1
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [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: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
2
|
Khan H, Naseem T, Kaushik P, Narang J, Khan R, Panwar S, Parvez S. Decoding paradoxical links of cytokine markers in cognition: Cross talk between physiology, inflammaging, and Alzheimer's disease- related cognitive decline. Ageing Res Rev 2024; 101:102535. [PMID: 39374831 DOI: 10.1016/j.arr.2024.102535] [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: 07/27/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Recent research has revolutionized our understanding of memory consolidation by emphasizing the critical role of astrocytes, microglia, and immune cells in through cytokine signaling. Cytokines, compact proteins, play pivotal roles in neuronal development, synaptic transmission, and normal aging. This review explores the cellular mechanisms contributing to cognitive decline in inflammaging and Alzheimer's disease, highlighting the paradoxical effects of most studied cytokines (IL-1, IL-6, TNF-α) in brain function, which act as a double-edged sword in brain physiology, acting both as facilitators of healthy cognitive function and as a potential contributor to cognitive decline.
Collapse
Affiliation(s)
- Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Talib Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Siddharth Panwar
- School of Computing and Electrical Engineering, Indian Institute of Technology, Mandi, Himachal Pradesh 175075, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
3
|
Benarroch E. What Is the Role of Cytokines in Synaptic Transmission? Neurology 2024; 103:e209928. [PMID: 39303183 DOI: 10.1212/wnl.0000000000209928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
|
4
|
Zeng J, Pei H, Wu H, Chen W, Du R, He Z. Palmatine attenuates LPS-induced neuroinflammation through the PI3K/Akt/NF-κB pathway. J Biochem Mol Toxicol 2024; 38:e23544. [PMID: 37815058 DOI: 10.1002/jbt.23544] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 08/16/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
To investigate the key molecular mechanisms of palmatine for the treatment of neuroinflammation through modulation of a pathway using molecular docking, molecular dynamics (MD) simulation combined with network pharmacology, and animal experiments. Five alkaloid components were obtained from the traditional Chinese medicine Huangteng through literature mining. Molecular docking and MD simulation with acetylcholinesterase were used to screen palmatine. At the animal level, mice were injected with LPS intracerebrally to cause a neuroinflammatory model, and the Morris water maze experiment was performed to examine the learning memory of mice. Anxiety levels were tested using the autonomous activity behavior experiment with the open field and elevated behavior experiments. HE staining and Niss staining were performed on brain tissue sections to observe morphological lesions and apoptosis; serum was examined for inflammatory factors TNF-α, IL-6, and IL-1β; Western blot was performed to detect the protein expression. The expression of PI3K/AKT/NFkB signaling pathway-related proteins was examined by Western blot. The results of network pharmacology showed that the screening of palmatine activation containing the PI3K/Akt/NFkB signaling pathway exerts antineuroinflammatory effects. Results from behavioral experiments showed that Pal enhanced learning memory in model mice, improved anxiety behavior, and significantly improved brain damage caused by neuroinflammation. The results of HE staining and Niss staining of brain tissue sections showed that palmatine could alleviate morphological lesions and nucleus damage in brain tissue. Palmatine improved the levels of serum inflammatory factors TNF-α, IL-6, and IL-1β. SOD, MDA, CAT, ACH, and ACHE in the hippocampus were improved. Western blot results showed that palmatine administration ameliorated LPS-induced neuroinflammation through the PI3K/Akt/NFkB pathway.
Collapse
Affiliation(s)
- Jianning Zeng
- Department of Agriculture and Rural Affairs of Jilin Province Pharmacy and Pharmaceutical Science and Technology, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Hongyan Pei
- Department of Agriculture and Rural Affairs of Jilin Province Pharmacy and Pharmaceutical Science and Technology, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Hong Wu
- Department of Agriculture and Rural Affairs of Jilin Province Pharmacy and Pharmaceutical Science and Technology, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Weijia Chen
- Department of Agriculture and Rural Affairs of Jilin Province Pharmacy and Pharmaceutical Science and Technology, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Rui Du
- Department of Science and Technology of Jilin Province, Department of Traditional Chinese Medicine Industry in Xinjiang, Engineering Research Center for High-Efficiency Breeding and Product Development Technology of Sika Deer, Jilin, China
| | - Zhongmei He
- Department of Agriculture and Rural Affairs of Jilin Province Pharmacy and Pharmaceutical Science and Technology, College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, China
- Department of Science and Technology of Jilin Province, Department of Traditional Chinese Medicine Industry in Xinjiang, Engineering Research Center for High-Efficiency Breeding and Product Development Technology of Sika Deer, Jilin, China
| |
Collapse
|
5
|
Zipp F, Bittner S, Schafer DP. Cytokines as emerging regulators of central nervous system synapses. Immunity 2023; 56:914-925. [PMID: 37163992 PMCID: PMC10233069 DOI: 10.1016/j.immuni.2023.04.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/12/2023]
Abstract
Cytokines are key messengers by which immune cells communicate, and they drive many physiological processes, including immune and inflammatory responses. Early discoveries demonstrated that cytokines, such as the interleukin family members and TNF-α, regulate synaptic scaling and plasticity. Still, we continue to learn more about how these traditional immune system cytokines affect neuronal structure and function. Different cytokines shape synaptic function on multiple levels ranging from fine-tuning neurotransmission, to regulating synapse number, to impacting global neuronal networks and complex behavior. These recent findings have cultivated an exciting and growing field centered on the importance of immune system cytokines for regulating synapse and neural network structure and function. Here, we highlight the latest findings related to cytokines in the central nervous system and their regulation of synapse structure and function. Moreover, we explore how these mechanisms are becoming increasingly important to consider in diseases-especially those with a large neuroinflammatory component.
Collapse
Affiliation(s)
- Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
6
|
Neuroinflammation microenvironment sharpens seizure circuit. Neurobiol Dis 2023; 178:106027. [PMID: 36736598 DOI: 10.1016/j.nbd.2023.106027] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
A large set of inflammatory molecules and their receptors are induced in epileptogenic foci of patients with pharmacoresistant epilepsies of structural etiologies or with refractory status epilepticus. Studies in animal models mimicking these clinical conditions have shown that the activation of specific inflammatory signallings in forebrain neurons or glial cells may modify seizure thresholds, thus contributing to both ictogenesis and epileptogenesis. The search for mechanisms underlying these effects has highlighted that inflammatory mediators have CNS-specific neuromodulatory functions, in addition to their canonical activation of immune responses for pathogen recognition and clearance. This review reports the neuromodulatory effects of inflammatory mediators and how they contribute to alter the inhibitory/excitatory balance in neural networks that underlie seizures. In particular, we describe key findings related to the ictogenic role of prototypical inflammatory cytokines (IL-1β and TNF) and danger signals (HMGB1), their modulatory effects of neuronal excitability, and the mechanisms underlying these effects. It will be discussed how harnessing these neuromodulatory properties of immune mediators may lead to novel therapies to control drug-resistant seizures.
Collapse
|
7
|
Nagayach A, Bhaskar R, Patro I. Microglia activation and inflammation in hippocampus attenuates memory and mood functions during experimentally induced diabetes in rat. J Chem Neuroanat 2022; 125:102160. [PMID: 36089179 DOI: 10.1016/j.jchemneu.2022.102160] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/28/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
Incidence of cognitive and emotional alterations are reportedly two times more in diabetic patients than in non-diabetic population with hitherto unexplained causation and mechanism. Purview of the hippocampus functional diversity sanctions the accessibility and the necessity to investigate the regional neuro-immunological aspects of neurodegeneration and related functional alterations following diabetes. We examined the possible involvement of microglia activation, macrophage response, oxidative stress and inflammatory stature in both ventral and dorsal hippocampus of rats rendered diabetic by a single injection of streptozotocin (STZ; 45 mg/ kg body weight; intraperitoneal). Cognitive and behavioural alterations were studied using open field test (locomotor activity), elevated plus maze (anxiety), Barnes maze (spatial cognition) and T maze (working memory) at 2nd, 4th, 6th, 8th, 10th and 12th week post diabetic confirmation. Oxidative stress was investigated via measuring the level of lipid peroxidation biochemically. Scenario of microglia activation, macrophage response and inflammation was gauged using qualitative and quantitative analysis. Pronounced macrophage expression and activation directed microglia phenotypic switching was prominent in both ventral and dorsal hippocampus indicating the impact of oxidative stress following diabetes in hippocampus. The resultant inflammatory response was also progressive and persistent in both ventral and dorsal hippocampus parallel to the altered cognitive, locomotor ability and anxiety behaviour in diabetic rats. Conclusively, present data not only comprehends the microglia, macrophage physiology and related immune response in functionally different hippocampal regions associated cognitive and behavioural deficits, but also offers a suggestive region-specific cellular mechanism pathway for developing an imminent therapeutic approach during particular diabetes deficits.
Collapse
Affiliation(s)
- Aarti Nagayach
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, Madhya Pradesh, India; Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, South Korea
| | - Ishan Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, Madhya Pradesh, India; School of Studies in Zoology, Jiwaji University, Gwalior 474011, Madhya Pradesh, India
| |
Collapse
|
8
|
Pan JX, Sun D, Lee D, Xiong L, Ren X, Guo HH, Yao LL, Lu Y, Jung C, Xiong WC. Osteoblastic Swedish mutant APP expedites brain deficits by inducing endoplasmic reticulum stress-driven senescence. Commun Biol 2021; 4:1326. [PMID: 34824365 PMCID: PMC8617160 DOI: 10.1038/s42003-021-02843-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/03/2021] [Indexed: 11/09/2022] Open
Abstract
Patients with Alzheimer’s disease (AD) often have osteoporosis or osteopenia. However, their direct link and relationship remain largely unclear. Previous studies have detected osteoporotic deficits in young adult Tg2576 and TgAPPsweOCN mice, which express APPswe (Swedish mutant) ubiquitously and selectively in osteoblast (OB)-lineage cells. This raises the question, whether osteoblastic APPswe contributes to AD development. Here, we provide evidence that TgAPPsweOCN mice also exhibit AD-relevant brain pathologies and behavior phenotypes. Some brain pathologies include age-dependent and regional-selective increases in glial activation and pro-inflammatory cytokines, which are accompanied by behavioral phenotypes such as anxiety, depression, and altered learning and memory. Further cellular studies suggest that APPswe, but not APPwt or APPlon (London mutant), in OB-lineage cells induces endoplasmic reticulum-stress driven senescence, driving systemic and cortex inflammation as well as behavioral changes in 6-month-old TgAPPsweOCN mice. These results therefore reveal an unrecognized function of osteoblastic APPswe to brain axis in AD development. Jin-Xiu Pan et al. report that an osteoblast-specific expression of Swedish mutant amyloid precursor protein (APPswe) induces ER stress-driven senescence, leading to systemic inflammation and inflammation in the cortex that drives behavioral changes. The results demonstrate a previously unrecognized function of osteoblastic APPswe to brain axis in AD development.
Collapse
Affiliation(s)
- Jin-Xiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Hao-Han Guo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ling-Ling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yuyi Lu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Caroline Jung
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA.
| |
Collapse
|
9
|
Woodburn SC, Bollinger JL, Wohleb ES. The semantics of microglia activation: neuroinflammation, homeostasis, and stress. J Neuroinflammation 2021; 18:258. [PMID: 34742308 PMCID: PMC8571840 DOI: 10.1186/s12974-021-02309-6] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia are emerging as critical regulators of neuronal function and behavior in nearly every area of neuroscience. Initial reports focused on classical immune functions of microglia in pathological contexts, however, immunological concepts from these studies have been applied to describe neuro-immune interactions in the absence of disease, injury, or infection. Indeed, terms such as 'microglia activation' or 'neuroinflammation' are used ubiquitously to describe changes in neuro-immune function in disparate contexts; particularly in stress research, where these terms prompt undue comparisons to pathological conditions. This creates a barrier for investigators new to neuro-immunology and ultimately hinders our understanding of stress effects on microglia. As more studies seek to understand the role of microglia in neurobiology and behavior, it is increasingly important to develop standard methods to study and define microglial phenotype and function. In this review, we summarize primary research on the role of microglia in pathological and physiological contexts. Further, we propose a framework to better describe changes in microglia1 phenotype and function in chronic stress. This approach will enable more precise characterization of microglia in different contexts, which should facilitate development of microglia-directed therapeutics in psychiatric and neurological disease.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Robinson S, Mogul AS, Taylor-Yeremeeva EM, Khan A, Tirabassi AD, Wang HY. Stress Diminishes BDNF-stimulated TrkB Signaling, TrkB-NMDA Receptor Linkage and Neuronal Activity in the Rat Brain. Neuroscience 2021; 473:142-158. [PMID: 34298123 PMCID: PMC8455453 DOI: 10.1016/j.neuroscience.2021.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/21/2022]
Abstract
Exposure to intense or repeated stressors can lead to depression or post-traumatic stress disorder (PTSD). Neurological changes induced by stress include impaired neurotrophin signaling, which is known to influence synaptic integrity and plasticity. The present study used an ex vivo approach to examine the impact of acute or repeated stress on BDNF-stimulated TrkB signaling in hippocampus (HIPPO) and prefrontal cortex (PFC). Rats in an acute multiple stressor group experienced five stressors in one day whereas rats in a repeated unpredictable stressor group experienced 20 stressors across 10 days. After stress exposure, slices were incubated with vehicle or BDNF, followed by immunoprecipitation and immunoblot assays to assess protein levels, activation states and protein-protein linkage associated with BDNF-TrkB signaling. Three key findings are (1) exposure to stressors significantly diminished BDNF-stimulated TrkB signaling in HIPPO and PFC such that reductions in TrkB activation, diminished recruitment of adaptor proteins to TrkB, reduced activation of downstream signaling molecules, disruption of TrkB-NMDAr linkage, and changes in basal and BDNF-stimulated Arc expression were observed. (2) After stress, BDNF stimulation enhanced TrkB-NMDAr linkage in PFC, suggestive of compensatory mechanisms in this region. (3) We discovered an uncoupling between TrkB signaling, TrkB-NMDAr linkage and Arc expression in PFC and HIPPO. In addition, a robust surge in pro-inflammatory cytokines was observed in both regions after repeated exposure to stressors. Collectively, these data provide therapeutic targets for future studies that investigate how to reverse stress-induced downregulation of BDNF-TrkB signaling and underscore the need for functional studies that examine stress-related TrkB-NMDAr activities in PFC.
Collapse
Affiliation(s)
- Siobhan Robinson
- Department of Psychology and Program in Neuroscience, Hamilton College, Clinton, NY, USA.
| | - Allison S Mogul
- Department of Psychology and Program in Neuroscience, Hamilton College, Clinton, NY, USA
| | | | - Amber Khan
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA; Department of Biology, Neuroscience Program, Graduate School of the City University of New York, New York, NY, USA
| | - Anthony D Tirabassi
- Department of Psychology and Program in Neuroscience, Hamilton College, Clinton, NY, USA
| | - Hoau-Yan Wang
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA; Department of Biology, Neuroscience Program, Graduate School of the City University of New York, New York, NY, USA
| |
Collapse
|
11
|
Wang Y, Emre C, Gyllenhammar-Schill H, Fjellman K, Eyjolfsdottir H, Eriksdotter M, Schultzberg M, Hjorth E. Cerebrospinal Fluid Inflammatory Markers in Alzheimer's Disease: Influence of Comorbidities. Curr Alzheimer Res 2021; 18:157-170. [PMID: 33784960 DOI: 10.2174/1567205018666210330162207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/20/2021] [Accepted: 03/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) develops into dementia after several years, and subjective cognitive impairment (SCI) and mild cognitive impairment (MCI) are used as intermediary diagnoses of increasing severity. Inflammation is an important part of AD pathology and provides potential novel biomarkers and treatment targets. OBJECTIVE To identify novel potential biomarkers of AD in cerebrospinal fluid (CSF) and create a molecular pattern of inflammatory factors providing differentiation between AD and SCI. METHODS We analyzed 43 inflammatory-related mediators in CSF samples from a cohort of SCI and AD cases vetted for confounding factors (Training cohort). Using multivariate analysis (MVA), a model for discrimination between SCI and AD was produced, which we then applied to a larger nonvetted cohort (named Test cohort). The data were analyzed for factors showing differences between diagnostic groups and factors that differed between the vetted and non-vetted cohorts. The relationship of the factors to the agreement between model and clinical diagnosis was investigated. RESULTS A good MVA model able to discriminate AD from SCI without including tangle and plaque biomarkers was produced from the Training cohort. The model showed 50% agreement with clinical diagnosis in the Test cohort. Comparison of the cohorts indicated different patterns of factors distinguishing SCI from AD. As an example, soluble interleukin (IL)-6Rα showed lower levels in AD cases in the Training cohort, whereas placental growth factor (PlGF) and serum amyloid A (SAA) levels were higher in AD cases of the Test cohort. The levels of p-tau were also higher in the Training cohort. CONCLUSION This study provides new knowledge regarding the involvement of inflammation in AD by indicating different patterns of factors in CSF depending on whether potential confounding comorbidities are present or not, and presents sIL-6Rα as a potential new biomarker for improved diagnosis of AD.
Collapse
Affiliation(s)
- Ying Wang
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| | - Ceren Emre
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| | | | - Karin Fjellman
- Karolinska University Hospital, Theme Clinical Pharmacology, SE-141 86 Huddinge, Sweden
| | | | - Maria Eriksdotter
- Karolinska University Hospital, Theme Aging, SE-141 86 Huddinge, Sweden
| | - Marianne Schultzberg
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| | - Erik Hjorth
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| |
Collapse
|
12
|
Banqueri M, Martínez JA, Prieto MJ, Cid-Duarte S, Méndez M, Arias JL. Photobiomodulation rescues cognitive flexibility in early stressed subjects. Brain Res 2019; 1720:146300. [PMID: 31226326 DOI: 10.1016/j.brainres.2019.146300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
Abstract
Neglectful parenting is one of the most prevalent forms of child mistreatment. This early life stress leads to long-term alterations in all the psychological domains, as well as brain alterations. Animal models have been developed to emulate and further study this early life stress, and one of most widely used models is maternal separation. In both human and animal models, cognitive flexibility has been found to be altered. In this study, we performed maternal separation (10 days, 4 h per day) in rats, and in adulthood, we tested their spatial navigation and cognitive flexibility. In addition, we delivered photobiomodulation treatment (Low-level light therapy: 1064 nm, 30 mW, 60 cycles) on the rats' brains, and we tested energy oxidative metabolism using cytochrome c oxidase histochemistry. Early life stress delivered in the form of maternal separation on the first 10 postnatal days leads to cognitive flexibility impairment and a general increase in energy metabolism in adulthood. Low-level light therapy seems to be useful for treating these unwanted outcomes, because it rescued cognitive flexibility and returned the oxidative energy metabolism to balanced scores, without harming controls' brains or behavior. Photobiomodulation is a promising tool in the treatment of chronic stress-related consequences because it rescued cognitive flexibility.
Collapse
Affiliation(s)
- María Banqueri
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Plaza Feijoo, s/n, E-33003 Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Spain.
| | - Juan A Martínez
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Spain; Electronic Technology Area, University of Oviedo, Torres Quevedo Building (West), 2, Gijón, Spain
| | - Miguel J Prieto
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Spain; Electronic Technology Area, University of Oviedo, Torres Quevedo Building (West), 2, Gijón, Spain
| | - Sandra Cid-Duarte
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Plaza Feijoo, s/n, E-33003 Oviedo, Spain
| | - Marta Méndez
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Plaza Feijoo, s/n, E-33003 Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Spain; Electronic Technology Area, University of Oviedo, Torres Quevedo Building (West), 2, Gijón, Spain
| | - Jorge L Arias
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Plaza Feijoo, s/n, E-33003 Oviedo, Spain; Instituto de Neurociencias del Principado de Asturias (INEUROPA), Spain
| |
Collapse
|
13
|
Taoro-González L, Cabrera-Pastor A, Sancho-Alonso M, Arenas YM, Meseguer-Estornell F, Balzano T, ElMlili N, Felipo V. Differential role of interleukin-1β in neuroinflammation-induced impairment of spatial and nonspatial memory in hyperammonemic rats. FASEB J 2019; 33:9913-9928. [PMID: 31162953 DOI: 10.1096/fj.201900230rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Activated microglia and increased brain IL-1β play a main role in cognitive impairment in much pathology. We studied the role of IL-1β in neuroinflammation-induced impairment of the following different types of learning and memory: novel object recognition (NOR), novel object location (NOL), spatial learning, reference memory (RM), and working memory (WM). All these processes are impaired in hyperammonemic rats. We assessed which of these types of learning and memory are restored by blocking the IL-1 receptor in vivo in hyperammonemic rats and the possible mechanisms involved. Blocking the IL-1 receptor reversed microglial activation in the hippocampus, perirhinal cortex, and prefrontal cortex but not in the postrhinal cortex. This was associated with the restoration of NOR and WM but not of tasks involving a spatial component (NOL and RM). This suggests that IL-1β would be involved in neuroinflammation-induced nonspatial memory impairment, whereas spatial memory impairment would be IL-1β-independent and would be mediated by other proinflammatory factors.-Taoro-González, L., Cabrera-Pastor, A., Sancho-Alonso, M., Arenas, Y. M., Meseguer-Estornell, F., Balzano, T., ElMlili, N., Felipo, V. Differential role of interleukin-1β in neuroinflammation-induced impairment of spatial and nonspatial memory in hyperammonemic rats.
Collapse
Affiliation(s)
- Lucas Taoro-González
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Andrea Cabrera-Pastor
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain.,Fundación Investigación Hospital Clínico, Instituto de Investigación Sanitaria (INCLIVA), Valencia, Spain
| | - María Sancho-Alonso
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Yaiza M Arenas
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | - Tiziano Balzano
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Nisrin ElMlili
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
14
|
Zhou Y, Peng W, Wang J, Zhou W, Zhou Y, Ying B. Plasma levels of IL-1Ra are associated with schizophrenia. Psychiatry Clin Neurosci 2019; 73:109-115. [PMID: 30375100 DOI: 10.1111/pcn.12794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/20/2018] [Accepted: 10/24/2018] [Indexed: 02/05/2023]
Abstract
AIM Although peripheral low-grade inflammation and brain-derived neurotrophic factor (BDNF) levels have been implicated in schizophrenia (SCZ), the interactions between them remain to be fully revealed. We aimed to compare BDNF and cytokines in patients with SCZ and healthy controls (HC). Additionally, we aimed to investigate the association between peripheral levels of cytokines and BDNF in patients with SCZ. METHODS Plasma levels of BDNF, interferon gamma, interleukin (IL)-10, IL-12, IL-1, IL-6, IL-8, tumor necrosis factor alpha, macrophage migration inhibitory factor, IL-1 receptor antagonist (IL-1Ra), and CD40 Ligand were compared in 45 SCZ patients and 38 HC using Luminex technology. RESULTS Compared to HC, patients had significantly higher IL-1Ra levels (P = 0.031). We found a strong positive association between BDNF and CD40 Ligand in the patient group (rho = 0.858, P < 0.001) as well as in the HC group (rho = 0.822, P < 0.001), respectively. Furthermore, there was a negative association between BDNF and tumor necrosis factor alpha in patients (rho = -0.429, P = 0.030) as well as in HC (rho = -0.649, P < 0.001). CONCLUSION These results suggest that the cytokine IL-1Ra may play a role in SCZ pathophysiology. Additionally, the interaction between cytokines and BDNF levels further indicated the diverse actions of these cytokines.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wu Peng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - WenJing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - YanHong Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - BinWu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Katche C, Medina JH. Requirement of an Early Activation of BDNF/c-Fos Cascade in the Retrosplenial Cortex for the Persistence of a Long-Lasting Aversive Memory. Cereb Cortex 2018; 27:1060-1067. [PMID: 26646512 DOI: 10.1093/cercor/bhv284] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During the past few years, there has been growing interest in the role of the retrosplenial cortex (RSC) in memory processing. However, little is known about the molecular changes that take place in this brain region during memory formation. In the present work, we studied the early post-training participation of RSC in the formation of a long-lasting memory in rats. We found an increase in c-Fos levels in the anterior part of the RSC (aRSC) after inhibitory avoidance (IA) training. Interestingly, this increase was associated with memory durability, since blocking c-Fos expression using specific antisense oligonucleotides (ASO) impaired long-lasting retention 7 days after training without affecting memory expression 2 days after training. In addition, we showed that BDNF is one of the upstream signals for c-Fos expression required for memory persistence, since blocking BDNF synthesis prevents IA training-induced increase in c-Fos levels in aRSC and affects memory persistence. In addition, we found that injection of BDNF into aRSC around training was sufficient to establish a persistent memory and that this effect was prevented by c-fos ASO infusion into the same structure. These findings reveal an early post-training involvement of aRSC in the processing of a long-lasting aversive memory.
Collapse
Affiliation(s)
- Cynthia Katche
- Laboratorio de Memoria, IBCN, Facultad de Medicina, Universidad de Buenos, Ciudad Autónoma de Buenos AiresC1121ABG, Argentina
| | - Jorge H Medina
- Laboratorio de Memoria, IBCN, Facultad de Medicina, Universidad de Buenos, Ciudad Autónoma de Buenos Aires C1121ABG, Argentina.,Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos, Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| |
Collapse
|
16
|
Werneburg S, Feinberg PA, Johnson KM, Schafer DP. A microglia-cytokine axis to modulate synaptic connectivity and function. Curr Opin Neurobiol 2017; 47:138-145. [PMID: 29096242 DOI: 10.1016/j.conb.2017.10.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/25/2017] [Accepted: 10/07/2017] [Indexed: 02/05/2023]
Abstract
Microglia have recently been recognized as key regulators of synapse development, function, and plasticity. Critical to progressing the field is the identification of molecular underpinnings necessary for microglia to carry out these important functions within neural circuits. Here, we focus a review specifically on roles for microglial cytokine signaling within developing and mature neural circuits. We review exciting new studies demonstrating essential roles for microglial cytokine signaling in axon outgrowth, synaptogenesis and synapse maturation during development, as well as synaptic transmission and plasticity in adulthood. Together, these studies identify microglia and cytokines as critical modulators of neural circuits within the healthy brain, with implications for a broad range of neurological disorders with disruptions in synaptic structure and function.
Collapse
Affiliation(s)
- Sebastian Werneburg
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA
| | - Philip A Feinberg
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA
| | - Kasey M Johnson
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA
| | - Dorothy P Schafer
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester MA 01605, USA.
| |
Collapse
|
17
|
Takemiya T, Fumizawa K, Yamagata K, Iwakura Y, Kawakami M. Brain Interleukin-1 Facilitates Learning of a Water Maze Spatial Memory Task in Young Mice. Front Behav Neurosci 2017; 11:202. [PMID: 29123474 PMCID: PMC5662897 DOI: 10.3389/fnbeh.2017.00202] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 10/06/2017] [Indexed: 01/11/2023] Open
Abstract
The proinflammatory cytokine interleukin-1 (IL-1) is produced by many types of cells, including immune cells in the periphery and glia and neurons in the brain. The type I IL-1 receptor (IL-1r1) is primarily responsible for transmitting the inflammatory effects of IL-1 and mediates several biological functions by binding to either IL-1α or IL-1β. IL-1β activation is associated with hippocampus-dependent memory tasks. Although IL-1β impairs spatial memory under certain pathophysiological conditions, IL-1β may be required for the normal physiological regulation of hippocampal plasticity and memory. In addition, brain IL-1β levels are thought to change in the hippocampus in an age-dependent manner. These findings suggest that IL-1β may have a beneficial, temporary effect on learning and memory in young mice, but the matter remains unclear. Therefore, we hypothesized that hippocampal IL-1β has a beneficial effect on spatial learning and memory in young mice via IL-1r1, which is diminished in adults. We investigated the performance of young (3-month-old) and adult (6-month-old) wild-type mice, IL-1β knockout mice (IL-1βko) and IL-1r1 knockout mice (IL-1r1ko) in learning a spatial memory task with a fixed platform in a water maze (WM) and measured the levels of IL-1β and IL-1α in the hippocampus and cortex of adult and young mice by using homogeneous time-resolved fluorescence (HTRF). Learning was significantly impaired in the training trials of the WM spatial memory task in young IL-1βko and IL-1r1ko mice but not in adult IL-1βko and IL-1r1ko mice. Moreover, young IL-1r1ko mice but not IL-1βko mice showed an impairment in long-term memory extinction, suggesting that IL-1α might facilitate memory extinction. In this study, the cytokine assay using HTRF did not indicate a higher expression of hippocampal IL-1 in young mice but cortical IL-1β and IL-1α were significantly increased in adult mice. We need to investigate the role of cortical IL-1 and the local IL-1 expression in the hippocampal neurons in the future.
Collapse
Affiliation(s)
- Takako Takemiya
- Medical Research Institute, Tokyo Women's Medical University, Tokyo, Japan
| | - Kumiko Fumizawa
- Medical Research Institute, Tokyo Women's Medical University, Tokyo, Japan
| | - Kanato Yamagata
- Synaptic Plasticity Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoichiro Iwakura
- Center for Experimental Animal Models, Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Marumi Kawakami
- Medical Research Institute, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
18
|
Prieto GA, Cotman CW. Cytokines and cytokine networks target neurons to modulate long-term potentiation. Cytokine Growth Factor Rev 2017; 34:27-33. [PMID: 28377062 PMCID: PMC5491344 DOI: 10.1016/j.cytogfr.2017.03.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 03/28/2017] [Indexed: 12/20/2022]
Abstract
Cytokines play crucial roles in the communication between brain cells including neurons and glia, as well as in the brain-periphery interactions. In the brain, cytokines modulate long-term potentiation (LTP), a cellular correlate of memory. Whether cytokines regulate LTP by direct effects on neurons or by indirect mechanisms mediated by non-neuronal cells is poorly understood. Elucidating neuron-specific effects of cytokines has been challenging because most brain cells express cytokine receptors. Moreover, cytokines commonly increase the expression of multiple cytokines in their target cells, thus increasing the complexity of brain cytokine networks even after single-cytokine challenges. Here, we review evidence on both direct and indirect-mediated modulation of LTP by cytokines. We also describe novel approaches based on neuron- and synaptosome-enriched systems to identify cytokines able to directly modulate LTP, by targeting neurons and synapses. These approaches can test multiple samples in parallel, thus allowing the study of multiple cytokines simultaneously. Hence, a cytokine networks perspective coupled with neuron-specific analysis may contribute to delineation of maps of the modulation of LTP by cytokines.
Collapse
Affiliation(s)
- G Aleph Prieto
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA.
| | - Carl W Cotman
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
19
|
Tronson NC, Collette KM. (Putative) sex differences in neuroimmune modulation of memory. J Neurosci Res 2017; 95:472-486. [PMID: 27870428 PMCID: PMC5120654 DOI: 10.1002/jnr.23921] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/11/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022]
Abstract
The neuroimmune system is significantly sexually dimorphic, with sex differences evident in the number and activation states of microglia, in the activation of astrocytes, and in cytokine release and function. Neuroimmune cells and signaling are now recognized as critical for many neural functions throughout the life span, including synaptic plasticity and memory function. Here we address the question of how cytokines, astrocytes, and microglia contribute to memory, and specifically how neuroimmune modulation of memory differentially affects males and females. Understanding sex differences in both normal memory processes and dysregulation of memory in psychiatric and neurological disorders is critical for developing treatment and preventive strategies for memory disorders that are effective for both men and women. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, Michigan
| | - Katie M Collette
- Department of Psychology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
20
|
Del Rey A, Verdenhalven M, Lörwald AC, Meyer C, Hernangómez M, Randolf A, Roggero E, König AM, Heverhagen JT, Guaza C, Besedovsky HO. Brain-borne IL-1 adjusts glucoregulation and provides fuel support to astrocytes and neurons in an autocrine/paracrine manner. Mol Psychiatry 2016; 21:1309-20. [PMID: 26643538 DOI: 10.1038/mp.2015.174] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/26/2015] [Accepted: 09/29/2015] [Indexed: 12/13/2022]
Abstract
It is still controversial which mediators regulate energy provision to activated neural cells, as insulin does in peripheral tissues. Interleukin-1β (IL-1β) may mediate this effect as it can affect glucoregulation, it is overexpressed in the 'healthy' brain during increased neuronal activity, and it supports high-energy demanding processes such as long-term potentiation, memory and learning. Furthermore, the absence of sustained neuroendocrine and behavioral counterregulation suggests that brain glucose-sensing neurons do not perceive IL-1β-induced hypoglycemia. Here, we show that IL-1β adjusts glucoregulation by inducing its own production in the brain, and that IL-1β-induced hypoglycemia is myeloid differentiation primary response 88 protein (MyD88)-dependent and only partially counteracted by Kir6.2-mediated sensing signaling. Furthermore, we found that, opposite to insulin, IL-1β stimulates brain metabolism. This effect is absent in MyD88-deficient mice, which have neurobehavioral alterations associated to disorders in glucose homeostasis, as during several psychiatric diseases. IL-1β effects on brain metabolism are most likely maintained by IL-1β auto-induction and may reflect a compensatory increase in fuel supply to neural cells. We explore this possibility by directly blocking IL-1 receptors in neural cells. The results showed that, in an activity-dependent and paracrine/autocrine manner, endogenous IL-1 produced by neurons and astrocytes facilitates glucose uptake by these cells. This effect is exacerbated following glutamatergic stimulation and can be passively transferred between cell types. We conclude that the capacity of IL-1β to provide fuel to neural cells underlies its physiological effects on glucoregulation, synaptic plasticity, learning and memory. However, deregulation of IL-1β production could contribute to the alterations in brain glucose metabolism that are detected in several neurologic and psychiatric diseases.
Collapse
Affiliation(s)
- A Del Rey
- Division of Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - M Verdenhalven
- Division of Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - A C Lörwald
- Division of Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - C Meyer
- Division of Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - M Hernangómez
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain
| | - A Randolf
- Division of Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| | - E Roggero
- Instituto de Inmunologia, Facultad de Medicina, Universidad Nacional de Rosario and Universidad Abierta Interamericana, Rosario, Argentina
| | - A M König
- Centre of Imaging Research (ZebiF), University Institute of Diagnostic and Interventional Radiology, Marburg, Germany
| | - J T Heverhagen
- University Institute of Diagnostic, Interventional and Pediatric Radiology, Inselspital, University of Bern, Bern, Switzerland
| | - C Guaza
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain
| | - H O Besedovsky
- Division of Immunophysiology, Department Neurophysiology, Institute of Physiology and Pathophysiology, Marburg, Germany
| |
Collapse
|
21
|
Malashenkova IK, Krynskiy SA, Khailov NA, Kazanova GV, Velichkovsky BB, Didkovsky NA. The role of cytokines in memory consolidation. ACTA ACUST UNITED AC 2016. [DOI: 10.1134/s2079086416020055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
22
|
Malan-Müller S, Fairbairn L, Daniels WMU, Dashti MJS, Oakeley EJ, Altorfer M, Kidd M, Seedat S, Gamieldien J, Hemmings SMJ. Molecular mechanisms of D-cycloserine in facilitating fear extinction: insights from RNAseq. Metab Brain Dis 2016; 31:135-56. [PMID: 26400817 DOI: 10.1007/s11011-015-9727-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/02/2015] [Indexed: 01/24/2023]
Abstract
D-cycloserine (DCS) has been shown to be effective in facilitating fear extinction in animal and human studies, however the precise mechanisms whereby the co-administration of DCS and behavioural fear extinction reduce fear are still unclear. This study investigated the molecular mechanisms of intrahippocampally administered D-cycloserine in facilitating fear extinction in a contextual fear conditioning animal model. Male Sprague Dawley rats (n = 120) were grouped into four experimental groups (n = 30) based on fear conditioning and intrahippocampal administration of either DCS or saline. The light/dark avoidance test was used to differentiate maladapted (MA) (anxious) from well-adapted (WA) (not anxious) subgroups. RNA extracted from the left dorsal hippocampus was used for RNA sequencing and gene expression data was compared between six fear-conditioned + saline MA (FEAR + SALINE MA) and six fear-conditioned + DCS WA (FEAR + DCS WA) animals. Of the 424 significantly downregulated and 25 significantly upregulated genes identified in the FEAR + DCS WA group compared to the FEAR + SALINE MA group, 121 downregulated and nine upregulated genes were predicted to be relevant to fear conditioning and anxiety and stress-related disorders. The majority of downregulated genes transcribed immune, proinflammatory and oxidative stress systems molecules. These molecules mediate neuroinflammation and cause neuronal damage. DCS also regulated genes involved in learning and memory processes, and genes associated with anxiety, stress-related disorders and co-occurring diseases (e.g., cardiovascular diseases, digestive system diseases and nervous system diseases). Identifying the molecular underpinnings of DCS-mediated fear extinction brings us closer to understanding the process of fear extinction.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | - Lorren Fairbairn
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Willie M U Daniels
- Department of Human Physiology, University of KwaZulu-Natal, Durban, South Africa
| | | | - Edward J Oakeley
- Novartis Institutes for BioMedical Research, Biomarker Development - Human Genetics and Genomics, Genome Technologies, Basel, Switzerland
| | - Marc Altorfer
- Novartis Institutes for BioMedical Research, Biomarker Development - Human Genetics and Genomics, Genome Technologies, Basel, Switzerland
| | - Martin Kidd
- Centre for Statistical Consultation, Stellenbosch University, Stellenbosch, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Junaid Gamieldien
- University of the Western Cape, South African National Bioinformatics Institute, Cape Town, South Africa
| | - Sîan Megan Joanna Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
23
|
Honig G, Mader S, Chen H, Porat A, Ochani M, Wang P, Volpe BT, Diamond B. Blood-Brain Barrier Deterioration and Hippocampal Gene Expression in Polymicrobial Sepsis: An Evaluation of Endothelial MyD88 and the Vagus Nerve. PLoS One 2016; 11:e0144215. [PMID: 26790027 PMCID: PMC4720404 DOI: 10.1371/journal.pone.0144215] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/16/2015] [Indexed: 12/29/2022] Open
Abstract
Systemic infection can initiate or exacerbate central nervous system (CNS) pathology, even in the absence of overt invasion of bacteria into the CNS. Recent epidemiological studies have demonstrated that human survivors of sepsis have an increased risk of long-term neurocognitive decline. There is thus a need for improved understanding of the physiological mechanisms whereby acute sepsis affects the CNS. In particular, MyD88-dependent activation of brain microvascular endothelial cells and a resulting loss of blood-brain barrier integrity have been proposed to play an important role in the effects of systemic inflammation on the CNS. Signaling through the vagus nerve has also been considered to be an important component of CNS responses to systemic infection. Here, we demonstrate that blood-brain barrier permeabilization and hippocampal transcriptional responses during polymicrobial sepsis occur even in the absence of MyD88-dependent signaling in cerebrovascular endothelial cells. We further demonstrate that these transcriptional responses can occur without vagus nerve input. These results suggest that redundant signals mediate CNS responses in sepsis. Either endothelial or vagus nerve activation may be individually sufficient to transmit systemic inflammation to the central nervous system. Transcriptional activation in the forebrain in sepsis may be mediated by MyD88-independent endothelial mechanisms or by non-vagal neuronal pathways.
Collapse
Affiliation(s)
- Gerard Honig
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Simone Mader
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Huiyi Chen
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Amit Porat
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Mahendar Ochani
- Center for Translational Research, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Ping Wang
- Center for Translational Research, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Bruce T. Volpe
- Laboratory of Biomedical Science, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- * E-mail:
| |
Collapse
|
24
|
Synapse-specific IL-1 receptor subunit reconfiguration augments vulnerability to IL-1β in the aged hippocampus. Proc Natl Acad Sci U S A 2015; 112:E5078-87. [PMID: 26305968 DOI: 10.1073/pnas.1514486112] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the aged brain, synaptic plasticity and memory show increased vulnerability to impairment by the inflammatory cytokine interleukin 1β (IL-1β). In this study, we evaluated the possibility that synapses may directly undergo maladaptive changes with age that augment sensitivity to IL-1β impairment. In hippocampal neuronal cultures, IL-1β increased the expression of the IL-1 receptor type 1 and the accessory coreceptor AcP (proinflammatory), but not of the AcPb (prosurvival) subunit, a reconfiguration that potentiates the responsiveness of neurons to IL-1β. To evaluate whether synapses develop a similar heightened sensitivity to IL-1β with age, we used an assay to track long-term potentiation (LTP) in synaptosomes. We found that IL-1β impairs LTP directly at the synapse and that sensitivity to IL-1β is augmented in aged hippocampal synapses. The increased synaptic sensitivity to IL-1β was due to IL-1 receptor subunit reconfiguration, characterized by a shift in the AcP/AcPb ratio, paralleling our culture data. We suggest that the age-related increase in brain IL-1β levels drives a shift in IL-1 receptor configuration, thus heightening the sensitivity to IL-1β. Accordingly, selective blocking of AcP-dependent signaling with Toll-IL-1 receptor domain peptidomimetics prevented IL-1β-mediated LTP suppression and blocked the memory impairment induced in aged mice by peripheral immune challenge (bacterial lipopolysaccharide). Overall, this study demonstrates that increased AcP signaling, specifically at the synapse, underlies the augmented vulnerability to cognitive impairment by IL-1β that occurs with age.
Collapse
|
25
|
Estes ML, McAllister AK. Alterations in immune cells and mediators in the brain: it's not always neuroinflammation! Brain Pathol 2015; 24:623-30. [PMID: 25345893 DOI: 10.1111/bpa.12198] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/02/2023] Open
Abstract
Neuroinflammation was once a clearly defined term denoting pathological immune processes within the central nervous system (CNS). Historically, this term was used to indicate the four hallmarks of peripheral inflammaton that occur following severe CNS injuries, such as stroke, injury or infection. Recently, however, the definition of neuroinflammation has relaxed to the point that it is often now assumed to be present when even only a single classical hallmark of inflammation is measured. As a result, a wide range of disorders, from psychiatric to degenerative diseases, are now assumed to have an integral inflammatory component. Ironically, at the same time, research has revealed unexpected nonclassical immune actions of immune mediators and cells in the CNS in the absence of pathology, increasing the likelihood that homeostatic and adaptive immune processes in the CNS will be mistaken for neuroinflammation. Thus, we suggest reserving the term neuroinflammation for contexts where multiple signs of inflammation are present to avoid erroneously classifying disorders as inflammatory when they may instead be caused by nonimmune etiologies or secondary immune processes that serve adaptive roles.
Collapse
|
26
|
Abstract
Glia are starting to be accepted as the equal of neurons. Their impact on intelligence, environmental enrichment, and cerebral dominance forms the basis for understanding the role of glia in stress. Along with neurons, astrocytes, microglia, NG2 cells, and oligodendrocytes all contribute. Glia can even be protective against drug abuse. Glial effects on depression, mood disorders and schizophrenia are reviewed.
Collapse
|
27
|
Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology 2014; 96:70-82. [PMID: 25445483 DOI: 10.1016/j.neuropharm.2014.10.027] [Citation(s) in RCA: 443] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/24/2014] [Accepted: 10/29/2014] [Indexed: 01/01/2023]
Abstract
Increasing evidence underlines that prototypical inflammatory cytokines (IL-1β, TNF-α and IL-6) either synthesized in the central (CNS) or peripheral nervous system (PNS) by resident cells, or imported by immune blood cells, are involved in several pathophysiological functions, including an unexpected impact on synaptic transmission and neuronal excitability. This review describes these unconventional neuromodulatory properties of cytokines, that are distinct from their classical action as effector molecules of the immune system. In addition to the role of cytokines in brain physiology, we report evidence that dysregulation of their biosynthesis and cellular release, or alterations in receptor-mediated intracellular pathways in target cells, leads to neuronal cell dysfunction and modifications in neuronal network excitability. As a consequence, targeting of these cytokines, and related signalling molecules, is considered a novel option for the development of therapies in various CNS or PNS disorders associated with an inflammatory component. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Annamaria Vezzani
- IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Department of Neuroscience, Milano, Italy.
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy.
| |
Collapse
|
28
|
Donzis EJ, Tronson NC. Modulation of learning and memory by cytokines: signaling mechanisms and long term consequences. Neurobiol Learn Mem 2014; 115:68-77. [PMID: 25151944 PMCID: PMC4250287 DOI: 10.1016/j.nlm.2014.08.008] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 11/16/2022]
Abstract
This review describes the role of cytokines and their downstream signaling cascades on the modulation of learning and memory. Immune proteins are required for many key neural processes and dysregulation of these functions by systemic inflammation can result in impairments of memory that persist long after the resolution of inflammation. Recent research has demonstrated that manipulations of individual cytokines can modulate learning, memory, and synaptic plasticity. The many conflicting findings, however, have prevented a clear understanding of the precise role of cytokines in memory. Given the complexity of inflammatory signaling, understanding its modulatory role requires a shift in focus from single cytokines to a network of cytokine interactions and elucidation of the cytokine-dependent intracellular signaling cascades. Finally, we propose that whereas signal transduction and transcription may mediate short-term modulation of memory, long-lasting cellular and molecular mechanisms such as epigenetic modifications and altered neurogenesis may be required for the long lasting impact of inflammation on memory and cognition.
Collapse
Affiliation(s)
- Elissa J Donzis
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
29
|
Dominy SS, Brown JN, Ryder MI, Gritsenko M, Jacobs JM, Smith RD. Proteomic analysis of saliva in HIV-positive heroin addicts reveals proteins correlated with cognition. PLoS One 2014; 9:e89366. [PMID: 24717448 PMCID: PMC3981673 DOI: 10.1371/journal.pone.0089366] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 01/21/2014] [Indexed: 02/07/2023] Open
Abstract
The prevalence of HIV-associated neurocognitive disorders (HAND) remains high despite effective antiretroviral therapies. Multiple etiologies have been proposed over the last several years to account for this phenomenon, including the neurotoxic effects of antiretrovirals and co-morbid substance abuse; however, no underlying molecular mechanism has been identified. Emerging evidence in several fields has linked the gut to brain diseases, but the effect of the gut on the brain during HIV infection has not been explored. Saliva is the most accessible gut biofluid, and is therefore of great scientific interest for diagnostic and prognostic purposes. This study presents a longitudinal, liquid chromatography-mass spectrometry-based quantitative proteomics study investigating saliva samples taken from 8 HIV-positive (HIV+), 11 −negative (HIV−) heroin addicts. In addition, saliva samples were investigated from 11 HIV−, non-heroin addicted healthy controls. In the HIV+ group, 58 proteins were identified that show significant correlations with cognitive scores, implicating disruption of protein quality control pathways by HIV. Notably, only one protein from the HIV− heroin addict cohort showed a significant correlation with cognitive scores, and no proteins correlated with cognitive scores in the healthy control group. In addition, the majority of correlated proteins have been shown to be associated with exosomes, allowing us to propose that the salivary glands and/or oral epithelium may modulate brain function during HIV infection through the release of discrete packets of proteins in the form of exosomes.
Collapse
Affiliation(s)
- Stephen S. Dominy
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (SD); (RS)
| | - Joseph N. Brown
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Mark I. Ryder
- Division of Periodontology, Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Marina Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Jon M. Jacobs
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National Laboratories, Richland, Washington, United States of America
- * E-mail: (SD); (RS)
| |
Collapse
|
30
|
Hertz L, Song D, Li B, Yan E, Peng L. Importance of ‘inflammatory molecules’, but not necessarily of inflammation, in the pathophysiology of bipolar disorder and in the mechanisms of action of anti-bipolar drugs. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.npbr.2013.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
31
|
Abstract
The nervous system and the immune system are two main regulators of homeostasis in the body. Communication between them ensures normal functioning of the organism. Immune cells and molecules are required for sculpting the circuitry and determining the activity of the nervous system. Within the parenchyma of the central nervous system (CNS), microglia constantly monitor synapses and participate in their pruning during development and possibly also throughout life. Classical inflammatory cytokines, such as interleukin (IL)-1β and tumor necrosis factor (TNF), are released during neuronal activity and play a crucial role in regulating the strength of synaptic transmission. Systemically, proper functioning of the immune system is critical for maintaining normal nervous system function. Disruption of the immune system functioning leads to impairments in cognition and in neurogenesis. In this review we provide examples of the communication between the nervous and the immune systems in the interest of normal CNS development and function.
Collapse
Affiliation(s)
- Ioana Marin
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, Neuroscience Graduate Program, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|
32
|
Fidalgo AR. Experimental insights into age-exacerbated cognitive dysfunction after peripheral surgery. Aging Cell 2013; 12:523-4. [PMID: 23461580 DOI: 10.1111/acel.12066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2013] [Indexed: 01/29/2023] Open
Abstract
Here I comment on the recent contribution by Barrientos et al. J. Neurosci. 32, 14641-14648 (2012) addressing treatment possibilities for surgery-induced cognitive dysfunction. It has been over 15 years since the publication of a landmark study that indicated age as a major risk factor for postoperative cognitive dysfunction (POCD) (Moller et al., Lancet 351, 857-861 1998). With increasing life expectancy, surgical procedures conducted in elderly persons are becoming more common. The prevalence of POCD may mean that some patients will exchange the incapacitating condition that led them to surgery in the first instance for another such condition, which has been created by the surgical procedure itself. The report by Barrientos and collaborators (2012) is a timely and welcome study that further examines treatment possibilities for surgery-induced cognitive dysfunction. Future studies should address issues such as intensity and onset of inflammation within the brain and additional treatments possibilities beyond IL-1-ra.
Collapse
Affiliation(s)
- Antonio R. Fidalgo
- Research Center for Advanced Science and Technology The University of Tokyo 4‐6‐1, Komaba, Meguro‐kuTokyo 153‐8904Japan
| |
Collapse
|
33
|
Vezzani A, Friedman A, Dingledine RJ. The role of inflammation in epileptogenesis. Neuropharmacology 2013; 69:16-24. [PMID: 22521336 PMCID: PMC3447120 DOI: 10.1016/j.neuropharm.2012.04.004] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/19/2012] [Accepted: 04/02/2012] [Indexed: 12/20/2022]
Abstract
One compelling challenge in the therapy of epilepsy is to develop anti-epileptogenic drugs with an impact on the disease progression. The search for novel targets has focused recently on brain inflammation since this phenomenon appears to be an integral part of the diseased hyperexcitable brain tissue from which spontaneous and recurrent seizures originate. Although the contribution of specific proinflammatory pathways to the mechanism of ictogenesis in epileptic tissue has been demonstrated in experimental models, the role of these pathways in epileptogenesis is still under evaluation. We review the evidence conceptually supporting the involvement of brain inflammation and the associated blood-brain barrier damage in epileptogenesis, and describe the available pharmacological evidence where post-injury intervention with anti-inflammatory drugs has been attempted. Our review will focus on three main inflammatory pathways, namely the IL-1 receptor/Toll-like receptor signaling, COX-2 and the TGF-β signaling. The mechanisms underlying neuronal-glia network dysfunctions induced by brain inflammation are also discussed, highlighting novel neuromodulatory effects of classical inflammatory mediators such as cytokines and prostaglandins. The increase in knowledge about a role of inflammation in disease progression, may prompt the use of specific anti-inflammatory drugs for developing disease-modifying treatments. This article is part of the Special Issue entitled 'New Targets and Approaches to the Treatment of Epilepsy'.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Laboratory Experimental Neurology, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Via G. La Masa 19, 20156 Milano, Italy.
| | | | | |
Collapse
|
34
|
Interleukin 1 receptor antagonist and soluble tumor necrosis factor receptor 1 are associated with general severity and psychotic symptoms in schizophrenia and bipolar disorder. Schizophr Res 2013; 145:36-42. [PMID: 23403415 DOI: 10.1016/j.schres.2012.12.023] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 11/13/2012] [Accepted: 12/17/2012] [Indexed: 11/24/2022]
Abstract
BACKGROUND Previous studies suggest elevated inflammation in schizophrenia and bipolar disorder, with increased activity of the Interleukin 1 (IL-1), interleukin 6 (IL-6), tumor necrosis factor (TNF)-alpha, von Willebrand factor (vWf) and osteoprotegerin (OPG). It is unclear how immune activation is involved in the psychopathology. We investigated if elevated inflammation was associated with disease severity (trait) or current symptom level (state), comparing psychotic with general characteristics. METHODS Plasma levels of sTNF receptor 1 (sTNF-R1), IL-1 receptor antagonist (IL-1Ra), IL-6, vWf and OPG were measured with ELISA techniques in 322 patients with schizophrenia spectrum and bipolar disorder. Current symptom level (state) was measured with Global Assessment of Functioning (GAF) and Positive and Negative Syndrome Scale (PANSS). Disease severity (trait) was measured with premorbid adjustment scale (PAS), age at onset, number of psychotic episodes and number and length of hospitalizations. RESULTS After controlling for confounders, IL-1Ra and TNF-R1 were independently associated with GAF, and significantly correlated with PANSS negative and positive, respectively. In addition, Il-1Ra was associated with PAS, and sTNF-R1 with number of hospitalizations and psychotic episodes. VWf was significantly correlated with psychotic episodes, OPG with hospitalizations and IL-6 with history of psychosis. Linear regression analysis showed that GAF remained associated with sTNF-R1 and IL-1Ra with PANSS, after controlling for the other clinical measures. CONCLUSIONS This supports that inflammatory markers, particularly IL-1Ra and sTNF-R1 are associated with both general disease severity and psychotic features. This supports a role of immune activation in the core pathological mechanisms of severe mental disorders.
Collapse
|
35
|
Identification of functional circuitry between retrosplenial and postrhinal cortices during fear conditioning. J Neurosci 2012; 32:12076-86. [PMID: 22933791 DOI: 10.1523/jneurosci.2814-12.2012] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The retrosplenial cortex (RSP) and postrhinal cortex (POR) are heavily interconnected with medial temporal lobe structures involved in learning and memory. Previous studies indicate that RSP and POR are necessary for contextual fear conditioning, but it remains unclear whether these regions contribute individually or instead work together as a functional circuit to modulate learning and/or memory. In Experiment 1, learning-related neuronal activity was assessed in RSP from home cage, shock-only, context-only, or fear-conditioned rats using real-time PCR and immunohistochemical methods to quantify immediate-early gene expression. A significant increase in activity-regulated cytoskeleton-associated protein (Arc) mRNA and Arc and c-Fos protein expression was detected in RSP from fear-conditioned rats compared with all other groups. In Experiment 2, retrograde tracing combined with immunohistochemistry revealed that, compared with controls, a significant proportion of cells projecting from RSP to POR were immunopositive for c-Fos in fear-conditioned rats. These results demonstrate that neurons projecting from RSP to POR are indeed active during fear conditioning. In Experiment 3, a functional disconnection paradigm was used to further examine the interaction between RSP and POR during fear conditioning. Compared with controls, rats with unilateral lesions of RSP and POR on opposite sides of the brain exhibited impaired contextual fear memory, whereas rats with unilateral lesions in the same hemisphere displayed intermediate levels of freezing compared with controls and rats with contralateral lesions. Collectively, these results are the first to show that RSP and POR function as a cortical network necessary for contextual fear learning and memory.
Collapse
|
36
|
Fatjó-Vilas M, Pomarol-Clotet E, Salvador R, Monté GC, Gomar JJ, Sarró S, Ortiz-Gil J, Aguirre C, Landín-Romero R, Guerrero-Pedraza A, Papiol S, Blanch J, McKenna PJ, Fañanás L. Effect of the interleukin-1β gene on dorsolateral prefrontal cortex function in schizophrenia: a genetic neuroimaging study. Biol Psychiatry 2012; 72:758-65. [PMID: 22763186 DOI: 10.1016/j.biopsych.2012.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 04/11/2012] [Accepted: 04/30/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Genetic studies have found that the interleukin-1β gene (IL1B, 2q13) influences the risk for schizophrenia, but the underlying biological mechanisms of the association are still unclear. Investigation of the effects of genetic variability in this gene on brain function could provide more information about its role in the disorder. METHODS The present study examined the effects of a functional polymorphism at IL1B gene promoter (-511C/T; rs16944) on brain correlates of working memory performance in schizophrenia. Forty-eight schizophrenia patients and 46 control subjects underwent functional magnetic resonance imaging while performing the n-back task. RESULTS In the pooled sample, genetic variability at this locus was associated with differential brain activation in a bilateral frontal region including the dorsolateral prefrontal cortex. There was also a significant diagnosis × genotype interaction effect in an overlapping frontal region: the IL1B polymorphism did not affect activation in the control subjects in this area, but the schizophrenia patients who were T carriers showed significantly higher activation than the CC homozygotes. CONCLUSIONS The findings support a role for IL1B variability in the dorsolateral prefrontal cortex dysfunction classically associated with schizophrenia.
Collapse
Affiliation(s)
- Mar Fatjó-Vilas
- Unitat d'Antropologia, Departament de Biologia Animal, Facultat de Biologia, Universitat de Barcelona, Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Bilbo SD, Schwarz JM. The immune system and developmental programming of brain and behavior. Front Neuroendocrinol 2012; 33:267-86. [PMID: 22982535 PMCID: PMC3484177 DOI: 10.1016/j.yfrne.2012.08.006] [Citation(s) in RCA: 402] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/28/2012] [Accepted: 08/29/2012] [Indexed: 12/16/2022]
Abstract
The brain, endocrine, and immune systems are inextricably linked. Immune molecules have a powerful impact on neuroendocrine function, including hormone-behavior interactions, during health as well as sickness. Similarly, alterations in hormones, such as during stress, can powerfully impact immune function or reactivity. These functional shifts are evolved, adaptive responses that organize changes in behavior and mobilize immune resources, but can also lead to pathology or exacerbate disease if prolonged or exaggerated. The developing brain in particular is exquisitely sensitive to both endogenous and exogenous signals, and increasing evidence suggests the immune system has a critical role in brain development and associated behavioral outcomes for the life of the individual. Indeed, there are associations between many neuropsychiatric disorders and immune dysfunction, with a distinct etiology in neurodevelopment. The goal of this review is to describe the important role of the immune system during brain development, and to discuss some of the many ways in which immune activation during early brain development can affect the later-life outcomes of neural function, immune function, mood and cognition.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, 572 Research Drive, Box 91050, Durham, NC 27708, USA.
| | | |
Collapse
|
38
|
Song D, Li B, Yan E, Man Y, Wolfson M, Chen Y, Peng L. Chronic Treatment with Anti-bipolar Drugs Causes Intracellular Alkalinization in Astrocytes, Altering Their Functions. Neurochem Res 2012; 37:2524-40. [DOI: 10.1007/s11064-012-0837-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 07/03/2012] [Accepted: 07/08/2012] [Indexed: 12/26/2022]
|
39
|
Central nervous system inflammation in disease related conditions: Mechanistic prospects. Brain Res 2012; 1446:144-55. [DOI: 10.1016/j.brainres.2012.01.061] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 12/24/2022]
|
40
|
Bilbo SD, Smith SH, Schwarz JM. A lifespan approach to neuroinflammatory and cognitive disorders: a critical role for glia. J Neuroimmune Pharmacol 2012; 7:24-41. [PMID: 21822589 PMCID: PMC3267003 DOI: 10.1007/s11481-011-9299-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/14/2011] [Indexed: 12/15/2022]
Abstract
Cognitive decline is a common problem of aging. Whereas multiple neural and glial mechanisms may account for these declines, microglial sensitization and/or dystrophy has emerged as a leading culprit in brain aging and dysfunction. However, glial activation is consistently observed in normal brain aging as well, independent of frank neuroinflammation or functional impairment. Such variability suggests the existence of additional vulnerability factors that can impact neuronal-glial interactions and thus overall brain and cognitive health. The goal of this review is to elucidate our working hypothesis that an individual's risk or resilience to neuroinflammatory disorders and poor cognitive aging may critically depend on their early life experience, which can change immune reactivity within the brain for the remainder of the lifespan. For instance, early-life infection in rats can profoundly disrupt memory function in young adulthood, as well as accelerate age-related cognitive decline, both of which are linked to enduring changes in glial function that occur in response to the initial infection. We discuss these findings within the context of the growing literature on the role of immune molecules and neuroimmune crosstalk in normal brain development. We highlight the intrinsic factors (e.g., chemokines, hormones) that regulate microglial development and their colonization of the embryonic and postnatal brain, and the capacity for disruption or "re-programming" of this crucial process by external events (e.g., stress, infection). An impact on glia, which in turn alters neural development, has the capacity to profoundly impact cognitive and mental health function at all stages of life.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology & Neuroscience, Duke University, Durham, NC 27708, USA.
| | | | | |
Collapse
|
41
|
Neuron-specific effects of interleukin-1β are mediated by a novel isoform of the IL-1 receptor accessory protein. J Neurosci 2012; 31:18048-59. [PMID: 22159118 DOI: 10.1523/jneurosci.4067-11.2011] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the CNS, interleukin-1β (IL-1β) is synthesized and released during injury, infection, and disease, mediating inflammatory responses. However, IL-1β is also present in the brain under physiological conditions, and can influence hippocampal neuronal function. Several cell-specific IL-1-mediated signaling pathways and functions have been identified in neurons and astrocytes, but their mechanisms have not been fully defined. In astrocytes, IL-1β induced both the p38 MAPK and NF-κB (nuclear factor κB) pathways regulating inflammatory responses, however in hippocampal neurons IL-1β activated p38 but not NF-κB. Additionally, IL-1β induced Src phosphorylation at 0.01 ng/ml in hippocampal neurons, a dose 1000-fold lower than that used to stimulate inflammatory responses. IL-1 signaling requires the type 1 IL-1 receptor and the IL-1 receptor accessory protein (IL-1RAcP) as a receptor partner. We previously reported a novel isoform of the IL-1RAcP, IL-1RAcPb, found exclusively in CNS neurons. In this study, we demonstrate that AcPb specifically mediates IL-1β activation of p-Src and potentiation of NMDA-induced calcium influx in mouse hippocampal neurons in a dose-dependent manner. Mice lacking the AcPb, but retaining the AcP, isoform were deficient in IL-1β regulation of p-Src in neurons. AcPb also played a modulatory role in the activation of p38 MAPK, but had no effect on NF-κB signaling. The restricted expression of AcPb in CNS neurons, therefore, governs specific neuronal signaling and functional responses to IL-1β.
Collapse
|
42
|
Baker DG, Nievergelt CM, O'Connor DT. Biomarkers of PTSD: Neuropeptides and immune signaling. Neuropharmacology 2012; 62:663-73. [DOI: 10.1016/j.neuropharm.2011.02.027] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 02/23/2011] [Accepted: 02/27/2011] [Indexed: 02/07/2023]
|
43
|
Blednov YA, Ponomarev I, Geil C, Bergeson S, Koob GF, Harris RA. Neuroimmune regulation of alcohol consumption: behavioral validation of genes obtained from genomic studies. Addict Biol 2012; 17:108-20. [PMID: 21309947 DOI: 10.1111/j.1369-1600.2010.00284.x] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Analysis of mouse brain gene expression, using strains that differ in alcohol consumption, provided a number of novel candidate genes that potentially regulate alcohol consumption. We selected six genes [beta-2-microglobulin (B2m), cathepsin S (Ctss), cathepsin F (Ctsf), interleukin 1 receptor antagonist (Il1rn), CD14 molecule (Cd14) and interleukin 6 (Il6)] for behavioral validation using null mutant mice. These genes are known to be important for immune responses but were not specifically linked to alcohol consumption by previous research. Null mutant mice were tested for ethanol intake in three tests: 24-hour two-bottle choice, limited access two-bottle choice and limited access to one bottle of ethanol. Ethanol consumption and preference were reduced in all the null mutant mice in the 24-hour two-bottle choice test, the test that was the basis for selection of these genes. No major differences were observed in consumption of saccharin or quinine in the null mutant mice. Deletion of B2m, Ctss, Il1rn, Cd14 and Il6 also reduced ethanol consumption in the limited access two bottle choice test for ethanol intake; with the Il1rn and Ctss null mutants showing reduced intake in all three tests (with some variation between males and females). These results provide the most compelling evidence to date that global gene expression analysis can identify novel genetic determinants of complex behavioral traits. Specifically, they suggest a novel role for neuroimmune signaling in regulation of alcohol consumption.
Collapse
Affiliation(s)
- Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, Section of Neurobiology and Department Pharmacology/Toxicology, University of Texas, Austin, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
The proinflammatory cytokine interleukin-1β (IL-1β) is critical for normal hippocampus (HP)-dependent cognition, whereas high levels can disrupt memory and are implicated in neurodegeneration. However, the cellular source of IL-1β during learning has not been shown, and little is known about the risk factors leading to cytokine dysregulation within the HP. We have reported that neonatal bacterial infection in rats leads to marked HP-dependent memory deficits in adulthood. However, deficits are only observed if unmasked by a subsequent immune challenge [lipopolysaccharide (LPS)] around the time of learning. These data implicate a long-term change within the immune system that, upon activation with the "second hit," LPS, acutely impacts the neural processes underlying memory. Indeed, inhibiting brain IL-1β before the LPS challenge prevents memory impairment in neonatally infected (NI) rats. We aimed to determine the cellular source of IL-1β during normal learning and thereby lend insight into the mechanism by which this cytokine is enduringly altered by early-life infection. We show for the first time that CD11b(+) enriched cells are the source of IL-1β during normal HP-dependent learning. CD11b(+) cells from NI rats are functionally sensitized within the adult HP and produce exaggerated IL-1β ex vivo compared with controls. However, an exaggerated IL-1β response in vivo requires LPS before learning. Moreover, preventing microglial activation during learning prevents memory impairment in NI rats, even following an LPS challenge. Thus, early-life events can significantly modulate normal learning-dependent cytokine activity within the HP, via a specific, enduring impact on brain microglial function.
Collapse
|
45
|
Claycomb RJ, Hewett SJ, Hewett JA. Neuromodulatory role of endogenous interleukin-1β in acute seizures: possible contribution of cyclooxygenase-2. Neurobiol Dis 2011; 45:234-42. [PMID: 21856425 DOI: 10.1016/j.nbd.2011.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 07/20/2011] [Accepted: 08/03/2011] [Indexed: 10/17/2022] Open
Abstract
The function of endogenous interleukin-1β (IL-1β) signaling in acute seizure activity was examined using transgenic mice harboring targeted deletions in the genes for either IL-1β (Il1b) or its signaling receptor (Il1r1). Acute epileptic seizure activity was modeled using two mechanistically distinct chemoconvulsants, kainic acid (KA) and pentylenetetrazole (PTZ). KA-induced seizure activity was more severe in homozygous null (-/-) Il1b mice compared to their wild-type (+/+) littermate controls, as indicated by an increase in the incidence of sustained generalized convulsive seizure activity. In the PTZ seizure model, the incidence of acute convulsive seizures was increased in both Il1b and Il1r1-/- mice compared to their respective +/+ littermate controls. Interestingly, the selective cyclooxygenase (COX)-2 inhibitor, rofecoxib, mimicked the effect of IL-1β deficiency on PTZ-induced convulsions in Il1r1+/+ but not -/- mice. Together, these results suggest that endogenous IL-1β possesses anticonvulsive properties that may be mediated by arachidonic acid metabolites derived from the catalytic action of COX-2.
Collapse
Affiliation(s)
- Robert J Claycomb
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | | | | |
Collapse
|
46
|
Ben Menachem-Zidon O, Avital A, Ben-Menahem Y, Goshen I, Kreisel T, Shmueli EM, Segal M, Ben Hur T, Yirmiya R. Astrocytes support hippocampal-dependent memory and long-term potentiation via interleukin-1 signaling. Brain Behav Immun 2011; 25:1008-16. [PMID: 21093580 DOI: 10.1016/j.bbi.2010.11.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/10/2010] [Accepted: 11/11/2010] [Indexed: 02/04/2023] Open
Abstract
Recent studies indicate that astrocytes play an integral role in neural and synaptic functioning. To examine the implications of these findings for neurobehavioral plasticity we investigated the involvement of astrocytes in memory and long-term potentiation (LTP), using a mouse model of impaired learning and synaptic plasticity caused by genetic deletion of the interleukin-1 receptor type I (IL-1RI). Neural precursor cells (NPCs), derived from either wild type (WT) or IL-1 receptor knockout (IL-1rKO) neonatal mice, were labeled with bromodeoxyuridine (BrdU) and transplanted into the hippocampus of either IL-1rKO or WT adult host mice. Transplanted NPCs survived and differentiated into astrocytes (expressing GFAP and S100β), but not to neurons or oligodendrocytes. The NPCs-derived astrocytes from WT but not IL-1rKO mice displayed co-localization of GFAP with the IL-1RI. Four to twelve weeks post-transplantation, memory functioning was examined in the fear-conditioning and the water maze paradigms and LTP of perforant path-dentate gyrus synapses was assessed in anesthetized mice. As expected, IL-1rKO mice transplanted with IL-1rKO cells or sham operated displayed severe memory disturbances in both paradigms as well as a marked impairment in LTP. In contrast, IL-1rKO mice transplanted with WT NPCs displayed a complete rescue of the impaired memory functioning as well as partial restoration of LTP. These findings indicate that astrocytes play a critical role in memory functioning and LTP, and specifically implicate astrocytic IL-1 signaling in these processes. The results suggest novel conceptualization and therapeutic targets for neuropsychiatric disorders characterized by impaired astrocytic functioning concomitantly with disturbed memory and synaptic plasticity.
Collapse
|
47
|
Central and peripheral cytokines mediate immune-brain connectivity. Neurochem Res 2010; 36:1-6. [PMID: 20820913 DOI: 10.1007/s11064-010-0252-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2010] [Indexed: 01/18/2023]
Abstract
The immune system is a homeostatic system that contributes to maintain the constancy of the molecular and cellular components of the organism. Immune cells can detect the intrusion of foreign antigens or alteration of self-components and send information to the central nervous system (CNS) about this kind of perturbations, acting as a receptor sensorial organ. The brain can respond to such signals by emitting neuro/endocrine signals capable of affecting immune reactivity. Thus, the immune system, as other physiologic systems, is under brain control. Under disease conditions, when priorities for survival change, the immune system can, within defined limits, reset brain-integrated neuro-endocrine mechanisms in order to favour immune processes at the expenses of other physiologic systems. In addition, some cytokines initially conceived as immune products, such as IL-1 and IL-6, are also produced in the "healthy" brain by glial cells and even by some neurons. These and other cytokines have the capacity to affect synaptic plasticity acting as mediators of interactions between astrocytes and pre- and post-synaptic neurons that constitute what is actually defined as a tripartite synapse. Since the production of cytokines in the brain is affected by peripheral immune and central neural signals, it is conceivable that tripartite synapses can, in turn, serve as a relay system in immune-CNS communication.
Collapse
|
48
|
Bilbo SD, Tsang V. Enduring consequences of maternal obesity for brain inflammation and behavior of offspring. FASEB J 2010; 24:2104-15. [PMID: 20124437 DOI: 10.1096/fj.09-144014] [Citation(s) in RCA: 368] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Obesity is well characterized as a systemic inflammatory condition, and is also associated with cognitive disruption, suggesting a link between the two. We assessed whether peripheral inflammation in maternal obesity may be transferred to the offspring brain, in particular, the hippocampus, and thereby result in cognitive dysfunction. Rat dams were fed a high-saturated-fat diet (SFD), a high-trans-fat diet (TFD), or a low-fat diet (LFD) for 4 wk prior to mating, and remained on the diet throughout pregnancy and lactation. SFD/TFD exposure significantly increased body weight in both dams and pups compared to controls. Microglial activation markers were increased in the hippocampus of SFD/TFD pups at birth. At weaning and in adulthood, proinflammatory cytokine expression was strikingly increased in the periphery and hippocampus following a bacterial challenge [lipopolysaccharide (LPS)] in the SFD/TFD groups compared to controls. Microglial activation within the hippocampus was also increased basally in SFD rats, suggesting a chronic priming of the cells. Finally, there were marked changes in anxiety and spatial learning in SFD/TFD groups. These effects were all observed in adulthood, even after the pups were placed on standard chow at weaning, suggesting these outcomes were programmed early in life.
Collapse
Affiliation(s)
- Staci D Bilbo
- Duke University, Department of Psychology and Neuroscience, Durham, NC 27708, USA.
| | | |
Collapse
|
49
|
Connection between inflammatory processes and transmittor function—Modulatory effects of interleukin-1. Prog Neurobiol 2010; 90:256-62. [DOI: 10.1016/j.pneurobio.2009.10.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 07/27/2009] [Accepted: 10/09/2009] [Indexed: 12/27/2022]
|
50
|
Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior: a critical role for the immune system. Front Behav Neurosci 2009; 3:14. [PMID: 19738918 PMCID: PMC2737431 DOI: 10.3389/neuro.08.014.2009] [Citation(s) in RCA: 458] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 07/22/2009] [Indexed: 12/12/2022] Open
Abstract
The immune system is well characterized for its critical role in host defense. Far beyond this limited role however, there is mounting evidence for the vital role the immune system plays within the brain, in both normal, “homeostatic” processes (e.g., sleep, metabolism, memory), as well as in pathology, when the dysregulation of immune molecules may occur. This recognition is especially critical in the area of brain development. Microglia and astrocytes, the primary immunocompetent cells of the CNS, are involved in every major aspect of brain development and function, including synaptogenesis, apoptosis, and angiogenesis. Cytokines such as tumor necrosis factor (TNF)α, interleukin [IL]-1β, and IL-6 are produced by glia within the CNS, and are implicated in synaptic formation and scaling, long-term potentiation, and neurogenesis. Importantly, cytokines are involved in both injury and repair, and the conditions underlying these distinct outcomes are under intense investigation and debate. Evidence from both animal and human studies implicates the immune system in a number of disorders with known or suspected developmental origins, including schizophrenia, anxiety/depression, and cognitive dysfunction. We review the evidence that infection during the perinatal period of life acts as a vulnerability factor for later-life alterations in cytokine production, and marked changes in cognitive and affective behaviors throughout the remainder of the lifespan. We also discuss the hypothesis that long-term changes in brain glial cell function underlie this vulnerability.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology & Neuroscience, Duke University Durham, NC, USA.
| | | |
Collapse
|