1
|
Šimončičová E, Henderson Pekarik K, Vecchiarelli HA, Lauro C, Maggi L, Tremblay MÈ. Adult Neurogenesis, Learning and Memory. ADVANCES IN NEUROBIOLOGY 2024; 37:221-242. [PMID: 39207695 DOI: 10.1007/978-3-031-55529-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neural plasticity can be defined as the ability of neural circuits to be shaped by external and internal factors. It provides the brain with a capacity for functional and morphological remodelling, with many lines of evidence indicating that these changes are vital for learning and memory formation. The basis of this brain plasticity resides in activity- and experience-driven modifications of synaptic strength, including synaptic formation, elimination or weakening, as well as of modulation of neuronal population, which drive the structural reorganization of neural networks. Recent evidence indicates that brain-resident glial cells actively participate in these processes, suggesting that mechanisms underlying plasticity in the brain are multifaceted. Establishing the 'tripartite' synapse, the role of astrocytes in modulating synaptic transmission in response to neuronal activity was recognized first. Further redefinition of the synapse as 'quad-partite' followed to acknowledge the contribution of microglia which were revealed to affect numerous brain functions via dynamic interactions with synapses, acting as 'synaptic sensors' that respond to neuronal activity and neurotransmitter release, as well as crosstalk with astrocytes. Early studies identified microglial ability to dynamically survey their local brain environment and established their integral role in the active interfacing of environmental stimuli (both internal and external), with brain plasticity and remodelling. Following the introduction to neurogenesis, this chapter details the role that microglia play in regulating neurogenesis in adulthood, specifically as it relates to learning and memory, as well as factors involved in modulation of microglia. Further, a microglial perspective is introduced for the context of environmental enrichment impact on neurogenesis, learning and memory across states of stress, ageing, disease and injury.
Collapse
Affiliation(s)
- Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | | | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
2
|
Church KA, Cardona AE, Hopp SC. Roles in Innate Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:263-286. [PMID: 39207697 DOI: 10.1007/978-3-031-55529-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.
Collapse
Affiliation(s)
- Kaira A Church
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Astrid E Cardona
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, Biggs Institute for Alzheimer's and Neurodegenerative Disease, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
3
|
Wang H, Yu R, Wang M, Wang S, Ouyang X, Yan Z, Chen S, Wang W, Wu F, Fan C. Insulin-like growth factor binding protein 4 loaded electrospun membrane ameliorating tendon injury by promoting retention of IGF-1. J Control Release 2023; 356:162-174. [PMID: 36868516 DOI: 10.1016/j.jconrel.2023.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Tendon injury is one of the most common musculoskeletal disorders that impair joint mobility and lower quality of life. The limited regenerative capacity of tendon remains a clinical challenge. Local delivery of bioactive protein is a viable therapeutic approach for tendon healing. Insulin-like growth factor binding protein 4 (IGFBP-4) is a secreted protein capable of binding and stabilizing insulin-like growth factor 1 (IGF-1). Here, we applied an aqueous-aqueous freezing-induced phase separation technology to obtain the IGFBP4-encapsulated dextran particles. Then, we added the particles into poly (L-lactic acid) (PLLA) solution to fabricate IGFBP4-PLLA electrospun membrane for efficient IGFBP-4 delivery. The scaffold showed excellent cytocompatibility and a sustained release of IGFBP-4 for nearly 30 days. In cellular experiments, IGFBP-4 promoted tendon-related and proliferative markers expression. In a rat Achilles tendon injury model, immunohistochemistry and quantitative real-time polymerase chain reaction confirmed better outcomes by using the IGFBP4-PLLA electrospun membrane at the molecular level. Furthermore, the scaffold effectively promoted tendon healing in functional performance, ultrastructure and biomechanical properties. We found addition of IGFBP-4 promoted IGF-1 retention in tendon postoperatively and then facilitated protein synthesis via IGF-1/AKT signaling pathway. Overall, our IGFBP4-PLLA electrospun membrane provides a promising therapeutic strategy for tendon injury.
Collapse
Affiliation(s)
- Hui Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Ruyue Yu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Meng Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shikun Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Xingyu Ouyang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Zhiwen Yan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Shuai Chen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Wei Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China.
| | - Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China.
| |
Collapse
|
4
|
Abdi IY, Ghanem SS, El-Agnaf OM. Immune-related biomarkers for Parkinson's disease. Neurobiol Dis 2022; 170:105771. [DOI: 10.1016/j.nbd.2022.105771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/13/2022] Open
|
5
|
García-Magro N, Zegarra-Valdivia JA, Troyas-Martinez S, Torres-Aleman I, Nuñez A. Response Facilitation Induced by Insulin-Like Growth Factor-I in the Primary Somatosensory Cortex of Mice Was Reduced in Aging. Cells 2022; 11:cells11040717. [PMID: 35203366 PMCID: PMC8870291 DOI: 10.3390/cells11040717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
Aging is accompanied by a decline in cognition that can be due to a lower IGF-I level. We studied response facilitation induced in primary somatosensory (S1) cortical neurons by repetitive stimulation of whiskers in young and old mice. Layer 2/3 and 5/6 neurons were extracellularly recorded in young (≤ 6 months of age) and old (≥ 20 month of age) anesthetized mice. IGF-I injection in S1 cortex (10 nM; 0.2 μL) increased whisker responses in young and old animals. A stimulation train at 8 Hz induced a long-lasting response facilitation in only layer 2/3 neurons of young animals. However, all cortical neurons from young and old animals showed long-lasting response facilitation when IGF-I was applied in the S1 cortex. The reduction in response facilitation in old animals can be due to a reduction in the IGF-I receptors as was indicated by the immunohistochemistry study. Furthermore, a reduction in the performance of a whisker discrimination task was observed in old animals. In conclusion, our findings indicate that there is a reduction in the synaptic plasticity of S1 neurons during aging that can be recovered by IGF-I. Therefore, it opens the possibility of use IGF-I as a therapeutic tool to ameliorate the effects of heathy aging.
Collapse
Affiliation(s)
- Nuria García-Magro
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Jonathan A. Zegarra-Valdivia
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
- Cajal Institute, Cibernet (CSIC), 28002 Madrid, Spain;
- Universidad Señor de Sipán, Chiclayo 02001, Peru
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Foundation for Science, 48009 Bilbao, Spain
| | - Sara Troyas-Martinez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
| | - Ignacio Torres-Aleman
- Cajal Institute, Cibernet (CSIC), 28002 Madrid, Spain;
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Foundation for Science, 48009 Bilbao, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
- Correspondence:
| |
Collapse
|
6
|
Carpenter JM, Brown KA, Diaz AN, Dockman RL, Benbow RA, Harn DA, Norberg T, Wagner JJ, Filipov NM. Delayed treatment with the immunotherapeutic LNFPIII ameliorates multiple neurological deficits in a pesticide-nerve agent prophylactic mouse model of Gulf War Illness. Neurotoxicol Teratol 2021; 87:107012. [PMID: 34256162 DOI: 10.1016/j.ntt.2021.107012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022]
Abstract
Residual effects of the 1990-1991 Gulf War (GW) still plague veterans 30 years later as Gulf War Illness (GWI). Thought to stem mostly from deployment-related chemical overexposures, GWI is a disease with multiple neurological symptoms with likely immunological underpinnings. Currently, GWI remains untreatable, and the long-term neurological disease manifestation is not characterized fully. The present study sought to expand and evaluate the long-term implications of prior GW chemicals exposure on neurological function 6-8 months post GWI-like symptomatology induction. Additionally, the beneficial effects of delayed treatment with the glycan immunotherapeutic lacto-N-fucopentaose III (LNFPIII) were evaluated. Male C57BL/6J mice underwent a 10-day combinational exposure (i.p.) to GW chemicals, the nerve agent prophylactic pyridostigmine bromide (PB) and the insecticide permethrin (PM; 0.7 and 200 mg/kg, respectively). Beginning 4 months after PB/PM exposure, a subset of the mice were treated twice a week until study completion with LNFPIII. Evaluation of cognition/memory, motor function, and mood was performed beginning 1 month after LNFPIII treatment initiation. Prior exposure to PB/PM produced multiple locomotor, neuromuscular, and sensorimotor deficits across several motor tests. Subtle anxiety-like behavior was also present in PB/PM mice in mood tests. Further, PB/PM-exposed mice learned at a slower rate, mostly during early phases of the learning and memory tests employed. LNFPIII treatment restored or improved many of these behaviors, particularly in motor and cognition/memory domains. Electrophysiology data collected from hippocampal slices 8 months post PB/PM exposure revealed modest aberrations in basal synaptic transmission and long-term potentiation in the dorsal or ventral hippocampus that were improved by LNFPIII treatment. Immunohistochemical analysis of tyrosine hydroxylase (TH), a dopaminergic marker, did not detect major PB/PM effects along the nigrostriatal pathway, but LNFPIII increased striatal TH. Additionally, neuroinflammatory cells were increased in PB/PM mice, an effect reduced by LNFPIII. Collectively, long-term neurobehavioral and neurobiological dysfunction associated with prior PB/PM exposure was characterized; delayed LNFPIII treatment provided multiple behavioral and biological beneficial effects in the context of GWI, highlighting its potential as a GWI therapeutic.
Collapse
Affiliation(s)
- Jessica M Carpenter
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Neuroscience Program, University of Georgia, Athens, GA, United States
| | - Kyle A Brown
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, United States
| | - Alexa N Diaz
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Rachel L Dockman
- Department of Microbiology, University of Georgia, Athens, GA, United States
| | - Robert A Benbow
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Donald A Harn
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States; Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Thomas Norberg
- Department of Chemistry, University of Uppsala, Uppsala, Sweden
| | - John J Wagner
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Neuroscience Program, University of Georgia, Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, United States.
| | - Nikolay M Filipov
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States; Neuroscience Program, University of Georgia, Athens, GA, United States; Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, United States.
| |
Collapse
|
7
|
Yegla B, Boles J, Kumar A, Foster TC. Partial microglial depletion is associated with impaired hippocampal synaptic and cognitive function in young and aged rats. Glia 2021; 69:1494-1514. [PMID: 33586813 DOI: 10.1002/glia.23975] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
The role of microglia in mediating age-related changes in cognition and hippocampal synaptic function was examined by microglial depletion and replenishment using PLX3397. We observed age-related differences in microglial number and morphology, as well as increased Iba-1 expression, indicating microglial activation. PLX3397 treatment decreased microglial number, with aged rats exhibiting the lowest density. Young rats exhibited increased expression of pro-inflammatory cytokines during depletion and repopulation and maintenance of Iba-1 levels despite reduced microglial number. For aged rats, several cytokines increased with depletion and recovered during repopulation; however, aged rats did not fully recover microglial cell number or Iba-1 expression during repopulation, with a recovery comparable to young control levels rather than aged controls. Hippocampal CA3-CA1 synaptic transmission was impaired with age, and microglial depletion was associated with decreased total synaptic transmission in young and aged rats. A robust decline in N-methyl-d-aspartate-receptor-mediated synaptic transmission arose in young depleted rats specifically. Microglial replenishment normalized depletion-induced synaptic function to control levels; however, recovery of aged animals did not mirror young. Microglial depletion was associated with decreased context-object discrimination memory in both age groups, which recovered with microglial repopulation. Aged rats displayed impaired contextual and cued fear memory, and microglial replenishment did not recover their memory to the level of young. The current study indicates that cognitive function and synaptic transmission benefit from the support of aged microglia and are hindered by removal of these cells. Replenishment of microglia in aging did not ameliorate age-related cognitive impairments or senescent synaptic function.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Jake Boles
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics Program, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
8
|
Brombacher T, Ajonijebu D, Scibiorek M, Berkiks I, Moses B, Mpotje T, Brombacher F. IL-4Rα deletion disrupts psychomotor performance and reference memory in mice while sparing behavioural phenotype associated with spatial learning. Brain Behav Immun 2021; 92:157-164. [PMID: 33301870 PMCID: PMC7909383 DOI: 10.1016/j.bbi.2020.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/02/2020] [Accepted: 12/04/2020] [Indexed: 11/20/2022] Open
Abstract
Contribution of immune mediators, interleukin-4 and interferon gamma to cognitive functioning is receiving increasing attention. However, the fundamental question about how heterodimeric interleukin-4 receptor alpha- and interferon gamma- producing myeloid cells converge to influence hippocampal-dependent spatial memory tasks through immunomodulation of multisensory inputs from other brain areas remains unexplored. Here, we show that mice lacking interleukin-4 receptor alpha are able to successfully learn spatial tasks, while reference memory is impaired. Moreover, the absence of interleukin-4 receptor alpha leads to simultaneous increase in proportions of CD11b + myeloid cells in the hippocampus and thalamus, but not the brainstem during acquisition. Interleukin-4 receptor alpha deletion significantly decreased expression of myeloid cell-derived interferon gamma in the thalamus during the acquisition phase and simultaneously increased brain-derived neurotrophic factor production in the thalamus and brainstem of trained mice. We provide evidence that interleukin-4 receptor alpha is essential for cognitive performance while training-induced alterations in interferon gamma activity and brain-derived neurotrophic factor signalling may contribute to neuromodulation of learned tasks and consequently affect systems-level memory encoding and consolidation.
Collapse
Affiliation(s)
- T.M. Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa,Corresponding author at: University of Cape Town, Wernher & Beit Building, South, 7925 Cape Town, South Africa.
| | - D.C. Ajonijebu
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa
| | - M. Scibiorek
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa
| | - I. Berkiks
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa
| | - B.O. Moses
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa
| | - T. Mpotje
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa
| | - F. Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, South Africa,Division of Immunology, Institute of Infectious Disease and Molecular Medicine (IDM), Health Science Faculty, University of Cape Town, 7925, South Africa
| |
Collapse
|
9
|
Flood L, Korol SV, Ekselius L, Birnir B, Jin Z. Interferon-γ potentiates GABA A receptor-mediated inhibitory currents in rat hippocampal CA1 pyramidal neurons. J Neuroimmunol 2019; 337:577050. [PMID: 31505410 DOI: 10.1016/j.jneuroim.2019.577050] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/19/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022]
Abstract
The neural transmission and plasticity can be differentially modulated by various elements of the immune system. Interferon-γ (IFN-γ) is a "pro-inflammatory" cytokine mainly produced by T lymphocytes, activates its corresponding receptor and plays important roles under both homeostatic and inflammatory conditions. However, the impact of IFN-γ on the γ-aminobutyric acid (GABA)-mediated currents in the hippocampus, a major brain region involved in the cognitive function, has not been investigated. Here we detected abundant expression of both IFN-γ receptor subunit gene transcripts (Ifngr1 and Ifngr2) in the rat hippocampus by quantitative PCR. In addition, we pre-incubated rat hippocampal slices with IFN-γ (100 ng/ml) and recorded GABA-activated spontaneous and miniature postsynaptic inhibitory currents (sIPSCs and mIPSCs) and tonic currents in hippocampal CA1 pyramidal neurons by the whole-cell patch-clamp method. The pre-incubation with IFN-γ increased the frequency but not the mean amplitude, rise time or decay time of both sIPSCs and mIPSCs in hippocampal CA1 pyramidal neurons, suggesting a presynaptic effect of IFN-γ. Moreover, the GABA-activated tonic currents were enhanced by IFN-γ. In conclusion, the potentiation of GABAergic currents in hippocampal neurons by IFN-γ may contribute to the disturbed neuronal excitability and cognitive dysfunction during neuroinflammation.
Collapse
Affiliation(s)
- Louise Flood
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala, Sweden
| | - Sergiy V Korol
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala, Sweden
| | - Lisa Ekselius
- Department of Neuroscience, Psychiatry, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - Bryndis Birnir
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala, Sweden
| | - Zhe Jin
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala, Sweden; Department of Neurosurgery, General Hospital of NingXia Medical University, Yinchuan, NingXia, China.
| |
Collapse
|
10
|
Haq N, Schmidt-Hieber C, Sialana FJ, Ciani L, Heller JP, Stewart M, Bentley L, Wells S, Rodenburg RJ, Nolan PM, Forsythe E, Wu MC, Lubec G, Salinas P, Häusser M, Beales PL, Christou-Savina S. Loss of Bardet-Biedl syndrome proteins causes synaptic aberrations in principal neurons. PLoS Biol 2019; 17:e3000414. [PMID: 31479441 PMCID: PMC6743795 DOI: 10.1371/journal.pbio.3000414] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/13/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023] Open
Abstract
Bardet-Biedl syndrome (BBS), a ciliopathy, is a rare genetic condition characterised by retinal degeneration, obesity, kidney failure, and cognitive impairment. In spite of progress made in our general understanding of BBS aetiology, the molecular and cellular mechanisms underlying cognitive impairment in BBS remain elusive. Here, we report that the loss of BBS proteins causes synaptic dysfunction in principal neurons, providing a possible explanation for the cognitive impairment phenotype observed in BBS patients. Using synaptosomal proteomics and immunocytochemistry, we demonstrate the presence of Bbs proteins in the postsynaptic density (PSD) of hippocampal neurons. Loss of Bbs results in a significant reduction of dendritic spines in principal neurons of Bbs mouse models. Furthermore, we show that spine deficiency correlates with events that destabilise spine architecture, such as impaired spine membrane receptor signalling, known to be involved in the maintenance of dendritic spines. Our findings suggest a role for BBS proteins in dendritic spine homeostasis that may be linked to the cognitive phenotype observed in BBS.
Collapse
Affiliation(s)
- Naila Haq
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Christoph Schmidt-Hieber
- Wolfson Institute for Biomedical Research and Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Fernando J. Sialana
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Lorenza Ciani
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Janosch P. Heller
- Institute of Neurology, University College London, London, United Kingdom
| | - Michelle Stewart
- MRC Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, United Kingdom
| | - Liz Bentley
- MRC Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, United Kingdom
| | - Sara Wells
- MRC Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, United Kingdom
| | - Richard J. Rodenburg
- Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Department of Pediatrics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Patrick M. Nolan
- MRC Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, United Kingdom
| | - Elizabeth Forsythe
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Michael C. Wu
- Neurodigitech, LLC, San Diego, California, United States of America
| | - Gert Lubec
- Programme in Proteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - P. Salinas
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Michael Häusser
- Wolfson Institute for Biomedical Research and Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Philip L. Beales
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sofia Christou-Savina
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
11
|
Rêgo MLM, Cabral DAR, Costa EC, Fontes EB. Physical Exercise for Individuals with Hypertension: It Is Time to Emphasize its Benefits on the Brain and Cognition. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2019; 13:1179546819839411. [PMID: 30967748 PMCID: PMC6444761 DOI: 10.1177/1179546819839411] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/27/2019] [Indexed: 01/01/2023]
Abstract
Hypertension affects more than 40% of adults worldwide and is associated with stroke, myocardial infarction, heart failure, and other cardiovascular diseases. It has also been shown to cause severe functional and structural damage to the brain, leading to cognitive impairment and dementia. Furthermore, it is believed that these cognitive impairments affect the mental ability to maintain productivity at work, ultimately causing social and economic problems. Because hypertension is a chronic condition that requires clinical treatment, strategies with fewer side effects and less-invasive procedures are needed. Physical exercise (PE) has proven to be an efficient and complementary tool for hypertension management, and its peripheral benefits have been widely supported by related studies. However, few studies have specifically examined the potential positive effects of PE on the brain in hypertensive individuals. This narrative review discusses the pathophysiological mechanisms that hypertension promotes in the brain, and suggests PE as an important tool to prevent and reduce cognitive damage caused by hypertension. We also provide PE recommendations for hypertensive individuals, as well as suggestions for promoting PE as a method for increasing cognitive abilities in the brain, particularly for hypertensive individuals.
Collapse
Affiliation(s)
- Maria LM Rêgo
- NEUROEX-Research Group in Physical
Activity, Cognition and Behavior, Health Science Center, Federal University of Rio
Grande do Norte, Natal/RN, Brazil
| | - Daniel AR Cabral
- NEUROEX-Research Group in Physical
Activity, Cognition and Behavior, Health Science Center, Federal University of Rio
Grande do Norte, Natal/RN, Brazil
| | - Eduardo C Costa
- GPEACE-Research Group on Acute and
Chronic Effects of Exercise, Health Science Center, Federal University of Rio Grande
do Norte, Natal/RN, Brazil
| | - Eduardo B Fontes
- NEUROEX-Research Group in Physical
Activity, Cognition and Behavior, Health Science Center, Federal University of Rio
Grande do Norte, Natal/RN, Brazil
| |
Collapse
|
12
|
Priming of microglia with IFN-γ slows neuronal gamma oscillations in situ. Proc Natl Acad Sci U S A 2019; 116:4637-4642. [PMID: 30782788 DOI: 10.1073/pnas.1813562116] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type II IFN (IFN-γ) is a proinflammatory T lymphocyte cytokine that serves in priming of microglia-resident CNS macrophages-during the complex microglial activation process under pathological conditions. Priming generally permits an exaggerated microglial response to a secondary inflammatory stimulus. The impact of primed microglia on physiological neuronal function in intact cortical tissue (in situ) is widely unknown, however. We explored the effects of chronic IFN-γ exposure on microglia in hippocampal slice cultures, i.e., postnatal parenchyma lacking leukocyte infiltration (adaptive immunity). We focused on fast neuronal network waves in the gamma-band (30-70 Hz). Such gamma oscillations are fundamental to higher brain functions, such as perception, attention, and memory, and are exquisitely sensitive to metabolic and oxidative stress. IFN-γ induced substantial morphological changes and cell population expansion in microglia as well as moderate up-regulation of activation markers, MHC-II, CD86, IL-6, and inducible nitric oxide synthase (iNOS), but not TNF-α. Cytoarchitecture and morphology of pyramidal neurons and parvalbumin-positive inhibitory interneurons were well-preserved. Notably, gamma oscillations showed a specific decline in frequency of up to 8 Hz, which was not mimicked by IFN-α or IL-17 exposure. The rhythm disturbance was caused by moderate microglial nitric oxide (NO) release demonstrated by pharmacological microglia depletion and iNOS inhibition. In conclusion, IFN-γ priming induces substantial proliferation and moderate activation of microglia that is capable of slowing neural information processing. This mechanism might contribute to cognitive impairment in chronic brain disease featuring elevated IFN-γ levels, blood-brain barrier leakage, and/or T cell infiltration, well before neurodegeneration occurs.
Collapse
|
13
|
Prieto GA, Tong L, Smith ED, Cotman CW. TNFα and IL-1β but not IL-18 Suppresses Hippocampal Long-Term Potentiation Directly at the Synapse. Neurochem Res 2018; 44:49-60. [PMID: 29619614 DOI: 10.1007/s11064-018-2517-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 12/13/2022]
Abstract
CNS inflammatory responses are linked to cognitive impairment in humans. Research in animal models supports this connection by showing that inflammatory cytokines suppress long-term potentiation (LTP), the best-known cellular correlate of memory. Cytokine-induced modulation of LTP has been previously studied in vivo or in brain slices, two experimental approaches containing multiple cell populations responsive to cytokines. In their target cells, cytokines commonly increase the expression of multiple cytokines, thus increasing the complexity of brain cytokine networks even after single-cytokine challenges. Whether cytokines suppress LTP by direct effects on neurons or by indirect mechanisms is still an open question. Here, we evaluated the effect of a major set of inflammatory cytokines including tumor necrosis factor-α (TNFα), interleukin-1β (IL-1β) and interleukin-18 (IL-18) on chemically-induced LTP (cLTP) in isolated hippocampal synaptosomes of mice, using fluorescence analysis of single-synapse long-term potentiation (FASS-LTP). We found that TNFα and IL-1β suppress synaptosomal cLTP. In contrast, cLTP was not affected by IL-18, at a concentration previously shown to block LTP in hippocampal slices. We also found that IL-18 does not impair cLTP or brain-derived neurotrophic factor (BDNF) signaling in primary hippocampal neuronal cultures. Thus, using both synaptosomes and neuron cultures, our data suggest that IL-18 impairs LTP by indirect mechanisms, which may depend on non-neuronal cells, such as glia. Notably, our results demonstrate that TNFα and IL-1β directly suppress hippocampal plasticity via neuron-specific mechanisms. A better understanding of the brain's cytokine networks and their final molecular effectors is crucial to identify specific targets for intervention.
Collapse
Affiliation(s)
- G Aleph Prieto
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
| | - Liqi Tong
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Erica D Smith
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Carl W Cotman
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| |
Collapse
|
14
|
Gutiérrez-Martos M, Girard B, Mendonça-Netto S, Perroy J, Valjent E, Maldonado R, Martin M. Cafeteria diet induces neuroplastic modifications in the nucleus accumbens mediated by microglia activation. Addict Biol 2018; 23:735-749. [PMID: 28872733 DOI: 10.1111/adb.12541] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/22/2017] [Accepted: 06/28/2017] [Indexed: 12/28/2022]
Abstract
High-palatable and caloric foods are widely overconsumed due to hedonic mechanisms that prevail over caloric necessities leading to overeating and overweight. The nucleus accumbens (NAc) is a key brain area modulating the reinforcing effects of palatable foods and is crucially involved in the development of eating disorders. We describe that prolonged exposure to high-caloric chocolate cafeteria diet leads to overeating and overweight in mice. NAc functionality was altered in these mice, presenting structural plasticity modifications in medium spiny neurons, increased expression of neuroinflammatory factors and activated microglia, and abnormal responses after amphetamine-induced hyperlocomotion. Chronic inactivation of microglia normalized these neurobiological and behavioural alterations exclusively in mice exposed to cafeteria diet. Our data suggest that prolonged exposure to cafeteria diet produces neuroplastic and functional changes in the NAc that can modify feeding behaviour. Microglia activation and neuroinflammation play an important role in the development of these neurobiological alterations.
Collapse
Affiliation(s)
- Miriam Gutiérrez-Martos
- Laboratory of Neuropharmacology (DCEXS); Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra (PRBB/UPF); Spain
| | - Benoit Girard
- Pathophysiology of Synaptic Transmission Laboratory; Institut de Génomique Fonctionnelle; France
| | - Sueli Mendonça-Netto
- Laboratory of Neuropharmacology (DCEXS); Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra (PRBB/UPF); Spain
| | - Julie Perroy
- Pathophysiology of Synaptic Transmission Laboratory; Institut de Génomique Fonctionnelle; France
| | - Emmanuel Valjent
- Inserm U1191, CNRS UMR5203, Laboratory of Neural Circuit and Signal Transduction; University Montpellier; France
| | - Rafael Maldonado
- Laboratory of Neuropharmacology (DCEXS); Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra (PRBB/UPF); Spain
- IMIM (Hospital del Mar Medical Research Institute); Barcelona Spain
| | - Miquel Martin
- Laboratory of Neuropharmacology (DCEXS); Parc de Recerca Biomèdica de Barcelona/Universitat Pompeu Fabra (PRBB/UPF); Spain
- IMIM (Hospital del Mar Medical Research Institute); Barcelona Spain
| |
Collapse
|
15
|
Brain interference: Revisiting the role of IFNγ in the central nervous system. Prog Neurobiol 2017; 156:149-163. [DOI: 10.1016/j.pneurobio.2017.05.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 01/28/2023]
|
16
|
Hypertension-induced synapse loss and impairment in synaptic plasticity in the mouse hippocampus mimics the aging phenotype: implications for the pathogenesis of vascular cognitive impairment. GeroScience 2017; 39:385-406. [PMID: 28664509 DOI: 10.1007/s11357-017-9981-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Strong epidemiological and experimental evidence indicates that hypertension has detrimental effects on the cerebral microcirculation and thereby promotes accelerated brain aging. Hypertension is an independent risk factor for both vascular cognitive impairment (VCI) and Alzheimer's disease (AD). However, the pathophysiological link between hypertension-induced cerebromicrovascular injury (e.g., blood-brain barrier disruption, increased microvascular oxidative stress, and inflammation) and cognitive decline remains elusive. The present study was designed to characterize neuronal functional and morphological alterations induced by chronic hypertension and compare them to those induced by aging. To achieve that goal, we induced hypertension in young C57BL/6 mice by chronic (4 weeks) infusion of angiotensin II. We found that long-term potentiation (LTP) of performant path synapses following high-frequency stimulation of afferent fibers was decreased in hippocampal slices obtained from hypertensive mice, mimicking the aging phenotype. Hypertension and advanced age were associated with comparable decline in synaptic density in the stratum radiatum of the mouse hippocampus. Hypertension, similar to aging, was associated with changes in mRNA expression of several genes involved in regulation of neuronal function, including down-regulation of Bdnf, Homer1, and Dlg4, which may have a role in impaired synaptic plasticity. Collectively, hypertension impairs synaptic plasticity, reduces synaptic density, and promotes dysregulation of genes involved in synaptic function in the mouse hippocampus mimicking the aging phenotype. These hypertension-induced neuronal alterations may impair establishment of memories in the hippocampus and contribute to the pathogenesis and clinical manifestation of both vascular cognitive impairment (VCI) and Alzheimer's disease (AD).
Collapse
|
17
|
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
|
18
|
Tylee DS, Hess JL, Quinn TP, Barve R, Huang H, Zhang-James Y, Chang J, Stamova BS, Sharp FR, Hertz-Picciotto I, Faraone SV, Kong SW, Glatt SJ. Blood transcriptomic comparison of individuals with and without autism spectrum disorder: A combined-samples mega-analysis. Am J Med Genet B Neuropsychiatr Genet 2017; 174:181-201. [PMID: 27862943 PMCID: PMC5499528 DOI: 10.1002/ajmg.b.32511] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/21/2016] [Indexed: 12/25/2022]
Abstract
Blood-based microarray studies comparing individuals affected with autism spectrum disorder (ASD) and typically developing individuals help characterize differences in circulating immune cell functions and offer potential biomarker signal. We sought to combine the subject-level data from previously published studies by mega-analysis to increase the statistical power. We identified studies that compared ex vivo blood or lymphocytes from ASD-affected individuals and unrelated comparison subjects using Affymetrix or Illumina array platforms. Raw microarray data and clinical meta-data were obtained from seven studies, totaling 626 affected and 447 comparison subjects. Microarray data were processed using uniform methods. Covariate-controlled mixed-effect linear models were used to identify gene transcripts and co-expression network modules that were significantly associated with diagnostic status. Permutation-based gene-set analysis was used to identify functionally related sets of genes that were over- and under-expressed among ASD samples. Our results were consistent with diminished interferon-, EGF-, PDGF-, PI3K-AKT-mTOR-, and RAS-MAPK-signaling cascades, and increased ribosomal translation and NK-cell related activity in ASD. We explored evidence for sex-differences in the ASD-related transcriptomic signature. We also demonstrated that machine-learning classifiers using blood transcriptome data perform with moderate accuracy when data are combined across studies. Comparing our results with those from blood-based studies of protein biomarkers (e.g., cytokines and trophic factors), we propose that ASD may feature decoupling between certain circulating signaling proteins (higher in ASD samples) and the transcriptional cascades which they typically elicit within circulating immune cells (lower in ASD samples). These findings provide insight into ASD-related transcriptional differences in circulating immune cells. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniel S. Tylee
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Jonathan L. Hess
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Thomas P. Quinn
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Rahul Barve
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yanli Zhang-James
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Jeffrey Chang
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY, U.S.A
| | - Boryana S. Stamova
- Department of Neurology, UC Davis School of Medicine, Sacramento, CA, USA
| | - Frank R. Sharp
- Department of Neurology, UC Davis School of Medicine, Sacramento, CA, USA
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences and UC Davis MIND Institute, School of Medicine, Davis, CA
| | - Stephen V. Faraone
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A,K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children’s Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA, U.S.A
| | - Stephen J. Glatt
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A,To whom correspondence should be addressed: SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, Phone: (315) 464-7742,
| |
Collapse
|
19
|
Ma G, Chen C, Jiang H, Qiu Y, Li Y, Li X, Zhang X, Liu J, Zhu T. Ribonuclease attenuates hepatic ischemia reperfusion induced cognitive impairment through the inhibition of inflammatory cytokines in aged mice. Biomed Pharmacother 2017; 90:62-68. [PMID: 28343072 DOI: 10.1016/j.biopha.2017.02.094] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Elderly patients undergoing major surgery often develop cognitive dysfunction, and no optimum treatment exists for this postoperative complication. Ribonuclease, the counterpart of ribonucleic acid, has mostly been reported in terms of its use as a potential modality in anticancer therapy, and recent studies have demonstrated that ribonuclease can exert organ-protective effects in several pathological conditions. Our study also demonstrated that ribonuclease protects the liver against ischemia reperfusion injury. Nevertheless, it is unknown whether ribonuclease can attenuate the cognitive dysfunction that is induced by liver ischemia reperfusion. In this study, we aimed to evaluate the effect of ribonuclease on cognitive function after liver ischemia reperfusion. METHODS Aged mice underwent sham surgery or 60min of hepatic ischemia reperfusion, vehicle or ribonuclease, which were administered subcutaneously. The primary observation endpoint was the Morris water maze; following 24h, 3days, and 7days of reperfusion, the levels of serum and hippocampus proinflammatory cytokines were measured to reveal the underlying mechanism. RESULTS A probe test was conducted on day 3 and a reversal probe test was conducted on day 7 after surgery; the results demonstrated a reduction in cognitive function after liver ischemia reperfusion and that ribonuclease treatment attenuated cognitive impairment. The levels of serum and hippocampus proinflammatory cytokines (interleukin-6 and interleukin-1β) and extracellular ribonucleic acid were significantly increased at 24h after reperfusion, but ribonuclease treatment markedly reduced the proinflammatory cytokine increase. CONCLUSION The results of the study suggested that hepatic ischemia reperfusion leads to cognitive impairment in aged mice and an increase in inflammatory cytokine expression in both serum and the hippocampus; more importantly, ribonuclease showed protective effects against cognitive impairment through inhibiting the release of inflammatory cytokines.
Collapse
Affiliation(s)
- Gang Ma
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China; Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Haixia Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanhua Qiu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Yansong Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Xiaoqiang Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Xiyang Zhang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Tao Zhu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041 China.
| |
Collapse
|
20
|
Singh A, Abraham WC. Astrocytes and synaptic plasticity in health and disease. Exp Brain Res 2017; 235:1645-1655. [PMID: 28299411 DOI: 10.1007/s00221-017-4928-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/20/2017] [Indexed: 12/22/2022]
Abstract
Activity-dependent synaptic plasticity phenomena such as long-term potentiation and long-term depression are candidate mechanisms for storing information in the brain. Regulation of synaptic plasticity is critical for healthy cognition and learning and this is provided in part by metaplasticity, which can act to maintain synaptic transmission within a dynamic range and potentially prevent excitotoxicity. Metaplasticity mechanisms also allow neurons to integrate plasticity-associated signals over time. Interestingly, astrocytes appear to be critical for certain forms of synaptic plasticity and metaplasticity mechanisms. Synaptic dysfunction is increasingly viewed as an early feature of AD that is correlated with the severity of cognitive decline, and the development of these pathologies is correlated with a rise in reactive astrocytes. This review focuses on the contributions of astrocytes to synaptic plasticity and metaplasticity in normal tissue, and addresses whether astroglial pathology may lead to aberrant engagement of these mechanisms in neurological diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- A Singh
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
21
|
Zhu D, Yang N, Liu YY, Zheng J, Ji C, Zuo PP. M2 Macrophage Transplantation Ameliorates Cognitive Dysfunction in Amyloid-β-Treated Rats Through Regulation of Microglial Polarization. J Alzheimers Dis 2017; 52:483-95. [PMID: 27003214 DOI: 10.3233/jad-151090] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly population. Neuroinflammation induced by amyloid-β (Aβ) aggregation is considered to be the critical factor underlying AD pathological mechanisms. Alternatively activated (M2) macrophages/microglia have been reported to have neuroprotective effects in neurodegenerative disease. In this study, we characterized the neuroprotective effects of M2 macrophage transplantation in AD model rats and investigated the underlying mechanisms. Intracerebroventricular injection of Aβ1 - 42 to rats was used to model AD and resulted in cognitive impairment, neuronal damage, and inflammatory changes in the brain microenvironment. We observed an increased interferon regulatory factor (IRF) 5/IRF4 ratio, resulting in greater production of classically activated (M1) versus M2 microglia. M2 macrophage transplantation attenuated inflammation in the brain, reversed Aβ1 - 42-induced changes in the IRF4-IRF5 ratio, drove endogenous microglial polarization toward the M2 phenotype, and ameliorated cognitive impairment. Nerve growth factor (NGF) treatment reduced the IRF5/IRF4 ratio and induced primary microglial polarization to the M2 phenotype in vitro; these effects were prevented by tyrosine Kinase Receptor A (TrkA) inhibition. M2 macrophage transplantation restored the balance of IRF4-IRF5 by affecting the expression of NGF and inflammatory cytokines in the brains of AD model rats. This drove microglial polarization to the M2 phenotype, promoted termination of neuroinflammation, and resulted in improved cognitive abilities.
Collapse
|
22
|
With mouse age comes wisdom: A review and suggestions of relevant mouse models for age-related conditions. Mech Ageing Dev 2016; 160:54-68. [DOI: 10.1016/j.mad.2016.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/07/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
|
23
|
Abstract
The functions of adult hippocampal neurogenesis have been extensively investigated during the past decade. Numerous studies have shown that adult neurogenesis may play an important role in the hippocampal-dependent learning and memory. This study evaluated the influence of exercise on hippocampal neurogenesis, neural plasticity, neurotrophic factors, and cognition. Areas of research focused on enhancing effect of exercise for adult hippocampal neurogenesis and protective role of exercise against brain diseases. The present study suggests that exercise improves brain functions and prevents decline of cognition across the lifespan. Understanding of neurobiological mechanisms of exercise on brain functions may lead to the development of novel therapeutic strategy for neurodegenerative disorders.
Collapse
Affiliation(s)
- Seung-Soo Baek
- Department of Sport & Health Science, College of Natural Science, Sangmyung University, Seoul, Korea
| |
Collapse
|
24
|
Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience 2016; 325:89-99. [DOI: 10.1016/j.neuroscience.2016.03.056] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/29/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
|
25
|
Cornejo F, von Bernhardi R. Age-Dependent Changes in the Activation and Regulation of Microglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:205-226. [DOI: 10.1007/978-3-319-40764-7_10] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
26
|
Monteiro S, Ferreira FM, Pinto V, Roque S, Morais M, de Sá-Calçada D, Mota C, Correia-Neves M, Cerqueira JJ. Absence of IFNγ promotes hippocampal plasticity and enhances cognitive performance. Transl Psychiatry 2016; 6:e707. [PMID: 26731444 PMCID: PMC5073154 DOI: 10.1038/tp.2015.194] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 01/31/2023] Open
Abstract
Cognitive functioning can be differentially modulated by components of the immune system. Interferon-γ (IFNγ) is a pro-inflammatory cytokine whose production is altered in many conditions displaying some degree of cognitive deficits, although its role in cognitive functioning is still unclear. Here we show that the absence of IFNγ selectively enhances cognitive behaviours in tasks in which the hippocampus is implicated. Moreover, the absence of IFNγ leads to volumetric and cell density changes that are restricted to the dorsal part of the hippocampus. In the dorsal hippocampus, the absence of this pro-inflammatory cytokine leads to an increase in the numbers of newly born neurons in the subgranular zone of the dentate gyrus (DG), an adult neurogenic niche known to support learning and memory, and to an enlargement of the dendritic arborization of DG granule and cornu ammonis (CA)1 pyramidal neurons. Moreover, it also modestly impacts synaptic plasticity, by decreasing the paired-pulse facilitation in the Schaffer collateral to CA1 pyramidal cell synapses. Taken together, our results provide evidence that IFNγ is a negative regulator of hippocampal functioning, as its absence positively impacts on dorsal hippocampus structure, cell density, neuronal morphology and synaptic plasticity. Importantly, these neuroplastic changes are associated with improved performance in learning and memory tasks. Therefore, blockage of the IFNγ signalling may present as promising therapeutic targets for the treatment of inflammation-associated cognitive dysfunction.
Collapse
Affiliation(s)
- S Monteiro
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - F M Ferreira
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - V Pinto
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - S Roque
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - M Morais
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - D de Sá-Calçada
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - C Mota
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - M Correia-Neves
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J J Cerqueira
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus de Gualtar, Braga, Portugal,Life and Health Sciences Research Institute, 3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal,Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, ICVS/3B's - PT Government Associate Laboratory, 4710-057 Braga, Portugal. E-mail:
| |
Collapse
|
27
|
von Bernhardi R, Eugenín-von Bernhardi L, Eugenín J. Microglial cell dysregulation in brain aging and neurodegeneration. Front Aging Neurosci 2015; 7:124. [PMID: 26257642 PMCID: PMC4507468 DOI: 10.3389/fnagi.2015.00124] [Citation(s) in RCA: 390] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/22/2015] [Indexed: 12/29/2022] Open
Abstract
Aging is the main risk factor for neurodegenerative diseases. In aging, microglia undergoes phenotypic changes compatible with their activation. Glial activation can lead to neuroinflammation, which is increasingly accepted as part of the pathogenesis of neurodegenerative diseases, including Alzheimer’s disease (AD). We hypothesize that in aging, aberrant microglia activation leads to a deleterious environment and neurodegeneration. In aged mice, microglia exhibit an increased expression of cytokines and an exacerbated inflammatory response to pathological changes. Whereas LPS increases nitric oxide (NO) secretion in microglia from young mice, induction of reactive oxygen species (ROS) predominates in older mice. Furthermore, there is accumulation of DNA oxidative damage in mitochondria of microglia during aging, and also an increased intracellular ROS production. Increased ROS activates the redox-sensitive nuclear factor kappa B, which promotes more neuroinflammation, and can be translated in functional deficits, such as cognitive impairment. Mitochondria-derived ROS and cathepsin B, are also necessary for the microglial cell production of interleukin-1β, a key inflammatory cytokine. Interestingly, whereas the regulatory cytokine TGFβ1 is also increased in the aged brain, neuroinflammation persists. Assessing this apparent contradiction, we have reported that TGFβ1 induction and activation of Smad3 signaling after inflammatory stimulation are reduced in adult mice. Other protective functions, such as phagocytosis, although observed in aged animals, become not inducible by inflammatory stimuli and TGFβ1. Here, we discuss data suggesting that mitochondrial and endolysosomal dysfunction could at least partially mediate age-associated microglial cell changes, and, together with the impairment of the TGFβ1-Smad3 pathway, could result in the reduction of protective activation and the facilitation of cytotoxic activation of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, Chile
| | | | - Jaime Eugenín
- Laboratory of Neural Systems, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile (USACH) Santiago, Chile
| |
Collapse
|
28
|
Eimerbrink M, White J, Pendry R, Hodges S, Sadler L, Wiles J, Weintraub M, Chumley M, Boehm G. Administration of the inverse benzodiazepine agonist MRK-016 rescues acquisition and memory consolidation following peripheral administration of bacterial endotoxin. Behav Brain Res 2015; 288:50-3. [DOI: 10.1016/j.bbr.2015.03.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/23/2015] [Accepted: 03/22/2015] [Indexed: 12/17/2022]
|
29
|
Growth hormone, insulin-like growth factor-1 and the aging brain. Exp Gerontol 2014; 68:76-81. [PMID: 25300732 DOI: 10.1016/j.exger.2014.10.002] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Abstract
Growth hormone (GH) and insulin-like growth factor (IGF)-1 regulate the development and function of cells throughout the body. Several clinical diseases that result in a decline in physical and mental functions are marked by mutations that disrupt GH or IGF-1 signaling. During the lifespan there is a robust decrease in both GH and IGF-1. Because GH and IGF-1 are master regulators of cellular function, impaired GH and IGF-1 signaling in aging/disease states leads to significant alterations in tissue structure and function, especially within the brain. This review is intended to highlight the effects of the GH and IGF-1 on neuronal structure, function, and plasticity. Furthermore, we address several potential mechanisms through which the age-related reductions in GH and IGF-1 affect cognition. Together, the studies reviewed here highlight the importance of maintaining GH and IGF-1 signaling in order to sustain proper brain function throughout the lifespan.
Collapse
|
30
|
Neuroinflammatory changes negatively impact on LTP: A focus on IL-1β. Brain Res 2014; 1621:197-204. [PMID: 25193603 DOI: 10.1016/j.brainres.2014.08.040] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/14/2014] [Indexed: 02/08/2023]
Abstract
In recent years it has become clear that neuroinflammatory changes develop in the brain with age and that similar, though more profound changes, occur in neurodegenerative conditions and in animal models of neurodegeneration. These changes are linked with deterioration in plasticity and the evidence suggests that a key causative factor is microglial activation and the associated increase in production and release of inflammatory cytokines. Several groups have reported that interleukin (IL)-1β negatively impacts on hippocampal-dependent learning and has an inhibitory effect on LTP although this is concentration-dependent. Similarly other inflammatory cytokines, which are also produced by microglia similarly decrease LTP. The evidence supporting these findings will be reviewed here and will be discussed in the context of considering mechanisms by which the negative impact of neuroinflammation can be ameliorated. This article is part of a Special Issue entitled SI: Brain and Memory.
Collapse
|
31
|
Jones RS, Lynch MA. How dependent is synaptic plasticity on microglial phenotype? Neuropharmacology 2014; 96:3-10. [PMID: 25168262 DOI: 10.1016/j.neuropharm.2014.08.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 11/27/2022]
Abstract
Microglia are particularly plastic cells which can be shifted from their resting state by numerous factors and adopt distinct phenotypes. The cells are multifunctional, though their main role is probably maintenance of homoeostasis. Resting cells are responsible for surveillance, whereas activation induces the cells to adopt neuroprotective or neurodetrimental roles, which are anti-inflammatory or pro-inflammatory respectively. The evidence indicates that activated cells with a pro-inflammatory phenotype predominate in neurodegenerative diseases and models of neurodegeneration and that this may significantly contribute to the deteriorating neuronal function. This question is considered in this review, in particular in the context of animal models of Alzheimer's disease (AD). This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Raasay S Jones
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College, Dublin 2, Ireland.
| | - Marina A Lynch
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College, Dublin 2, Ireland
| |
Collapse
|
32
|
Lynch MA. The impact of neuroimmune changes on development of amyloid pathology; relevance to Alzheimer's disease. Immunology 2014; 141:292-301. [PMID: 23876085 DOI: 10.1111/imm.12156] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/17/2013] [Accepted: 07/18/2013] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammatory changes are a characteristic of several, if not all, neurodegenerative diseases including Alzheimer's disease and are typified by increased microglial activation. Microglia express several receptors making them highly reactive and plastic cells, and, at least in vitro, they adopt different phenotypes in a manner analogous to their peripheral counterparts, macrophages. Microglia also express numerous cell surface proteins enabling them to interact with cells and the evidence indicates that maintenance of microglia in a quiescent state relies, at least to some extent, on an interaction with neurons by means of specific ligand-receptor pairs, for example CD200-CD200R. It is clear that microglia also interact with T cells and recent evidence indicates that co-incubation of microglia with T helper type 1 cells markedly increases their activation. Under normal conditions, small numbers of activated T cells gain entry to the brain and are involved in immune surveillance but infiltration of significant numbers of T cells occurs in disease and following injury. The consequences of T cell infiltration appear to depend on the conditions, with descriptions of both neurodestructive and neuroprotective effects in animal models of different diseases. This review will discuss the modulatory effect of T cells on microglia and the impact of infiltration of T cells into the brain with a focus on Alzheimer's disease, and will propose that infiltration of interferon-γ-producing cells may be an important factor in triggering inflammation that is pathogenic and destructive.
Collapse
Affiliation(s)
- Marina A Lynch
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
33
|
Immune responses in Parkinson's disease: interplay between central and peripheral immune systems. BIOMED RESEARCH INTERNATIONAL 2014; 2014:275178. [PMID: 24822191 PMCID: PMC4005076 DOI: 10.1155/2014/275178] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/16/2014] [Indexed: 02/06/2023]
Abstract
The etiology of Parkinson's disease (PD) is complex and most likely involves numerous environmental and heritable risk factors. Recent studies establish that central and peripheral inflammation occurs in the prodromal stage of the disease and sustains disease progression. Aging, heritable risk factors, or environmental exposures may contribute to the initiation of central or peripheral inflammation. One emerging hypothesis is that inflammation plays a critical role in PD neuropathology. Increasing evidence suggest that activation of the peripheral immune system exacerbates the discordant central inflammatory response and synergistically drives neurodegeneration. We provide an overview of current knowledge on the temporal profile of central and peripheral immune responses in PD and discuss the potential synergistic effects of the central and peripheral inflammation in disease development. The understanding of the nature of the chronic inflammation in disease progression and the possible risk factors that contribute to altered central and peripheral immune responses will offer mechanistic insights into PD etiology and pathology and benefit the development of effective tailored therapeutics for human PD.
Collapse
|
34
|
Immunity and Alzheimer's disease: immunological perspectives on the development of novel therapies. Drug Discov Today 2013; 18:1212-20. [DOI: 10.1016/j.drudis.2013.07.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 07/19/2013] [Accepted: 07/30/2013] [Indexed: 02/07/2023]
|
35
|
Classical activation of microglia in CD200-deficient mice is a consequence of blood brain barrier permeability and infiltration of peripheral cells. Brain Behav Immun 2013; 34:86-97. [PMID: 23916893 DOI: 10.1016/j.bbi.2013.07.174] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 01/08/2023] Open
Abstract
The interaction between CD200, expressed on several cell types, and its receptor CD200R, expressed on cells of the myeloid lineage, has been shown to be an important factor in modulating inflammation in macrophage function in several conditions including colitis and arthritis. More recently its modulatory effect on microglial activation has been identified and CD200-deficiency has been associated with increased microglial activation accompanied by increased production of inflammatory cytokines. The response of glia prepared from CD200-deficient mice to stimuli like lipopolysaccharide (LPS) is markedly greater than the response of cells prepared from wildtype mice and, consistent with this, is the recent observation that expression of Toll-like receptor (TLR)4 and signalling through NFκB are increased in microglia prepared from CD200-deficient mice. Here we show that glia from CD200-deficient mice are also more responsive to interferon-γ (IFNγ) which triggers classical activation of microglia. We investigated the effects of CD200-deficiency in vivo and report that there is an increase in expression of several markers of microglial activation including tumor necrosis factor (TNF)-α, which is a hallmark of classically-activated microglia. These changes are accompanied by increased IFNγ, and the evidence suggests that this is produced by infiltrating cells including T cells and macrophages. We propose that these cells enter the brain as a consequence of increased blood brain barrier (BBB) permeability in CD200-deficient mice and that infiltration is assisted by increased expression of the chemokines, monocyte chemotactic protein-1 (MCP-1), IFNγ-induced protein-10 (IP-10) and RANTES. This may have implications in neurodegenerative diseases where BBB permeability is compromised.
Collapse
|
36
|
Microglial phenotype and adaptation. J Neuroimmune Pharmacol 2013; 8:807-23. [PMID: 23881706 DOI: 10.1007/s11481-013-9490-4] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 07/08/2013] [Indexed: 12/14/2022]
Abstract
Microglia are the prime innate immune cells of the central nervous system. They can transit from a (so-called) resting state under homeostatic conditions towards a pro-inflammatory activation state upon homeostatic disturbances. Under neurodegenerative conditions, microglia have been largely perceived as neurotoxic cells. It is now becoming clear that resting microglia are not inactive but that they serve house-keeping functions. Moreover, microglia activity is not limited to proinflammatory responses, but covers a spectrum of reactive profiles. Depending on the actual situation, activated microglia display specific effector functions supporting inflammation, tissue remodeling, synaptic plasticity and neurogenesis. Many of these functions not only relate to the current state of the local neural environment but also depend on previous experience. In this review, we address microglia functions with respect to determining factors, phenotypic presentations, adaptation to environmental signals and aging. Finally, we point out primary mechanisms of microglia activation, which may comprise therapeutic targets to control neuro-inflammatory and neurodegenerative activity.
Collapse
|
37
|
Increased micro-RNA 29b in the aged brain correlates with the reduction of insulin-like growth factor-1 and fractalkine ligand. Neurobiol Aging 2013; 34:2748-58. [PMID: 23880139 DOI: 10.1016/j.neurobiolaging.2013.06.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/28/2013] [Accepted: 06/14/2013] [Indexed: 12/13/2022]
Abstract
Microglia develop an inflammatory phenotype during normal aging. The mechanism by which this occurs is not well understood, but might be related to impairments in several key immunoregulatory systems. Here we show that micro-RNA (miR)-29a and miR-29b, 2 immunoregulatory micro-RNAs, were increased in the brain of aged BALB/c mice compared with adults. Insulin-like growth factor-1 (IGF-1) and fractalkine ligand (CX3CL1) are negative modulators of microglial activation and were identified as targets of miR-29a and miR-29b using luciferase assay and primary microglia transfection. Indeed, higher expression of miR-29b in the brain of aged mice was associated with reduced messenger RNA (mRNA) levels of IGF-1 and CX3CL1. Parallel to these results in mice, miR-29a and miR-29b were also markedly increased in cortical brain tissue of older individuals (mean, 77 years) compared with middle-aged adults (mean, 45 years). Moreover, increased expression of miR-29b in human cortical tissue was negatively correlated with IGF-1 and CX3CL1 expression. Collectively, these data indicate that an age-associated increase in miR-29 corresponded with the reduction of 2 important regulators of microglia, IGF-1 and CX3CL1.
Collapse
|
38
|
Hung IC, Chang SS, Chang PC, Lee CC, Chen CYC. Memory enhancement by traditional Chinese medicine? J Biomol Struct Dyn 2012; 31:1411-39. [PMID: 23249175 DOI: 10.1080/07391102.2012.741052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Cognitive repair by insulin-like growth factor-I (IGF-I) through activation of insulin-like growth factor-I receptor (IGF-IR) is well established, but not used for clinical therapy due to its link to cancer. We hypothesize that IGF-IR activation rather than IGF-I per se may be essential for cognitive repair and attempted to identify ligands from traditional Chinese medicine (TCM) with drug-like potential towards IGF-IR. TCM ligands, 3-(2-carboxyphenyl)-4(3H)-quinazolinone from Isatisin digotica, (+)-N-methyllaurotetanine from Lindera aggregate, and (+)-1(R)-Coclaurine from Nelumbonucifera Gaertn, exhibited high binding affinities and good blood brain barrier (BBB) penetration crucial for accessing IGF-IR. Stable complex formation of the candidates was observed during molecular dynamics (MD) simulation. Interactions with Leu975 and Gly1055 or Asp1056 were important for ligand binding. Amino acid distance analysis revealed residues 974/975, 984-986, 996-1006, 1040-1056, and 1122-1135 as "hotspots" for ligand binding in IGF-IR. Versatile entry pathways for the TCM candidates suggest high accessibility to the binding site. Blockage of the binding site opening by the TCM candidates limits binding site access by other compounds. Multiple linear regression (R² = 0.9715), support vector machine (R² = 0.9084), Bayesian network (R² =0.8233) comparative molecular field analysis (CoMFA, R² = 0.9941), and comparative molecular similarity indices analysis (CoMSIA, R² = 0.9877) models consistently suggest that the TCM candidates might exert bioactivity on IGF-IR. Contour of representative MD conformations to CoMFA and CoMSIA maps exhibits similar results. Properties including BBB passage, evidence of ability to form stable complexes with IGF-IR by MD simulation, and predicted bioactivity suggest that the TCM candidates have drug-like properties and might have potential as cognitive-enhancing drugs.
Collapse
Affiliation(s)
- I-Chi Hung
- a School of Pharmacy, China Medical University , Taichung , 40402 , Taiwan
| | | | | | | | | |
Collapse
|
39
|
Varnum MM, Ikezu T. The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimer's disease brain. Arch Immunol Ther Exp (Warsz) 2012; 60:251-66. [PMID: 22710659 PMCID: PMC4429536 DOI: 10.1007/s00005-012-0181-2] [Citation(s) in RCA: 308] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/20/2012] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline of cognitive function. There is no therapy that can halt or reverse its progression. Contemporary research suggests that age-dependent neuroinflammatory changes may play a significant role in the decreased neurogenesis and cognitive impairments in AD. The innate immune response is characterized by pro-inflammatory (M1) activation of macrophages and subsequent production of specific cytokines, chemokines, and reactive intermediates, followed by resolution and alternative activation for anti-inflammatory signaling (M2a) and wound healing (M2c). We propose that microglial activation phenotypes are analogous to those of macrophages and that their activation plays a significant role in regulating neurogenesis in the brain. Microglia undergo a switch from an M2- to an M1-skewed activation phenotype during aging. This review will assess the neuroimmunological studies that led to characterization of the different microglial activation states in AD mouse models. It will also discuss the roles of microglial activation on neurogenesis in AD and propose anti-inflammatory molecules as exciting therapeutic targets for research. Molecules such as interleukin-4 and CD200 have proven to be important anti-inflammatory mediators in the regulation of neuroinflammation in the brain, which will be discussed in detail for their therapeutic potential.
Collapse
Affiliation(s)
- Megan M. Varnum
- Laboratory of Molecular NeuroTherapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
- Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Tsuneya Ikezu
- Laboratory of Molecular NeuroTherapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Kohman RA, DeYoung EK, Bhattacharya TK, Peterson LN, Rhodes JS. Wheel running attenuates microglia proliferation and increases expression of a proneurogenic phenotype in the hippocampus of aged mice. Brain Behav Immun 2012; 26:803-10. [PMID: 22056294 PMCID: PMC3275652 DOI: 10.1016/j.bbi.2011.10.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/17/2011] [Accepted: 10/18/2011] [Indexed: 11/23/2022] Open
Abstract
Aging is associated with low-grade neuroinflammation including primed microglia that may contribute to deficits in neural plasticity and cognitive function. The current study evaluated whether exercise modulates division and/or activation state of microglia in the dentate gyrus of the hippocampus, as activated microglia can express a classic inflammatory or an alternative neuroprotective phenotype. We also assessed hippocampal neurogenesis to determine whether changes in microglia were associated with new neuron survival. Adult (3.5 months) and aged (18 months) male BALB/c mice were individually housed with or without running wheels for 8 weeks. Mice received bromodeoxyuridine injections during the first or last 10 days of the experiment to label dividing cells. Immunofluorescence was conducted to measure microglia division, co-expression of the neuroprotective indicator insulin-like growth factor (IGF-1), and new neuron survival. The proportion of new microglia was increased in aged mice, and decreased from wheel running. Running increased the proportion of microglia expressing IGF-1 suggesting exercise shifts microglia phenotype towards neuroprotection. Additionally, running enhanced survival of new neurons in both age groups. Findings suggest that wheel running may attenuate microglia division and promote a proneurogenic phenotype in aged mice.
Collapse
Affiliation(s)
- Rachel A Kohman
- Department of Psychology, University of Illinois, Beckman Institute, Urbana, IL 61801-3873, USA.
| | | | | | | | | |
Collapse
|
41
|
Deak F, Sonntag WE. Aging, synaptic dysfunction, and insulin-like growth factor (IGF)-1. J Gerontol A Biol Sci Med Sci 2012; 67:611-25. [PMID: 22503992 PMCID: PMC3348499 DOI: 10.1093/gerona/gls118] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/16/2012] [Indexed: 01/05/2023] Open
Abstract
Insulin-like growth factor (IGF)-1 is an important neurotrophic hormone. Deficiency of this hormone has been reported to influence the genesis of cognitive impairment and dementia in the elderly patients. Nevertheless, there are studies indicating that cognitive function can be maintained into old age even in the absence of circulating IGF-1 and studies that link IGF-1 to an acceleration of neurological diseases. Although IGF-1 has a complex role in brain function, synaptic effects appear to be central to the IGF-1-induced improvement in learning and memory. In this review, synaptic mechanisms of learning and memory and the effects of IGF-1 on synaptic communication are discussed. The emerging data indicate that synaptic function decreases with age and that IGF-1 contributes to information processing in the brain. Further studies that detail the specific actions of this important neurotrophic hormone will likely lead to therapies that result in improved cognitive function for the elderly patients.
Collapse
Affiliation(s)
- Ferenc Deak
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - William E. Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, Oklahoma City, Oklahoma
| |
Collapse
|
42
|
Prefrontal dopaminergic and enkephalinergic synaptic accommodation in HIV-associated neurocognitive disorders and encephalitis. J Neuroimmune Pharmacol 2012; 7:686-700. [PMID: 22391864 PMCID: PMC3419353 DOI: 10.1007/s11481-012-9345-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 02/06/2012] [Indexed: 12/15/2022]
Abstract
Changes in synapse structure occur in frontal neocortex with HIV encephalitis (HIVE) and may contribute to HIV-associated neurocognitive disorders (HAND). A postmortem survey was conducted to determine if mRNAs involved in synaptic transmission are perturbed in dorsolateral prefrontal cortex (DLPFC) in subjects with HIVE or HAND. Expression of the opioid neurotransmitter preproenkephalin mRNA (PENK) was significantly decreased in a sampling of 446 brain specimens from HIV-1 infected people compared to 67 HIV negative subjects. Decreased DLPFC PENK was most evident in subjects with HIVE and/or increased expression of interferon regulatory factor 1 mRNA (IRF1). Type 2 dopamine receptor mRNA (DRD2L) was decreased significantly, but not in the same set of subjects with PENK dysregulation. DRD2L downregulation occurred primarily in the subjects without HIVE or neurocognitive impairment. Subjects with neurocognitive impairment often failed to significantly downregulate DRD2L and had abnormally high IRF1 expression. Conclusion: Dysregulation of synaptic preproenkephalin and DRD2L in frontal neocortex can occur with and without neurocognitive impairment in HIV-infected people. Downregulation of DRD2L in the prefrontal cortex was associated with more favorable neuropsychological and neuropathological outcomes; the failure to downregulate DRD2L was significantly less favorable. PENK downregulation was related neuropathologically to HIVE, but was not related to neuropsychological outcome independently. Emulating endogenous synaptic plasticity pharmacodynamically could enhance synaptic accommodation and improve neuropsychological and neuropathological outcomes in HIV/AIDS.
Collapse
|
43
|
Abstract
Over the years it has become evident that the immune system can affect the function of the central nervous system (CNS), including altering cognitive processes. The impact of immune activation on the CNS is particularly important for aged individuals, as the brain's resident immune cells, microglia, acquire a pro-inflammatory profile. The low-grade chronic neuroinflammation that develops with normal aging likely contributes to the susceptibility to cognitive deficits and a host of age-related pathologies. Understanding why microglia show increased inflammatory activity (i.e., neuroinflammation) and identifying effective treatments to reduce microglia activation is expected to have beneficial effects on cognitive performance and measures of neural plasticity. However, microglia also promote regeneration after injury. Therefore, effective treatments must dampen inflammatory activity while preserving microglia's neuroprotective function. Discovering factors that induce neuroinflammation and investigating potential preventative therapies is expected to uncover the ways of maintaining normal microglia activity in the aged brain.
Collapse
Affiliation(s)
- Rachel A Kohman
- Department of Psychology, University of Illinois at Urbana-Champaign, Beckman Institute, Urbana, IL, USA.
| |
Collapse
|
44
|
Lyons A, Murphy KJ, Clarke R, Lynch MA. Atorvastatin prevents age-related and amyloid-β-induced microglial activation by blocking interferon-γ release from natural killer cells in the brain. J Neuroinflammation 2011; 8:27. [PMID: 21453520 PMCID: PMC3077319 DOI: 10.1186/1742-2094-8-27] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/31/2011] [Indexed: 12/01/2022] Open
Abstract
Background Microglial function is modulated by several factors reflecting the numerous receptors expressed on the cell surface, however endogenous factors which contribute to the age-related increase in microglial activation remain largely unknown. One possible factor which may contribute is interferon-γ (IFNγ). IFNγ has been shown to increase in the aged brain and potently activates microglia, although its endogenous cell source in the brain remains unidentified. Methods Male Wistar rats were used to assess the effect of age and amyloid-β (Aβ) on NK cell infiltration into the brain. The effect of the anti-inflammatory compound, atorvastatin was also assessed under these conditions. We measured cytokine and chemokine (IFNγ, IL-2, monocyte chemoattractant protein-1 (MCP-1) and IFNγ-induced protein 10 kDa (IP-10)), expression in the brain by appropriate methods. We also looked at NK cell markers, CD161, NKp30 and NKp46 using flow cytometry and western blot. Results Natural killer (NK) cells are a major source of IFNγ in the periphery and here we report the presence of CD161+ NKp30+ cells and expression of CD161 and NKp46 in the brain of aged and Aβ-treated rats. Furthermore, we demonstrate that isolated CD161+ cells respond to interleukin-2 (IL-2) by releasing IFNγ. Atorvastatin, the HMG-CoA reductase inhibitor, attenuates the increase in CD161 and NKp46 observed in hippocampus of aged and Aβ-treated rats. This was paralleled by a decrease in IFNγ, markers of microglial activation and the chemokines, MCP-1 and IP-10 which are chemotactic for NK cells. Conclusions We propose that NK cells contribute to the age-related and Aβ-induced neuroinflammatory changes and demonstrate that these changes can be modulated by atorvastatin treatment.
Collapse
Affiliation(s)
- Anthony Lyons
- Physiology Department, Trinity College Institute for Neuroscience, Trinity College, Dublin 2, Ireland.
| | | | | | | |
Collapse
|
45
|
Park SE, Dantzer R, Kelley KW, McCusker RH. Central administration of insulin-like growth factor-I decreases depressive-like behavior and brain cytokine expression in mice. J Neuroinflammation 2011; 8:12. [PMID: 21306618 PMCID: PMC3045937 DOI: 10.1186/1742-2094-8-12] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 02/09/2011] [Indexed: 01/12/2023] Open
Abstract
Exogenous administration of insulin-like growth factor (IGF)-I has anti-depressant properties in rodent models of depression. However, nothing is known about the anti-depressant properties of IGF-I during inflammation, nor have mechanisms by which IGF-I alters behavior following activation of the innate immune system been clarified. We hypothesized that central IGF-I would diminish depressive-like behavior on a background of an inflammatory response and that it would do so by inducing expression of the brain-derived neurotrophic factor (BDNF) while decreasing pro-inflammatory cytokine expression in the brain. IGF-I (1,000 ng) was administered intracerebroventricularly (i.c.v.) to CD-1 mice. Mice were subsequently given lipopolysaccharide i.c.v. (LPS, 10 ng). Sickness and depressive-like behaviors were assessed followed by analysis of brain steady state mRNA expression. Central LPS elicited typical transient signs of sickness of mice, including body weight loss, reduced feed intake and decreased social exploration toward a novel juvenile. Similarly, LPS increased time of immobility in the tail suspension test (TST). Pretreatment with IGF-I or antidepressants significantly decreased duration of immobility in the TST in both the absence and presence of LPS. To elucidate the mechanisms underlying the anti-depressant action of IGF-I, we quantified steady-state mRNA expression of inflammatory mediators in whole brain using real-time RT-PCR. LPS increased, whereas IGF-I decreased, expression of inflammatory markers interleukin-1ß (IL-1ß), tumor necrosis factor-(TNF)α, inducible nitric oxide synthase (iNOS) and glial fibrillary acidic protein (GFAP). Moreover, IGF-I increased expression of BDNF. These results indicate that IGF-I down regulates glial activation and induces expression of an endogenous growth factor that shares anti-depressant activity. These actions of IGF-I parallel its ability to diminish depressive-like behavior.
Collapse
Affiliation(s)
- Sook-Eun Park
- Integrated Immunology and Behavior Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801-3873, USA
| | | | | | | |
Collapse
|
46
|
Cytokines and neuronal channels: A molecular basis for age-related decline of neuronal function? Exp Gerontol 2011; 46:199-206. [DOI: 10.1016/j.exger.2010.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/14/2010] [Accepted: 09/15/2010] [Indexed: 01/09/2023]
|
47
|
Wong-Goodrich SJ, Pfau ML, Flores CT, Fraser JA, Williams CL, Jones LW. Voluntary running prevents progressive memory decline and increases adult hippocampal neurogenesis and growth factor expression after whole-brain irradiation. Cancer Res 2010; 70:9329-38. [PMID: 20884629 PMCID: PMC2982943 DOI: 10.1158/0008-5472.can-10-1854] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Whole-brain irradiation (WBI) therapy produces progressive learning and memory deficits in patients with primary or secondary brain tumors. Exercise enhances memory and adult hippocampal neurogenesis in the intact brain, so we hypothesized that exercise may be an effective treatment to alleviate consequences of WBI. Previous studies using animal models to address this issue have yielded mixed results and have not examined potential molecular mechanisms. We investigated the short- and long-term effects of WBI on spatial learning and memory retention and determined whether voluntary running after WBI aids recovery of brain and cognitive function. Forty adult female C57Bl/6 mice given a single dose of 5 Gy or sham WBI were trained 2.5 weeks and up to 4 months after WBI in a Barnes maze. Half of the mice received daily voluntary wheel access starting 1 month after sham or WBI. Daily running following WBI prevented the marked decline in spatial memory retention observed months after irradiation. Bromodeoxyuridine (BrdUrd) immunolabeling and enzyme-linked immunosorbent assay indicated that this behavioral rescue was accompanied by a partial restoration of newborn BrdUrd+/NeuN+ neurons in the dentate gyrus and increased hippocampal expression of brain-derived vascular endothelial growth factor and insulin-like growth factor-1, and occurred despite irradiation-induced elevations in hippocampal proinflammatory cytokines. WBI in adult mice produced a progressive memory decline consistent with what has been reported in cancer patients receiving WBI therapy. Our findings show that running can abrogate this memory decline and aid recovery of adult hippocampal plasticity, thus highlighting exercise as a potential therapeutic intervention.
Collapse
Affiliation(s)
| | - Madeline L. Pfau
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Catherine T. Flores
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jennifer A. Fraser
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Lee W. Jones
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
48
|
Zhong Y, Zhou LJ, Ren WJ, Xin WJ, Li YY, Zhang T, Liu XG. The direction of synaptic plasticity mediated by C-fibers in spinal dorsal horn is decided by Src-family kinases in microglia: the role of tumor necrosis factor-alpha. Brain Behav Immun 2010; 24:874-80. [PMID: 20116424 DOI: 10.1016/j.bbi.2010.01.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 01/16/2010] [Accepted: 01/19/2010] [Indexed: 12/27/2022] Open
Abstract
Previous studies have shown that Src-family kinases (SFKs) are selectively activated in spinal microglia following peripheral nerve injury and the activated SFKs play a key role for the development of neuropathic pain. To investigate the underlying mechanism, in the present study the effect of SFKs on long-term potentiation (LTP) at C-fiber synapses in spinal dorsal horn, which is believed as central mechanism of neuropathic pain, was investigated in adult rats. Electrophysiological data revealed that pretreatment with either microglia inhibitor (minocycline, 200 microM) or SFKs inhibitors (PP2, 100 microM and SU6656, 200 microM) reversed the effect of high frequency stimulation (HFS), that is, HFS, which induces long-term potentiation (LTP) normally, induced long-term depression (LTD) after inhibition of either microglia or SFKs. Western blotting analysis showed that the level of phosphorylated SFKs (p-SFKs) in ipsilateral spinal dorsal horn was transiently increased after LTP induced by HFS, starting at 15 min and returning to control level at 60 min after HFS. Double-labeled immunofluorescence staining demonstrated that p-SFKs were highly restricted to microglia. Furthermore, we found that the inhibitory effects of minocycline or SU6656 on spinal LTP were reversed by spinal application of rat recombinant tumor necrosis factor-alpha (TNF-alpha 0.5 ng/ml, 200 microl). HFS failed to induce LTP of C-fiber evoked field potentials in TNF receptor-1 knockout mice and in rats pretreated with TNF-alpha neutralization antibody (0.6 microg/ml, 200 microl). The results suggested that in spinal dorsal horn activation of SFKs in microglia might control the direction of plastic changes at C-fiber synapses and TNF-alpha might be involved in the process.
Collapse
Affiliation(s)
- Yi Zhong
- Pain Research Center, Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Profenno LA, Porsteinsson AP, Faraone SV. Meta-analysis of Alzheimer's disease risk with obesity, diabetes, and related disorders. Biol Psychiatry 2010; 67:505-12. [PMID: 19358976 DOI: 10.1016/j.biopsych.2009.02.013] [Citation(s) in RCA: 486] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 02/10/2009] [Accepted: 02/19/2009] [Indexed: 12/15/2022]
Abstract
BACKGROUND Late-onset Alzheimer's disease (AD) is a multifactorial and heterogeneous disorder with major risk factors including advanced age, presence of an apolipoprotein E epsilon4 (APOE4) allele, and family history of AD. Other risk factors may be obesity and diabetes and related disorders, which are highly prevalent. METHODS We reviewed longitudinal epidemiological studies of body mass, diabetes, metabolic syndrome, and glucose and insulin levels on risk for AD. We conducted meta-analyses of the results from these studies. RESULTS For obesity assessed by body mass index, the pooled effect size for AD was 1.59 (95% confidence interval [CI] 1.02-2.5; z = 2.0; p = .042), and for diabetes, the pooled effect size for AD was 1.54 (95% CI 1.33-1.79; z = 5.7; p < .001). Egger's test did not find significant evidence for publication bias in the meta-analysis for obesity (t = -1.4, p = .21) or for diabetes (t = -.86, p = .42). Since these disorders are highly comorbid, we conducted a meta-analysis combining all studies of obesity, diabetes, and abnormal glucose or insulin levels, which yielded a highly significant pooled effect size for AD of 1.63 (95% CI 1.39-1.92; z = 5.9; p < .001). CONCLUSIONS Obesity and diabetes significantly and independently increase risk for AD. Though the level of risk is less than that with the APOE4 allele, the high prevalence of these disorders may result in substantial increases in future incidence of AD. Physiological changes common to obesity and diabetes plausibly promote AD.
Collapse
Affiliation(s)
- Louis A Profenno
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, New York, USA.
| | | | | |
Collapse
|
50
|
Lynch MA. Age-related neuroinflammatory changes negatively impact on neuronal function. Front Aging Neurosci 2010; 1:6. [PMID: 20552057 PMCID: PMC2874409 DOI: 10.3389/neuro.24.006.2009] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 12/08/2009] [Indexed: 12/25/2022] Open
Abstract
Neuroinflammatory changes, characterized by an increase in microglial activation and often accompanied by upregulation of inflammatory cytokines like interleukin-1β (IL-1β), are common to many, if not all, neurodegenerative diseases. Similar, though less dramatic neuroinflammatory changes, are also known to occur with age. Among the consequences of these changes is an impairment in synaptic function and the evidence suggests that inflammatory cytokines may be the primary contributory factor responsible for the deficits in synaptic plasticity which have been identified in aged rodents. Specifically a decrease in the ability of aged rats to sustain long-term potentiation (LTP) in perforant path-granule cells of the hippocampus is associated with increased microglial activation. This review considers the evidence which suggests a causal relationship between these changes and the factors which contribute to the age-related microglial activation, and reflects on data which demonstrate that agents which inhibit microglial activation also improve ability of rats to sustain LTP.
Collapse
Affiliation(s)
- Marina A Lynch
- Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| |
Collapse
|