1
|
Lawrence AB, Brown SM, Bradford BM, Mabbott NA, Bombail V, Rutherford KMD. Non-neuronal brain biology and its relevance to animal welfare. Neurosci Biobehav Rev 2025; 173:106136. [PMID: 40185375 DOI: 10.1016/j.neubiorev.2025.106136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-neuronal cells constitute a significant portion of brain tissue and are seen as having key roles in brain homeostasis and responses to challenges. This review illustrates how non-neuronal biology can bring new perspectives to animal welfare through understanding mechanisms that determine welfare outcomes and highlighting interventions to improve welfare. Most obvious in this respect is the largely unrecognised relevance of neuroinflammation to animal welfare which is increasingly found to have roles in determining how animals respond to challenges. We start by introducing non-neuronal cells and review their involvement in affective states and cognition often seen as core psychological elements of animal welfare. We find that the evidence for a causal involvement of glia in cognition is currently more advanced than the corresponding evidence for affective states. We propose that translational research on affective disorders could usefully apply welfare science derived approaches for assessing affective states. Using evidence from translational research, we illustrate the involvement of non-neuronal cells and neuroinflammatory processes as mechanisms modulating resilience to welfare challenges including disease, pain, and social stress. We review research on impoverished environments and environmental enrichment which suggests that environmental conditions which improve animal welfare also improve resilience to challenges through balancing pro- and anti-inflammatory non-neuronal processes. We speculate that non-neuronal biology has relevance to animal welfare beyond neuro-inflammation including facilitating positive affective states. We acknowledge the relevance of neuronal biology to animal welfare whilst proposing that non-neuronal biology provides additional and relevant insights to improve animals' lives.
Collapse
Affiliation(s)
- Alistair B Lawrence
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Scotland's Rural College (SRUC), Edinburgh EH9 3JG, UK.
| | - Sarah M Brown
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | | | | |
Collapse
|
2
|
Mancin E, Maltecca C, Jiang J, Huang YJ, Tiezzi F. Capturing resilience from phenotypic deviations: a case study using feed consumption and whole genome data in pigs. BMC Genomics 2024; 25:1128. [PMID: 39574040 PMCID: PMC11583387 DOI: 10.1186/s12864-024-11052-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND In recent years, interest has grown in quantifying resilience in livestock by examining deviations in target phenotypes. This method is based on the idea that variability in these phenotypes reflects an animal's ability to adapt to external factors. By utilizing routinely collected time-series feed intake data in pigs, researchers can obtain a broad measure of resilience. This measure extends beyond specific conditions, capturing the impact of various unknown external factors that influence phenotype variations. Importantly, this method does not require additional phenotyping investments. Despite growing interest, the relationship between resilience indicators-calculated as deviations from longitudinally recorded target traits-and the mean of those traits remains largely unexplored. This gap raises the risk of inadvertently selecting for the mean rather than accurately capturing true resilience. Additionally, distinguishing between random phenotype fluctuations (white noise) and structural variations linked to resilience poses a challenge. With the aim of developing general resilience indicators applicable to commercial swine populations, we devised four resilience indicators utilizing daily feed consumption as the target trait. These include a canonical resilience indicator (BALnVar) and three novel ones (BAMaxArea, SPLnVar, and SPMaxArea), designed to minimize noise and ensure independence from daily feed consumption. We subsequently integrated these indicators with Whole Genome Sequencing using SLEMM algorithm, data from 1,250 animals to assess their efficacy in capturing resilience and their independence from the mean of daily feed consumption. RESULTS Our findings revealed that conventional resilience indicators failed to differentiate from the mean of daily feed consumption, underscoring potential limitations in accurately capturing true resilience. Notably, significant associations involving conventional resilience indicators were identified on chromosome 1, which is commonly linked to body weight. CONCLUSION We observed that deviations in feed consumption can effectively serve as indicators for selecting resilience in commercial pig farming, as confirmed by the identification of genes such as PKN1 and GYPC. However, the identification of other genes, such as RNF152, related to growth, suggests that common resilience quantification methods may be more closely related to the mean of daily feed consumption rather than capturing true resilience.
Collapse
Affiliation(s)
- Enrico Mancin
- Department of Agronomy, Natural Resources, Animals and Environment, (DAFNAE), University of Padova, Viale del Università 14, Legnaro (Padova), Food, 35020, Italy
| | - Christian Maltecca
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Piazzale delle Cascine 18, Firenze, 50144, Italy
| | - Jicaj Jiang
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
| | - Yi Jian Huang
- Smithfield Premium Genetics, Rose Hill, NC, 28458, USA
| | - Francesco Tiezzi
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Piazzale delle Cascine 18, Firenze, 50144, Italy.
| |
Collapse
|
3
|
Louie AY, Drnevich J, Johnson JL, Woodard M, Kukekova AV, Johnson RW, Steelman AJ. Respiratory infection with influenza A virus delays remyelination and alters oligodendrocyte metabolism. iScience 2024; 27:110464. [PMID: 39104416 PMCID: PMC11298649 DOI: 10.1016/j.isci.2024.110464] [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: 02/09/2024] [Revised: 05/31/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Peripheral viral infection disrupts oligodendrocyte (OL) homeostasis such that endogenous remyelination may be affected. Here, we demonstrate that influenza A virus infection perpetuated a demyelination- and disease-associated OL phenotype following cuprizone-induced demyelination that resulted in delayed OL maturation and remyelination in the prefrontal cortex. Furthermore, we assessed cellular metabolism ex vivo, and found that infection altered brain OL and microglia metabolism in a manner that opposed the metabolic profile induced by remyelination. Specifically, infection increased glycolytic capacity of OLs and microglia, an effect that was recapitulated by lipopolysaccharide (LPS) stimulation of mixed glia cultures. In contrast, mitochondrial dependence was increased in OLs during remyelination, which was similarly observed in OLs of myelinating P14 mice compared to adult and aged mice. Collectively, our data indicate that respiratory viral infection is capable of suppressing remyelination, and suggest that metabolic dysfunction of OLs is implicated in remyelination impairment.
Collapse
Affiliation(s)
- Allison Y. Louie
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jennifer L. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Meagan Woodard
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Anna V. Kukekova
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rodney W. Johnson
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Andrew J. Steelman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
4
|
Rodriguez-Zas SL, Nowak RA, Antonson AM, Rund L, Bhamidi S, Gomez AN, Southey BR, Johnson RW. Immune and metabolic challenges induce changes in pain sensation and related pathways in the hypothalamus. Physiol Genomics 2024; 56:343-359. [PMID: 38189117 PMCID: PMC11283907 DOI: 10.1152/physiolgenomics.00134.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/06/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024] Open
Abstract
The hypothalamic molecular processes participate in the regulation of the neuro-immune-endocrine system, including hormone, metabolite, chemokine circulation, and corresponding physiological and behavioral responses. RNA-sequencing profiles were analyzed to understand the effect of juvenile immune and metabolic distress 100 days after virally elicited maternal immune activation during gestation in pigs. Over 1,300 genes exhibited significant additive or interacting effects of gestational immune activation, juvenile distress, and sex. One-third of these genes presented multiple effects, emphasizing the complex interplay of these factors. Key functional categories enriched among affected genes included sensory perception of pain, steroidogenesis, prolactin, neuropeptide, and inflammatory signaling. These categories underscore the intricate relationship between gestational immune activation during gestation, distress, and the response of hypothalamic pathways to insults. These effects were sex-dependent for many genes, such as Prdm12, Oprd1, Isg20, Prl, Oxt, and Vip. The prevalence of differentially expressed genes annotated to proinflammatory and cell cycle processes suggests potential implications for synaptic plasticity and neuronal survival. The gene profiles affected by immune activation, distress, and sex pointed to the action of transcription factors SHOX2, STAT1, and REST. These findings underscore the importance of considering sex and postnatal challenges when studying causes of neurodevelopmental disorders and highlight the complexity of the "two-hit" hypothesis in understanding their etiology. Our study furthers the understanding of the intricate molecular responses in the hypothalamus to gestational immune activation and subsequent distress, shedding light on the sex-specific effects and the potential long-lasting consequences on pain perception, neuroendocrine regulation, and inflammatory processes.NEW & NOTEWORTHY The interaction of infection during gestation and insults later in life influences the molecular mechanisms in the hypothalamus that participate in pain sensation. The response of the hypothalamic transcriptome varies between sexes and can also affect synapses and immune signals. The findings from this study assist in the identification of agonists or antagonists that can guide pretranslational studies to ameliorate the effects of gestational insults interacting with postnatal challenges on physiological or behavioral disorders.
Collapse
Affiliation(s)
- Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Carl R. Woese Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Romana A Nowak
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Carl R. Woese Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Adrienne M Antonson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Laurie Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Sreelaya Bhamidi
- School of Integrative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andrea N Gomez
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Rodney W Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Carl R. Woese Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| |
Collapse
|
5
|
Wangler LM, Godbout JP. Microglia moonlighting after traumatic brain injury: aging and interferons influence chronic microglia reactivity. Trends Neurosci 2023; 46:926-940. [PMID: 37723009 PMCID: PMC10592045 DOI: 10.1016/j.tins.2023.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 09/20/2023]
Abstract
Most of the individuals who experience traumatic brain injury (TBI) develop neuropsychiatric and cognitive complications that negatively affect recovery and health span. Activation of multiple inflammatory pathways persists after TBI, but it is unclear how inflammation contributes to long-term behavioral and cognitive deficits. One outcome of TBI is microglial priming and subsequent hyper-reactivity to secondary stressors, injuries, or immune challenges that further augment complications. Additionally, microglia priming with aging contributes to exaggerated glial responses to TBI. One prominent inflammatory pathway, interferon (IFN) signaling, is increased after TBI and may contribute to microglial priming and subsequent reactivity. This review discusses the contributions of microglia to inflammatory processes after TBI, as well as the influence of aging and IFNs on microglia reactivity and chronic inflammation after TBI.
Collapse
Affiliation(s)
- Lynde M Wangler
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10th Ave, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 W 10th Ave, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, 460 Medical Center Drive, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, Columbus, OH, USA.
| |
Collapse
|
6
|
Louie AY, Kim JS, Drnevich J, Dibaeinia P, Koito H, Sinha S, McKim DB, Soto-Diaz K, Nowak RA, Das A, Steelman AJ. Influenza A virus infection disrupts oligodendrocyte homeostasis and alters the myelin lipidome in the adult mouse. J Neuroinflammation 2023; 20:190. [PMID: 37596606 PMCID: PMC10439573 DOI: 10.1186/s12974-023-02862-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Recent data suggest that myelin may be altered by physiological events occurring outside of the central nervous system, which may cause changes to cognition and behavior. Similarly, peripheral infection by non-neurotropic viruses is also known to evoke changes to cognition and behavior. METHODS Mice were inoculated with saline or influenza A virus. Bulk RNA-seq, lipidomics, RT-qPCR, flow cytometry, immunostaining, and western blots were used to determine the effect of infection on OL viability, protein expression and changes to the lipidome. To determine if microglia mediated infection-induced changes to OL homeostasis, mice were treated with GW2580, an inhibitor of microglia activation. Additionally, conditioned medium experiments using primary glial cell cultures were also used to test whether secreted factors from microglia could suppress OL gene expression. RESULTS Transcriptomic and RT-qPCR analyses revealed temporal downregulation of OL-specific transcripts with concurrent upregulation of markers characteristic of cellular stress. OLs isolated from infected mice had reduced cellular expression of myelin proteins compared with those from saline-inoculated controls. In contrast, the expression of these proteins within myelin was not different between groups. Similarly, histological and immunoblotting analysis performed on various brain regions indicated that infection did not alter OL viability, but increased expression of a cellular stress marker. Shot-gun lipidomic analysis revealed that infection altered the lipid profile within the prefrontal cortex as well as in purified brain myelin and that these changes persisted after recovery from infection. Treatment with GW2580 during infection suppressed the expression of genes associated with glial activation and partially restored OL-specific transcripts to baseline levels. Finally, conditioned medium from activated microglia reduced OL-gene expression in primary OLs without altering their viability. CONCLUSIONS These findings show that peripheral respiratory viral infection with IAV is capable of altering OL homeostasis and indicate that microglia activation is likely involved in the process.
Collapse
Affiliation(s)
- Allison Y Louie
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA
| | - Justin S Kim
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 3306, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, 315 Fernst Dr. NW, Atlanta, GA, 30332, USA
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, 3516 Veterinary Medicine Basic Sciences Bldg., 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Payam Dibaeinia
- Department of Computer Science, University of Illinois at Urbana-Champaign, 201 North Goodwin Avenue, Urbana, IL, 61801, USA
| | - Hisami Koito
- Department of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi, Saitama, 350-0295, Japan
| | - Saurabh Sinha
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
- Department of Computer Science, University of Illinois at Urbana-Champaign, 201 North Goodwin Avenue, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Dr., Urbana, IL, 61801, USA
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Daniel B McKim
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
| | - Katiria Soto-Diaz
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA
| | - Romana A Nowak
- Department of Computer Science, University of Illinois at Urbana-Champaign, 201 North Goodwin Avenue, Urbana, IL, 61801, USA
- Department of Bioengineering, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL, 61801, USA
| | - Aditi Das
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA.
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 3306, IBB, Parker H. Petit Institute for Bioengineering and Biosciences, 315 Fernst Dr. NW, Atlanta, GA, 30332, USA.
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, 3516 Veterinary Medicine Basic Sciences Bldg., 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Dr., Urbana, IL, 61801, USA.
| | - Andrew J Steelman
- Neuroscience Program, 2325/21 Beckman Institute, 405 North Mathews Ave., Urbana, IL, 61801, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Dr., Urbana, IL, 61801, USA.
- Department of Bioengineering, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL, 61801, USA.
| |
Collapse
|
7
|
Saddala MS, Yang X, Tang S, Huang H. Transcriptome-wide analysis reveals core sets of transcriptional regulators of sensome and inflammation genes in retinal microglia. Genomics 2021; 113:3058-3071. [PMID: 34242709 DOI: 10.1016/j.ygeno.2021.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Retinal microglial cells (RMCs) play crucial roles in maintaining normal visual functions in a healthy eye. However, the underlying mechanisms of RMCs over-activation manifesting the alterations of sensome profile and inflammation state, which contribute to various retinal neurodegenerative diseases, remain elusive. Here, we aimed to identify the core set of sensome and pro-inflammatory genes and their regulators using transcriptome and data mining approaches. METHODS We performed paired-end RNA-sequencing in primary microglial cell cultures treated with TNFα/IFNϒ (10 ng/ml for 12 h) and PBS as a control. Gene enrichment analysis and hierarchical clustering for the differentially expressed transcripts highlight functional pathways and network perturbations. We examined overlaps of the mouse microglial gene expression profiles with the data-mined human sensome and pro-inflammatory marker genes. The core sets of sensome and pro-inflammatory genes were selected and predicted for transcription factors (TFs). The identified TFs in RNA-Seq are validated by the quantitative PCR method. RESULTS TNFα/IFNϒ induced 668 differentially expressed transcripts in retinal microglial cells relative to the control. Furthermore, gene enrichment analysis and the gene expression network revealed activated microglial genes, biological, molecular and inflammatory pathways. The overlapping analysis of the TNFα/IFNϒ-activated microglia genes and the data-mined human gene sets revealed 22 sensome and 61 pro-inflammatory genes. Based on network analysis, we determined 10 genes as the core sets of sensome and pro-inflammatory genes and predicted the top ten TFs that regulate them. The SP110, IRF1, FLI1, SP140 (sensome) and RELB, BATF2, NFKB2, TRAFD1, SP100, NFKB1 (inflammation) are differentially expressed between the TNFα/IFNϒ activated and the non-activated microglia which were validated by quantitative PCR. The outcomes indicate that these transcriptional regulators are highly expressed and may regulate the sensome and inflammatory genes of RMCs and switch them to over-activation. CONCLUSION Our results comprise a powerful, cross-species functional genomics resource for sensome and inflammation of RMCs, which may provide novel therapeutic approaches to prevent retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Madhu Sudhana Saddala
- University of Missouri School of Medicine, Columbia, Missouri, United States of America; Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xu Yang
- University of Missouri School of Medicine, Columbia, Missouri, United States of America; Aier Eye Institute, Aier Eye Hospital Group, Changsha, Hunan, China
| | - Shibo Tang
- Aier Eye Institute, Aier Eye Hospital Group, Changsha, Hunan, China
| | - Hu Huang
- University of Missouri School of Medicine, Columbia, Missouri, United States of America.
| |
Collapse
|
8
|
Comparative Analysis Identifies Similarities between the Human and Murine Microglial Sensomes. Int J Mol Sci 2021; 22:ijms22031495. [PMID: 33540859 PMCID: PMC7867338 DOI: 10.3390/ijms22031495] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/31/2022] Open
Abstract
One of the essential functions of microglia is to continuously sense changes in their environment and adapt to those changes. For this purpose, they use a set of genes termed the sensome. This sensome is comprised of the most abundantly expressed receptors on the surface of microglia. In this study, we updated previously identified mouse microglial sensome by incorporating an additional published RNAseq dataset into the data-analysis pipeline. We also identified members of the human microglial sensome using two independent human microglia RNAseq data sources. Using both the mouse and human microglia sensomes, we identified a key set of genes conserved between the mouse and human microglial sensomes as well as some differences between the species. We found a key set of 57 genes to be conserved in both mouse and human microglial sensomes. We define these genes as the “microglia core sensome”. We then analyzed expression of genes in this core sensome in five different datasets from two neurodegenerative disease models at various stages of the diseases and found that, overall, changes in the level of expression of microglial sensome genes are specific to the disease or condition studied. Our results highlight the relevance of data generated in mice for understanding the biology of human microglia, but also stress the importance of species-specific gene sets for the investigation of diseases involving microglia. Defining this microglial specific core sensome may help identify pathological changes in microglia in humans and mouse models of human disease.
Collapse
|
9
|
Traumatic Brain Injury Causes Chronic Cortical Inflammation and Neuronal Dysfunction Mediated by Microglia. J Neurosci 2021; 41:1597-1616. [PMID: 33452227 DOI: 10.1523/jneurosci.2469-20.2020] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023] Open
Abstract
Traumatic brain injury (TBI) can lead to significant neuropsychiatric problems and neurodegenerative pathologies, which develop and persist years after injury. Neuroinflammatory processes evolve over this same period. Therefore, we aimed to determine the contribution of microglia to neuropathology at acute [1 d postinjury (dpi)], subacute (7 dpi), and chronic (30 dpi) time points. Microglia were depleted with PLX5622, a CSF1R antagonist, before midline fluid percussion injury (FPI) in male mice and cortical neuropathology/inflammation was assessed using a neuropathology mRNA panel. Gene expression associated with inflammation and neuropathology were robustly increased acutely after injury (1 dpi) and the majority of this expression was microglia independent. At 7 and 30 dpi, however, microglial depletion reversed TBI-related expression of genes associated with inflammation, interferon signaling, and neuropathology. Myriad suppressed genes at subacute and chronic endpoints were attributed to neurons. To understand the relationship between microglia, neurons, and other glia, single-cell RNA sequencing was completed 7 dpi, a critical time point in the evolution from acute to chronic pathogenesis. Cortical microglia exhibited distinct TBI-associated clustering with increased type-1 interferon and neurodegenerative/damage-related genes. In cortical neurons, genes associated with dopamine signaling, long-term potentiation, calcium signaling, and synaptogenesis were suppressed. Microglial depletion reversed the majority of these neuronal alterations. Furthermore, there was reduced cortical dendritic complexity 7 dpi, reduced neuronal connectively 30 dpi, and cognitive impairment 30 dpi. All of these TBI-associated functional and behavioral impairments were prevented by microglial depletion. Collectively, these studies indicate that microglia promote persistent neuropathology and long-term functional impairments in neuronal homeostasis after TBI.SIGNIFICANCE STATEMENT Millions of traumatic brain injuries (TBIs) occur in the United States alone each year. Survivors face elevated rates of cognitive and psychiatric complications long after the inciting injury. Recent studies of human brain injury link chronic neuroinflammation to adverse neurologic outcomes, suggesting that evolving inflammatory processes may be an opportunity for intervention. Here, we eliminate microglia to compare the effects of diffuse TBI on neurons in the presence and absence of microglia and microglia-mediated inflammation. In the absence of microglia, neurons do not undergo TBI-induced changes in gene transcription or structure. Microglial elimination prevented TBI-induced cognitive changes 30 d postinjury (dpi). Therefore, microglia have a critical role in disrupting neuronal homeostasis after TBI, particularly at subacute and chronic timepoints.
Collapse
|
10
|
Kurakin A, Bredesen DE. Alzheimer's disease as a systems network disorder: chronic stress/dyshomeostasis, innate immunity, and genetics. Aging (Albany NY) 2020; 12:17815-17844. [PMID: 32957083 PMCID: PMC7585078 DOI: 10.18632/aging.103883] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/25/2020] [Indexed: 01/24/2023]
Abstract
Ineffective results of clinical trials of over 200 anti-Alzheimer's drug candidates, with a 99.6% attrition rate, suggest that the current paradigm of Alzheimer's disease (AD) may be incomplete, necessitating exploration of alternative and complementary frameworks.Using algorithms for hypothesis independent search and expert-assisted synthesis of heterogeneous data, we attempted to reconcile multimodal clinical profiles of early-stage AD patients and accumulated research data within a parsimonious framework. Results of our analysis suggest that Alzheimer's may not be a brain disease but a progressive system-level network disorder, which is driven by chronic network stress and dyshomeostasis. The latter can be caused by various endogenous and exogenous factors, such as chronic inflammatory conditions, infections, vascular dysfunction, head trauma, environmental toxicity, and immune disorders. Whether originating in the brain or on the periphery, chronic stress, toxicity, and inflammation are communicated to the central nervous system (CNS) via humoral and neural routes, preferentially targeting high-centrality regulatory nodes and circuits of the nervous system, and eventually manifesting as a neurodegenerative CNS disease.In this report, we outline an alternative perspective on AD as a systems network disorder and discuss biochemical and genetic evidence suggesting the central role of chronic tissue injury/dyshomeostasis, innate immune reactivity, and inflammation in the etiopathobiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Alexei Kurakin
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Dale E. Bredesen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA,Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
11
|
Bordeleau M, Lacabanne C, Fernández de Cossío L, Vernoux N, Savage JC, González-Ibáñez F, Tremblay MÈ. Microglial and peripheral immune priming is partially sexually dimorphic in adolescent mouse offspring exposed to maternal high-fat diet. J Neuroinflammation 2020; 17:264. [PMID: 32891154 PMCID: PMC7487673 DOI: 10.1186/s12974-020-01914-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background Maternal nutrition is critical for proper fetal development. While increased nutrient intake is essential during pregnancy, an excessive consumption of certain nutrients, like fat, can lead to long-lasting detrimental consequences on the offspring. Animal work investigating the consequences of maternal high-fat diet (mHFD) revealed in the offspring a maternal immune activation (MIA) phenotype associated with increased inflammatory signals. This inflammation was proposed as one of the mechanisms causing neuronal circuit dysfunction, notably in the hippocampus, by altering the brain-resident macrophages—microglia. However, the understanding of mechanisms linking inflammation and microglial activities to pathological brain development remains limited. We hypothesized that mHFD-induced inflammation could prime microglia by altering their specific gene expression signature, population density, and/or functions. Methods We used an integrative approach combining molecular (i.e., multiplex-ELISA, rt-qPCR) and cellular (i.e., histochemistry, electron microscopy) techniques to investigate the effects of mHFD (saturated and unsaturated fats) vs control diet on inflammatory priming, as well as microglial transcriptomic signature, density, distribution, morphology, and ultrastructure in mice. These analyses were performed on the mothers and/or their adolescent offspring at postnatal day 30. Results Our study revealed that mHFD results in MIA defined by increased circulating levels of interleukin (IL)-6 in the mothers. This phenotype was associated with an exacerbated inflammatory response to peripheral lipopolysaccharide in mHFD-exposed offspring of both sexes. Microglial morphology was also altered, and there were increased microglial interactions with astrocytes in the hippocampus CA1 of mHFD-exposed male offspring, as well as decreased microglia-associated extracellular space pockets in the same region of mHFD-exposed offspring of the two sexes. A decreased mRNA expression of the inflammatory-regulating cytokine Tgfb1 and microglial receptors Tmem119, Trem2, and Cx3cr1 was additionally measured in the hippocampus of mHFD-exposed offspring, especially in males. Conclusions Here, we described how dietary habits during pregnancy and nurturing, particularly the consumption of an enriched fat diet, can influence peripheral immune priming in the offspring. We also found that microglia are affected in terms of gene expression signature, morphology, and interactions with the hippocampal parenchyma, in a partially sexually dimorphic manner, which may contribute to the adverse neurodevelopmental outcomes on the offspring.
Collapse
Affiliation(s)
- 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.,Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Chloé Lacabanne
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | | | - Nathalie Vernoux
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Julie C Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Fernando González-Ibáñez
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - 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. .,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Ambrose N, Rodriguez M, Waters KA, Machaalani R. Microglia in the human infant brain and factors that affect expression. Brain Behav Immun Health 2020; 7:100117. [PMID: 34589874 PMCID: PMC8474518 DOI: 10.1016/j.bbih.2020.100117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
The present study reports on the microglial populations present in 34 regions of the human infant brain (1-11 months), and whether developmental parameters or extrinsic factors such as cigarette smoke exposure, prone sleeping and an upper respiratory tract infection (URTI) influence their expression. Further, we compare microglia populations amongst three sudden unexpected death in infancy (SUDI) sub-groups: explained SUDI (eSUDI, n = 7), sudden infant death syndrome (SIDS) I (n = 8) and SIDS II (n = 13). Ionised calcium binding adaptor molecule-1 (Iba1) was used to determine the morphology and area covered by microglia in a given brain region. Activation was explored using cluster-of-differentiation factor 68 (CD68) and human leukocyte antigen-DP,DQ,DR (HLA). We found regional heterogeneity in the area covered and activation status of microglia across the infant brain. The hippocampus, basal ganglia, white matter and dentate nucleus of the cerebellum showed larger areas of Iba1, while the brainstem had the smallest. Microglia in regions of the basal ganglia and cortex demonstrated positive correlations with infant developmental parameters, while in nuclei of the rostral medulla, negative correlations between microglia parameters were seen. URTI and cigarette smoke exposure were associated with a reduced microglial area in regions of the hippocampus and cortex (parietal and occipital), respectively. In the context of SIDS, a reduced microglial area was seen in SIDS II and fewer SIDS I infants demonstrated activated phenotypes in the hippocampus. Overall, we identify the distribution of microglia in the infant brain to be heterogenous, and influenced by intrinsic and extrinsic factors, and that the SIDS I group is a useful control group for future research into other infant CNS pathologies.
Collapse
Affiliation(s)
- Natalie Ambrose
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Michael Rodriguez
- Department of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Karen A. Waters
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
- Discipline of Child and Adolescent Health, Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Rita Machaalani
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
- Discipline of Child and Adolescent Health, Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
13
|
Prinz M, Jung S, Priller J. Microglia Biology: One Century of Evolving Concepts. Cell 2020; 179:292-311. [PMID: 31585077 DOI: 10.1016/j.cell.2019.08.053] [Citation(s) in RCA: 902] [Impact Index Per Article: 180.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 01/05/2023]
Abstract
Microglia were first recognized as a distinct cell population in the CNS one century ago. For a long time, they were primarily considered to be phagocytes responsible for removing debris during CNS development and disease. More recently, advances in imaging and genetics and the advent of single-cell technologies provided new insights into the much more complex and fascinating biology of microglia. The ontogeny of microglia was identified, and their functions in health and disease were better defined. Although many questions about microglia and their roles in human diseases remain unanswered, the prospect of targeting microglia for the treatment of neurological and psychiatric disorders is tantalizing.
Collapse
Affiliation(s)
- Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany; DZNE and BIH, Berlin, Germany; University of Edinburgh and UK DRI, Edinburgh, UK.
| |
Collapse
|
14
|
Schachtschneider KM, Welge ME, Auvil LS, Chaki S, Rund LA, Madsen O, Elmore MR, Johnson RW, Groenen MA, Schook LB. Altered Hippocampal Epigenetic Regulation Underlying Reduced Cognitive Development in Response to Early Life Environmental Insults. Genes (Basel) 2020; 11:genes11020162. [PMID: 32033187 PMCID: PMC7074491 DOI: 10.3390/genes11020162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is involved in learning and memory and undergoes significant growth and maturation during the neonatal period. Environmental insults during this developmental timeframe can have lasting effects on brain structure and function. This study assessed hippocampal DNA methylation and gene transcription from two independent studies reporting reduced cognitive development stemming from early life environmental insults (iron deficiency and porcine reproductive and respiratory syndrome virus (PRRSv) infection) using porcine biomedical models. In total, 420 differentially expressed genes (DEGs) were identified between the reduced cognition and control groups, including genes involved in neurodevelopment and function. Gene ontology (GO) terms enriched for DEGs were associated with immune responses, angiogenesis, and cellular development. In addition, 116 differentially methylated regions (DMRs) were identified, which overlapped 125 genes. While no GO terms were enriched for genes overlapping DMRs, many of these genes are known to be involved in neurodevelopment and function, angiogenesis, and immunity. The observed altered methylation and expression of genes involved in neurological function suggest reduced cognition in response to early life environmental insults is due to altered cholinergic signaling and calcium regulation. Finally, two DMRs overlapped with two DEGs, VWF and LRRC32, which are associated with blood brain barrier permeability and regulatory T-cell activation, respectively. These results support the role of altered hippocampal DNA methylation and gene expression in early life environmentally-induced reductions in cognitive development across independent studies.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Michael E. Welge
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Loretta S. Auvil
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Sulalita Chaki
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Laurie A. Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Monica R.P. Elmore
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Rodney W. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Martien A.M. Groenen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
- Correspondence:
| |
Collapse
|
15
|
Soto-Díaz K, Juda MB, Blackmore S, Walsh C, Steelman AJ. TAK1 inhibition in mouse astrocyte cultures ameliorates cytokine-induced chemokine production and neutrophil migration. J Neurochem 2019; 152:697-709. [PMID: 31782806 DOI: 10.1111/jnc.14930] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/21/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022]
Abstract
Systemic inflammation can exacerbate symptoms of many neurological diseases. This effect may be facilitated by glial cells of the central nervous system (CNS) that alter their transcriptional responses and up-regulate cytokine and chemokine expression which can, in turn trigger immune surveillance. In this study, we sought to determine the effects of pro-inflammatory cytokine stimulation (TNF, IL-1α, IL-1β) on astrocyte and microglia chemokine secretion. Primary cultures of astrocytes or microglia were stimulated with the recombinant cytokines and the levels of secreted chemokines were semi-quantitatively determined using a chemokine-specific proteome profiler array and densitometry. Pharmacological inhibitors were used to determine the effects of p38 MAPK, JNK, ERK1/2, NFkB, and transforming growth factor beta-associated kinase 1 (TAK1) in controlling chemokine production. Finally, neutrophil migration assays were performed to demonstrate functionality. Our data show that stimulated astrocytes secrete at least eight chemokines as a response to cytokine stimulation. These include those involved in neutrophil chemo-attraction and proved capable of promoting neutrophil migration in vitro. In contrast, microglia up-regulated few chemokines in response to cytokine stimulation and did not promote neutrophil migration. However, microglia readily secreted chemokines following stimulation with the toll-like receptor agonists. Finally, we show that both the production of chemokines and neutrophil migration resulting from cytokine stimulation of astrocytes was dependent on TAK1 signaling. Collectively, this study adds to the understanding of how astrocytes and microglia respond to stimuli and their role in promoting neutrophil migration to the CNS during inflammatory conditions.
Collapse
Affiliation(s)
- Katiria Soto-Díaz
- Neuroscience Program, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA
| | - Michal B Juda
- Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA
| | - Stephen Blackmore
- Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA
| | - Claire Walsh
- Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA
| | - Andrew J Steelman
- Neuroscience Program, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA.,Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA.,Division of Nutritional Sciences, University of Illinois Urbana-Champaign Urbana, Urbana, IL, USA
| |
Collapse
|
16
|
Caputo MP, Radlowski EC, Lawson M, Antonson A, Watson JE, Matt SM, Leyshon BJ, Das A, Johnson RW. Herring roe oil supplementation alters microglial cell gene expression and reduces peripheral inflammation after immune activation in a neonatal piglet model. Brain Behav Immun 2019; 81:455-469. [PMID: 31271868 PMCID: PMC6754775 DOI: 10.1016/j.bbi.2019.06.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/20/2019] [Accepted: 06/29/2019] [Indexed: 01/29/2023] Open
Abstract
Neonatal brain development can be disrupted by infection that results in microglial cell activation and neuroinflammation. Studies indicate that polyunsaturated fatty acids (PUFAs) and their metabolites can resolve inflammation. It is not known if dietary PUFA increases lipid metabolites in brain or reduces neuroinflammation in neonates. We hypothesized that dietary PUFAs might suppress neuroinflammation by inhibiting pro-inflammatory cytokine over-production and promoting inflammatory resolution in the periphery and brain. Piglets were obtained on postnatal day (PD) 2 and randomly assigned to herring roe oil (HRO) or control (CON) diet. HRO was included at 2 g/kg powdered diet. HRO increased DHA levels in occipital lobe and the DHA to arachidonic acid (ARA) ratio in hippocampal tissue. HRO decreased ARA metabolites in occipital lobe. HRO failed to attenuate microglial pro-inflammatory cytokine production ex vivo. HRO did not affect fever or circulating resolvin D1 levels. HRO decreased circulating neutrophils and liver inflammatory gene expression, but increased resolution marker gene expression in liver post LPS. HRO upregulated CXCL16, TGFBR1, and C1QA in microglial cells. HRO supplementation exerted beneficial effects on inflammation in the periphery, but further studies are needed to evaluate the specific effects of omega-3 supplementation on microglial cell physiology in the neonate.
Collapse
Affiliation(s)
- Megan P. Caputo
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA,Veterinary Medical Scholars Program, Office of Research and Advanced Studies, University of Illinois at Urbana-Champaign, College of Veterinary Medicine, 3505 VMBSB, 2001 South Lincoln Ave, Urbana, IL, 61802 USA
| | - Emily C. Radlowski
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Marcus Lawson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Adrienne Antonson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Josephine E. Watson
- Department of Biochemistry, School of Molecular & Cellular Biology, University of Illinois at Urbana-Champaign, 393 Morrill Hall, 505 South Goodwin Ave, Urbana, IL, 61802 USA
| | - Stephanie M. Matt
- Neuroscience Program, University of Illinois at Urbana-Champaign, 2325/21 Beckman Institute, 405 North Matthews Ave, Urbana, IL, 61801 USA
| | - Brian J. Leyshon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Aditi Das
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL 61802, USA; Department of Biochemistry, School of Molecular & Cellular Biology, University of Illinois at Urbana-Champaign, 393 Morrill Hall, 505 South Goodwin Ave, Urbana, IL 61802, USA; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, 3516 VMBSB, 2001 South Lincoln Ave, Urbana, IL 61802, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, 2325/21 Beckman Institute, 405 North Matthews Ave, Urbana, IL 61801, USA; Bioengineering Department, University of Illinois at Urbana-Champaign, 1102 Everitt Lab, MC-278, 1406 West Green St., Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Matthews Ave, M/C 251, Urbana, IL 61801, USA.
| | - Rodney W. Johnson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA,Neuroscience Program, University of Illinois at Urbana-Champaign, 2325/21 Beckman Institute, 405 North Matthews Ave, Urbana, IL, 61801 USA
| |
Collapse
|
17
|
Maternal viral infection causes global alterations in porcine fetal microglia. Proc Natl Acad Sci U S A 2019; 116:20190-20200. [PMID: 31527230 DOI: 10.1073/pnas.1817014116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Maternal infections during pregnancy are associated with increased risk of neurodevelopmental disorders, although the precise mechanisms remain to be elucidated. Previously, we established a maternal immune activation (MIA) model using swine, which results in altered social behaviors of piglet offspring. These behavioral abnormalities occurred in the absence of microglia priming. Thus, we examined fetal microglial activity during prenatal development in response to maternal infection with live porcine reproductive and respiratory syndrome virus. Fetuses were obtained by cesarean sections performed 7 and 21 d postinoculation (dpi). MIA fetuses had reduced brain weights at 21 dpi compared to controls. Furthermore, MIA microglia increased expression of major histocompatibility complex class II that was coupled with reduced phagocytic and chemotactic activity compared to controls. High-throughput gene-expression analysis of microglial-enriched genes involved in neurodevelopment, the microglia sensome, and inflammation revealed differential regulation in primary microglia and in whole amygdala tissue. Microglia density was increased in the fetal amygdala at 7 dpi. Our data also reveal widespread sexual dimorphisms in microglial gene expression and demonstrate that the consequences of MIA are sex dependent. Overall, these results indicate that fetal microglia are significantly altered by maternal viral infection, presenting a potential mechanism through which MIA impacts prenatal brain development and function.
Collapse
|
18
|
Gomes GF, Peixoto RDDF, Maciel BG, Santos KFD, Bayma LR, Feitoza Neto PA, Fernandes TN, de Abreu CC, Casseb SMM, de Lima CM, de Oliveira MA, Diniz DG, Vasconcelos PFDC, Sosthenes MCK, Diniz CWP. Differential Microglial Morphological Response, TNFα, and Viral Load in Sedentary-like and Active Murine Models After Systemic Non-neurotropic Dengue Virus Infection. J Histochem Cytochem 2019; 67:419-439. [PMID: 30924711 DOI: 10.1369/0022155419835218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Peripheral inflammatory stimuli increase proinflammatory cytokines in the bloodstream and central nervous system and activate microglial cells. Here we tested the hypothesis that contrasting environments mimicking sedentary and active lives would be associated with differential microglial morphological responses, inflammatory cytokines concentration, and virus load in the peripheral blood. For this, mice were maintained either in standard (standard environment) or enriched cages (enriched environment) and then subjected to a single (DENV1) serotype infection. Blood samples from infected animals showed higher viral loads and higher tumor necrosis factor-α (TNFα) mRNA concentrations than control subjects. Using an unbiased stereological sampling approach, we selected 544 microglia from lateral septum for microscopic 3D reconstruction. Morphological complexity contributed most to cluster formation. Infected groups exhibited significant increase in the microglia morphological complexity and number, despite the absence of dengue virus antigens in the brain. Two microglial phenotypes (type I with lower and type II with higher morphological complexity) were found in both infected and control groups. However, microglia from infected mice maintained in enriched environment showed only one morphological phenotype. Two-way ANOVA revealed that environmental changes and infection influenced type-I and II microglial morphologies and number. Environmental enrichment and infection interactions may contribute to microglial morphological change to a point that type-I and II morphological phenotypes could no longer be distinguished in infected mice from enriched environment. Significant linear correlation was found between morphological complexity and TNFα peripheral blood. Our findings demonstrated that sedentary-like and active murine models exhibited differential microglial responses and peripheral inflammation to systemic non-neurotropic infections with DENV1 virus.
Collapse
Affiliation(s)
- Giovanni Freitas Gomes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Railana Deise da Fonseca Peixoto
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Brenda Gonçalves Maciel
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Kedma Farias Dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Lohrane Rosa Bayma
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Pedro Alves Feitoza Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Taiany Nogueira Fernandes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Cintya Castro de Abreu
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | | | - Camila Mendes de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Marcus Augusto de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | | | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| |
Collapse
|
19
|
Leyshon BJ, Ji P, Caputo MP, Matt SM, Johnson RW. Dietary Iron Deficiency Impaired Peripheral Immunity but Did Not Alter Brain Microglia in PRRSV-Infected Neonatal Piglets. Front Immunol 2019; 9:3150. [PMID: 30778359 PMCID: PMC6369153 DOI: 10.3389/fimmu.2018.03150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/20/2018] [Indexed: 11/13/2022] Open
Abstract
During the postnatal period the developing brain is vulnerable to insults including nutrient insufficiency and infection that may lead to disrupted development and cognitive dysfunction. Since iron deficiency (ID) often presents with immunodeficiency, the objective of this study was to investigate peripheral viremia and inflammation as well as brain microglial phenotype and function when ID and respiratory infection occur simultaneously in a neonatal piglet model. On postnatal day 2 (PD 2) male and female piglets were assigned to one of four treatments and fed either control or ID milk replacer. On PD 8 half the pigs on each diet were inoculated with either vehicle or porcine reproductive and respiratory syndrome virus (PRRSV; P-129). Blood samples were collected prior to inoculation (PD 7) and repeated once weekly. Rectal temperature, feeding score, and sickness behavior were measured daily until PD 28. Hematocrit, hemoglobin, and serum iron were reduced by ID but not PRRSV infection. PRRSV-infected piglets displayed viremia by PD 14; however, those fed control diet had lower viral titer on PD 28, while circulating virus remained elevated in those fed an ID diet, suggesting that ID either impaired immune function necessary for viral clearance or increased viral replication. ID piglets infected with PRRSV displayed reduced sickness behavior compared to those fed control diet on PD 13-15 and 18-20. While ID piglet sickness behavior progressively worsened, piglets fed control diet displayed improved sickness score after PD 21. Microglia isolated from PRRSV piglets had increased MHCII expression and phagocytic activity ex vivo compared to uninfected piglets. ID did not alter microglial activation or phagocytic activity. Similarly, microglial cytokine expression was increased by PRRSV but unaffected by ID, in stark contrast to peripheral blood mononuclear cell (PBMC) cytokine expression, which was increased by infection and generally decreased by ID. Taken together, these data suggest that ID decreases peripheral immune function leading to increased viremia, but immune activity in the brain is protected from acute ID.
Collapse
Affiliation(s)
- Brian J Leyshon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Peng Ji
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Megan P Caputo
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Stephanie M Matt
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rodney W Johnson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
20
|
McQuade A, Coburn M, Tu CH, Hasselmann J, Davtyan H, Blurton-Jones M. Development and validation of a simplified method to generate human microglia from pluripotent stem cells. Mol Neurodegener 2018; 13:67. [PMID: 30577865 PMCID: PMC6303871 DOI: 10.1186/s13024-018-0297-x] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Microglia, the principle immune cells of the brain, play important roles in neuronal development, homeostatic function and neurodegenerative disease. Recent genetic studies have further highlighted the importance of microglia in neurodegeneration with the identification of disease risk polymorphisms in many microglial genes. To better understand the role of these genes in microglial biology and disease, we, and others, have developed methods to differentiate microglia from human induced pluripotent stem cells (iPSCs). While the development of these methods has begun to enable important new studies of microglial biology, labs with little prior stem cell experience have sometimes found it challenging to adopt these complex protocols. Therefore, we have now developed a greatly simplified approach to generate large numbers of highly pure human microglia. RESULTS iPSCs are first differentiated toward a mesodermal, hematopoietic lineage using commercially available media. Highly pure populations of non-adherent CD43+ hematopoietic progenitors are then simply transferred to media that includes three key cytokines (M-CSF, IL-34, and TGFβ-1) that promote differentiation of homeostatic microglia. This updated approach avoids the prior requirement for hypoxic incubation, complex media formulation, FACS sorting, or co-culture, thereby significantly simplifying human microglial generation. To confirm that the resulting cells are equivalent to previously developed iPSC-microglia, we performed RNA-sequencing, functional testing, and transplantation studies. Our findings reveal that microglia generated via this simplified method are virtually identical to iPS-microglia produced via our previously published approach. To also determine whether a small molecule activator of TGFβ signaling (IDE1) can be used to replace recombinant TGFβ1, further reducing costs, we examined growth kinetics and the transcriptome of cells differentiated with IDE1. These data demonstrate that a microglial cell can indeed be produced using this alternative approach, although transcriptional differences do occur that should be considered. CONCLUSION We anticipate that this new and greatly simplified protocol will enable many interested labs, including those with little prior stem cell or flow cytometry experience, to generate and study human iPS-microglia. By combining this method with other advances such as CRISPR-gene editing and xenotransplantation, the field will continue to improve our understanding of microglial biology and their important roles in human development, homeostasis, and disease.
Collapse
Affiliation(s)
- Amanda McQuade
- Department of Neurobiology & Behavior, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Sue and Bill Gross Stem Cell Research Center, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA
| | - Morgan Coburn
- Department of Neurobiology & Behavior, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Sue and Bill Gross Stem Cell Research Center, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA
| | - Christina H Tu
- Sue and Bill Gross Stem Cell Research Center, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA
| | - Jonathan Hasselmann
- Department of Neurobiology & Behavior, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Sue and Bill Gross Stem Cell Research Center, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA
| | - Hayk Davtyan
- Sue and Bill Gross Stem Cell Research Center, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA. .,Institute for Memory Impairments and Neurological Disorders, University of California, 3014 Gross Hall, 845 Health Science Rd, Irvine, CA, 92697-4545, USA.
| |
Collapse
|
21
|
Witcher KG, Bray CE, Dziabis JE, McKim DB, Benner BN, Rowe RK, Kokiko-Cochran ON, Popovich PG, Lifshitz J, Eiferman DS, Godbout JP. Traumatic brain injury-induced neuronal damage in the somatosensory cortex causes formation of rod-shaped microglia that promote astrogliosis and persistent neuroinflammation. Glia 2018; 66:2719-2736. [PMID: 30378170 DOI: 10.1002/glia.23523] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022]
Abstract
Microglia undergo dynamic structural and transcriptional changes during the immune response to traumatic brain injury (TBI). For example, TBI causes microglia to form rod-shaped trains in the cerebral cortex, but their contribution to inflammation and pathophysiology is unclear. The purpose of this study was to determine the origin and alignment of rod microglia and to determine the role of microglia in propagating persistent cortical inflammation. Here, diffuse TBI in mice was modeled by midline fluid percussion injury (FPI). Bone marrow chimerism and BrdU pulse-chase experiments revealed that rod microglia derived from resident microglia with limited proliferation. Novel data also show that TBI-induced rod microglia were proximal to axotomized neurons, spatially overlapped with dense astrogliosis, and aligned with apical pyramidal dendrites. Furthermore, rod microglia formed adjacent to hypertrophied microglia, which clustered among layer V pyramidal neurons. To better understand the contribution of microglia to cortical inflammation and injury, microglia were eliminated prior to TBI by CSF1R antagonism (PLX5622). Microglial elimination did not affect cortical neuron axotomy induced by TBI, but attenuated rod microglial formation and astrogliosis. Analysis of 262 immune genes revealed that TBI caused profound cortical inflammation acutely (8 hr) that progressed in nature and complexity by 7 dpi. For instance, gene expression related to complement, phagocytosis, toll-like receptor signaling, and interferon response were increased 7 dpi. Critically, these acute and chronic inflammatory responses were prevented by microglial elimination. Taken together, TBI-induced neuronal injury causes microglia to structurally associate with neurons, augment astrogliosis, and propagate diverse and persistent inflammatory/immune signaling pathways.
Collapse
Affiliation(s)
| | - Chelsea E Bray
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Julia E Dziabis
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Daniel B McKim
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Brooke N Benner
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, Arizona
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, Arizona
| | | | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| |
Collapse
|
22
|
Liu JYW, Matarin M, Reeves C, McEvoy AW, Miserocchi A, Thompson P, Sisodiya SM, Thom M. Doublecortin-expressing cell types in temporal lobe epilepsy. Acta Neuropathol Commun 2018; 6:60. [PMID: 30005693 PMCID: PMC6045867 DOI: 10.1186/s40478-018-0566-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/03/2018] [Indexed: 12/14/2022] Open
Abstract
Doublecortin (DCX) is widely regarded as a marker of immature and migrating neurons during development. While DCX expression persists in adults, particularly in the temporal lobe and neurogenic regions, it is unknown how seizures influence its expression. The aim of the present study was to explore the distribution and characteristics of DCX-expressing cells in surgical and postmortem samples from 40 adult and paediatric patients, with epilepsy and with or without hippocampal sclerosis (HS), compared to post mortem controls. The hippocampus (pes and body), parahippocampal gyrus, amygdala, temporal pole and temporal cortex were examined with DCX immunohistochemistry using four commercially-available DCX antibodies, labelled cells were quantified in different regions of interest as well as their co-expression with cell type specific markers (CD68, Iba1, GFAP, GFAP∂, nestin, SOX2, CD34, OLIG2, PDGFRβ, NeuN) and cell cycle marker (MCM2). Histological findings were compared with clinical data, as well as gene expression data obtained from the temporal cortex of 83 temporal lobe epilepsy cases with HS. DCX immunohistochemistry identified immature (Nestin−/NeuN−) neurons in layer II of the temporal neocortex in patients with and without epilepsy. Their number declined significantly with age but was not associated with the presence of hippocampal sclerosis, seizure semiology or memory dysfunction. DCX+ cells were prominent in the paralaminar nuclei and periamygdalar cortex and these declined with age but were not significantly associated with epilepsy history. DCX expressing cells with ramified processes were prominent in all regions, particularly in the hippocampal subgranular zone, where significantly increased numbers were observed in epilepsy samples compared to controls. DCX ramified cells co-expressed Iba1, CD68 and PDGFRβ, and less frequently MCM2, OLIG2 and SOX2, but no co-localization was observed with CD34, nestin or GFAP/GFAP ∂. Gene expression data from neocortical samples in patients with TLE and HS supported ongoing DCX expression in adults. We conclude that DCX identifies a range of morphological cell types in temporal lobe epilepsy, including immature populations, glial and microglial cell types. Their clinical relevance and biological function requires further study but we show some evidence for alteration with age and in epilepsy.
Collapse
|
23
|
Ayana R, Singh S, Pati S. Deconvolution of Human Brain Cell Type Transcriptomes Unraveled Microglia-Specific Potential Biomarkers. Front Neurol 2018; 9:266. [PMID: 29755398 PMCID: PMC5932158 DOI: 10.3389/fneur.2018.00266] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/05/2018] [Indexed: 12/19/2022] Open
Abstract
Microglial cells form a context-dependent network of brain immunoeffector cells. Despite their indispensable roles, unresolved questions exist around biomarker discovery relevant to their cellular localization, self-renewing potential, and brain developmental dynamics. To resolve the existent gap in the annotation of candidate biomarkers, we conducted a meta-analysis of brain cells using available high-throughput data sets for deciphering microglia-specific expression profiles. We have identified 3,290 significant genes specific to microglia and further selected the top 20 dysregulated genes on the basis of p-value and log2FC. To this list, we added 7 known microglia-specific markers making the candidate list comprising 27 genes for further downstream analyses. Next, we established a connectome of these potential markers with their putative protein partners, which demonstrated strong associations of upregulated genes like Dedicator of cytokinesis 2 (DOCK2) with early/mature microglial markers such as Sphingosine kinase 1 (SPHK1), CD68, and CD45. To elucidate their respective brain anatomical location, we deconvoluted the BrainSpan Atlas expression data. This analysis showed high expression of the majority of candidate genes in microglia-dense regions (Amygdala, Hippocampus, Striatum) in the postnatal brain. Furthermore, to decipher their localized expression across brain ages, we constructed a developmental dynamics map (DDM) comprising extensive gene expression profiles throughout prenatal to postnatal stages, which resulted in the discovery of novel microglia-specific gene signatures. One of the interesting readout from DDM is that all the microglia-dense regions exhibit dynamic regulation of few genes at 37 post conception week (pcw), the transition period between pre- and postnatal stages. To validate these findings and correlate them as potential biomarkers, we analyzed the expression of corresponding proteins in hESC-derived human microglia precursors. The cultured microglial precursors showed expression of Pentraxin 3 (PTX3) and SPHK1 as well as several known markers like CD68, Allograft inflammatory factor 1 (AIF1/IBA1). In summary, this study has furnished critical insights into microglia dynamics across human brain ages and cataloged potential transcriptomic fingerprints that can be further exploited for designing novel neurotherapeutics.
Collapse
Affiliation(s)
- R Ayana
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Lucknow, India
| | - Shailja Singh
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Lucknow, India.,Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Lucknow, India
| |
Collapse
|
24
|
Antonson AM, Balakrishnan B, Radlowski EC, Petr G, Johnson RW. Altered Hippocampal Gene Expression and Morphology in Fetal Piglets following Maternal Respiratory Viral Infection. Dev Neurosci 2018. [PMID: 29539630 DOI: 10.1159/000486850] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Maternal infection during pregnancy increases the risk of neurobehavioral problems in offspring. Evidence from rodent models indicates that the maternal immune response to infection can alter fetal brain development, particularly in the hippocampus. However, information on the effects of maternal viral infection on fetal brain development in gyrencephalic species is limited. Thus, the objective of this study was to assess several effects of maternal viral infection in the last one-third of gestation on hippocampal gene expression and development in fetal piglets. Pregnant gilts were inoculated with porcine reproductive and respiratory syndrome virus (PRRSV) at gestational day (GD) 76 and the fetuses were removed by cesarean section at GD 111 (3 days before anticipated parturition). The gilts infected with PRRSV had elevated plasma interleukin-6 levels and developed transient febrile and anorectic responses lasting approximately 21 days. Despite having a similar overall body weight, fetuses from the PRRSV-infected gilts had a decreased brain weight and altered hippocampal gene expression compared to fetuses from control gilts. Notably, maternal infection caused a reduction in estimated neuronal numbers in the fetal dentate gyrus and subiculum. The number of proliferative Ki-67+ cells was not altered, but the relative integrated density of GFAP+ staining was increased, in addition to an increase in GFAP gene expression, indicating astrocyte-specific gliosis. Maternal viral infection caused an increase in fetal hippocampal gene expression of the inflammatory cytokines TNF-α and IFN-γ and the myelination marker myelin basic protein. MHCII protein, a classic monocyte activation marker, was reduced in microglia, while expression of the MHCII gene was decreased in hippocampal tissue of the fetuses from PRRSV-infected gilts. Together, these data suggest that maternal viral infection at the beginning of the last trimester results in a reduction in fetal hippocampal neurons that is evident 5 weeks after infection, when fetal piglets are near full term. The neuronal reduction was not accompanied by pronounced neuroinflammation at GD 111, indicating that any activation of classic neuroinflammatory pathways by maternal viral infection, if present, is mostly resolved by parturition.
Collapse
Affiliation(s)
- Adrienne M Antonson
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Bindu Balakrishnan
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Emily C Radlowski
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Geraldine Petr
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Rodney W Johnson
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.,Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.,Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
25
|
Hanamsagar R, Bilbo SD. Environment matters: microglia function and dysfunction in a changing world. Curr Opin Neurobiol 2017; 47:146-155. [PMID: 29096243 DOI: 10.1016/j.conb.2017.10.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/21/2017] [Accepted: 10/12/2017] [Indexed: 01/29/2023]
Abstract
The immune system is our interface with the environment, and immune molecules such as cytokines and chemokines and the cells that produce them within the brain, notably microglia, are critical for normal brain development. This recognition has in recent years led to the working hypothesis that inflammatory events during pregnancy or the early postnatal period, for example, in response to infection, may disrupt the normal developmental trajectory of microglia and consequently their interactions with neurons, thereby contributing to the risk for neurological disorders. The current article outlines recent findings on the impact of diverse, pervasive environmental challenges, beyond infection, including air pollution and maternal stress; and their impact on microglial development and its broad implications for neural pathologies.
Collapse
Affiliation(s)
- Richa Hanamsagar
- Department of Pediatrics, Harvard Medical School, and Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02129, United States
| | - Staci D Bilbo
- Department of Pediatrics, Harvard Medical School, and Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02129, United States.
| |
Collapse
|
26
|
Schachtschneider KM, Schwind RM, Newson J, Kinachtchouk N, Rizko M, Mendoza-Elias N, Grippo P, Principe DR, Park A, Overgaard NH, Jungersen G, Garcia KD, Maker AV, Rund LA, Ozer H, Gaba RC, Schook LB. The Oncopig Cancer Model: An Innovative Large Animal Translational Oncology Platform. Front Oncol 2017; 7:190. [PMID: 28879168 PMCID: PMC5572387 DOI: 10.3389/fonc.2017.00190] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/10/2017] [Indexed: 12/20/2022] Open
Abstract
Despite an improved understanding of cancer molecular biology, immune landscapes, and advancements in cytotoxic, biologic, and immunologic anti-cancer therapeutics, cancer remains a leading cause of death worldwide. More than 8.2 million deaths were attributed to cancer in 2012, and it is anticipated that cancer incidence will continue to rise, with 19.3 million cases expected by 2025. The development and investigation of new diagnostic modalities and innovative therapeutic tools is critical for reducing the global cancer burden. Toward this end, transitional animal models serve a crucial role in bridging the gap between fundamental diagnostic and therapeutic discoveries and human clinical trials. Such animal models offer insights into all aspects of the basic science-clinical translational cancer research continuum (screening, detection, oncogenesis, tumor biology, immunogenicity, therapeutics, and outcomes). To date, however, cancer research progress has been markedly hampered by lack of a genotypically, anatomically, and physiologically relevant large animal model. Without progressive cancer models, discoveries are hindered and cures are improbable. Herein, we describe a transgenic porcine model—the Oncopig Cancer Model (OCM)—as a next-generation large animal platform for the study of hematologic and solid tumor oncology. With mutations in key tumor suppressor and oncogenes, TP53R167H and KRASG12D, the OCM recapitulates transcriptional hallmarks of human disease while also exhibiting clinically relevant histologic and genotypic tumor phenotypes. Moreover, as obesity rates increase across the global population, cancer patients commonly present clinically with multiple comorbid conditions. Due to the effects of these comorbidities on patient management, therapeutic strategies, and clinical outcomes, an ideal animal model should develop cancer on the background of representative comorbid conditions (tumor macro- and microenvironments). As observed in clinical practice, liver cirrhosis frequently precedes development of primary liver cancer or hepatocellular carcinoma. The OCM has the capacity to develop tumors in combination with such relevant comorbidities. Furthermore, studies on the tumor microenvironment demonstrate similarities between OCM and human cancer genomic landscapes. This review highlights the potential of this and other large animal platforms as transitional models to bridge the gap between basic research and clinical practice.
Collapse
Affiliation(s)
| | - Regina M Schwind
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | | | | | - Mark Rizko
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nasya Mendoza-Elias
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Paul Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Daniel R Principe
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Alex Park
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nana H Overgaard
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gregers Jungersen
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kelly D Garcia
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL, United States
| | - Ajay V Maker
- Department of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, United States
| | - Laurie A Rund
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Howard Ozer
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Ron C Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States.,Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| |
Collapse
|
27
|
García-Pupo L, Sánchez JR, Ratman D, Pérez-Novo C, Declerck K, De Bosscher K, Markakis MN, Beemster G, Zaldo A, Nuñez Figueredo Y, Delgado-Hernández R, Vanden Berghe W. Semi-synthetic sapogenin exerts neuroprotective effects by skewing the brain ischemia reperfusion transcriptome towards inflammatory resolution. Brain Behav Immun 2017; 64:103-115. [PMID: 28390980 DOI: 10.1016/j.bbi.2017.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 10/19/2022] Open
Abstract
Stroke represents one of the first causes of mortality and morbidity worldwide. We evaluated the therapeutic potential of a novel semi-synthetic spirosteroid sapogenin derivative "S15" in a transient middle cerebral artery occlusion (tMCAO) focal ischemia model in rat. S15-treated rats had significantly reduced infarct volumes and improved neurological functions at 24h post-reperfusion, compared with ischemia. Corresponding gene expression changes in brain were characterized by mRNA sequencing and qPCR approaches. Next, we applied geneset, pathway and transcription factor motif enrichment analysis to identify relevant signaling networks responsible for neuronal damage upon ischemia-reperfusion or neuroprotection upon pretreatment with S15. As expected, ischemia-reperfusion brain damage strongly modulates transcriptional programs associated with immune responses, increased differentiation of immune cells as well as reduced (cat)ion transport and synaptic activity. Interestingly, S15-dependent neuroprotection regulates inflammation-associated genes involved in phagosome specific resolution of tissue damage, chemotaxis and anti-inflammatory alternative activation of microglia. Altogether our transcriptome wide RNA sequencing and integrated pathway analysis provides new clues in the neuroprotective properties of a novel spirosteroid S15 or neuronal damage in rat brains subjected to ischemia, which opens new perspectives for successful treatment of stroke.
Collapse
Affiliation(s)
- Laura García-Pupo
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), BioCubaFarma, Ave 26, No. 1605 Boyeros y Puentes Grandes, CP 10600 La Habana, Cuba.
| | - Jeney Ramírez Sánchez
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), BioCubaFarma, Ave 26, No. 1605 Boyeros y Puentes Grandes, CP 10600 La Habana, Cuba.
| | - Dariusz Ratman
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, Department of Biochemistry, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Claudina Pérez-Novo
- Proteinscience, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ken Declerck
- Proteinscience, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, Medical Biotechnology Center, VIB, Department of Biochemistry, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Marios Nektarios Markakis
- Integrated Molecular Plant Physiology Research (IMPRES), Department of Biology, University of Antwerp, Campus Groenenborger, Groenenborgerlaan 171 G.U.613, 2020 Antwerp, Belgium
| | - Gerrit Beemster
- Integrated Molecular Plant Physiology Research (IMPRES), Department of Biology, University of Antwerp, Campus Groenenborger, Groenenborgerlaan 171 G.U.613, 2020 Antwerp, Belgium
| | - Armando Zaldo
- Centro de Estudios de Productos Naturales, Facultad de Química, Universidad de la Habana, Zapata s/n entre G y Carlitos Aguirre, Vedado, Plaza de la Revolución, CP 10400 La Habana, Cuba.
| | - Yanier Nuñez Figueredo
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), BioCubaFarma, Ave 26, No. 1605 Boyeros y Puentes Grandes, CP 10600 La Habana, Cuba.
| | - René Delgado-Hernández
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), BioCubaFarma, Ave 26, No. 1605 Boyeros y Puentes Grandes, CP 10600 La Habana, Cuba.
| | - Wim Vanden Berghe
- Proteinscience, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.
| |
Collapse
|
28
|
Blackmore S, Hernandez J, Juda M, Ryder E, Freund GG, Johnson RW, Steelman AJ. Influenza infection triggers disease in a genetic model of experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2017; 114:E6107-E6116. [PMID: 28696309 PMCID: PMC5544260 DOI: 10.1073/pnas.1620415114] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. Most MS patients experience periods of symptom exacerbation (relapses) followed by periods of partial recovery (remission). Interestingly, upper-respiratory viral infections increase the risk for relapse. Here, we used an autoimmune-prone T-cell receptor transgenic mouse (2D2) and a mouse-adapted human influenza virus to test the hypothesis that upper-respiratory viral infection can cause glial activation, promote immune cell trafficking to the CNS, and trigger disease. Specifically, we inoculated 2D2 mice with influenza A virus (Puerto Rico/8/34; PR8) and then monitored them for symptoms of inflammatory demyelination. Clinical and histological experimental autoimmune encephalomyelitis was observed in ∼29% of infected 2D2 mice. To further understand how peripheral infection could contribute to disease onset, we inoculated wild-type C57BL/6 mice and measured transcriptomic alterations occurring in the cerebellum and spinal cord and monitored immune cell surveillance of the CNS by flow cytometry. Infection caused temporal alterations in the transcriptome of both the cerebellum and spinal cord that was consistent with glial activation and increased T-cell, monocyte, and neutrophil trafficking to the brain at day 8 post infection. Finally, Cxcl5 expression was up-regulated in the brains of influenza-infected mice and was elevated in cerebrospinal fluid of MS patients during relapse compared with specimens acquired during remission. Collectively, these data identify a mechanism by which peripheral infection may exacerbate MS as well as other neurological diseases.
Collapse
Affiliation(s)
- Stephen Blackmore
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Jessica Hernandez
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Michal Juda
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Emily Ryder
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Gregory G Freund
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Department of Pathology, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Rodney W Johnson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801;
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
29
|
Feng PP, Deng P, Liu LH, Ai Q, Yin J, Liu Z, Wang GM. Electroacupuncture Alleviates Postoperative Cognitive Dysfunction in Aged Rats by Inhibiting Hippocampal Neuroinflammation Activated via Microglia/TLRs Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2017; 2017:6421260. [PMID: 28684969 PMCID: PMC5480055 DOI: 10.1155/2017/6421260] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/18/2017] [Accepted: 04/30/2017] [Indexed: 01/01/2023]
Abstract
Neuroinflammation has been suggested to be involved in the pathogenesis of postoperative cognitive dysfunction (POCD). Electroacupuncture (EA) is an irreplaceable method in traditional Chinese medicine that is used for treating neurodegenerative diseases in clinical and experimental studies. The aim of this study was to examine whether EA improves cognitive dysfunction caused by surgery and to investigate the pathological mechanism of TLR2 and TLR4 in the hippocampus of aged rats. A rat model of POCD was established and treated with EA or minocycline. Both EA- and minocycline-treated rats performed significantly better than untreated operated rats in spatial memory tasks of the Morris water maze (MWM) test, spending comparatively greater amounts of time in the target zone during the probe test. Additionally, decreased levels of proinflammatory cytokines (IL-1β, IL-6, TNF-α, and HMGB1) and decreased TLR2 and TLR4 protein expression in the hippocampus of EA- and minocycline-treated rats were detected. Our data suggested that EA treatment alleviated the cognition performance deficit and neuroinflammation in aged rats following surgery, which may be mediated by inhibiting the expression of hippocampal neuroinflammatory cytokines through the microglia/TLR2/4 pathway.
Collapse
Affiliation(s)
- Pei-pei Feng
- Department of Neurobiology & Acupuncture Research, The Third Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Pu Deng
- Department of Neurobiology & Acupuncture Research, The Third Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Li-hua Liu
- Department of Neurobiology & Acupuncture Research, The Third Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qi Ai
- Department of Neurobiology & Acupuncture Research, The Third Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jie Yin
- Department of Neurobiology & Acupuncture Research, The Third Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhe Liu
- Department of Neurobiology & Acupuncture Research, The Third Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Gai-mei Wang
- Medical Department for Senior Cadres, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| |
Collapse
|
30
|
Antonson AM, Radlowski EC, Lawson MA, Rytych JL, Johnson RW. Maternal viral infection during pregnancy elicits anti-social behavior in neonatal piglet offspring independent of postnatal microglial cell activation. Brain Behav Immun 2017; 59:300-312. [PMID: 27650113 DOI: 10.1016/j.bbi.2016.09.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/31/2016] [Accepted: 09/16/2016] [Indexed: 02/03/2023] Open
Abstract
Maternal infection during pregnancy increases risk for neurodevelopmental disorders and reduced stress resilience in offspring, but the mechanisms are not fully understood. We hypothesized that piglets born from gilts infected with a respiratory virus during late gestation would exhibit aberrant microglia activity, cognitive deficits and reduced sociability. Pregnant gilts were inoculated with porcine reproductive and respiratory syndrome virus (PRRSV; 5×105 TCID50 of live PRRSV) or saline at gestational day 76. Gilts infected with PRRSV exhibited fever (p<0.01) and reduced appetite (p<0.001) for 2weekspost-inoculation and were PRRSV-positive at parturition. Piglets born from infected and control gilts were weaned at postnatal day (PD) 1 and assigned to two groups. Group 1 was challenged with lipopolysaccharide (LPS, 5μg/kg body weight i.p.) or saline on PD 14 and tissues were collected. Group 2 was tested in a T-maze task to assess spatial learning and in a 3-chamber arena with unfamiliar conspecifics to assess social behavior from PD 14-27. Microglia (CD11b+ CD45low) isolated from Group 2 piglets at PD 28 were challenged ex vivo with LPS; a subset of cells was analyzed for MHCII expression. Maternal infection did not affect offspring circulating TNFα, IL-10, or cortisol levels basally or 4h post-LPS challenge. While performance in the T-maze task was not affected by maternal infection, both sociability and preference for social novelty were decreased in piglets from infected gilts. There was no effect of maternal infection on microglial MHCII expression or LPS-induced cytokine production. Taken together, these results suggest the reduced social behavior elicited by maternal infection is not due to aberrant microglia activity postnatally.
Collapse
Affiliation(s)
- Adrienne M Antonson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Integrative Immunology and Behavior Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Emily C Radlowski
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Integrative Immunology and Behavior Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Marcus A Lawson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Integrative Immunology and Behavior Program, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jennifer L Rytych
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Integrative Immunology and Behavior Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Rodney W Johnson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA; Integrative Immunology and Behavior Program, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|