1
|
Lymer J, Bergman H, Yang S, Mallick R, Galea LAM, Choleris E, Fergusson D. The effects of estrogens on spatial learning and memory in female rodents - A systematic review and meta-analysis. Horm Behav 2024; 164:105598. [PMID: 38968677 DOI: 10.1016/j.yhbeh.2024.105598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 07/07/2024]
Abstract
Estrogens have inconsistent effects on learning and memory in both the clinical and preclinical literature. Preclinical literature has the advantage of investigating an array of potentially important factors contributing to the varied effects of estrogens on learning and memory, with stringently controlled studies. This study set out to identify specific factors in the animal literature that influence the effects of estrogens on cognition, for possible translation back to clinical practice. The literature was screened and studies meeting strict inclusion criteria were included in the analysis. Eligible studies included female ovariectomized rodents with an adequate vehicle for the estrogen treatment, with an outcome of spatial learning and memory in the Morris water maze. Training days of the Morris water maze were used to assess acquisition of spatial learning, and the probe trial was used to evaluate spatial memory recall. Continuous outcomes were pooled using a random effects inverse variance method and reported as standardized mean differences with 95 % confidence intervals. Subgroup analyses were developed a priori to assess important factors. The overall analysis favoured treatment for the later stages of training and for the probe trial. Factors including the type of estrogen, route, schedule of administration, age of animals, timing relative to ovariectomy, and duration of treatment were all found to be important. The subgroup analyses showed that chronic treatment with 17β-estradiol, either cyclically or continuously, to young animals improved spatial recall. These results, observed in animals, can inform and guide further clinical research on hormone replacement therapy for cognitive benefits.
Collapse
Affiliation(s)
- Jennifer Lymer
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.
| | - Hailey Bergman
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Sabrina Yang
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | | | - Liisa A M Galea
- Department of Psychiatry, University of Toronto, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada.
| | - Dean Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Mukhina KA, Mitkevich VA, Popova IY. Cannula Implantation Reduces the Severity of the Beta Amyloid Effect on Peroxidized Lipids and Glutathione Levels in the Brain of BALB/c Mice. Acta Naturae 2024; 16:51-59. [PMID: 39555175 PMCID: PMC11569840 DOI: 10.32607/actanaturae.27439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/06/2024] [Indexed: 11/19/2024] Open
Abstract
Sporadic Alzheimer's disease (sAD) is the most common of neurodegenerative disorders. The lack of effective therapy indicates that the mechanisms of sAD development remain poorly understood. To investigate this pathology in animals, intracerebroventricular injection of β-amyloid peptide (Aβ) using a Hamilton syringe, either during stereotactic surgery or through a pre-implanted cannula, is used. In this study, we analyzed the effect of chronic cannula implantation on the severity of Aβ effects at the behavioral, histological, and biochemical levels. The results showed that the local damage to neural tissue caused by cannulation has no bearing on the effect of Aβ on animal behavior and the microglial parameters of the unilateral hippocampus two weeks after the Aβ administration. However, cannula implantation fundamentally modifies some biochemical markers of the oxidative stress that occurs in the brain tissue in response to Aβ administration. Thus, the presence of a cannula reduces the severity of the Aβ impact on the levels of peroxidized lipids and glutathione two- and 10-fold, respectively. It is important to note that the detected changes are chronic and systemic. This is known because the homogenate of the entire contralateral (in relation to the cannula implantation site) hemisphere was analyzed, and the analysis was performed two weeks after implantation. At the same time, cannulation does not affect the rate of reactive oxygen species production. The obtained data indicate that chronic implantation of a cannula into the brain of experimental animals fundamentally distorts some parameters of oxidative stress in the neural tissue, which are widely used to assess the severity of experimental Alzheimer's-type diseases.
Collapse
Affiliation(s)
- K. A. Mukhina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991 Russian Federation
| | - V. A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991 Russian Federation
| | - I. Yu. Popova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991 Russian Federation
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Russian Federation
| |
Collapse
|
3
|
Frank MG, Baratta MV. Use of an immunocapture device to detect cytokine release in discrete brain regions. Neural Regen Res 2024; 19:703-704. [PMID: 37843193 PMCID: PMC10664115 DOI: 10.4103/1673-5374.382237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
- Matthew G. Frank
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Michael V. Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
4
|
González Olmo BM, Bettes MN, DeMarsh JW, Zhao F, Askwith C, Barrientos RM. Short-term high-fat diet consumption impairs synaptic plasticity in the aged hippocampus via IL-1 signaling. NPJ Sci Food 2023; 7:35. [PMID: 37460765 DOI: 10.1038/s41538-023-00211-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
More Americans are consuming diets higher in saturated fats and refined sugars than ever before. These trends could have serious consequences for the older population because high-fat diet (HFD) consumption, known to induce neuroinflammation, has been shown to accelerate and aggravate memory declines. We have previously demonstrated that short-term HFD consumption, which does not evoke obesity-related comorbidities, produced profound impairments to hippocampal-dependent memory in aged rats. These impairments were precipitated by increases in proinflammatory cytokines, primarily interleukin-1 beta (IL-1β). Here, we explored the extent to which short-term HFD consumption disrupts hippocampal synaptic plasticity, as measured by long-term potentiation (LTP), in young adult and aged rats. We demonstrated that (1) HFD disrupted late-phase LTP in the hippocampus of aged, but not young adult rats, (2) HFD did not disrupt early-phase LTP, and (3) blockade of the IL-1 receptor rescued L-LTP in aged HFD-fed rats. These findings suggest that hippocampal memory impairments in aged rats following HFD consumption occur through the deterioration of synaptic plasticity and that IL-1β is a critical driver of that deterioration.
Collapse
Affiliation(s)
- Brigitte M González Olmo
- Department of Biomedical Education & Anatomy, Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Fangli Zhao
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Candice Askwith
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Yamazaki A, Shue F, Yamazaki Y, Martens YA, Bu G, Liu CC. Preparation of single cell suspensions enriched in mouse brain vascular cells for single-cell RNA sequencing. STAR Protoc 2021; 2:100715. [PMID: 34401781 PMCID: PMC8353357 DOI: 10.1016/j.xpro.2021.100715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cerebral blood vessels supply oxygen and nutrients, remove metabolic waste, and play a critical role in maintaining brain homeostasis. Cerebrovasculature is composed of heterogeneous populations of brain vascular cells (BVCs). A major challenge in effective cerebrovascular transcriptional profiling is high-quality BVC procurement, permitting high sequencing depth. Here, we establish cell isolation procedures for glio-vascular cell-enriched single-cell RNA sequencing enabling unbiased characterization of BVC transcriptional heterogeneity. Our approach can be used to address vascular-specific contribution to brain diseases. For complete details on the use and execution of this protocol, please refer to Yamazaki et al. (2021).
Collapse
Affiliation(s)
- Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuka A. Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
6
|
Perkins AE, Piazza MK, Vore AS, Deak MM, Varlinskaya EI, Deak T. Assessment of neuroinflammation in the aging hippocampus using large-molecule microdialysis: Sex differences and role of purinergic receptors. Brain Behav Immun 2021; 91:546-555. [PMID: 33166661 PMCID: PMC8454272 DOI: 10.1016/j.bbi.2020.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/10/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Aging is associated with an enhanced neuroinflammatory response to acute immune challenge, often termed "inflammaging." However, there are conflicting reports about whether baseline levels of inflammatory markers are elevated under ambient conditions in the aging brain, or whether such changes are observed predominantly in response to acute challenge. The present studies utilized two distinct approaches to assess inflammatory markers in young and aging Fischer 344 rats. Experiment 1 examined total tissue content of inflammatory markers from hippocampus of adult (3 month), middle-aged (12 month), and aging (18 month) male Fischer (F) 344 rats using multiplex analysis (23-plex). Though trends emerged for several cytokines, no significant differences in basal tissue content were observed across the 3 ages examined. Experiment 2 measured extracellular concentrations of inflammatory factors in the hippocampus from adult (3 month) and aging (18 month) males and females using large-molecule in vivo microdialysis. Although few significant aging-related changes were observed, robust sex differences were observed in extracellular concentrations of CCL3, CCL20, and IL-1α. Experiment 2 also evaluated the involvement of the P2X7 purinergic receptor in neuroinflammation using reverse dialysis of the selective agonist BzATP. BzATP produced an increase in IL-1α and IL-1β release and rapidly suppressed the release of CXCL1, CCL2, CCL3, CCL20, and IL-6. Other noteworthy sex by aging trends were observed in CCL3, IL-1β, and IL-6. Together, these findings provide important new insight into late-aging and sex differences in neuroinflammation, and their regulation by the P2X7 receptor.
Collapse
Affiliation(s)
- Amy E. Perkins
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton, NY 13902-6000,Department of Psychology, Purdue University Fort Wayne, Fort Wayne, IN 46805
| | - Michelle K. Piazza
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton, NY 13902-6000,Department of Psychology, Purdue University Fort Wayne, Fort Wayne, IN 46805
| | - Andrew S. Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton, NY 13902-6000
| | - Molly M. Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton, NY 13902-6000
| | - Elena I. Varlinskaya
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University—SUNY, Binghamton, NY 13902-6000
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY 13902-6000, United States.
| |
Collapse
|
7
|
Frank MG, Baratta MV, Zhang K, Fallon IP, Pearson MA, Liu G, Hutchinson MR, Watkins LR, Goldys EM, Maier SF. Acute stress induces the rapid and transient induction of caspase-1, gasdermin D and release of constitutive IL-1β protein in dorsal hippocampus. Brain Behav Immun 2020; 90:70-80. [PMID: 32750541 PMCID: PMC7544655 DOI: 10.1016/j.bbi.2020.07.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 01/31/2023] Open
Abstract
The proinflammatory cytokine interleukin (IL)-1β plays a pivotal role in the behavioral manifestations (i.e., sickness) of the stress response. Indeed, exposure to acute and chronic stressors induces the expression of IL-1β in stress-sensitive brain regions. Thus, it is typically presumed that exposure to stressors induces the extra-cellular release of IL-1β in the brain parenchyma. However, this stress-evoked neuroimmune phenomenon has not been directly demonstrated nor has the cellular process of IL-1β release into the extracellular milieu been characterized in brain. This cellular process involves a form of inflammatory cell death, termed pyroptosis, which involves: 1) activation of caspase-1, 2) caspase-1 maturation of IL-1β, 3) caspase-1 cleavage of gasdermin D (GSDMD), and 4) GSDMD-induced permeability of the cell membrane through which IL-1β is released into the extracellular space. Thus, the present study examined whether stress induces the extra-cellular release of IL-1β and engages the above cellular process in mediating IL-1β release in the brain. Male Sprague-Dawley rats were exposed to inescapable tailshock (IS). IL-1β extra-cellular release, caspase-1 activity and cleavage of GSDMD were measured in dorsal hippocampus. We found that exposure to IS induced a transient increase in the release of IL-1β into the extracellular space immediately after termination of the stressor. IS also induced a transient increase in caspase-1 activity prior to IL-1β release, while activation of GSDMD was observed immediately after termination of the stressor. IS also increased mRNA and protein expression of the ESCRTIII protein CHMP4B, which is involved in cellular repair. The present results suggest that exposure to an acute stressor induces the hallmarks of pyroptosis in brain, which might serve as a key cellular process involved in the release of IL-1β into the extracellular milieu of the brain parenchyma.
Collapse
Affiliation(s)
- Matthew G. Frank
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO;,Corresponding Author: Department of Psychology and Neuroscience, Center for Neuroscience, Campus Box 603, University of Colorado Boulder, Boulder, CO, 80301, USA, Tel: +1-303-919-8116,
| | - Michael V. Baratta
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Kaixin Zhang
- ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Macquarie University, North Ryde, Australia;,Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, Australia
| | - Isabella P. Fallon
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Mikayleigh A. Pearson
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, Australia;,International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, China
| | - Mark R. Hutchinson
- Adelaide Medical School & ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), The University of Adelaide, Adelaide, Australia
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Ewa M. Goldys
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, Australia
| | - Steven F. Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
8
|
Banerjee P, Paza E, Perkins EM, James OG, Kenkhuis B, Lloyd AF, Burr K, Story D, Yusuf D, He X, Backofen R, Dando O, Chandran S, Priller J. Generation of pure monocultures of human microglia-like cells from induced pluripotent stem cells. Stem Cell Res 2020; 49:102046. [PMID: 33096385 DOI: 10.1016/j.scr.2020.102046] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 01/28/2023] Open
Abstract
Microglia are resident tissue macrophages of the central nervous system (CNS) that arise from erythromyeloid progenitors during embryonic development. They play essential roles in CNS development, homeostasis and response to disease. Since microglia are difficult to procure from the human brain, several protocols have been developed to generate microglia-like cells from human induced pluripotent stem cells (hiPSCs). However, some concerns remain over the purity and quality of in vitro generated microglia. Here, we describe a new protocol that does not require co-culture with neural cells and yields cultures of 100% P2Y12+ 95% TMEM119+ ramified human microglia-like cells (hiPSC-MG). In the presence of neural precursor cell-conditioned media, hiPSC-MG expressed high levels of human microglia signature genes, including SALL1, CSF1R, P2RY12, TMEM119, TREM2, HEXB and SIGLEC11, as revealed by whole-transcriptome analysis. Stimulation of hiPSC-MG with lipopolysaccharide resulted in downregulation of P2Y12 expression, induction of IL1B mRNA expression and increase in cell capacitance. HiPSC-MG were phagocytically active and maintained their cell identity after transplantation into murine brain slices and human brain spheroids. Together, our new protocol for the generation of microglia-like cells from human iPSCs will facilitate the study of human microglial function in health and disease.
Collapse
Affiliation(s)
- Poulomi Banerjee
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Evdokia Paza
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Emma M Perkins
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Owen G James
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Boyd Kenkhuis
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Amy F Lloyd
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Karen Burr
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - David Story
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Dilmurat Yusuf
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
| | - Xin He
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Owen Dando
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Josef Priller
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin, BIH and DZNE, Berlin, Germany.
| |
Collapse
|
9
|
Zhang K, Baratta MV, Liu G, Frank MG, Leslie NR, Watkins LR, Maier SF, Hutchinson MR, Goldys EM. A novel platform for in vivo detection of cytokine release within discrete brain regions. Brain Behav Immun 2018; 71:18-22. [PMID: 29678795 DOI: 10.1016/j.bbi.2018.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/02/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
Abstract
Mounting evidence indicates that cytokines secreted by innate immune cells in the brain play a central role in regulating neural circuits that subserve mood, cognition, and sickness responses. A major impediment to the study of neuroimmune signaling in healthy and disease states is the absence of tools for in vivo detection of cytokine release in the brain. Here we describe the design and application of a cytokine detection device capable of serial monitoring of local cytokine release in discrete brain regions. The immunocapture device consisted of a modified optical fiber labeled with a capture antibody specific for the pro-inflammatory cytokine interleukin-1 beta (IL-1β). Using a sandwich immunoassay method, in vitro data demonstrate that the sensing interface of the modified optical fiber has a linear detection range of 3.9 pg mL-1-500 pg mL-1 and spatial resolution on the order of 200-450 μm. Finally, we show that the immunocapture device can be introduced into a perforated guide cannula for repeated analyte measurements in vivo. An increase in fluorescence detection of spatially localized intrahippocampal IL-1β release was observed following a peripheral lipopolysaccharide challenge in Sprague-Dawley rats. This novel immunosensing technology represents an opportunity for unlocking the function of neuroimmune signaling.
Collapse
Affiliation(s)
- Kaixin Zhang
- ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Macquarie University, Sydney, Australia.
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
| | - Guozhen Liu
- ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Macquarie University, Sydney, Australia; Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, Australia; International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, China.
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
| | - Nathan R Leslie
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
| | - Linda R Watkins
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
| | - Mark R Hutchinson
- ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), The University of Adelaide, Adelaide, Australia.
| | - Ewa M Goldys
- ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Macquarie University, Sydney, Australia; Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, Australia.
| |
Collapse
|
10
|
Deak T, Kudinova A, Lovelock DF, Gibb BE, Hennessy MB. A multispecies approach for understanding neuroimmune mechanisms of stress. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 28566946 PMCID: PMC5442363 DOI: 10.31887/dcns.2017.19.1/tdeak] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The relationship between stress challenges and adverse health outcomes, particularly for the development of affective disorders, is now well established. The highly conserved neuroimmune mechanisms through which responses to stressors are transcribed into effects on males and females have recently garnered much attention from researchers and clinicians alike. The use of animal models, from mice to guinea pigs to primates, has greatly increased our understanding of these mechanisms on the molecular, cellular, and behavioral levels, and research in humans has identified particular brain regions and connections of interest, as well as associations between stress-induced inflammation and psychiatric disorders. This review brings together findings from multiple species in order to better understand how the mechanisms of the neuroimmune response to stress contribute to stress-related psychopathologies, such as major depressive disorder, schizophrenia, and bipolar disorder.
Collapse
Affiliation(s)
- Terrence Deak
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | - Anastacia Kudinova
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | - Dennis F Lovelock
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | - Brandon E Gibb
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | | |
Collapse
|
11
|
Deak T. A multispecies approach for understanding neuroimmune mechanisms of stress. DIALOGUES IN CLINICAL NEUROSCIENCE 2017; 19:37-53. [PMID: 28566946 PMCID: PMC5442363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2023]
Abstract
The relationship between stress challenges and adverse health outcomes, particularly for the development of affective disorders, is now well established. The highly conserved neuroimmune mechanisms through which responses to stressors are transcribed into effects on males and females have recently garnered much attention from researchers and clinicians alike. The use of animal models, from mice to guinea pigs to primates, has greatly increased our understanding of these mechanisms on the molecular, cellular, and behavioral levels, and research in humans has identified particular brain regions and connections of interest, as well as associations between stress-induced inflammation and psychiatric disorders. This review brings together findings from multiple species in order to better understand how the mechanisms of the neuroimmune response to stress contribute to stress-related psychopathologies, such as major depressive disorder, schizophrenia, and bipolar disorder.
Collapse
Affiliation(s)
- Terrence Deak
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| |
Collapse
|
12
|
Frank MG, Weber MD, Fonken LK, Hershman SA, Watkins LR, Maier SF. The redox state of the alarmin HMGB1 is a pivotal factor in neuroinflammatory and microglial priming: A role for the NLRP3 inflammasome. Brain Behav Immun 2016; 55:215-224. [PMID: 26482581 PMCID: PMC4837097 DOI: 10.1016/j.bbi.2015.10.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/02/2015] [Accepted: 10/15/2015] [Indexed: 01/05/2023] Open
Abstract
The alarmin high mobility group box-1 (HMGB1) has been implicated as a key factor mediating neuroinflammatory processes. Recent findings suggest that the redox state of HMGB1 is a critical molecular feature of HMGB1 such that the reduced form (fr-HMGB1) is chemotactic, while the disulfide form (ds-HMGB1) is pro-inflammatory. The present study examined the neuroinflammatory effects of these molecular forms as well as the ability of these forms to prime the neuroinflammatory and microglial response to an immune challenge. To examine the neuroinflammatory effects of these molecular forms in vivo, animals were administered intra-cisterna magna (ICM) a single dose of fr-HMGB1 (10μg), ds-HMGB1 (10μg) or vehicle and basal pro-inflammatory effects were measured 2 and 24h post-injection in hippocampus. Results of this initial experiment demonstrated that ds-HMGB1 increased hippocampal pro-inflammatory mediators at 2h (NF-κBIα mRNA, NLRP3 mRNA and IL-1β protein) and 24h (NF-κBIα mRNA, TNFα mRNA, and NLRP3 protein) after injection. fr-HMGB1 had no effect on these mediators. These neuroinflammatory effects of ds-HMGB1 suggested that ds-HMGB1 may function to prime the neuroinflammatory response to a subsequent immune challenge. To assess the neuroinflammatory priming effects of these molecular forms, animals were administered ICM a single dose of fr-HMGB1 (10μg), ds-HMGB1 (10μg) or vehicle and 24h after injection, animals were challenged with LPS (10μg/kg IP) or vehicle. Neuroinflammatory mediators and the sickness response (3, 8 and 24h after injection) were measured 2h after immune challenge. We found that ds-HMGB1 potentiated the neuroinflammatory (NF-κBIα mRNA, TNFα mRNA, IL-1β mRNA, IL-6 mRNA, NLRP3 mRNA and IL-1β protein) and sickness response (reduced social exploration) to LPS challenge. fr-HMGB1 failed to potentiate the neuroinflammatory response to LPS. To examine whether these molecular forms of HMGB1 directly induce neuroinflammatory effects in isolated microglia, whole brain microglia were isolated and treated with fr-HMGB1 (0, 1, 10, 100, or 1000ng/ml) or ds-HMGB1 (0, 1, 10, 100, or 1000ng/ml) for 4h and pro-inflammatory mediators measured. To assess the effects of these molecular forms on microglia priming, whole brain microglia were pre-exposed to these forms of HMGB1 (0, 1, 10, 100, or 1000ng/ml) and subsequently challenged with LPS (10ng/ml). We found that ds-HMGB1 increased expression of NF-κBIα mRNA and NLRP3 mRNA in isolated microglia, and potentiated the microglial pro-inflammatory response (TNFα mRNA, IL-1β mRNA and IL-1β protein) to LPS. fr-HMGB1 failed to potentiate the microglial pro-inflammatory response to LPS. Consistent with prior reports, the present findings demonstrate that the disulfide form of HMGB1 not only potentiates the neuroinflammatory response to a subsequent immune challenge in vivo, but also potentiates the sickness response to that challenge. Moreover, the present findings demonstrate for the first time that ds-HMGB1 directly potentiates the microglia pro-inflammatory response to an immune challenge, a finding that parallels the effects of ds-HMGB1 in vivo. In addition, ds-HMGB1 induced expression of NLRP3 and NF-κBIα in vivo and in vitro suggesting that the NLRP3 inflammasome may play role in the priming effects of ds-HMGB1. Taken together, the present results suggest that the redox state of HMGB1 is a critical determinant of the priming properties of HMGB1 such that the disulfide form of HMGB1 induces a primed immunophenotype in the CNS, which may result in an exacerbated neuroinflammatory response upon exposure to a subsequent pro-inflammatory stimulus.
Collapse
Affiliation(s)
- Matthew G. Frank
- Corresponding Author: Department of Psychology and Neuroscience, Center for Neuroscience, Campus Box 345, University of Colorado Boulder Boulder, CO, 80309-0345, USA, Tel: +1-303-919-8116, Fax: +1-303-492-2967,
| | | | | | | | | | | |
Collapse
|
13
|
Verma D, Wood J, Lach G, Mietzsch M, Weger S, Heilbronn R, Herzog H, Bonaventure P, Sperk G, Tasan RO. NPY Y2 receptors in the central amygdala reduce cued but not contextual fear. Neuropharmacology 2015; 99:665-74. [PMID: 26314208 DOI: 10.1016/j.neuropharm.2015.08.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/18/2015] [Accepted: 08/21/2015] [Indexed: 10/23/2022]
Abstract
The amygdala is fundamental for associative fear and extinction learning. Recently, also the central nucleus of the amygdala (CEA) has emerged as a site of plasticity actively controlling efferent connections to downstream effector brain areas. Although synaptic transmission is primarily mediated by glutamate and GABA, neuropeptides critically influence the overall response. While neuropeptide Y (NPY) acting via postsynaptic Y1 receptors exerts an important anxiolytic and fear-reducing action, the role of the predominantly presynaptic Y2 receptors is less defined. To investigate the role of Y2 receptors in the CEA we employed viral-vector mediated over-expression of the Y2 selective agonist NPY3-36 in fear conditioning and extinction experiments. NPY3-36 over-expression in the CEA resulted in reduced fear expression during fear acquisition and recall. Interestingly, this effect was blocked by intraperitoneal injection of a brain-penetrant Y2 receptor antagonist. Furthermore, over-expression of NPY3-36 in the CEA also reduced fear expression during fear extinction of CS-induced but not context-related fear. Again, fear extinction appeared delayed by peripheral injection of a Y2 receptor antagonist JNJ-31020028. Importantly, mice with over-expression of NPY3-36 in the CEA also displayed reduced spontaneous recovery and reinstatement, suggesting that Y2 receptor activation supports a permanent suppression of fear. Local deletion of Y2 receptors in the CEA, on the other hand, increased the expression of CS-induced freezing during fear recall and fear extinction. Thus, NPY inhibits fear learning and promotes cued extinction by reducing fear expression also via activation of presynaptic Y2 receptors on CEA neurons.
Collapse
Affiliation(s)
- D Verma
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - J Wood
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - G Lach
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria; Capes Foundation, Ministry of Education of Brazil, 70040-020 Brasília, DF, Brazil
| | - M Mietzsch
- Institute of Virology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - S Weger
- Institute of Virology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - R Heilbronn
- Institute of Virology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - H Herzog
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - P Bonaventure
- Janssen Research & Development, LLC, San Diego, CA, USA
| | - G Sperk
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - R O Tasan
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
14
|
Intraventricular Delivery of siRNA Nanoparticles to the Central Nervous System. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e242. [PMID: 25965552 DOI: 10.1038/mtna.2015.15] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/08/2015] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease currently lacking effective treatment. Efficient delivery of siRNA via nanoparticles may emerge as a viable therapeutic approach to treat AD and other central nervous system disorders. We report here the use of a linear polyethyleneimine (LPEI)-g-polyethylene glycol (PEG) copolymer-based micellar nanoparticle system to deliver siRNA targeting BACE1 and APP, two therapeutic targets of AD. Using LPEI-siRNA nanoparticles against either BACE1 or APP in cultured mouse neuroblastoma (N2a) cells, we observe selective knockdown, respectively, of BACE1 or APP. The encapsulation of siRNA by LPEI-g-PEG carriers, with different grafting degrees of PEG, leads to the formation of micellar nanoparticles with distinct morphologies, including worm-like, rod-like, or spherical nanoparticles. By infusing these shaped nanoparticles into mouse lateral ventricles, we show that rod-shaped nanoparticles achieved the most efficient knockdown of BACE1 in the brain. Furthermore, such knockdown is evident in spinal cords of these treated mice. Taken together, our findings indicate that the shape of siRNA-encapsulated nanoparticles is an important determinant for their delivery and gene knockdown efficiency in the central nervous system.
Collapse
|
15
|
Stress induces the danger-associated molecular pattern HMGB-1 in the hippocampus of male Sprague Dawley rats: a priming stimulus of microglia and the NLRP3 inflammasome. J Neurosci 2015; 35:316-24. [PMID: 25568124 DOI: 10.1523/jneurosci.3561-14.2015] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Exposure to acute and chronic stressors sensitizes the proinflammatory response of microglia to a subsequent immune challenge. However, the proximal signal by which stressors prime microglia remains unclear. Here, high mobility group box-1 (HMGB-1) protein was explored as a potential mediator of stress-induced microglial priming and whether HMGB-1 does so via the nucleotide-binding domain, leucine-rich repeat, pyrin domain containing protein 3 (NLRP3) inflammasome. Exposure to 100 inescapable tail shocks (ISs) increased HMGB-1 and NLRP3 protein in the hippocampus and led isolated microglia to release HMGB-1 ex vivo. To determine whether HMGB-1 signaling is necessary for stress-induced sensitization of microglia, the HMGB-1 antagonist BoxA was injected into the cisterna magna before IS. Hippocampal microglia were isolated 24 h later and stimulated with LPS ex vivo to probe for stress-induced sensitization of proinflammatory responses. Previous IS potentiated gene expression of NLRP3 and proinflammatory cytokines to LPS, that is, microglia were sensitized. Treatment with BoxA abolished this effect. To determine whether HMGB-1 is sufficient to prime microglia, IS was replaced with intracerebral administration of disulfide or fully reduced HMGB-1. Intracerebral disulfide HMGB-1 mimicked the effect of the stressor, because microglia isolated from HMGB-1-treated rats expressed exaggerated NLRP3 and proinflammatory cytokine expression after LPS treatment, whereas fully reduced HMGB-1 had no effect. The present results suggest that the CNS innate immune system can respond to an acute stressor as if it were cellular damage, thereby releasing the danger signal HMGB-1 in the brain to prime microglia by acting on the NLRP3 inflammasome, in preparation for a later immune challenge.
Collapse
|
16
|
Sobesky JL, Barrientos RM, De May HS, Thompson BM, Weber MD, Watkins LR, Maier SF. High-fat diet consumption disrupts memory and primes elevations in hippocampal IL-1β, an effect that can be prevented with dietary reversal or IL-1 receptor antagonism. Brain Behav Immun 2014; 42:22-32. [PMID: 24998196 PMCID: PMC5652296 DOI: 10.1016/j.bbi.2014.06.017] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/05/2014] [Accepted: 06/25/2014] [Indexed: 12/11/2022] Open
Abstract
High-fat diet (HFD)-induced obesity is reaching worldwide proportions. In addition to causing obesity, HFDs also induce a variety of health disorders, which includes cognitive decline. Hippocampal function may be particularly vulnerable to the negative consequences of HFD, and it is suspected that 'primed' neuroinflammatory processes may mediate this response. To examine the link between diet, hippocampal function and neuroinflammation, male Wistar rats were fed a medium or HFD. Hippocampal memory function was measured using contextual pre-exposure fear conditioning (CPE-FC). Rats fed a HFD demonstrated impaired memory, an effect that was augmented with longer duration of HFD consumption. HFD-induced memory impairments were linked to potentiated levels of interleukin-1 beta (IL-1β) protein in the hippocampus 2h after the foot-shock that occurs during CPE-FC. Central IL-1 receptor antagonism, with intracisterna magna (ICM) administration of hIL-1RA prior to the foot-shock prevented the diet-induced memory disruption, suggesting a critical role for IL-1β in this phenomenon. Additionally, obese animals whose diet regimen was reversed from HFD back to standard chow recovered memory function and did not demonstrate a foot-shock-induced hippocampal IL-1β increase. Interestingly, dietary reversal neutralized the negative impact of HFD on memory and IL-1β, yet animals maintained physiological evidence of obesity (increased body mass and serum leptin), indicating that dietary components, not body mass, may mediate the negative effects on memory.
Collapse
Affiliation(s)
- Julia L. Sobesky
- Corresponding author. Address: Department of Psychology and Neuroscience, Campus Box 345, University of Colorado, Boulder, CO 80309, USA. Tel: +1 616 403 5401. (J.L. Sobesky)
| | | | | | | | | | | | | |
Collapse
|
17
|
Weber MD, Frank MG, Sobesky JL, Watkins LR, Maier SF. Blocking toll-like receptor 2 and 4 signaling during a stressor prevents stress-induced priming of neuroinflammatory responses to a subsequent immune challenge. Brain Behav Immun 2013; 32:112-21. [PMID: 23500798 PMCID: PMC3810175 DOI: 10.1016/j.bbi.2013.03.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 02/26/2013] [Accepted: 03/08/2013] [Indexed: 11/16/2022] Open
Abstract
Acute and chronic stressors sensitize or prime the neuroinflammatory response to a subsequent peripheral or central immunologic challenge. However, the neuroimmune process(es) by which stressors prime or sensitize subsequent neuroinflammatory responses remains unclear. Prior evidence suggested that toll-like receptors (TLRs) might be involved in the mediation of primed neuroinflammatory responses, but the role of TLRs during a stressor has never been directly tested. Here, a novel TLR2 and TLR4 antagonist, OxPAPC, was used to probe the contribution of TLRs in the stress sensitization phenomenon. OxPAPC has not previously been administered to the brain, and so its action in blocking TLR2 and TLR4 action in brain was first verified. Administration of OxPAPC into the CNS prior to stress prevented the stress-induced potentiation of hippocampal pro-inflammatory response to a subsequent peripheral LPS challenge occurring 24 h later. In addition, in vivo administration of OxPAPC prior to stress prevented the sensitized pro-inflammatory response from isolated microglia following administration of LPS ex vivo, further implicating microglia as a key neuroimmune substrate that mediates stress-induced sensitized neuroinflammation.
Collapse
Affiliation(s)
- Michael D. Weber
- Corresponding Author: Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0345, USA. Phone number: 614-937-2613. Fax number: 303-492-2967,
| | | | | | | | | |
Collapse
|
18
|
Blandino P, Hueston CM, Barnum CJ, Bishop C, Deak T. The impact of ventral noradrenergic bundle lesions on increased IL-1 in the PVN and hormonal responses to stress in male sprague dawley rats. Endocrinology 2013; 154:2489-500. [PMID: 23671261 DOI: 10.1210/en.2013-1075] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The impact of acute stress on inflammatory signaling within the central nervous system is of interest because these factors influence neuroendocrine function both directly and indirectly. Exposure to certain stressors increases expression of the proinflammatory cytokine, Il-1β in the hypothalamus. Increased IL-1 is reciprocally regulated by norepinephrine (stimulatory) and corticosterone (inhibitory), yet neural pathways underlying increased IL-1 have not been clarified. These experiments explored the impact of bilateral lesions of the ventral noradrenergic bundle (VNAB) on IL-1 expression in the paraventricular nucleus of the hypothalamus (PVN) after foot shock. Adult male Sprague Dawley rats received bilateral 6-hydroxydopamine lesions of the VNAB (VNABx) and were exposed to intermittent foot shock. VNABx depleted approximately 64% of norepinephrine in the PVN and attenuated the IL-1 response produced by foot shock. However, characterization of the hypothalamic-pituitary-adrenal response, a crucial prerequisite for interpreting the effect of VNABx on IL-1 expression, revealed a profound dissociation between ACTH and corticosterone. Specifically, VNABx blocked the intronic CRH response in the PVN and the increase in plasma ACTH, whereas corticosterone was unaffected at all time points examined. Additionally, foot shock led to a rapid and profound increase in cyclooxygenase-2 and IL-1 expression within the adrenal glands, whereas more subtle effects were observed in the pituitary gland. Together the findings were the 1) demonstration that exposure to acute stress increased expression of inflammatory factors more broadly throughout the hypothalamic-pituitary-adrenal axis; 2) implication of a modest role for norepinephrine-containing fibers of the VNAB as an upstream regulator of PVN IL-1; and 3) suggestion of an ACTH-independent mechanism controlling the release of corticosterone in VNABx rats.
Collapse
Affiliation(s)
- Peter Blandino
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, USA
| | | | | | | | | |
Collapse
|
19
|
Johnson JD, Zimomra ZR, Stewart LT. Beta-adrenergic receptor activation primes microglia cytokine production. J Neuroimmunol 2013; 254:161-4. [DOI: 10.1016/j.jneuroim.2012.08.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 08/15/2012] [Accepted: 08/17/2012] [Indexed: 12/22/2022]
|
20
|
Forcelli PA, Sweeney CT, Kammerich AD, Lee BCW, Rubinson LH, Kayinamura YP, Gale K, Rubinson JF. Histocompatibility and in vivo signal throughput for PEDOT, PEDOP, P3MT, and polycarbazole electrodes. J Biomed Mater Res A 2012; 100:3455-62. [PMID: 22821813 DOI: 10.1002/jbm.a.34285] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 04/02/2012] [Accepted: 05/14/2012] [Indexed: 12/22/2022]
Abstract
Stimulation and recording of the in vivo electrical activity of neurons are critical functions in contemporary biomedical research and in treatment of patients with neurological disorders. The electrodes presently in use tend to exhibit short effective lifespans due to degradation of signal transmission resulting from the tissue response at the electrode-brain interface, with signal throughput suffering most at the low frequencies relevant for biosignals. To overcome these limitations, new electrode designs to minimize tissue responses, including conducting polymers (CPs) have been explored. Here, we report the short-term histocompatibility and signal throughput results comparing platinum and CP-modified platinum electrodes in a Sprague-Dawley rat model. Two of the polymers tested elicited significantly decreased astrocyte responses relative to platinum. These polymers also showed improved signal throughput at low frequencies and comparable signal-to-noise ratios during targeted intracranial electroencephalograms. These results suggest that CP electrodes may present viable alternatives to the metal electrodes that are currently in use.
Collapse
Affiliation(s)
- Patrick A Forcelli
- Department of Pharmacology and Physiology, Georgetown University, Washington, District of Columbia 20057, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Tabrizian K, Najafi S, Belaran M, Hosseini-Sharifabad A, Azami K, Soodi M, Kazemi A, Kebriaeezadeh A, Sharifzadeh M. Effects of Selective iNOS Inhibitor on Spatial Memory in Recovered and Non-Recovered Ketamine Induced Anesthesia. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2011; 10:861-8. [PMID: 24250424 PMCID: PMC3813073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nitric oxide (NO) is thought to be involved in spatial learning and memory in several brain areas such as hippocampus. This study examined the effects of post-training intrahippocampal microinjections of 1400W as a selective iNOS inhibitor on spatial memory, in anesthetized and non-anesthetized situations in rats. In the present work, 4-day training trials of animals were conducted. Spatial memory was tested 48 hours after the drug infusions. For microinjection of 1400W into CA1 region of the hippocampus in conscious animals, guide cannula was implanted into the CA1 area and 1400W was infused after recovery from surgical anesthesia. In anesthetized animals, 1400W was microinjected directly into CA1 region by Hamilton syringe during anesthesia. After completion of training, 1400W (10, 50 and 100 μM/side) were microinjected bilaterally (1 μL/side) and testing trials were performed 48 h after drug infusions in both groups of cannulated and non-cannulated rats. Significant reduction was observed in escape latency and traveled distance in animals that received 1400W (100 μM/side, *p < 0.05) via cannula after recovery in comparison with control group. Also, microinjection of 1400W (100 μM/side) in post recovery phase caused a significant (***p < 0.001) reduction in time and distance of finding the hidden platform in comparison with anesthetized situation. These findings suggest that 1400W has a significant improvement on spatial memory and memory enhancement induced by iNOS inhibitor can be affected by anesthesia in a period of time.
Collapse
Affiliation(s)
| | | | | | | | - Kian Azami
- Department of Pharmacology and Toxicology.
| | - Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Ali Kazemi
- Department of Pharmacology and Toxicology.
| | | | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology.,Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran, Iran,Corresponding author: E-mail:
| |
Collapse
|
22
|
Curia G, Levitt M, Fender JS, Miller JW, Ojemann J, D'Ambrosio R. Impact of injury location and severity on posttraumatic epilepsy in the rat: role of frontal neocortex. Cereb Cortex 2010; 21:1574-92. [PMID: 21112931 DOI: 10.1093/cercor/bhq218] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Human posttraumatic epilepsy (PTE) is highly heterogeneous, ranging from mild remitting to progressive disabling forms. PTE results in simple partial, complex partial, and secondarily generalized seizures with a wide spectrum of durations and semiologies. PTE variability is thought to depend on the heterogeneity of head injury and patient's age, gender, and genetic background. To better understand the role of these factors, we investigated the seizures resulting from calibrated fluid percussion injury (FPI) to adolescent male Sprague-Dawley rats with video electrocorticography. We show that PTE incidence and the frequency and severity of chronic seizures depend on the location and severity of FPI. The frontal neocortex was more prone to epileptogenesis than the parietal and occipital, generating earlier, longer, and more frequent partial seizures. A prominent limbic focus developed in most animals, regardless of parameters of injury. Remarkably, even with carefully controlled injury parameters, including type, severity, and location, the duration of posttraumatic apnea and the age and gender of outbred rats, there was great subject-to-subject variability in frequency, duration, and rate of progression of seizures, indicating that other factors, likely the subjects' genetic background and physiological states, have critical roles in determining the characteristics of PTE.
Collapse
Affiliation(s)
- Giulia Curia
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | | | | | | | | | | |
Collapse
|
23
|
Tabrizian K, Najafi S, Belaran M, Hosseini-Sharifabad A, Azami K, Hosseini A, Soodi M, Kazemi A, Abbas A, Sharifzadeh M. Effects of Selective iNOS Inhibitor on Spatial Memory in Recovered and Non-recovered Ketamine Induced-anesthesia in Wistar Rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2010; 9:313-20. [PMID: 24363743 PMCID: PMC3863448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nitric oxide (NO) is thought to be involved in spatial learning and memory in several brain areas such as hippocampus. This study examined the effects of post-training intrahippocampal microinjections of 1400W as a selective inducible nitric oxide synthase (iNOS) inhibitor on spatial memory, in both anesthetized and non-anesthetized situations in rats. In the present work, 4-day training trials of animals were conducted. Spatial memory was tested 48 h after the drug infusions. For microinjection of 1400W into CA1 region of the hippocampus in conscious animals, guide cannula was implanted into the CA1 area and 1400W was infused after recovery from surgical anesthesia. In anesthetized animals, 1400W was microinjected directly into CA1 region by Hamilton syringe during anesthesia. After completion of training, 1400W (10, 50 and 100 μM/side) were microinjected bilaterally (1 μL/side) and testing trials were performed 48 h after drug infusions in both groups of cannulated and non-cannulated rats. Significant reduction was observed in escape latency and traveled distance in animals that received 1400W (100 μM/side, * P < 0.05) via cannula after recovery in comparison with control group. Moreover, microinjection of 1400W (100 μM/side) in post recovery phase also caused a significant (*** P < 0.001) reduction in time and distance of finding the hidden platform in comparison with anesthetized situation. These results suggest that 1400W has a significant improvement on spatial memory, and memory enhancement induced by iNOS inhibitor can be affected by anesthesia in a period of time.
Collapse
Affiliation(s)
- Kaveh Tabrizian
- Department of Pharmacology and Toxicology,, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran.
| | | | | | | | - Kian Azami
- Department of Pharmacology and Toxicology,
| | | | - Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,
| | - Ali Kazemi
- Department of Pharmacology and Toxicology,
| | | | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology,, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran,,Corresponding author: E-mail:
| |
Collapse
|
24
|
Konsman JP, Veeneman J, Combe C, Poole S, Luheshi GN, Dantzer R. Central nervous action of interleukin-1 mediates activation of limbic structures and behavioural depression in response to peripheral administration of bacterial lipopolysaccharide. Eur J Neurosci 2009; 28:2499-510. [PMID: 19087175 DOI: 10.1111/j.1460-9568.2008.06549.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although receptors for the pro-inflammatory cytokine interleukin-1 have long been known to be expressed in the brain, their role in fever and behavioural depression observed during the acute phase response (APR) to tissue infection remains unclear. This may in part be due to the fact that interleukin-1 in the brain is bioactive only several hours after peripheral administration of bacterial lipopolysaccharide (LPS). To study the role of cerebral interleukin-1 action in temperature and behavioural changes, and activation of brain structures during the APR, interleukin-1 receptor antagonist (IL-1ra; 100 microg) was infused into the lateral brain ventricle 4 h after intraperitoneal (i.p.) LPS injection (250 microg/kg) in rats. I.p. LPS administration induced interleukin-1beta (IL-1beta) production in systemic circulation as well as in brain circumventricular organs and the choroid plexus. Intracerebroventricular (i.c.v.) infusion of IL-1ra 4 h after i.p. LPS injection attenuated the reduction in social interaction, a cardinal sign of behavioural depression during sickness, and c-Fos expression in the amygdala and bed nucleus of the stria terminalis. However, LPS-induced fever, rises in plasma corticosterone, body weight loss and c-Fos expression in the hypothalamus and caudal brainstem were not altered by i.c.v. infusion of IL-1ra. These findings, together with our previous observations showing that i.c.v. infused IL-1ra diffuses throughout perivascular spaces, where macrophages express interleukin-1 receptors, can be interpreted to suggest that circulating or locally produced brain IL-1beta acts on these cells to bring about behavioural depression and activation of limbic structures during the APR after peripheral LPS administration.
Collapse
Affiliation(s)
- J P Konsman
- PsychoNeuroImmunologie, Nutrition et Génétique, CNRS UMR 5526/INRA UMR 1286, Université Victor Ségalen Bordeaux 2, Bordeaux 33076, France.
| | | | | | | | | | | |
Collapse
|
25
|
Hutchinson MR, Lewis SS, Coats BD, Skyba DA, Crysdale NY, Berkelhammer DL, Brzeski A, Northcutt A, Vietz CM, Judd CM, Maier SF, Watkins LR, Johnson KW. Reduction of opioid withdrawal and potentiation of acute opioid analgesia by systemic AV411 (ibudilast). Brain Behav Immun 2009; 23:240-50. [PMID: 18938237 PMCID: PMC2662518 DOI: 10.1016/j.bbi.2008.09.012] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 09/18/2008] [Accepted: 09/28/2008] [Indexed: 12/26/2022] Open
Abstract
Morphine-induced glial proinflammatory responses have been documented to contribute to tolerance to opioid analgesia. Here, we examined whether drugs previously shown to suppress glial proinflammatory responses can alter other clinically relevant opioid effects; namely, withdrawal or acute analgesia. AV411 (ibudilast) and minocycline, drugs with distinct mechanisms of action that result in attenuation of glial proinflammatory responses, each reduced naloxone-precipitated withdrawal. Analysis of brain nuclei associated with opioid withdrawal revealed that morphine altered expression of glial activation markers, cytokines, chemokines, and a neurotrophic factor. AV411 attenuated many of these morphine-induced effects. AV411 also protected against spontaneous withdrawal-induced hyperactivity and weight loss recorded across a 12-day timecourse. Notably, in the spontaneous withdrawal study, AV411 treatment was delayed relative to the start of the morphine regimen so to also test whether AV411 could still be effective in the face of established morphine dependence, which it was. AV411 did not simply attenuate all opioid effects, as co-administering AV411 with morphine or oxycodone caused three-to-five-fold increases in acute analgesic potency, as revealed by leftward shifts in the analgesic dose response curves. Timecourse analyses revealed that plasma morphine levels were not altered by AV411, suggestive that potentiated analgesia was not simply due to prolongation of morphine exposure or increased plasma concentrations. These data support and extend similar potentiation of acute opioid analgesia by minocycline, again providing converging lines of evidence of glial involvement. Hence, suppression of glial proinflammatory responses can significantly reduce opioid withdrawal, while improving analgesia.
Collapse
Affiliation(s)
- Mark R. Hutchinson
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA,Discipline of Pharmacology, School of Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Susannah S. Lewis
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Benjamen D. Coats
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - David A. Skyba
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV, USA
| | - Nicole Y. Crysdale
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Debra L. Berkelhammer
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Anita Brzeski
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Alexis Northcutt
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | | | - Charles M. Judd
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Steven F. Maier
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Linda R. Watkins
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA,Corresponding author: Linda R. Watkins, Department of Psychology, Campus Box 345, University of Colorado at Boulder, Boulder, Colorado, USA 80309-0345, , Fax: 303 492 2967, Ph: 303 492-7034
| | | |
Collapse
|
26
|
Toro-Nieves DM, Rodriguez Y, Plaud M, Ciborowski P, Duan F, Pérez Laspiur J, Wojna V, Meléndez LM. Proteomic analyses of monocyte-derived macrophages infected with human immunodeficiency virus type 1 primary isolates from Hispanic women with and without cognitive impairment. J Neurovirol 2008; 15:36-50. [PMID: 19115125 DOI: 10.1080/13550280802385505] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The signature for human immunodeficiency virus type 1 (HIV-1) neurovirulence remains a subject of intense debate. Macrophage viral tropism is one prerequisite but others, including virus-induced alterations in innate and adaptive immunity, remain under investigation. HIV-1-infected mononuclear phagocytes (MPs; perivascular macrophages and microglia) secrete toxins that affect neurons. The authors hypothesize that neurovirulent HIV-1 variants affect the MP proteome by inducing a signature of neurotoxic proteins and thus affect cognitive function. To test this hypothesis, HIV-1 isolates obtained from peripheral blood of women with normal cognition (NC) were compared to isolates obtained from women with cognitive impairment (CI) and to the laboratory adapted SF162, a spinal fluid R5 isolate from a patient with HIV-1-associated dementia. HIV-1 isolates were used to infect monocyte-derived macrophages (MDMs) and infection monitored by secreted HIV-1 p24 by enzyme-linked immunosorbent assay (ELISA). Cell lysates of uninfected and HIV-1-infected MDMs at 14 days post infection were fractionated by cationic exchange chromatography and analyzed by surface enhanced laser desorption ionization time of flight (SELDI-TOF) using generalized estimating equations statistics. Proteins were separated by one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis (1D SDS-PAGE) and identified by tandem mass spectrometry. Levels of viral replication were similar amongst the HIV-1 isolates, although higher levels were obtained from one viral strain obtained from a patient with CI. Significant differences were found in protein profiles between virus-infected MDMs with NC, CI, and SF162 isolates (adjusted P value after multiple testing corrections, or q value <.10). The authors identified 6 unique proteins in NC, 7 in SF162, and 20 in CI. Three proteins were common to SF162 and CI strains. The MDM proteins linked to infection with CI strains were related to apoptosis, chemotaxis, inflammation, and redox metabolism. These findings support the hypothesis that the macrophage proteome differ when infected with viral isolates of women with and without CI.
Collapse
Affiliation(s)
- D M Toro-Nieves
- Department of Microbiology and Medical Zoology, Specialized Neurosciences Research Program, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ben Menachem-Zidon O, Goshen I, Kreisel T, Ben Menahem Y, Reinhartz E, Ben Hur T, Yirmiya R. Intrahippocampal transplantation of transgenic neural precursor cells overexpressing interleukin-1 receptor antagonist blocks chronic isolation-induced impairment in memory and neurogenesis. Neuropsychopharmacology 2008; 33:2251-62. [PMID: 17987063 DOI: 10.1038/sj.npp.1301606] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The proinflammatory cytokine interleukin-1 (IL-1) within the brain is critically involved in mediating the memory impairment induced by acute inflammatory challenges and psychological stress. However, the role of IL-1 in memory impairment and suppressed neurogenesis induced by chronic stress exposure has not been investigated before now. We report here that mice that were isolated for 4 weeks displayed a significant elevation in hippocampal IL-1beta levels concomitantly with body weight loss, specific impairment in hippocampal-dependent memory, and decreased hippocampal neurogenesis. To examine the causal role of IL-1 in these effects, we developed a novel approach for long-term delivery of IL-1 receptor antagonist (IL-1ra) into the brain, using transplantation of neural precursor cells (NPCs), obtained from neonatal mice with transgenic overexpression of IL-1ra (IL-1raTG) under the glial fibrillary acidic protein promoter. Four weeks following intrahippocampal transplantation of IL-1raTG NPCs labeled with PKH-26, the transplanted cells were incorporated within the dentate gyrus and expressed mainly astrocytic markers. IL-1ra levels were markedly elevated in the hippocampus, but not in other brain regions, by 10 days and for at least 4 weeks post-transplantation. Transplantation of IL-1raTG NPCs completely rescued the chronic isolation-induced body weight loss, memory impairment, and suppressed hippocampal neurogenesis, compared with isolated mice transplanted with WT cells or sham operated. The transplantation had no effect in group-housed mice. These findings elucidate the role of IL-1 in the pathophysiology of chronic isolation and suggest that transplantation of IL-1raTG NPCs may provide a useful therapeutic procedure for IL-1-mediated memory disturbances in chronic inflammatory and neurological conditions.
Collapse
Affiliation(s)
- Ofra Ben Menachem-Zidon
- Department of Psychology, The Hebrew University of Jerusalem, Department of Neurology, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
28
|
Barnum CJ, Blandino P, Deak T. Social status modulates basal IL-1 concentrations in the hypothalamus of pair-housed rats and influences certain features of stress reactivity. Brain Behav Immun 2008; 22:517-27. [PMID: 18037266 DOI: 10.1016/j.bbi.2007.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 09/27/2007] [Accepted: 10/04/2007] [Indexed: 11/17/2022] Open
Abstract
Recent findings from our laboratory and others indicate that exposure to stress can increase expression of the pro-inflammatory cytokine interleukin-1 (IL-1). In a series of studies examining this response, we observed pronounced differences in baseline levels of hypothalamic IL-1 of pair-housed rats. We hypothesized that these pair-wise differences might be a result of prolonged social stress associated with dominance/submissiveness, and that the submissive animal would show heightened baseline levels of IL-1. In order to test this hypothesis, we utilized a food competition paradigm (access to cheerios) to assess dominance within a dyad prior to the assessment of hypothalamic IL-1 levels. Based on the results of this test, clear dominance hierarchies were observed in approximately 50% of the dyads, a ratio comparable to what has been reported previously. More importantly, this dominant/submissive categorization could be used to predict pair-wise differences in hypothalamic IL-1 with greater than 90% accuracy. Specifically, the submissive rat in each dyad (determined a priori) consistently evinced hypothalamic IL-1 levels that were nearly double that of its dominant cage mate. Further studies demonstrated that submissive rats showed a more rapid and pronounced hyperthermic response to novel environment stress relative to dominant rats. Interestingly, social status had no effect on corticosterone reactivity, even when the nature and intensity of the stressor was varied. Finally, maintenance of a clear dominance hierarchy obfuscated hypothalamic IL-1 responses to footshock exposure, with the most robust increases in hypothalamic IL-1 provoked by footshock being observed in pairs where there was no clear dominance hierarchy. Together, these findings suggest that social status can have a significant impact on stress reactivity and neuroimmune consequences of stressor exposure even in the unperturbed home cage environment.
Collapse
Affiliation(s)
- Christopher J Barnum
- Behavioral Neuroscience Program, Department of Psychology, State University of New York at Binghamton, Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | | | | |
Collapse
|
29
|
McCluskey L, Campbell S, Anthony D, Allan SM. Inflammatory responses in the rat brain in response to different methods of intra-cerebral administration. J Neuroimmunol 2008; 194:27-33. [PMID: 18191461 DOI: 10.1016/j.jneuroim.2007.11.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 11/06/2007] [Accepted: 11/07/2007] [Indexed: 12/21/2022]
Abstract
Direct intra-cerebral administration of substances into the brain parenchyma is a common technique used by researchers in neuroscience. However, inflammatory responses to the needle may confound the results obtained following injection of these substances. In this paper we show that the use of a glass micro-needle for intra-cerebral injection reduces mechanical injury, blood-brain barrier breakdown and neutrophil recruitment in response to the injection of vehicle or interleukin-1, compared to using a 26-gauge Hamilton syringe. Therefore, the use of a glass micro-needle to inject substances intra-cerebrally appears to cause minimal injection artefact and should be the method of choice.
Collapse
Affiliation(s)
- Lisa McCluskey
- Faculty of Life Sciences, Michael Smith Building, The University of Manchester, Manchester, M13 9PT, UK
| | | | | | | |
Collapse
|
30
|
Watkins LR, Hutchinson MR, Milligan ED, Maier SF. "Listening" and "talking" to neurons: implications of immune activation for pain control and increasing the efficacy of opioids. BRAIN RESEARCH REVIEWS 2007; 56:148-69. [PMID: 17706291 PMCID: PMC2245863 DOI: 10.1016/j.brainresrev.2007.06.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 06/21/2007] [Accepted: 06/26/2007] [Indexed: 01/08/2023]
Abstract
It is recently become clear that activated immune cells and immune-like glial cells can dramatically alter neuronal function. By increasing neuronal excitability, these non-neuronal cells are now implicated in the creation and maintenance of pathological pain, such as occurs in response to peripheral nerve injury. Such effects are exerted at multiple sites along the pain pathway, including at peripheral nerves, dorsal root ganglia, and spinal cord. In addition, activated glial cells are now recognized as disrupting the pain suppressive effects of opioid drugs and contributing to opioid tolerance and opioid dependence/withdrawal. While this review focuses on regulation of pain and opioid actions, such immune-neuronal interactions are broad in their implications. Such changes in neuronal function would be expected to occur wherever immune-derived substances come in close contact with neurons.
Collapse
Affiliation(s)
- Linda R Watkins
- Department of Psychology and Center for Neuroscience, University of Colorado at Boulder, Boulder, CO 80309-0345, USA.
| | | | | | | |
Collapse
|