evidence strongly supports a link between exposure to a variety of maternal infections and an increased risk for offspring neurodevelopmental disorders. Inflammatory biomarkers identified from archived or prospectively collected maternal biospecimens suggest that the maternal immune response is the critical link between infection during pregnancy and altered offspring neurodevelopment. This maternal immune activation (MIA) hypothesis has been tested in animal models by artificially activating the immune system during pregnancy and evaluating the neurodevelopmental consequences in MIA-exposed offspring. Although the vast majority of MIA model research is carried out in rodents, the nonhuman primate model has emerged in recent years as an important translational tool. In this review, we briefly summarize human epidemiological studies that have prompted the development of translationally relevant MIA models. We then highlight notable similarities between humans and nonhuman primates, including placental structure, pregnancy physiology, gestational timelines, and offspring neurodevelopmental stages, that provide an opportunity to explore the MIA hypothesis in species more closely related to humans. Finally, we provide a comprehensive review of neurodevelopmental alterations reported in current nonhuman primate models of maternal infection and discuss future directions for this promising area of research.
Collapse
Affiliation(s)
- Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis, Davis, California; California National Primate Research Center, University of California Davis, Davis, California
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis, Davis, California; California National Primate Research Center, University of California Davis, Davis, California.
| |
Collapse
|
107
|
Abstract
Immunity could be viewed as the common factor in neurodevelopmental disorders and cancer. The immune and nervous systems coevolve as the embryo develops. Immunity can release cytokines that activate MAPK signaling in neural cells. In specific embryonic brain cell types, dysregulated signaling that results from germline or embryonic mutations can promote changes in chromatin organization and gene accessibility, and thus expression levels of essential genes in neurodevelopment. In cancer, dysregulated signaling can emerge from sporadic somatic mutations during human life. Neurodevelopmental disorders and cancer share similarities. In neurodevelopmental disorders, immunity, and cancer, there appears an almost invariable involvement of small GTPases (e.g., Ras, RhoA, and Rac) and their pathways. TLRs, IL-1, GIT1, and FGFR signaling pathways, all can be dysregulated in neurodevelopmental disorders and cancer. Although there are signaling similarities, decisive differentiating factors are timing windows, and cell type specific perturbation levels, pointing to chromatin reorganization. Finally, we discuss drug discovery.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Corresponding author
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
108
|
Mitchell AJ, Dunn GA, Sullivan EL. The Influence of Maternal Metabolic State and Nutrition on Offspring Neurobehavioral Development: A Focus on Preclinical Models. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:450-460. [PMID: 34915175 PMCID: PMC9086110 DOI: 10.1016/j.bpsc.2021.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022]
Abstract
The prevalence of both obesity and neurodevelopmental disorders has increased substantially over the last several decades. Early environmental factors, including maternal nutrition and metabolic state during gestation, influence offspring neurodevelopment. Both human and preclinical models demonstrate a link between poor maternal nutrition, altered metabolic state, and risk of behavioral abnormalities in offspring. This review aims to highlight evidence from the current literature connecting maternal nutrition and the associated metabolic changes with neural and behavioral outcomes in the offspring, as well as identify possible mechanisms underlying these neurodevelopmental outcomes. Owing to the highly correlated nature of poor nutrition and obesity in humans, preclinical animal models are important in distinguishing the unique effects of maternal nutrition and metabolic state on offspring brain development. We use a translational lens to highlight results from preclinical animal models of maternal obesogenic diet related to alterations in behavioral and neurodevelopmental outcomes in offspring. Specifically, we aim to highlight results that resemble behavioral phenotypes described in the diagnostic criteria of neurodevelopmental conditions in humans. Finally, we examine the proinflammatory nature of maternal obesity and consumption of a high-fat diet as a mechanism for neurodevelopmental alterations that may alter offspring behavior later in life. It is important that future studies examine potential therapeutic interventions and prevention strategies to interrupt the transgenerational transmission of the disease. Given the tremendous risk to the next generation, changes need to be made to ensure that all pregnant people have access to nutritious food and are informed about the optimal diet for their developing child.
Collapse
Affiliation(s)
- A J Mitchell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon
| | - Geoffrey A Dunn
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Elinor L Sullivan
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Department of Psychiatry, Oregon Health & Science University, Portland, Oregon; Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon; Department of Human Physiology, University of Oregon, Eugene, Oregon.
| |
Collapse
|
109
|
Antibiotic Treatment during Pregnancy Alters Offspring Gut Microbiota in a Sex-Dependent Manner. Biomedicines 2022; 10:biomedicines10051042. [PMID: 35625778 PMCID: PMC9138679 DOI: 10.3390/biomedicines10051042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
This study investigated the effect of antibiotics administered to pregnant dams on offspring gut microbiome composition and metabolic capabilities, and how these changes in the microbiota may influence their immune responses in both the periphery and the brain. We orally administered a broad-spectrum antibiotic (ABX) cocktail consisting of vancomycin 0.5 mg/mL, ampicillin 1 mg/mL, and neomycin 1 mg/mL to pregnant dams during late gestation through birth. Bacterial DNA was extracted from offspring fecal samples, and 16S ribosomal RNA gene was sequenced by Illumina, followed by analysis of gut microbiota composition and PICRUSt prediction. Serum and brain tissue cytokine levels were analyzed by Luminex. Our results indicate that the ABX-cocktail led to significant diversity and taxonomic changes to the offspring's gut microbiome. In addition, the predicted KEGG and MetaCyc pathways were significantly altered in the offspring. Finally, there were decreased innate inflammatory cytokines and chemokines and interleukin (IL)-17 seen in the brains of ABX-cocktail offspring in response to lipopolysaccharide (LPS) immune challenge. Our results suggest that maternal ABX can produce long-lasting effects on the gut microbiome and neuroimmune responses of offspring. These findings support the role of the early microbiome in the development of offspring gastrointestinal and immune systems.
Collapse
|
110
|
Molecular Signature of Neuroinflammation Induced in Cytokine-Stimulated Human Cortical Spheroids. Biomedicines 2022; 10:biomedicines10051025. [PMID: 35625761 PMCID: PMC9138619 DOI: 10.3390/biomedicines10051025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/04/2022] Open
Abstract
Crucial in the pathogenesis of neurodegenerative diseases is the process of neuroinflammation that is often linked to the pro-inflammatory cytokines Tumor necrosis factor alpha (TNFα) and Interleukin-1beta (IL-1β). Human cortical spheroids (hCSs) constitute a valuable tool to study the molecular mechanisms underlying neurological diseases in a complex three-dimensional context. We recently designed a protocol to generate hCSs comprising all major brain cell types. Here we stimulate these hCSs for three time periods with TNFα and with IL-1β. Transcriptomic analysis reveals that the main process induced in the TNFα- as well as in the IL-1β-stimulated hCSs is neuroinflammation. Central in the neuroinflammatory response are endothelial cells, microglia and astrocytes, and dysregulated genes encoding cytokines, chemokines and their receptors, and downstream NFκB- and STAT-pathway components. Furthermore, we observe sets of neuroinflammation-related genes that are specifically modulated in the TNFα-stimulated and in the IL-1β-stimulated hCSs. Together, our results help to molecularly understand human neuroinflammation and thus a key mechanism of neurodegeneration.
Collapse
|
111
|
Dash S, Syed YA, Khan MR. Understanding the Role of the Gut Microbiome in Brain Development and Its Association With Neurodevelopmental Psychiatric Disorders. Front Cell Dev Biol 2022; 10:880544. [PMID: 35493075 PMCID: PMC9048050 DOI: 10.3389/fcell.2022.880544] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome has a tremendous influence on human physiology, including the nervous system. During fetal development, the initial colonization of the microbiome coincides with the development of the nervous system in a timely, coordinated manner. Emerging studies suggest an active involvement of the microbiome and its metabolic by-products in regulating early brain development. However, any disruption during this early developmental process can negatively impact brain functionality, leading to a range of neurodevelopment and neuropsychiatric disorders (NPD). In this review, we summarize recent evidence as to how the gut microbiome can influence the process of early human brain development and its association with major neurodevelopmental psychiatric disorders such as autism spectrum disorders, attention-deficit hyperactivity disorder, and schizophrenia. Further, we discuss how gut microbiome alterations can also play a role in inducing drug resistance in the affected individuals. We propose a model that establishes a direct link of microbiome dysbiosis with the exacerbated inflammatory state, leading to functional brain deficits associated with NPD. Based on the existing research, we discuss a framework whereby early diet intervention can boost mental wellness in the affected subjects and call for further research for a better understanding of mechanisms that govern the gut-brain axis may lead to novel approaches to the study of the pathophysiology and treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Somarani Dash
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Yasir Ahmed Syed
- School of Biosciences and Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Cardiff, United Kingdom
| | - Mojibur R. Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, India
- *Correspondence: Mojibur R. Khan,
| |
Collapse
|
112
|
Zhao Q, Dai W, Chen HY, Jacobs RE, Zlokovic BV, Lund BT, Montagne A, Bonnin A. Prenatal disruption of blood-brain barrier formation via cyclooxygenase activation leads to lifelong brain inflammation. Proc Natl Acad Sci U S A 2022; 119:e2113310119. [PMID: 35377817 PMCID: PMC9169666 DOI: 10.1073/pnas.2113310119] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic–polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood–brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte–endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qiuying Zhao
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Weiye Dai
- Master of Science, Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, Los Angeles, CA 90089
| | - Hui Yu Chen
- Master of Medical Physiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Russell E. Jacobs
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Brett T. Lund
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, EH16 4SB Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, Edinburgh BioQuarter, EH16 4SB Edinburgh, United Kingdom
| | - Alexandre Bonnin
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
113
|
Michel V, Berk J, Bozakova N, van der Eijk J, Estevez I, Mircheva T, Relic R, Rodenburg TB, Sossidou EN, Guinebretière M. The Relationships between Damaging Behaviours and Health in Laying Hens. Animals (Basel) 2022; 12:986. [PMID: 35454233 PMCID: PMC9029779 DOI: 10.3390/ani12080986] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 01/23/2023] Open
Abstract
Since the ban in January 2012 of conventional cages for egg production in the European Union (Council Directive 1999/74/EC), alternative systems such as floor, aviary, free-range, and organic systems have become increasingly common, reaching 50% of housing for hens in 2019. Despite the many advantages associated with non-cage systems, the shift to a housing system where laying hens are kept in larger groups and more complex environments has given rise to new challenges related to management, health, and welfare. This review examines the close relationships between damaging behaviours and health in modern husbandry systems for laying hens. These new housing conditions increase social interactions between animals. In cases of suboptimal rearing and/or housing and management conditions, damaging behaviour or infectious diseases are likely to spread to the whole flock. Additionally, health issues, and therefore stimulation of the immune system, may lead to the development of damaging behaviours, which in turn may result in impaired body conditions, leading to health and welfare issues. This raises the need to monitor both behaviour and health of laying hens in order to intervene as quickly as possible to preserve both the welfare and health of the animals.
Collapse
Affiliation(s)
- Virginie Michel
- Direction de la Stratégie et des Programmes, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 94701 Maisons-Alfort, France
| | - Jutta Berk
- Institute for Animal Welfare and Animal Husbandry, Friedrich-Loeffler-Institut, 29223 Celle, Germany;
| | - Nadya Bozakova
- Department of General Animal Breeding, Animal Hygiene, Ethology and Animal Protection Section, Faculty of Veterinary Medicine, Student’s Campus, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Jerine van der Eijk
- Animal Health and Welfare, Wageningen Livestock Research, Wageningen University and Research, De Elst 1, 6708 Wageningen, The Netherlands;
| | - Inma Estevez
- Department of Animal Production, Neiker-Basque Institute for Agricultural Research and Development, 01080 Vitoria-Gasteiz, Spain;
| | - Teodora Mircheva
- Section of Biochemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Renata Relic
- Faculty of agriculture, University of Belgrade, 11080 Belgrade, Serbia;
| | - T. Bas Rodenburg
- Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584 Utrecht, The Netherlands;
| | - Evangelia N. Sossidou
- Laboratory of Farm Animal Health and Welfare, Veterinary Research Institute, Ellinikos Georgikos Or-Ganismos-DIMITRA (ELGO-DIMITRA), 57001 Thessaloniki, Greece;
| | - Maryse Guinebretière
- Epidemiology, Health and Welfare Unit, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), 22440 Ploufragan, France;
| |
Collapse
|
114
|
Brynge M, Gardner R, Sjöqvist H, Karlsson H, Dalman C. Maternal levels of acute phase proteins in early pregnancy and risk of autism spectrum disorders in offspring. Transl Psychiatry 2022; 12:148. [PMID: 35393396 PMCID: PMC8989993 DOI: 10.1038/s41398-022-01907-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023] Open
Abstract
Previous research supports a contribution of early-life immune disturbances in the etiology of autism spectrum disorders (ASD). Biomarker studies of the maternal innate (non-adaptive) immune status related to ASD risk have focused on one of the acute phase proteins (APP), C-reactive protein (CRP), with conflicting results. We evaluated levels of eight different APP in first-trimester maternal serum samples, from 318 mothers to ASD cases and 429 mothers to ASD-unaffected controls, nested within the register-based Stockholm Youth Cohort. While no overall associations between high levels of APP and ASD were observed, associations varied across diagnostic sub-groups based on co-occurring conditions. Maternal levels of CRP in the lowest compared to the middle tertile were associated with increased risk of ASD without ID or ADHD in offspring (OR = 1.92, 95% CI 1.08-3.42). Further, levels of maternal ferritin in the lowest (OR = 1.78, 95% CI 1.18-2.69) and highest (OR = 1.64, 95% CI 1.11-2.43) tertiles were associated with increased risk of any ASD diagnosis in offspring, with stronger associations still between the lowest (OR = 3.81, 95% CI 1.91-7.58) and highest (OR = 3.36, 95% CI 1.73-6.53) tertiles of ferritin and risk of ASD with ID. The biological interpretation of lower CRP levels among mothers to ASD cases is not clear but might be related to the function of the maternal innate immune system. The finding of aberrant levels of ferritin conferring risk of ASD-phenotypes indicates a plausibly important role of iron during neurodevelopment.
Collapse
Affiliation(s)
- Martin Brynge
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden.
| | - Renee Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Hugo Sjöqvist
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, 17129, Sweden
| |
Collapse
|
115
|
Santos J, Dalla PV, Milthorpe BK. Molecular Dynamics of Cytokine Interactions and Signalling of Mesenchymal Stem Cells Undergoing Directed Neural-like Differentiation. Life (Basel) 2022; 12:life12030392. [PMID: 35330143 PMCID: PMC8948714 DOI: 10.3390/life12030392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells are a continually expanding area in research and clinical applications. Their usefulness and capacity to differentiate into various cells, particularly neural types, has driven the research area for several years. Neural differentiation has considerable usefulness. There are several successful differentiation techniques of mesenchymal stem cells that employ the use of small molecules, growth factors and commercially available kits and supplements. Phenotyping, molecular biology, genomics and proteomics investigation revealed a wealth of data about these cells during neurogenic differentiation. However, there remain large gaps in the knowledge base, particularly related to cytokines and how their role, drive mechanisms and the downstream signalling processes change with their varied expression throughout the differentiation process. In this study, adult mesenchymal stem cells were induced with neurogenic differentiation media, the cellular changes monitored by live-cell microscopy and the changes in cytokine expression in the intracellular region, secretion into the media and in the extracellular vesicle cargo were examined and analysed bioinformatically. Through this analysis, the up-regulation of key cytokines was revealed, and several neuroprotective and neurotrophic roles were displayed. Statistically significant molecules IFN-G, IL1B, IL6, TNF-A, have roles in astrocyte development. Furthermore, the cytokine bioinformatics suggests the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway is upregulated, supporting differentiation toward an astroglial lineage.
Collapse
|
116
|
Kronsten VT, Tranah TH, Pariante C, Shawcross DL. Gut-derived systemic inflammation as a driver of depression in chronic liver disease. J Hepatol 2022; 76:665-680. [PMID: 34800610 DOI: 10.1016/j.jhep.2021.11.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023]
Abstract
Depression and chronic liver disease (CLD) are important causes of disability, morbidity and mortality worldwide and their prevalence continues to rise. The rate of depression in CLD is high compared to that of the general population and is comparable to the increased rates observed in other medical comorbidities and chronic inflammatory conditions. Notably, a comorbid diagnosis of depression has a detrimental effect on outcomes in cirrhosis. Systemic inflammation is pivotal in cirrhosis-associated immune dysfunction - a phenomenon present in advanced CLD (cirrhosis) and implicated in the development of complications, organ failure, disease progression, increased infection rates and poor outcome. The presence of systemic inflammation is also well-documented in a cohort of patients with depression; peripheral cytokine signals can result in neuroinflammation, behavioural change and depressive symptoms via neural mechanisms, cerebral endothelial cell and circumventricular organ signalling, and peripheral immune cell-to-brain signalling. Gut dysbiosis has been observed in both patients with cirrhosis and depression. It leads to intestinal barrier dysfunction resulting in increased bacterial translocation, in turn activating circulating immune cells, leading to cytokine production and systemic inflammation. A perturbed gut-liver-brain axis may therefore explain the high rates of depression in patients with cirrhosis. The underlying mechanisms explaining the critical relationship between depression and cirrhosis remain to be fully elucidated. Several other psychosocial and biological factors are likely to be involved, and therefore the cause is probably multifactorial. However, the role of the dysfunctional gut-liver-brain axis as a driver of gut-derived systemic inflammation requires further exploration and consideration as a target for the treatment of depression in patients with cirrhosis.
Collapse
Affiliation(s)
- Victoria T Kronsten
- Institute of Liver Studies, 1(st) Floor James Black Centre, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK.
| | - Thomas H Tranah
- Institute of Liver Studies, 1(st) Floor James Black Centre, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Carmine Pariante
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, The Maurice Wohl Clinical Neuroscience Institute, Cutcombe Road, London, SE5 9RT, UK
| | - Debbie L Shawcross
- Institute of Liver Studies, 1(st) Floor James Black Centre, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| |
Collapse
|
117
|
Che X, Hornig M, Bresnahan M, Stoltenberg C, Magnus P, Surén P, Mjaaland S, Reichborn-Kjennerud T, Susser E, Lipkin WI. Maternal mid-gestational and child cord blood immune signatures are strongly associated with offspring risk of ASD. Mol Psychiatry 2022; 27:1527-1541. [PMID: 34987169 PMCID: PMC9106807 DOI: 10.1038/s41380-021-01415-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022]
Abstract
Epidemiological studies and work in animal models indicate that immune activation may be a risk factor for autism spectrum disorders (ASDs). We measured levels of 60 cytokines and growth factors in 869 maternal mid-gestational (MMG) and 807 child cord blood (CB) plasma samples from 457 ASD (385 boys, 72 girls) and 497 control children (418 boys, 79 girls) from the Norwegian Autism Birth Cohort. We analyzed associations first using sex-stratified unadjusted and adjusted logistic regression models, and then employed machine learning strategies (LASSO + interactions, Random Forests, XGBoost classifiers) with cross-validation and randomly sampled test set evaluation to assess the utility of immune signatures as ASD biomarkers. We found prominent case-control differences in both boys and girls with alterations in a wide range of analytes in MMG and CB plasma including but not limited to IL1RA, TNFα, Serpin E1, VCAM1, VEGFD, EGF, CSF1, and CSF2. MMG findings were most striking, with particularly strong effect sizes in girls. Models did not change appreciably upon adjustment for maternal conditions, medication use, or emotional distress ratings. Findings were corroborated using machine learning approaches, with area under the receiver operating characteristic curve values in the test sets ranging from 0.771 to 0.965. Our results are consistent with gestational immunopathology in ASD, may provide insights into sex-specific differences, and have the potential to lead to biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Xiaoyu Che
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Mady Hornig
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Michaeline Bresnahan
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Camilla Stoltenberg
- Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health, University of Bergen, Bergen, Norway
| | - Per Magnus
- Norwegian Institute of Public Health, Oslo, Norway
| | - Pål Surén
- Norwegian Institute of Public Health, Oslo, Norway
| | | | - Ted Reichborn-Kjennerud
- Norwegian Institute of Public Health, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ezra Susser
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| | - W Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA.
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
118
|
Maigoro AY, An D, Lee S. Exploring the Link Between Vitamin D Deficiency and Cytokine Storms in COVID-19 Patients: An In Silico Analysis. J Med Food 2022; 25:130-137. [PMID: 35148193 DOI: 10.1089/jmf.2021.k.0085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has become a global infectious pandemic affecting the entire world with complications related to the lungs and compromised immune systems. Recently, cytokine storms, which are hallmarks of the disease, have been identified in most COVID-19 patients. In addition, vitamin D deficiency is increasingly appearing to be another element exposing COVID-19 patients to a preferential increase in their symptoms. In an effort to identify a possible link between cytokine storms and vitamin D deficiency to streamline a possible treatment, an in silico analysis using bioinformatics approach was performed using collections of highly expressed cytokines in both severe acute respiratory syndrome and COVID-19 patients (commonly elevated cytokines) as well as vitamin D deficiency-associated genes (VD). Gene Multiple Association Network Integration Algorithm was used for network interactions, whereas the Enrichr enrichment analysis tool was used for biological functions. The network analysis GLay clustering results indicated the vitamin D receptor as a possible link between these two groups. Furthermore, cell chemotaxis and chemotactic-related features were identified as significantly affected pathways, which serve as possible key players mitigating cytokine storms under low vitamin D availability.
Collapse
Affiliation(s)
- Abdulkadir Yusif Maigoro
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Dayoung An
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Soojin Lee
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
119
|
Increased Monocyte Production of IL-6 after Toll-like Receptor Activation in Children with Autism Spectrum Disorder (ASD) Is Associated with Repetitive and Restricted Behaviors. Brain Sci 2022; 12:brainsci12020220. [PMID: 35203983 PMCID: PMC8870658 DOI: 10.3390/brainsci12020220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 01/27/2023] Open
Abstract
The prevalence of autism spectrum disorder (ASD) has starkly increased, instigating research into risk factors for ASD. This research has identified immune risk factors for ASD, along with evidence of immune dysfunction and excess inflammation frequently experienced by autistic individuals. Increased innate inflammatory cytokines, including interleukin (IL)-6, are seen repeatedly in ASD; however, the origin of excess IL-6 in ASD has not been identified. Here we explore specific responses of circulating monocytes from autistic children. We isolated CD14+ monocytes from whole blood and stimulated them for 24 h under three conditions: media alone, lipoteichoic acid to activate TLR2, and lipopolysaccharide to activate TLR4. We then measured secreted cytokine concentrations in cellular supernatant using a human multiplex bead immunoassay. We found that after TLR4 activation, CD14+ monocytes from autistic children produce increased IL-6 compared to monocytes from children with typical development. IL-6 concentration also correlated with worsening restrictive and repetitive behaviors. These findings suggest dysfunctional activation of myeloid cells, and may indicate that other cells of this lineage, including macrophages, and microglia in the brain, might have a similar dysfunction. Further research on myeloid cells in ASD is warranted.
Collapse
|
120
|
Guzzardi MA, Ederveen THA, Rizzo F, Weisz A, Collado MC, Muratori F, Gross G, Alkema W, Iozzo P. Maternal pre-pregnancy overweight and neonatal gut bacterial colonization are associated with cognitive development and gut microbiota composition in pre-school-age offspring. Brain Behav Immun 2022; 100:311-320. [PMID: 34920092 DOI: 10.1016/j.bbi.2021.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/03/2021] [Accepted: 12/11/2021] [Indexed: 12/17/2022] Open
Abstract
Maternal gestational obesity is a risk factor for offspring's neurodevelopment and later neuro-cognitive disorders. Altered gut microbiota composition has been found in patients with neurocognitive disorders, and in relation to maternal metabolic health. We explored the associations between gut microbiota and cognitive development during infancy, and their link with maternal obesity. In groups of children from the Pisa birth Cohort (PISAC), we analysed faecal microbiota composition by 16S rRNA marker gene sequencing of first-pass meconium samples and of faecal samples collected at age 3, 6, 12, 24, 36 months, and its relationship with maternal gestational obesity or diabetes, and with cognitive development, as measured from 6 to 60 months of age by the Griffith's Mental Development Scales. Gut microbiota composition in the first phases of life is dominated by Bifidobacteria (Actinobacteria phylum), with contribution of Escherichia/Shigella and Klebsiella genera (Proteobacteria phylum), whereas Firmicutes become more dominant at 36 months of age. Maternal overweight leads to lower abundance of Bifidobacterium, Blautia and Ruminococcus, and lower practical reasoning scores in the offspring at the age of 36 months. In the whole population, microbiota in the first-pass meconium samples shows much higher alpha diversity compared to later samples, and its composition, particularly Bifidobacterium and Veillonella abundances, correlates with practical reasoning scores at 60 months of age. Maternal overweight correlates with bacterial colonization and with the development of reasoning skills at pre-school age. Associations between neonatal gut colonization and later cognitive function provide new perspectives of primary (antenatal) prevention of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Maria Angela Guzzardi
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.
| | - Thomas H A Ederveen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands.
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana, University of Salerno, Baronissi, SA, Italy; Genome Research Center for Health (CRGS), Baronissi, SA, Italy.
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana, University of Salerno, Baronissi, SA, Italy; Genome Research Center for Health (CRGS), Baronissi, SA, Italy.
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain.
| | | | - Gabriele Gross
- Medical and Scientific Affairs, Nutrition, RB Mead Johnson Nutrition Institute, Nijmegen, the Netherlands.
| | - Wynand Alkema
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands.
| | - Patricia Iozzo
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.
| |
Collapse
|
121
|
Rasmussen JM, Graham AM, Gyllenhammer LE, Entringer S, Chow DS, O’Connor TG, Fair DA, Wadhwa PD, Buss C. Neuroanatomical Correlates Underlying the Association Between Maternal Interleukin 6 Concentration During Pregnancy and Offspring Fluid Reasoning Performance in Early Childhood. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:24-33. [PMID: 33766778 PMCID: PMC8458517 DOI: 10.1016/j.bpsc.2021.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Maternal inflammation during pregnancy can alter offspring brain development and influence risk for disorders commonly accompanied by deficits in cognitive functioning. We therefore examined associations between maternal interleukin 6 (IL-6) concentrations during pregnancy and offspring cognitive ability and concurrent magnetic resonance imaging-based measures of brain anatomy in early childhood. We further examined newborn brain anatomy in secondary analyses to consider whether effects are evident soon after birth and to increase capacity to differentiate effects of pre- versus postnatal exposures. METHODS IL-6 concentrations were quantified in early (12.6 ± 2.8 weeks), mid (20.4 ± 1.5 weeks), and late (30.3 ± 1.3 weeks) pregnancy. Offspring nonverbal fluid intelligence (Gf) was assessed at 5.2 ± 0.6 years using a spatial reasoning task (Wechsler Preschool and Primary Scale of Intelligence-Matrix) (n = 49). T1-weighted magnetic resonance imaging scans were acquired at birth (n = 89, postmenstrual age = 42.9 ± 2.0 weeks) and in early childhood (n = 42, scan age = 5.1 ± 1.0 years). Regional cortical volumes were examined for a joint association between maternal IL-6 and offspring Gf performance. RESULTS Average maternal IL-6 concentration during pregnancy was inversely associated with offspring Gf performance after adjusting for socioeconomic status and the quality of the caregiving and learning environment (R2 = 13%; p = .02). Early-childhood pars triangularis volume was jointly associated with maternal IL-6 and childhood Gf (pcorrected < .001). An association also was observed between maternal IL-6 and newborn pars triangularis volume (R2 = 6%; p = .02). CONCLUSIONS These findings suggest that the origins of variation in child cognitive ability can, in part, trace back to maternal conditions during the intrauterine period of life and support the role of inflammation as an important component of this putative biological pathway.
Collapse
Affiliation(s)
- Jerod M. Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, California, USA 92697.,Department of Pediatrics, University of California, Irvine, California, USA 92697.,Corresponding Authors: Claudia Buss, PhD, Institute for Medical Psychology, Charité University Medicine, Luisenstr. 57, 10117 Berlin, Germany, Tel: +49 (0)30 450 529 222, Fax: +49 (0)30 450 529 990, ; Jerod M. Rasmussen, PhD., UC Irvine Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility (GNRF), 837 Health Sciences Road, Irvine, CA 92697,
| | - Alice M. Graham
- Department of Behavioral Neuroscience,Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, United States
| | - Lauren E. Gyllenhammer
- Development, Health and Disease Research Program, University of California, Irvine, California, USA 92697.,Department of Pediatrics, University of California, Irvine, California, USA 92697
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, California, USA 92697.,Department of Pediatrics, University of California, Irvine, California, USA 92697.,Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Medical Psychology, Berlin, Germany
| | - Daniel S. Chow
- Department of Radiology, University of California, Irvine, California, USA 92697
| | - Thomas G. O’Connor
- Departments of Psychiatry, Psychology, Neuroscience and Obstetrics & Gynecology, University of Rochester Medical Center, Rochester, New York, USA 14642
| | - Damien A. Fair
- Department of Behavioral Neuroscience,Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, United States
| | - Pathik D. Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, California, USA 92697.,Department of Pediatrics, University of California, Irvine, California, USA 92697.,Departments of Psychiatry and Human Behavior, Obstetrics & Gynecology, Epidemiology, University of California, Irvine, California, USA 92697
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, California, USA 92697.,Department of Pediatrics, University of California, Irvine, California, USA 92697.,Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Medical Psychology, Berlin, Germany.,Corresponding Authors: Claudia Buss, PhD, Institute for Medical Psychology, Charité University Medicine, Luisenstr. 57, 10117 Berlin, Germany, Tel: +49 (0)30 450 529 222, Fax: +49 (0)30 450 529 990, ; Jerod M. Rasmussen, PhD., UC Irvine Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility (GNRF), 837 Health Sciences Road, Irvine, CA 92697,
| |
Collapse
|
122
|
Ma H, Xu J, Li R, McIntyre RS, Teopiz KM, Cao B, Yang F. The Impact of Cognitive Behavioral Therapy on Peripheral Interleukin-6 Levels in Depression: A Systematic Review and Meta-Analysis. Front Psychiatry 2022; 13:844176. [PMID: 35633813 PMCID: PMC9136073 DOI: 10.3389/fpsyt.2022.844176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/28/2022] [Indexed: 11/28/2022] Open
Abstract
UNLABELLED There is interest in the role of peripheral interleukin-6 (IL-6) in depression and the effect of treatment (e. g., pharmacologic, psychosocial, neurostimulation). However, the relationship between cognitive behavioral therapy (CBT), IL-6 and depression has not yet been established. We conducted a meta-analysis to explore the association between CBT and change of peripheral IL-6 levels in depressive symptoms or major depressive disorder (MDD). A systematic search of online databases (i.e., PubMed, Web of Science, Google Scholar, PsycINFO, and Cochrane Library) was completed from inception to May 2021. In total, 10 eligible papers with 940 participants reporting peripheral IL-6 levels before and after CBT were included in the analysis. The main result indicates that peripheral levels of IL-6 were significantly lower after CBT intervention in individuals with depression, with a small effect (SMD = 0.38, 95% CI: 0.07, 0.69, p = 0.02). The results of subgroup analyses demonstrate that (1) there was a significant decrease in IL-6 for studies that were equal to or <8 weeks in duration vs. more than 8 weeks in duration, and (2) IL-6 was significantly reduced in the Diagnostic and Statistical Manual of Mental Disorders (DSM) diagnosis (i.e., DSM-IV, DSM-IV-TR, or DSM-V) of MDD, but not for the subgroup without DSM diagnosis. Publication year was identified as a potential contributor to heterogeneity of the results from our analysis. Taken together, our findings support the notion that CBT influences peripheral IL-6 in individuals with depression and represents a point of commonality with other antidepressant treatment modalities (e.g., antidepressants). SYSTEMATIC REVIEW REGISTRATION https://doi.org/10.17605/osf.io/tr9yh, identifier: 10.17605/osf.io/tr9yh.
Collapse
Affiliation(s)
- Haijing Ma
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China
| | - Jiatong Xu
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China
| | - Ruonan Li
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Kayla M Teopiz
- Mood Disorders Psychopharmacology Unit, Toronto, ON, Canada
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China.,National Demonstration Center for Experimental Psychology Education, Southwest University, Chongqing, China
| | - Fahui Yang
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China.,National Demonstration Center for Experimental Psychology Education, Southwest University, Chongqing, China
| |
Collapse
|
123
|
Feng Y, Ma L, Chen X, Zhang Y, Cao Z, Yuan Y, Xie Y, Liu H, Shi Y, Ren X. Relationship between serum brain-derived neurotrophic factor and cognitive impairment in children with sleep-disordered breathing. Front Pediatr 2022; 10:1027894. [PMID: 36683819 PMCID: PMC9849753 DOI: 10.3389/fped.2022.1027894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND As an important neuroprotective factor, the brain-derived neurotrophic factor (BDNF) may have a key role in cognitive impairment in children with sleep-disordered breathing (SDB). The main aim of this study was to compare the levels of BDNF and tyrosine kinase receptor B (TrkB) in normal children and those with obstructive sleep apnea (OSA) and primary snoring (PS) and to explore a possible link between BDNF/TrkB, inflammation, and SDB with cognitive impairment in children. METHODS A total of 44 OSA children and 35 PS children who completed polysomnography between October 2017 and October 2019 were enrolled. At the same time, 40 healthy children during the same period were included as a control. Enzyme-linked immunosorbent assay was used to measure serum indices of BDNF, TrkB, interleukin-1beta (IL-1β), and tumor necrosis factor-alpha (TNF-α). Correlation and pooled analyses were performed between the cognitive scores and four serological indicators. Logistic regression was used to analyze the risk factors for cognitive impairment. RESULTS Significant differences were found in serum BDNF, TrkB, IL-1β, and TNF-α between the three groups (all P < 0.01). The serum BDNF and TrkB in the OSA and PS groups were lower than those in the control group, whereas the serum IL-1β and TNF-α were higher than those in the control group (all P < 0.05). Moreover, among these four indices, the strongest correlation was found between BDNF and the Chinese Wechsler Intelligence Scale (all P < 0.05). Logistic regression analysis revealed a correlation between OSA status, TrkB, and course of mouth breathing and cognitive status. CONCLUSION The levels of serum BDNF and TrkB were related to cognitive impairment in children with SDB. Also, BDNF and TrkB could be used as noninvasive and objective candidate markers and predictive indices of cognitive impairment in children with SDB.
Collapse
Affiliation(s)
- Yani Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lina Ma
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yitong Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zine Cao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuqi Yuan
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yushan Xie
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haiqin Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yewen Shi
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyong Ren
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
124
|
Edem EE, Anyanwu CKC, Nebo KE, Akinluyi ET, Fafure AA, Ishola AO, Enye LA. Ketamine abrogates sensorimotor deficits and cytokine dysregulation in a chronic unpredictable mild stress model of depression. Psychopharmacology (Berl) 2022; 239:185-200. [PMID: 34792632 DOI: 10.1007/s00213-021-06021-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
Major depressive disorder (MDD) is a serious mental disorder with influence across the functional systems of the body. The pathogenesis of MDD has been known to involve the alteration of normal body functions responsible for the normal inflammation processes within the CNS; this along with other effects results in the depreciation of the sensorimotor performance of the body. Ketamine hydrochloride, a novel antidepressant agent, has been used as a therapeutic agent to treat MDD with its efficacy stretching as far as enhancing sensorimotor performance and restoring normal cytokine levels of the CNS. While these therapeutic actions of ketamine may or may not be related, this study made use of chronic unpredictable mild stress (CUMS) to generate the mouse model of depression. The efficacy of ketamine as an antidepressant following sequential exposure and co-administrative treatment protocols of administration was evaluated using behavioural tests for sensorimotor performance and depressive-like behaviours. Its effect in managing CNS inflammation was assessed via the biochemical analysis of inflammatory cytokine levels in the cerebrum, spinal cord and cerebellum; and immunohistochemical demonstration of microglial activity in the corpus striatum and cerebellum. The sensorimotor performance which had been diminished by CUMS showed greater improvement under the sequential exposure regimen of ketamine. Ketamine was also efficacious in decreasing the level of inflammation with an evident reduction in microglial activation and pro-inflammatory cytokines in the studied regions, following CUMS exposure. Taken together, our study indicates that ketamine therapy can improve sensorimotor deficits co-morbid with a depressive disorder in parallel with modulation of the inflammatory system.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Neuroscience Unit, Department of Human Anatomy, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria.
| | - Collins-Kevin Chukwudi Anyanwu
- Neuroscience Unit, Department of Human Anatomy, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
| | - Kate Eberechukwu Nebo
- Neuroscience Unit, Department of Human Anatomy, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
| | - Elizabeth Toyin Akinluyi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
| | - Adedamola Adediran Fafure
- Neuroscience Unit, Department of Human Anatomy, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
| | - Azeez Olakunle Ishola
- Neuroscience Unit, Department of Human Anatomy, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
| | - Linus Anderson Enye
- Neuroscience Unit, Department of Human Anatomy, College Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, P.M.B. 5454, Ado-Ekiti, Ekiti, Nigeria
| |
Collapse
|
125
|
McLellan J, Kim DHJ, Bruce M, Ramirez-Celis A, Van de Water J. Maternal Immune Dysregulation and Autism-Understanding the Role of Cytokines, Chemokines and Autoantibodies. Front Psychiatry 2022; 13:834910. [PMID: 35722542 PMCID: PMC9201050 DOI: 10.3389/fpsyt.2022.834910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is acknowledged as a highly heterogeneous, behaviorally defined neurodevelopmental disorder with multiple etiologies. In addition to its high heritability, we have come to recognize a role for maternal immune system dysregulation as a prominent risk factor for the development of ASD in the child. Examples of these risk factors include altered cytokine/chemokine activity and the presence of autoantibodies in mothers that are reactive to proteins in the developing brain. In addition to large clinical studies, the development of pre-clinical models enables the ability to evaluate the cellular and molecular underpinnings of immune-related pathology. For example, the novel animal models of maternal autoantibody-related (MAR) ASD described herein will serve as a preclinical platform for the future testing of targeted therapeutics for one 'type' of ASD. Identification of the cellular targets will advance precision medicine efforts toward tailored therapeutics and prevention. This minireview highlights emerging evidence for the role of maternal immune dysregulation as a potential biomarker, as well as a pathologically relevant mechanism for the development of ASD in offspring. Further, we will discuss the current limitations of these models as well as potential avenues for future research.
Collapse
Affiliation(s)
- Janna McLellan
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Danielle H J Kim
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Matthew Bruce
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Alexandra Ramirez-Celis
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Judy Van de Water
- Division of Rheumatology, Department of Internal Medicine, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA, United States.,MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
126
|
Carter M, Casey S, O'Keeffe GW, Gibson L, Gallagher L, Murray DM. Maternal Immune Activation and Interleukin 17A in the Pathogenesis of Autistic Spectrum Disorder and Why It Matters in the COVID-19 Era. Front Psychiatry 2022; 13:823096. [PMID: 35250672 PMCID: PMC8891512 DOI: 10.3389/fpsyt.2022.823096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
Abstract
Autism spectrum disorder (ASD) is the commonest neurodevelopmental disability. It is a highly complex disorder with an increasing prevalence and an unclear etiology. Consensus indicates that ASD arises as a genetically modulated, and environmentally influenced condition. Although pathogenic rare genetic variants are detected in around 20% of cases of ASD, no single factor is responsible for the vast majority of ASD cases or that explains their characteristic clinical heterogeneity. However, a growing body of evidence suggests that ASD susceptibility involves an interplay between genetic factors and environmental exposures. One such environmental exposure which has received significant attention in this regard is maternal immune activation (MIA) resulting from bacterial or viral infection during pregnancy. Reproducible rodent models of ASD are well-established whereby induction of MIA in pregnant dams, leads to offspring displaying neuroanatomical, functional, and behavioral changes analogous to those seen in ASD. Blockade of specific inflammatory cytokines such as interleukin-17A during gestation remediates many of these observed behavioral effects, suggesting a causative or contributory role. Here, we review the growing body of animal and human-based evidence indicating that interleukin-17A may mediate the observed effects of MIA on neurodevelopmental outcomes in the offspring. This is particularly important given the current corona virus disease-2019 (COVID-19) pandemic as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy is a potent stimulator of the maternal immune response, however the long-term effects of maternal SARS-CoV-2 infection on neurodevelopmental outcomes is unclear. This underscores the importance of monitoring neurodevelopmental outcomes in children exposed to SARS-CoV-2-induced MIA during gestation.
Collapse
Affiliation(s)
- Michael Carter
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,National Children's Research Centre, Dublin, Ireland
| | - Sophie Casey
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard W O'Keeffe
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Louise Gibson
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Deirdre M Murray
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| |
Collapse
|
127
|
Hoshikawa T, Okamoto N, Natsuyama T, Fujii R, Ikenouchi A, Honma Y, Harada M, Yoshimura R. Associations of Serum Cytokines, Growth Factors, and High-Sensitivity C-Reactive Protein Levels in Patients with Major Depression with and without Type 2 Diabetes Mellitus: An Explanatory Investigation. Neuropsychiatr Dis Treat 2022; 18:173-186. [PMID: 35140467 PMCID: PMC8820450 DOI: 10.2147/ndt.s350121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/04/2022] [Indexed: 12/26/2022] Open
Abstract
PURPOSE We investigated the serum levels of cytokines, including interleukin 1β (IL-β), IL-6, IL-8, IL-10, tumor necrosis factor-alpha (TNF-α), and growth factors, including brain-derived neurotrophic factor, vascular endothelial growth factor, and insulin-like growth factor 1, and their association with major depression in patients with and without type 2 diabetes mellitus. We also investigated the response to antidepressant treatment in both groups. PATIENTS AND METHODS Forty-one patients with major depression were recruited at the University Hospital of Occupational and Environmental Health. All patients were diagnosed with major depression using the Diagnostic and Statistical Manual for Mental Disorders, Fifth Edition. Type 2 diabetes mellitus was diagnosed according to the criteria of the Japan Diabetes Society. Six healthy controls with no history of psychiatric or physical diseases were also enrolled. Serum levels of several cytokines, growth factors, and high-sensitivity C-reactive protein (hs-CRP) were measured. The clinical symptoms of patients with major depression were assessed using the Montgomery-Asberg Depression Rating Scale. RESULTS Significant differences in cytokines, growth factors, and hs-CRP were observed between the major depression and healthy control groups. Serum TNF-α levels were significantly higher in patients with major depression and type 2 diabetes mellitus than in those without type 2 diabetes mellitus. In the major depression group, serum IL-6 and hs-CRP levels tended to be higher in patients with type 2 diabetes mellitus than in those without. Several correlations among cytokines, growth factors, and hs-CRP were observed in patients with major depression with and without type 2 diabetes mellitus. Responses to pharmacological interventions for major depression did not differ between patients with and without type 2 diabetes mellitus. CONCLUSION Serum levels of TNF-α, hs-CRP, and IL-6 were different between patients with major depression with and without type 2 diabetes mellitus. Also, correlations were found between serum levels of cytokines, growth factors, and hs-CRP in patients with major depression. Inflammatory factors, which may be associated with growth factors, may be involved in the pathophysiology of major depression, particularly among patients with comorbid type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Takashi Hoshikawa
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Naomichi Okamoto
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Tomoya Natsuyama
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Rintaro Fujii
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Atsuko Ikenouchi
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Yuichi Honma
- 3rd Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Masaru Harada
- 3rd Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| | - Reiji Yoshimura
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 8078555, Japan
| |
Collapse
|
128
|
Ayash TA, Vancolen SY, Segura M, Allard MJ, Sebire G. Protective Effects of Interleukin-1 Blockade on Group B Streptococcus-Induced Chorioamnionitis and Subsequent Neurobehavioral Impairments of the Offspring. Front Endocrinol (Lausanne) 2022; 13:833121. [PMID: 35846278 PMCID: PMC9283950 DOI: 10.3389/fendo.2022.833121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
Group B Streptococcus (GBS) is one of the most common bacteria isolated in human chorioamnionitis. Placental infection due to GBS is a major risk factor for fetal organ injuries, preterm birth, perinatal morbidity and mortality, and life-long multiorgan morbidities. Preclinical and clinical studies have shown that GBS-induced infection drives polymorphonuclear (PMN) cell infiltration within the placenta, the hallmark of human chorioamnionitis. In preclinical and clinical studies, the upregulation of interleukin(IL)-1β in the placenta and maternal/fetal blood was associated with a high risk of neurodevelopmental impairments in the progeny. We hypothesized that targeted IL-1 blockade administered to the dam alleviates GBS-induced chorioamnionitis and the downstream fetal inflammatory response syndrome (FIRS). IL-1 receptor antagonist (IL-1Ra) improved the gestational weight gain of GBS-infected dams and did not worsen the infectious manifestations. IL-1Ra reduced the IL-1β titer in the maternal sera of GBS-infected dams. IL-1Ra decreased the levels of IL-1β, IL-6, chemokine (C-X-C motif) ligand 1 (CXCL1), and polymorphonuclear (PMN) infiltration in GBS-infected placenta. IL-1Ra treatment reduced the IL-1β titer in the fetal sera of GBS-exposed fetuses. IL-1 blockade also alleviated GBS-induced FIRS and subsequent neurobehavioral impairments of the offspring without worsening the outcome of GBS infection. Altogether, these results showed that IL-1 plays a key role in the physiopathology of live GBS-induced chorioamnionitis and consequent neurobehavioral impairments.
Collapse
Affiliation(s)
| | | | - Mariela Segura
- Faculty of Veterinary Medicine, Université de Montreal, St-Hyacinthe, QC, Canada
| | | | - Guillaume Sebire
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- *Correspondence: Guillaume Sebire,
| |
Collapse
|
129
|
A Molecular Analysis of Cytokine Content across Extracellular Vesicles, Secretions, and Intracellular Space from Different Site-Specific Adipose-Derived Stem Cells. Int J Mol Sci 2021; 23:ijms23010397. [PMID: 35008824 PMCID: PMC8745205 DOI: 10.3390/ijms23010397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Cytokines are multifunctional small proteins that have a vital influence on inflammatory states of tissues and play a role in signalling and cellular control mechanisms. Cytokine expression has primarily been viewed as a form of direct secretion of molecules through an active transportation; however, other forms of active transport such as extracellular vesicles are at play. This is particularly important in stem cells where signalling molecules are key to communication managing the levels of proliferation, migration, and differentiation into mature cells. This study investigated cytokines from intracellular content, direct cellular secretions, and extracellular vesicles from adult adipose-derived stem cells isolated from three distinct anatomical locations: abdomen, thigh, and chin. The cells were cultured investigated using live cell microscopy, cytokine assays, and bioinformatics analysis. The cytokines quantified and examined from each sample type showed a distinct difference between niche areas and sample types. The varying levels of TNF-alpha, IL-6 and IL-8 cytokines were shown to play a crucial role in signalling pathways such as MAPK, ERK1/2 and JAK-STAT in cells. On the other hand, the chemotactic cytokines IL-1rn, Eotaxin, IP-10 and MCP-1 showed the most prominent changes across extracellular vesicles with roles in noncanonical signalling. By examining the local and tangential roles of cytokines in stem cells, their roles in signalling and in regenerative mechanisms may be further understood.
Collapse
|
130
|
Ganguli S, Chavali PL. Corrigendum: Intrauterine Viral Infections: Impact of Inflammation on Fetal Neurodevelopment. Front Neurosci 2021; 15:817697. [PMID: 34955742 PMCID: PMC8707729 DOI: 10.3389/fnins.2021.817697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
[This corrects the article DOI: 10.3389/fnins.2021.771557.].
Collapse
Affiliation(s)
- Sourav Ganguli
- CSIR-Center for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcCSIR), Ghaziabad, India
| | - Pavithra L Chavali
- CSIR-Center for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcCSIR), Ghaziabad, India
| |
Collapse
|
131
|
Lussier AA, Bodnar TS, Weinberg J. Intersection of Epigenetic and Immune Alterations: Implications for Fetal Alcohol Spectrum Disorder and Mental Health. Front Neurosci 2021; 15:788630. [PMID: 34924946 PMCID: PMC8680672 DOI: 10.3389/fnins.2021.788630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/02/2021] [Indexed: 01/15/2023] Open
Abstract
Prenatal alcohol exposure can impact virtually all body systems, resulting in a host of structural, neurocognitive, and behavioral abnormalities. Among the adverse impacts associated with prenatal alcohol exposure are alterations in immune function, including an increased incidence of infections and alterations in immune/neuroimmune parameters that last throughout the life-course. Epigenetic patterns are also highly sensitive to prenatal alcohol exposure, with widespread alcohol-related alterations to epigenetic profiles, including changes in DNA methylation, histone modifications, and miRNA expression. Importantly, epigenetic programs are crucial for immune system development, impacting key processes such as immune cell fate, differentiation, and activation. In addition to their role in development, epigenetic mechanisms are emerging as attractive candidates for the biological embedding of environmental factors on immune function and as mediators between early-life exposures and long-term health. Here, following an overview of the impact of prenatal alcohol exposure on immune function and epigenetic patterns, we discuss the potential role for epigenetic mechanisms in reprogramming of immune function and the consequences for health and development. We highlight a range of both clinical and animal studies to provide insights into the array of immune genes impacted by alcohol-related epigenetic reprogramming. Finally, we discuss potential consequences of alcohol-related reprogramming of immune/neuroimmune functions and their effects on the increased susceptibility to mental health disorders. Overall, the collective findings from animal models and clinical studies highlight a compelling relationship between the immune system and epigenetic pathways. These findings have important implications for our understanding of the biological mechanisms underlying the long-term and multisystem effects of prenatal alcohol exposure, laying the groundwork for possible novel interventions and therapeutic strategies to treat individuals prenatally exposed to alcohol.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Tamara S Bodnar
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
132
|
Tao D, Zhong T, Pang W, Li X. Saccharomyces boulardii improves the behaviour and emotions of spastic cerebral palsy rats through the gut-brain axis pathway. BMC Neurosci 2021; 22:76. [PMID: 34876019 PMCID: PMC8653608 DOI: 10.1186/s12868-021-00679-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 11/23/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cerebral palsy (CP) is a kind of disability that influences motion, and children with CP also exhibit depression-like behaviour. Inflammation has been recognized as a contributor to CP and depression, and some studies suggest that the gut-brain axis may be a contributing factor. Our team observed that Saccharomyces boulardii (S. boulardii) could reduce the inflammatory level of rats with hyperbilirubinemia and improve abnormal behaviour. Both CP and depression are related to inflammation, and probiotics can improve depression by reducing inflammation. Therefore, we hypothesize that S. boulardii may improve the behaviour and emotions of spastic CP rats through the gut-brain axis pathway. METHODS Our new rat model was produced by resecting the cortex and subcortical white matter. Seventeen-day-old CP rats were exposed to S. boulardii or vehicle control by gastric gavage for 9 days, and different behavioural domains and general conditions were tested. Inflammation was assessed by measuring the inflammatory markers IL-6 and TNF-α. Hypothalamic-pituitary-adrenal (HPA) axis activity was assessed by measuring adrenocorticotropic hormone and corticosterone in the serum. Changes in the gut microbiome were detected by 16S rRNA. RESULTS The hemiplegic spastic CP rats we made with typical spastic paralysis exhibited depression-like behaviour. S. boulardii treatment of hemiplegic spastic CP rats improves behaviour and general conditions and significantly reduces the level of inflammation, decreases HPA axis activity, and increases gut microbiota diversity. CONCLUSIONS The model developed in this study mimics a hemiplegic spastic cerebral palsy. Damage to the cortex and subcortical white matter of 17-day-old Sprague-Dawley (SD) rats led to spastic CP-like behaviour, and the rats exhibited symptoms of depression-like behaviour. Our results indicate that S. boulardii might have potential in treating hemiplegic spastic CP rat models or as an add-on therapy via the gut-brain axis pathway.
Collapse
Affiliation(s)
- Deshuang Tao
- College of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China
- Jiamusi Central Hospital, Jiamusi, Heilongjiang, China
| | - Tangwu Zhong
- College of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Wei Pang
- College of Rehab Medicine, Jiamusi University, Jiamusi, China
- Rehab Center for Child Cerebral Palsy, Jiamusi, Heilongjiang, China
- Institute of Pediatric Neurological Disorders, Jiamusi University, Jiamusi, China
| | - Xiaojie Li
- College of Rehab Medicine, Jiamusi University, Jiamusi, China.
- Rehab Center for Child Cerebral Palsy, Jiamusi, Heilongjiang, China.
- Institute of Pediatric Neurological Disorders, Jiamusi University, Jiamusi, China.
| |
Collapse
|
133
|
Ganguli S, Chavali PL. Intrauterine Viral Infections: Impact of Inflammation on Fetal Neurodevelopment. Front Neurosci 2021; 15:771557. [PMID: 34858132 PMCID: PMC8631423 DOI: 10.3389/fnins.2021.771557] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/18/2021] [Indexed: 12/22/2022] Open
Abstract
Intrauterine viral infections during pregnancy by pathogens such as Zika virus, Cytomegalovirus, Rubella and Herpes Simplex virus can lead to prenatal as well as postnatal neurodevelopmental disorders. Although maternal viral infections are common during pregnancy, viruses rarely penetrate the trophoblast. When they do cross, viruses can cause adverse congenital health conditions for the fetus. In this context, maternal inflammatory responses to these neurotropic pathogens play a significant role in negatively affecting neurodevelopment. For instance, intrauterine inflammation poses an increased risk of neurodevelopmental disorders such as microcephaly, schizophrenia, autism spectrum disorder, cerebral palsy and epilepsy. Severe inflammatory responses have been linked to stillbirths, preterm births, abortions and microcephaly. In this review, we discuss the mechanistic basis of how immune system shapes the landscape of the brain and how different neurotropic viral pathogens evoke inflammatory responses. Finally, we list the consequences of neuroinflammation on fetal brain development and discuss directions for future research and intervention strategies.
Collapse
Affiliation(s)
- Sourav Ganguli
- CSIR-Center for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcCSIR), Ghaziabad, India
| | - Pavithra L Chavali
- CSIR-Center for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcCSIR), Ghaziabad, India
| |
Collapse
|
134
|
Dufford AJ, Spann M, Scheinost D. How prenatal exposures shape the infant brain: Insights from infant neuroimaging studies. Neurosci Biobehav Rev 2021; 131:47-58. [PMID: 34536461 DOI: 10.1016/j.neubiorev.2021.09.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/30/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
Brain development during the prenatal period is rapid and unparalleled by any other time during development. Biological systems undergoing rapid development are at higher risk for disorganizing influences. Therefore, certain prenatal exposures impact brain development, increasing risk for negative neurodevelopmental outcome. While prenatal exposures have been associated with cognitive and behavioral outcomes later in life, the underlying macroscopic brain pathways remain unclear. Here, we review magnetic resonance imaging (MRI) studies investigating the association between prenatal exposures and infant brain development focusing on prenatal exposures via maternal physical health factors, maternal mental health factors, and maternal drug and medication use. Further, we discuss the need for studies to consider multiple prenatal exposures in parallel and suggest future directions for this body of research.
Collapse
Affiliation(s)
| | - Marisa Spann
- Columbia University Irving Medical Center, 622 West 168th Street, New York, NY, 10032, USA
| | - Dustin Scheinost
- Child Study Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, USA; Department of Statistics and Data Science, Yale University, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| |
Collapse
|
135
|
Lagodka S, Petrucci S, Moretti ML, Cabbad M, Lakhi NA. Fetal and maternal inflammatory response in the setting of maternal intrapartum fever with and without clinical and histologic chorioamnionitis. Am J Obstet Gynecol MFM 2021; 4:100539. [PMID: 34861429 DOI: 10.1016/j.ajogmf.2021.100539] [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: 06/16/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Both infectious and noninfectious causes of maternal fever have been linked to adverse neonatal outcomes including low Apg0ar scores, respiratory distress, hypotonia, and neonatal seizures. Even in the absence of infection, the occurrence of intrapartum fever is a strong risk factor for poor long-term neonatal developmental outcomes, including encephalopathy, cerebral palsy, and neonatal death. OBJECTIVE The primary objective of this study was to compare intrapartum and postpartum maternal and fetal umbilical cord serum levels of cytokines RANTES, interferon-ɣ, interleukin-1β, interleukin-2, interleukin-4, interleukin-6, interleukin-8, interleukin-10, interleukin-13, and tumor necrosis factor-α among nonfebrile patients, febrile patients without clinical chorioamnionitis, and febrile patient with clinical chorioamnionitis. STUDY DESIGN This study was conducted at the Richmond University Medical Center from May 15, 2020 to July 16, 2019. During this time, we recruited 30 nonfebrile patients at >36 gestational weeks who were in labor and collected umbilical cord and pre- and postdelivery maternal serum samples to evaluate the cytokine levels. Placentas were collected for pathologic review and to evaluate the histopathologic findings. These results were compared with 121 patients who developed a fever of >38°C during labor. The febrile patients were further divided based on the presence or absence of clinical chorioamnionitis. A secondary analysis was performed based on the presence of absence of histologic chorioamnionitis. Statistical analysis was performed using IBM Statistical Package for the Social Sciences version 25.0. For the 3 group comparisons, a P value of <.017 was considered statistically significant after application of a Bonferroni correction. RESULTS A total of 151 patients were included in the study; 30 were nonfebrile patients, 46 were febrile patients with a diagnosis of clinical chorioamnionitis, and 75 were febrile patients without clinical chorioamnionitis. Compared with nonfebrile patients, umbilical cord serum interferon-ɣ, interleukin-1β, interleukin-6, interleukin-8, RANTES, and tumor necrosis factor-α levels were elevated in the presence of maternal hyperthermia irrespective of the diagnosis of clinical chorioamnionitis. Interleukin-6 umbilical cord levels were more than doubled from 63.60 pg/mL (6.09-1769.03 pg/mL) in febrile patients with no clinical chorioamnionitis to 135.77 pg/mL (1.86-6004.78 pg/mL) in febrile patients with clinical chorioamnionitis, making it the only cytokine that was significantly different between these 2 groups. When comparing the intrapartum maternal serum, we found a significant elevation in the interleukin-10, RANTES, and tumor necrosis factor-α levels in the febrile group irrespective of the presence of clinical chorioamnionitis when compared with the nonfebrile group. In the postpartum maternal blood evaluations, tumor necrosis factor-α was the only cytokine that was significantly higher in febrile patients than in nonfebrile controls. CONCLUSION In the setting of intrapartum fever, maternal cytokine profiles were similar irrespective of the diagnosis of clinical chorioamnionitis. Even in the absence of clinical or histologic chorioamnionitis, maternal hyperthermia induced elevations in fetal cytokines.
Collapse
Affiliation(s)
- Sylvie Lagodka
- Department of Obstetrics and Gynecology, Richmond University Medical Center, Staten Island, NY
| | | | - Michael L Moretti
- Department of Obstetrics and Gynecology, The Brooklyn Hospital Center, Brooklyn, NY
| | - Michael Cabbad
- Department of Obstetrics and Gynecology, Richmond University Medical Center, Staten Island, NY
| | - Nisha A Lakhi
- Department of Obstetrics and Gynecology, Richmond University Medical Center, Staten Island, NY; Department of Obstetrics and Gynecology, New York Medical College, Valhalla, NY.
| |
Collapse
|
136
|
Serra I, Manusama OR, Kaiser FMP, Floriano II, Wahl L, van der Zalm C, IJspeert H, van Hagen PM, van Beveren NJM, Arend SM, Okkenhaug K, Pel JJM, Dalm VASH, Badura A. Activated PI3Kδ syndrome, an immunodeficiency disorder, leads to sensorimotor deficits recapitulated in a murine model. Brain Behav Immun Health 2021; 18:100377. [PMID: 34786564 PMCID: PMC8579111 DOI: 10.1016/j.bbih.2021.100377] [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: 06/04/2021] [Revised: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 02/08/2023] Open
Abstract
The phosphoinositide-3-kinase (PI3K) family plays a major role in cell signaling and is predominant in leukocytes. Gain-of-function (GOF) mutations in the PIK3CD gene lead to the development of activated PI3Kδ syndrome (APDS), a rare primary immunodeficiency disorder. A subset of APDS patients also displays neurodevelopmental delay symptoms, suggesting a potential role of PIK3CD in cognitive and behavioural function. However, the extent and nature of the neurodevelopmental deficits has not been previously quantified. Here, we assessed the cognitive functions of two APDS patients, and investigated the causal role of the PIK3CD GOF mutation in neurological deficits using a murine model of this disease. We used p110δE1020K knock-in mice, harbouring the most common APDS mutation in patients. We found that APDS patients present with visuomotor deficits, exacerbated by autism spectrum disorder comorbidity, whereas p110δE1020K mice exhibited impairments in motor behaviour, learning and repetitive behaviour patterning. Our data indicate that PIK3CD GOF mutations increase the risk for neurodevelopmental deficits, supporting previous findings on the interplay between the nervous and the immune system. Further, our results validate the knock-in mouse model, and offer an objective assessment tool for patients that could be incorporated in diagnosis and in the evaluation of treatments.
Collapse
Affiliation(s)
- Ines Serra
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Fabian M P Kaiser
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.,Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | | | - Lucas Wahl
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Hanna IJspeert
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - P Martin van Hagen
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.,Division of Clinical Immunology, Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | | | - Sandra M Arend
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Virgil A S H Dalm
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.,Division of Clinical Immunology, Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus MC, Rotterdam, the Netherlands
| | | |
Collapse
|
137
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
138
|
Airapetov MI, Eresko SO, Bychkov ER, Lebedev AA, Shabanov PD. [Prenatal exposure to alcohol alters TLR4 signaling in the prefrontal cortex in rats]. BIOMEDITSINSKAIA KHIMIIA 2021; 67:500-506. [PMID: 34964444 DOI: 10.18097/pbmc20216706500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Prenatal alcohol exposure (PAE) can lead to developmental disorders of the central nervous system (CNS) and mental retardation. Toll-like receptor (TLR) 4 plays an important role in the development of defects in the nervous system caused by PAE. However, how PAE affects the TLR4 response in the brain remains unclear. Using the model of semi-forced alcoholization of pregnant rats, we investigated TLR4-mediated signaling on the 30th day of postnatal development in their offspring. Rats exposed to PAE showed a higher expression of proinflammatory cytokines in the prefrontal cortex, but TLR4-mediated signaling in response to lipopolysaccharide (LPS) was weakened. These data suggest that PAE can lead to neuroinflammation and suppression of the TLR4-mediated response to LPS in the prefrontal cortex of young rats. Since innate immunity plays an important role in brain development, PAE-induced suppression of the TLR4-mediated response may be one of the mechanisms for the development of CNS pathology.
Collapse
Affiliation(s)
- M I Airapetov
- Institute of Experimental Medicine, Saint Petersburg, Russia; Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russia
| | - S O Eresko
- Institute of Experimental Medicine, Saint Petersburg, Russia; Saint Petersburg State University, Saint Petersburg, Russia
| | - E R Bychkov
- Institute of Experimental Medicine, Saint Petersburg, Russia
| | - A A Lebedev
- Institute of Experimental Medicine, Saint Petersburg, Russia
| | - P D Shabanov
- Institute of Experimental Medicine, Saint Petersburg, Russia; Kirov Military Medical Academy, Saint Petersburg, Russia
| |
Collapse
|
139
|
Effects of maternal psychological stress during pregnancy on offspring brain development: Considering the role of inflammation and potential for preventive intervention. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:461-470. [PMID: 34718150 PMCID: PMC9043032 DOI: 10.1016/j.bpsc.2021.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022]
Abstract
Heightened psychological stress during pregnancy has repeatedly been associated with increased risk for offspring development of behavior problems and psychiatric disorders. This review covers a rapidly growing body of research with the potential to advance a mechanistic understanding of these associations grounded in knowledge about maternal-placental-fetal stress biology and fetal brain development. Specifically, we highlight research employing magnetic resonance imaging to examine the infant brain soon after birth in relation to maternal psychological stress during pregnancy to increase capacity to identify specific alterations in brain structure and function and to differentiate between effects of pre- versus postnatal exposures. We then focus on heightened maternal inflammation during pregnancy as a mechanism through which maternal stress influences the developing fetal brain based on extensive preclinical literature and emerging research in humans. We place these findings in the context of recent work identifying psychotherapeutic interventions found to be effective for reducing psychological stress among pregnant individuals, which also show promise for reducing inflammation. We argue that a focus on inflammation, among other mechanistic pathways, has the potential to lead to a productive and necessary integration of research focused on the effects of maternal psychological stress on offspring brain development and prevention and intervention studies aimed at reducing maternal psychological stress during pregnancy. In addition to increasing capacity for common measurements and understanding potential mechanisms of action relevant to maternal mental health and fetal neurodevelopment, this focus can inform and broaden thinking about prevention and intervention strategies.
Collapse
|
140
|
Chang CY, Ting HC, Liu CA, Su HL, Chiou TW, Harn HJ, Lin SZ, Ho TJ. Differentiation of Human Pluripotent Stem Cells Into Specific Neural Lineages. Cell Transplant 2021; 30:9636897211017829. [PMID: 34665040 PMCID: PMC8529300 DOI: 10.1177/09636897211017829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are sources of several somatic cell
types for human developmental studies, in vitro disease modeling, and
cell transplantation therapy. Improving strategies of derivation of
high-purity specific neural and glial lineages from hPSCs is critical
for application to the study and therapy of the nervous system. Here,
we will focus on the principles behind establishment of neuron and
glia differentiation methods according to developmental studies. We
will also highlight the limitations and challenges associated with the
differentiation of several “difficult” neural lineages and delay in
neuronal maturation and functional integration. To overcome these
challenges, we will introduce strategies and novel technologies aimed
at improving the differentiation of various neural lineages to expand
the application potential of hPSCs to the study of the nervous
system.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Neuroscience Center, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Hsiao-Chien Ting
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ching-Ann Liu
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Neuroscience Center, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tzyy-Wen Chiou
- Department of Life Science, National Dong Hwa University, Hualien, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Pathology, Hualien Tzu Chi Hospital and Tzu Chi University, Hualien, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
141
|
Lins B. Maternal immune activation as a risk factor for psychiatric illness in the context of the SARS-CoV-2 pandemic. Brain Behav Immun Health 2021; 16:100297. [PMID: 34308388 PMCID: PMC8279925 DOI: 10.1016/j.bbih.2021.100297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/02/2021] [Accepted: 07/12/2021] [Indexed: 01/10/2023] Open
Abstract
Inflammation, due to infectious pathogens or other non-infectious stimuli, during pregnancy is associated with elevated risk for neurodevelopmental disorders such as schizophrenia and autism in the offspring. Although historically identified through retrospective epidemiologic studies, the relationship between maternal immune activation and offspring neurodevelopmental disease risk is now well established because of clinical studies which utilized prospective birth cohorts, serologically confirmed infection records, and subsequent long-term offspring follow-up. These efforts have been corroborated by preclinical research which demonstrates anatomical, biochemical, and behavioural alterations that resemble the clinical features of psychiatric illnesses. Intervention studies further demonstrate causal roles of inflammatory mediators, such as cytokines, in these long-lasting changes in behaviour and brain. This review summarizes a selection of maternal immune activation literature that explores the relationship between these inflammatory mediators and the neuropsychiatric-like effects later observed in the offspring. This literature is presented alongside emerging information regarding SARS-CoV-2 infection in pregnancy, with discussion of how these data may inform future research regarding the effects of the present coronavirus pandemic on emerging birth cohorts.
Collapse
Affiliation(s)
- Brittney Lins
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.
| |
Collapse
|
142
|
Fernández de Cossío L, Lacabanne C, Bordeleau M, Castino G, Kyriakakis P, Tremblay MÈ. Lipopolysaccharide-induced maternal immune activation modulates microglial CX3CR1 protein expression and morphological phenotype in the hippocampus and dentate gyrus, resulting in cognitive inflexibility during late adolescence. Brain Behav Immun 2021; 97:440-454. [PMID: 34343619 DOI: 10.1016/j.bbi.2021.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/26/2022] Open
Abstract
Inflammation during pregnancy can disturb brain development and lead to disorders in the progeny, including autism spectrum disorder and schizophrenia. However, the mechanism by which a prenatal, short-lived increase of cytokines results in adverse neurodevelopmental outcomes remains largely unknown. Microglia-the brain's resident immune-cells-stand as fundamental cellular mediators, being highly sensitive and responsive to immune signals, which also play key roles during normal development. The fractalkine signaling axis is a neuron-microglia communication mechanism used to regulate neurogenesis and network formation. Previously, we showed hippocampal reduction of fractalkine receptor (Cx3cr1) mRNA at postnatal day (P) 15 in male offspring exposed to maternal immune activation induced with lipopolysaccharide (LPS) during late gestation, which was concomitant to an increased dendritic spine density in the dentate gyrus, a neurogenic niche. The current study sought to evaluate the origin and impact of this reduced hippocampal Cx3cr1 mRNA expression on microglia and cognition. We found that microglial total cell number and density are not affected in the dorsal hippocampus and dentate gyrus, respectively, but that the microglial CX3CR1 protein is decreased in the hippocampus of LPS-male offspring at P15. Further characterization of microglial morphology in the dentate gyrus identified a more ameboid phenotype in LPS-exposed offspring, predominantly in males, at P15. We thus explored maternal plasma and fetal brain cytokines to understand the mechanism behind microglial priming, showing a robust immune activation in the mother at 2 and 4 hrs after LPS administration, while only IL-10 tended towards upregulation at 2 hrs after LPS in fetal brains. To evaluate the functional long-term consequences, we assessed learning and cognitive flexibility behavior during late adolescence, finding that LPS affects only the latter with a male predominance on perseveration. A CX3CR1 gene variant in humans that results in disrupted fractalkine signaling has been recently associated with an increased risk for neurodevelopmental disorders. We show that an acute immune insult during late gestation can alter fractalkine signaling by reducing the microglial CX3CR1 protein expression, highlighting neuron-microglial fractalkine signaling as a relevant target underlying the outcomes of environmental risk factors on neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lourdes Fernández de Cossío
- Department of Neurosciences, University of California, La Jolla, CA, USA; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
| | - Chloé Lacabanne
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada
| | - Garance Castino
- Department of Biology, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | | | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Colombia, Vancouver, BC, Canada.
| |
Collapse
|
143
|
Hameete BC, Fernández-Calleja JM, de Groot MW, Oppewal TR, Tiemessen MM, Hogenkamp A, de Vries RB, Groenink L. The poly(I:C)-induced maternal immune activation model; a systematic review and meta-analysis of cytokine levels in the offspring. Brain Behav Immun Health 2021; 11:100192. [PMID: 34589729 PMCID: PMC8474626 DOI: 10.1016/j.bbih.2020.100192] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022] Open
Abstract
The maternal polyinosinic:polycytidylic acid (poly(I:C)) animal model is frequently used to study how maternal immune activation may impact neuro development in the offspring. Here, we present the first systematic review and meta-analysis on the effects of maternal poly(I:C) injection on immune mediators in the offspring and provide an openly accessible systematic map of the data including methodological characteristics. Pubmed and EMBASE were searched for relevant publications, yielding 45 unique papers that met inclusion criteria. We extracted data on immune outcomes and methodological characteristics, and assessed the risk of bias. The descriptive summary showed that most studies reported an absence of effect, with an equal number of studies reporting an increase or decrease in the immune mediator being studied. Meta-analysis showed increased IL-6 concentrations in the offspring of poly(I:C) exposed mothers. This effect appeared larger prenatally than post-weaning. Furthermore, poly(I:C) administration during mid-gestation was associated with higher IL-6 concentrations in the offspring. Maternal poly(I:C) induced changes in IL-1β, Il-10 and TNF-α concentrations were small and could not be associated with age of offspring, gestational period or sampling location. Finally, quality of reporting of potential measures to minimize bias was low, which stresses the importance of adherence to publication guidelines. Since neurodevelopmental disorders in humans tend to be associated with lifelong changes in cytokine concentrations, the absence of these effects as identified in this systematic review may suggest that combining the model with other etiological factors in future studies may provide further insight in the mechanisms through which maternal immune activation affects neurodevelopment. Long-term effects of maternal poly(I:C) on immune mediators in the offspring appear limited. Prenatal measurements and mid gestation poly(I:C) injection are associated with increases in IL-6 concentrations. Variety in methodological conduct hampers identification of key elements that affect cytokine concentrations. The quality of reporting of potential measures to minimize bias is poor.
Collapse
Affiliation(s)
- Bart C. Hameete
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - José M.S. Fernández-Calleja
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Martje W.G.D.M. de Groot
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Titia Rixt Oppewal
- University College Utrecht (UCU), Campusplein 1, Utrecht, 3584 ED, the Netherlands
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, the Netherlands
| | - Machteld M. Tiemessen
- Research & Innovation, GCoE Immunology, Danone Nutricia Research, Uppsalalaan 12, Utrecht, 3584 CT, the Netherlands
| | - Astrid Hogenkamp
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Rob B.M. de Vries
- SYstematic Review Center for Laboratory (Animal) Experimentation, Department for Health Evidence, Radboud University Medical Center, Geert Grooteplein zuid 10, Nijmegen, 6525 GA, the Netherlands
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
- Corresponding author.
| |
Collapse
|
144
|
Cohrs G, Blumenröther AK, Sürie JP, Synowitz M, Held-Feindt J, Knerlich-Lukoschus F. Fetal and perinatal expression profiles of proinflammatory cytokines in the neuroplacodes of rats with myelomeningoceles: A contribution to the understanding of secondary spinal cord injury in open spinal dysraphism. J Neurotrauma 2021; 38:3376-3392. [PMID: 34541905 DOI: 10.1089/neu.2021.0091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The cellular and molecular mechanisms that presumably underlie the progressive functional decline of the myelomeningocele (MMC) placode are not well understood. We previously identified key players in posttraumatic spinal cord injury cascades in human MMC tissues obtained during postnatal repair. In this study we conducted experiments to further investigate these mediators in the prenatal time course under standardized conditions in a retinoic-acid-induced MMC rat model. A retinoic acid MMC model was established using time-dated Sprague-Dawley rats, which were gavage-fed with all-trans retinoic acid (RA; 60 mg/kg) dissolved in olive oil at E10. Control animals received olive oil only. Fetuses from both groups were obtained at E16, E18, E22. The spinal cords (SCs) of both groups were formalin-fixed or snap-frozen. Tissues were screened by real-time RT-PCR for the expression of cytokines and chemokines known to play a role in the lesion cascades of the central nervous system after trauma. MMC placodes exhibited inflammatory cells and glial activation in the later gestational stages. At the mRNA level, IL-1b, TNFa, and TNF-R1 exhibited significant induction at E22. IL1-R1 mRNA was induced significantly at E16 and E22. Double labeling experiments confirmed the costaining of these cytokines and their receptors with Iba1 (i.e., inflammatory cells), Vimentin, and Nestin in different anatomical SC areas and NeuN in ventral horn neurons. CXCL12 mRNA was elevated in control and MMC animals at E16 compared to E18 and E22. CX3CL1 mRNA was lower in MMC tissues than in control tissues on E16. The presented findings contribute to the concept that pathophysiological mechanisms, such as cytokine induction in the neuroplacode, in addition to the "first hit", promote secondary spinal cord injury with functional loss in the late fetal time course. Furthermore, these mediators should be taken into consideration in the development of new therapeutic approaches for open spinal dysraphism.
Collapse
Affiliation(s)
- Gesa Cohrs
- Universitatsklinikum Schleswig-Holstein Campus Kiel, 15056, Dept. of Neurosurgery, Arnold-Heller-Straße 3, Kiel, Germany, 24105;
| | - Ann-Kathrin Blumenröther
- Universitätsklinikum Schleswig-Holstein, 54186, Neurosurgery, Kiel, Schleswig-Holstein, Germany;
| | - Jan-Philip Sürie
- Universitätsklinikum Schleswig-Holstein, 54186, Neurosurgery, Kiel, Schleswig-Holstein, Germany;
| | - Michael Synowitz
- Universitatsklinikum Schleswig-Holstein Campus Kiel, 15056, Neurosurgery, Kiel, Schleswig-Holstein, Germany;
| | - Janka Held-Feindt
- Universitatsklinikum Schleswig-Holstein Campus Kiel, 15056, Neurosurgery, Kiel, Schleswig-Holstein, Germany;
| | - Friederike Knerlich-Lukoschus
- Universitätsklinikum Schleswig-Holstein, 54186, Neurosurgery, Kiel, Schleswig-Holstein, Germany.,Asklepios Kinderklinik Sankt Augustin, 248587, Pediatric Neurosurgery, Sankt Augustin, Nordrhein-Westfalen, Germany;
| |
Collapse
|
145
|
Maternal Mid-Gestation Cytokine Dysregulation in Mothers of Children with Autism Spectrum Disorder. J Autism Dev Disord 2021; 52:3919-3932. [PMID: 34505185 PMCID: PMC9349096 DOI: 10.1007/s10803-021-05271-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Autism spectrum disorder (ASD) is a developmental disorder characterised by deficits in social interactions and communication, with stereotypical and repetitive behaviours. Recent evidence suggests that maternal immune dysregulation may predispose offspring to ASD. Independent samples t-tests revealed downregulation of IL-17A concentrations in cases, when compared to controls, at both 15 weeks (p = 0.02), and 20 weeks (p = 0.02), which persisted at 20 weeks following adjustment for confounding variables. This adds to the growing body of evidence that maternal immune regulation may play a role in foetal neurodevelopment.
Collapse
|
146
|
Turner AD, Sullivan T, Drury K, Hall TA, Williams CN, Guilliams KP, Murphy S, Iqbal O’Meara AM. Cognitive Dysfunction After Analgesia and Sedation: Out of the Operating Room and Into the Pediatric Intensive Care Unit. Front Behav Neurosci 2021; 15:713668. [PMID: 34483858 PMCID: PMC8415404 DOI: 10.3389/fnbeh.2021.713668] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
In the midst of concerns for potential neurodevelopmental effects after surgical anesthesia, there is a growing awareness that children who require sedation during critical illness are susceptible to neurologic dysfunctions collectively termed pediatric post-intensive care syndrome, or PICS-p. In contrast to healthy children undergoing elective surgery, critically ill children are subject to inordinate neurologic stress or injury and need to be considered separately. Despite recognition of PICS-p, inconsistency in techniques and timing of post-discharge assessments continues to be a significant barrier to understanding the specific role of sedation in later cognitive dysfunction. Nonetheless, available pediatric studies that account for analgesia and sedation consistently identify sedative and opioid analgesic exposures as risk factors for both in-hospital delirium and post-discharge neurologic sequelae. Clinical observations are supported by animal models showing neuroinflammation, increased neuronal death, dysmyelination, and altered synaptic plasticity and neurotransmission. Additionally, intensive care sedation also contributes to sleep disruption, an important and overlooked variable during acute illness and post-discharge recovery. Because analgesia and sedation are potentially modifiable, understanding the underlying mechanisms could transform sedation strategies to improve outcomes. To move the needle on this, prospective clinical studies would benefit from cohesion with regard to datasets and core outcome assessments, including sleep quality. Analyses should also account for the wide range of diagnoses, heterogeneity of this population, and the dynamic nature of neurodevelopment in age cohorts. Much of the related preclinical evidence has been studied in comparatively brief anesthetic exposures in healthy animals during infancy and is not generalizable to critically ill children. Thus, complementary animal models that more accurately "reverse translate" critical illness paradigms and the effect of analgesia and sedation on neuropathology and functional outcomes are needed. This review explores the interactive role of sedatives and the neurologic vulnerability of critically ill children as it pertains to survivorship and functional outcomes, which is the next frontier in pediatric intensive care.
Collapse
Affiliation(s)
- Ashley D. Turner
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - Travis Sullivan
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Kurt Drury
- Department of Pediatrics, Division of Pediatric Critical Care, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Trevor A. Hall
- Department of Pediatrics, Division of Pediatric Psychology, Pediatric Critical Care and Neurotrauma Recovery Program, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Cydni N. Williams
- Department of Pediatrics, Division of Pediatric Critical Care, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
| | - Kristin P. Guilliams
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
- Division of Pediatric Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, MO, United States
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Sarah Murphy
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - A. M. Iqbal O’Meara
- Department of Pediatrics, Child Health Research Institute, Children’s Hospital of Richmond at Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
147
|
Xiao F, Zhang X, Ni P, Yu H, Gao Q, Li M, Huo P, Wei Z, Wang S, Zhang Y, Zhao R, Li A, Li Z, Li Y, Cheng H, Du L, Ren S, Yu Q, Liu Y, Zhao Y. Voltage-dependent potassium channel Kv4.2 alleviates the ischemic stroke impairments through activating neurogenesis. Neurochem Int 2021; 150:105155. [PMID: 34384853 DOI: 10.1016/j.neuint.2021.105155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 10/20/2022]
Abstract
As well as their ion transportation function, the voltage-dependent potassium channels could act as the cell signal inducer in a variety of pathogenic processes. However, their roles in neurogenesis after stroke insults have not been clearly illustrated. In our preliminary study, the expressions of voltage-dependent potassium channels Kv4.2 was significantly decreased after stroke in cortex, striatum and hippocampus by real-time quantitative PCR assay. To underlie the neuroprotection of Kv4.2 in stroke rehabilitation, recombinant plasmids encoding the cDNAs of mouse Kv4.2 was constructed. Behavioral tests showed that the increased Kv4.2 could be beneficial to the recovery of the sensory, the motor functions and the cognitive deficits after stroke. Temozolomide (TMZ), an inhibitor of neurogenesis, could partially abolish the mentioned protections of Kv4.2. The immunocytochemical staining showed that Kv4.2 could promote the proliferations of neural stem cells and induce the neural stem cells to differentiate into neurons in vitro and in vivo. And Kv4.2 could up-regulate the expressions of ERK1/2, p-ERK1/2, p-STAT3, NGF, p-TrkA, and BDNF, CAMKII and the concentration of intracellular Ca2+. Namely, we concluded that Kv4.2 promoted neurogenesis through ERK1/2/STAT3, NGF/TrkA, Ca2+/CAMKII signal pathways and rescued the ischemic impairments. Kv4.2 might be a potential drug target for ischemic stroke intervention.
Collapse
Affiliation(s)
- Fuyao Xiao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Xiaojie Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Pinfei Ni
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Haibo Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Qiming Gao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Mengyao Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Peiyun Huo
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Ziwei Wei
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Sihan Wang
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Yi Zhang
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Rui Zhao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Aixue Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Zhirui Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Yuejia Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Haixiao Cheng
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Libo Du
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Suping Ren
- Beijing Institute of Transfusion Medicine, Beijing, 100850, PR China
| | - Qun Yu
- Beijing Institute of Transfusion Medicine, Beijing, 100850, PR China
| | - Yang Liu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yuming Zhao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China.
| |
Collapse
|
148
|
Abstract
Interactions between the immune system and the nervous system have been described mostly in the context of diseases. More recent studies have begun to reveal how certain immune cell-derived soluble effectors, the cytokines, can influence host behaviour even in the absence of infection. In this Review, we contemplate how the immune system shapes nervous system function and how it controls the manifestation of host behaviour. Interactions between these two highly complex systems are discussed here also in the context of evolution, as both may have evolved to maximize an organism's ability to respond to environmental threats in order to survive. We describe how the immune system relays information to the nervous system and how cytokine signalling occurs in neurons. We also speculate on how the brain may be hardwired to receive and process information from the immune system. Finally, we propose a unified theory depicting a co-evolution of the immune system and host behaviour in response to the evolutionary pressure of pathogens.
Collapse
|
149
|
Cui J, Song W, Jin Y, Xu H, Fan K, Lin D, Hao Z, Lin J. Research Progress on the Mechanism of the Acupuncture Regulating Neuro-Endocrine-Immune Network System. Vet Sci 2021; 8:149. [PMID: 34437474 PMCID: PMC8402722 DOI: 10.3390/vetsci8080149] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
As one of the conventional treatment methods, acupuncture is an indispensable component of Traditional Chinese Medicine. Currently, acupuncture has been partly accepted throughout the world, but the mechanism of acupuncture is still unclear. Since the theory of the neuro-endocrine-immune network was put forward, new insights have been brought into the understanding of the mechanism of acupuncture. Studies have proven that acupuncture is a mechanical stimulus that can activate local cell functions and neuroreceptors. It also regulates the release of related biomolecules (peptide hormones, lipid hormones, neuromodulators and neurotransmitters, and other small and large biomolecules) in the microenvironment, where they can affect each other and further activate the neuroendocrine-immune network to achieve holistic regulation. Recently, growing efforts have been made in the research on the mechanism of acupuncture. Some researchers have transitioned from studying the mechanism of acupuncture as a single linear pathway to using systems approaches, including metabolomics, genomics, proteomics and biological pathway analysis. This review summarizes the research progress on the neuro-endocrine-immune network related mechanism of acupuncture and discusses its current challenges and future directions.
Collapse
Affiliation(s)
- Jingwen Cui
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, Beijing 100193, China
| | - Wanrong Song
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, Beijing 100193, China
| | - Yipeng Jin
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
| | - Huihao Xu
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
| | - Kai Fan
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
| | - Degui Lin
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
| | - Zhihui Hao
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, Beijing 100193, China
| | - Jiahao Lin
- College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China; (J.C.); (W.S.); (Y.J.); (H.X.); (K.F.); (D.L.)
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, Beijing 100193, China
| |
Collapse
|
150
|
Kamte YS, Chandwani MN, Michaels AC, O’Donnell LA. Neural Stem Cells: What Happens When They Go Viral? Viruses 2021; 13:v13081468. [PMID: 34452333 PMCID: PMC8402908 DOI: 10.3390/v13081468] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
Viruses that infect the central nervous system (CNS) are associated with developmental abnormalities as well as neuropsychiatric and degenerative conditions. Many of these viruses such as Zika virus (ZIKV), cytomegalovirus (CMV), and herpes simplex virus (HSV) demonstrate tropism for neural stem cells (NSCs). NSCs are the multipotent progenitor cells of the brain that have the ability to form neurons, astrocytes, and oligodendrocytes. Viral infections often alter the function of NSCs, with profound impacts on the growth and repair of the brain. There are a wide spectrum of effects on NSCs, which differ by the type of virus, the model system, the cell types studied, and the age of the host. Thus, it is a challenge to predict and define the consequences of interactions between viruses and NSCs. The purpose of this review is to dissect the mechanisms by which viruses can affect survival, proliferation, and differentiation of NSCs. This review also sheds light on the contribution of key antiviral cytokines in the impairment of NSC activity during a viral infection, revealing a complex interplay between NSCs, viruses, and the immune system.
Collapse
|