351
|
Calabrese M, Magliozzi R, Ciccarelli O, Geurts JJG, Reynolds R, Martin R. Exploring the origins of grey matter damage in multiple sclerosis. Nat Rev Neurosci 2015; 16:147-58. [PMID: 25697158 DOI: 10.1038/nrn3900] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis is characterized at the gross pathological level by the presence of widespread focal demyelinating lesions of the myelin-rich white matter. However, it is becoming clear that grey matter is not spared, even during the earliest phases of the disease. Furthermore, grey matter damage may have an important role both in physical and cognitive disability. Grey matter pathology involves both inflammatory and neurodegenerative mechanisms, but the relationship between the two is unclear. Histological, immunological and neuroimaging studies have provided new insight in this rapidly expanding field, and form the basis of the most recent hypotheses on the pathogenesis of grey matter damage.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Advanced Neuroimaging Laboratory of Neurology B, Department of Neurological and Movement Sciences, University Hospital Verona, Piazzale Ludovico Antonio Scuro 10, 37134, Verona, Italy
| | - Roberta Magliozzi
- 1] Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK. [2] Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
| | - Olga Ciccarelli
- 1] National Institute for Health Research, University College London/University College London Hospitals NHS Foundation Trust (NIHR UCL/UCLH) Biomedical Research Centre, 149 Tottenham Court Road, London W1T 7DN, UK. [2] Queen Square Multiple Sclerosis Centre, University College London, Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jeroen J G Geurts
- Section of Clinical Neuroscience, Department of Anatomy and Neurosciences, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Richard Reynolds
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
352
|
Lin C, Lin HY, Chen JH, Tseng WP, Ko PY, Liu YS, Yeh WL, Lu DY. Effects of paeonol on anti-neuroinflammatory responses in microglial cells. Int J Mol Sci 2015; 16:8844-60. [PMID: 25906473 PMCID: PMC4425112 DOI: 10.3390/ijms16048844] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/07/2015] [Accepted: 04/14/2015] [Indexed: 12/17/2022] Open
Abstract
Increasing studies suggest that inflammatory processes in the central nervous system mediated by microglial activation plays an important role in numerous neurodegenerative diseases. Development of planning for microglial suppression is considered a key strategy in the search for neuroprotection. Paeonol is a major phenolic component of Moutan Cortex, widely used as a nutrient supplement in Chinese medicine. In this study, we investigated the effects of paeonol on microglial cells stimulated by inflammagens. Paeonol significantly inhibited the release of nitric oxide (NO) and the expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Treatment with paeonol also reduced reactive oxygen species (ROS) production and inhibited an ATP-induced increased cell migratory activity. Furthermore, the inhibitory effects of neuroinflammation by paeonol were found to be regulated by phosphorylated adenosine monophosphate-activated protein kinase-α (AMPK-α) and glycogen synthase kinase 3 α/β (GSK 3α/β). Treatment with AMPK or GSK3 inhibitors reverse the inhibitory effect of neuroinflammation by paeonol in microglial cells. Furthermore, paeonol treatment also showed significant improvement in the rotarod performance and microglial activation in the mouse model as well. The present study is the first to report a novel inhibitory role of paeonol on neuroinflammation, and presents a new candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Hsiao-Yun Lin
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 40402, Taiwan.
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan.
| | - Wen-Pei Tseng
- Graduate Institute of Sports and Health, National Changhua University of Education, Changhua 500, Taiwan.
| | - Pei-Ying Ko
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Yu-Shu Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua 500, Taiwan.
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 40402, Taiwan.
- Department of Photonics and Communication Engineering, Asia University, Taichung 40402, Taiwan.
| |
Collapse
|
353
|
Abstract
Microglia are the resident immune cells in the CNS and play diverse roles in the maintenance of CNS homeostasis. Recent studies have shown that microglia continually survey the CNS microenvironment and scavenge cell debris and aberrant proteins by phagocytosis and pinocytosis, and that reactive microglia are capable to present antigens to T cells and initiate immune responses. However, how microglia process the endocytosed contents and evoke an immune response remain unclear. Here we report that a size-dependent selective transport of small soluble contents from the pinosomal lumen into lysosomes is critical for the antigen processing in microglia. Using fluorescent probes and water-soluble magnetic nanobeads of defined sizes, we showed in cultured rodent microglia, and in a cell-free reconstructed system that pinocytosed proteins become degraded immediately following pinocytosis and the resulting peptides are selectively delivered to major histocompatibility complex class II (MHC-II) containing lysosomes, whereas undegraded proteins are retained in the pinosomal lumen. This early size-based sorting of pinosomal contents relied on the formation of transient tunnel between pinosomes and lysosomes in a Rab7- and dynamin II-dependent manner, which allowed the small contents to pass through but restricted large ones. Inhibition of the size-based sorting markedly reduced proliferation and cytokine release of cocultured CD4(+) T cells, indicating that the size-based sorting is required for efficient antigen presentation by microglial cells. Together, these findings reveal a novel early sorting mechanism for pinosomal luminal contents in microglial cells, which may explain how microglia efficiently process protein antigens and evoke an immune response.
Collapse
|
354
|
Chang CZ, Wu SC, Lin CL, Kwan AL. Valproic acid attenuates intercellular adhesion molecule-1 and E-selectin through a chemokine ligand 5 dependent mechanism and subarachnoid hemorrhage induced vasospasm in a rat model. J Inflamm (Lond) 2015; 12:27. [PMID: 25908928 PMCID: PMC4407545 DOI: 10.1186/s12950-015-0074-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Up-regulation of regulated upon activation, normal T-cell expressed and secreted (RANTES/CCL5) and adhesion molecules is observed in the serum of animals following experimental subarachnoid hemorrhage (SAH). The present study was to examine the effect of valproic acid (VPA) on RANTES and alternation of adhesion molecules in this model. METHODS A rodent SAH model was employed. Animals were randomly assigned into six groups. Basilar artery (BA) was harvested for intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin evaluation (western blotting) and RANTES (rt-PCR). 1 ng CCL5 recombinant protein intrathecal injection was performed in the VPA + SAH groups. (N = 5). RESULTS Convoluted internal elastic lamina, distorted endothelial wall, and smooth muscle micro-necrosis was prominently observed in the SAH groups, which is absent in the VPA treatment and the healthy controls. Treatment with VPA dose-dependently reduced the ICAM-1, E-selectin and RANTES level, compared with the SAH group (p <0.01). The administration of CCL5 significantly increased CD45(+) glia and ICAM-1 level in the VPA treatment groups. CONCLUSION VPA exerts its anti-vasospastic effect through the dual effect of inhibiting RANTES expression and reduced adhesion molecules. Besides, VPA also decreased CD45(+) cells transmigrated to the vascular wall. The administration of CCL5 significantly reversed the inhibitory effect of this compound on CD45(+) monocytes, E-selectin, and ICAM-1 level. This study also lends credence to support this compound could attenuate SAH induced adhesion molecules and neuro-inflammation in a CCL5 dependent mechanism.
Collapse
Affiliation(s)
- Chih-Zen Chang
- />Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- />Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, No.100, Tzyou 1st Road, Kaohsiung, Taiwan
- />Department of Surgery, Kaohsiung Municipal Ta Tung Hospital, Kaohsiung, Taiwan
| | - Shu-Chuan Wu
- />Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, No.100, Tzyou 1st Road, Kaohsiung, Taiwan
| | - Chih-Lung Lin
- />Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- />Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, No.100, Tzyou 1st Road, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- />Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- />Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, No.100, Tzyou 1st Road, Kaohsiung, Taiwan
| |
Collapse
|
355
|
Dehkordi NG, Noorbakhshnia M, Ghaedi K, Esmaeili A, Dabaghi M. Omega-3 fatty acids prevent LPS-induced passive avoidance learning and memory and CaMKII-α gene expression impairments in hippocampus of rat. Pharmacol Rep 2015; 67:370-5. [DOI: 10.1016/j.pharep.2014.10.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 10/24/2022]
|
356
|
Mallucci G, Peruzzotti-Jametti L, Bernstock JD, Pluchino S. The role of immune cells, glia and neurons in white and gray matter pathology in multiple sclerosis. Prog Neurobiol 2015; 127-128:1-22. [PMID: 25802011 PMCID: PMC4578232 DOI: 10.1016/j.pneurobio.2015.02.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/24/2014] [Accepted: 02/27/2015] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis is one of the most common causes of chronic neurological disability beginning in early to middle adult life. Multiple sclerosis is idiopathic in nature, yet increasing correlative evidence supports a strong association between one's genetic predisposition, the environment and the immune system. Symptoms of multiple sclerosis have primarily been shown to result from a disruption in the integrity of myelinated tracts within the white matter of the central nervous system. However, recent research has also highlighted the hitherto underappreciated involvement of gray matter in multiple sclerosis disease pathophysiology, which may be especially relevant when considering the accumulation of irreversible damage and progressive disability. This review aims at providing a comprehensive overview of the interplay between inflammation, glial/neuronal damage and regeneration throughout the course of multiple sclerosis via the analysis of both white and gray matter lesional pathology. Further, we describe the common pathological mechanisms underlying both relapsing and progressive forms of multiple sclerosis, and analyze how current (as well as future) treatments may interact and/or interfere with its pathology. Understanding the putative mechanisms that drive disease pathogenesis will be key in helping to develop effective therapeutic strategies to prevent, mitigate, and treat the diverse morbidities associated with multiple sclerosis.
Collapse
Affiliation(s)
- Giulia Mallucci
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
- Department of Brain and Behavioural Sciences, National Neurological Institute C. Mondino, University of Pavia, 27100 Pavia, Italy
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
| | - Joshua D. Bernstock
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bldg10/Rm5B06, MSC 1401, 10 Center Drive, Bethesda, MD 20892, USA
| | - Stefano Pluchino
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, Wellcome Trust-MRC Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY, UK
| |
Collapse
|
357
|
Sil S, Ghosh R, Sanyal M, Guha D, Ghosh T. A comparison of neurodegeneration linked with neuroinflammation in different brain areas of rats after intracerebroventricular colchicine injection. J Immunotoxicol 2015; 13:181-90. [PMID: 25812625 DOI: 10.3109/1547691x.2015.1030804] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Colchicine induces neurodegeneration, but the extent of neurodegeneration in different areas of the brain in relation to neuroinflammation remains unclear. Such information may be useful to allow for the development of a model to compare colchicine-induced neurodegeneration with other neurodegenerative diseases such as Alzheimer's Disease (AD). The present study was designed to investigate the extent of neurodegeneration along with neuroinflammation in different areas of the brain, e.g. frontal cortex, parietal cortex, occipital cortex, corpus striatum, amygdala and hippocampus, in rats along with memory impairment 21 days after a single intracerebroventricular (icv) injection of colchicine. Memory parameters were measured before and after icv colchicine injection in all test groups of rats (control, sham-operated, colchicine-injected [ICIR] rats). On Day 21 post-injection, rats from all groups were anesthesized and tissues from the various brain areas were collected for assessment of biomarkers of neuroinflammation (i.e. levels of ROS, nitrite and proinflammatory cytokines TNFα and IL-1β) and neurodegeneration (assessed histologically). The single injection of colchicine resulted in impaired memory and neurodegeneration (significant presence of plaques, Nissl granule chromatolysis) in various brain areas (frontal cortex, amygdala, parietal cortex, corpus striatum), with maximum severity in the hippocampus. While IL-1β, TNFα, ROS and nitrite levels were altered in different brain areas in the ICIR rats, these parameters had their greatest change in the hippocampus. This study showed that icv injection of colchicine caused strong neurodegeneration and neuroinflammation in the hippocampus of rats and the increases in neurodegeneration were corroborated with those of neuroinflammation at the site. The present study also showed that the extent of neurodegeneration and neuroinflammation in different brain areas of the colchicine-injected rats were AD-like and supported the fact that such rats might have the ability to serve as a sporadic model of AD.
Collapse
Affiliation(s)
- Susmita Sil
- a Department of Physiology , University College of Science and Technology and
| | - Rupsa Ghosh
- a Department of Physiology , University College of Science and Technology and
| | - Moumita Sanyal
- a Department of Physiology , University College of Science and Technology and
| | - Debjani Guha
- b S. N. Pradhan Centre for Neurosciences, University of Calcutta , West Bengal , India
| | - Tusharkanti Ghosh
- a Department of Physiology , University College of Science and Technology and
| |
Collapse
|
358
|
Liu H, Wei X, Kong L, Liu X, Cheng L, Yan S, Zhang X, Chen L. NOD2 is involved in the inflammatory response after cerebral ischemia-reperfusion injury and triggers NADPH oxidase 2-derived reactive oxygen species. Int J Biol Sci 2015; 11:525-35. [PMID: 25892960 PMCID: PMC4400384 DOI: 10.7150/ijbs.10927] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/23/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Increasing evidences suggest that innate immunity is involved in cerebral ischemia-reperfusion (I/R) injury, but the liable innate immune receptors have not been completely elucidated. Here, we explored the role of the nucleotide-binding oligomerization domain (NOD)2, a member of the cytosolic NOD-like receptor family, in acute focal cerebral I/R injury. METHODS An in vivo middle cerebral artery occlusion (MCAO) model that in wild type (WT) and NOD2 deficient (NOD2(-/-)) mice and in vitro model of oxygen glucose deprivation and reoxygenation (OGD/R) in cultured primary microglia and astrocytes were used to investigate the expression of NOD2 and explore the roles of NOD2 in ischemic stroke. RESULTS Our results showed that NOD2 expression was significantly increased in microglia and astrocytes in response to the I/R insult. Pretreatment with muramyl dipeptide, an extrinsic ligand of NOD2, significantly increased the infarct volume and neurological dysfunction in mice subjected to MCAO. Genetic ablation of the NOD2 gene significantly improved stroke outcomes and reduced inflammation, as evidenced by a lower expression of the pro-inflammatory cytokines IL-1β, IL-6 and TNFα in conjunction with attenuated activation of nuclear factor κB (NF-κB), p38 mitogen activated protein kinases (MAPK) and JNK. Moreover, NOD2 deficiency prevented the upregulation of the NADPH oxidase (NOX) 2 and ROS generation induced by I/R. Mechanistically, NOD2-induced production of IL-6 in primary cultured microglia was mediated through activation of NOX2. CONCLUSIONS This study showed the contribution of NOD2 to inflammatory response and provided direct evidence that NOX2-mediated oxidative stress as an important target molecule linked NOD2 to inflammatory damage in ischemic stroke. Pharmacological targeting of NOD2-mediated inflammatory response at multiple levels may help design a new approach to develop therapeutic strategies for prevention of deterioration of cerebral function and for the treatment of stroke.
Collapse
Affiliation(s)
- Huiqing Liu
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Xinbing Wei
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Lingjun Kong
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Xiaoqian Liu
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China. ; 2. Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Qingquan Road 30, Yantai, Shandong 264005, P.R. China
| | - Li Cheng
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Shi Yan
- 3. Department of Obstetrics and Gynaecology, Qilu Hospital of Shandong University, Wenhua West Road 107, Jinan, Shandong 250012, P.R. China
| | - Xiumei Zhang
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Lin Chen
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
359
|
Doorn KJ, Brevé JJP, Drukarch B, Boddeke HW, Huitinga I, Lucassen PJ, van Dam AM. Brain region-specific gene expression profiles in freshly isolated rat microglia. Front Cell Neurosci 2015; 9:84. [PMID: 25814934 PMCID: PMC4357261 DOI: 10.3389/fncel.2015.00084] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/23/2015] [Indexed: 12/30/2022] Open
Abstract
Microglia are important cells in the brain that can acquire different morphological and functional phenotypes dependent on the local situation they encounter. Knowledge on the region-specific gene signature of microglia may hold valuable clues for microglial functioning in health and disease, e.g., Parkinson's disease (PD) in which microglial phenotypes differ between affected brain regions. Therefore, we here investigated whether regional differences exist in gene expression profiles of microglia that are isolated from healthy rat brain regions relevant for PD. We used an optimized isolation protocol based on a rapid isolation of microglia from discrete rat gray matter regions using density gradients and fluorescent-activated cell sorting. Application of the present protocol followed by gene expression analysis enabled us to identify subtle differences in region-specific microglial expression profiles and show that the genetic profile of microglia already differs between different brain regions when studied under control conditions. As such, these novel findings imply that brain region-specific microglial gene expression profiles exist that may contribute to the region-specific differences in microglia responsivity during disease conditions, such as seen in, e.g., PD.
Collapse
Affiliation(s)
- Karlijn J Doorn
- Department Structural and Functional Plasticity of the Nervous System, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands ; Neuroscience Campus Amsterdam, Department Anatomy and Neurosciences, VU University Medical Center Amsterdam, Netherlands
| | - John J P Brevé
- Neuroscience Campus Amsterdam, Department Anatomy and Neurosciences, VU University Medical Center Amsterdam, Netherlands
| | - Benjamin Drukarch
- Neuroscience Campus Amsterdam, Department Anatomy and Neurosciences, VU University Medical Center Amsterdam, Netherlands
| | - Hendrikus W Boddeke
- Section Medical Physiology, Department of Neuroscience, University Medical Centre Groningen Groningen, Netherlands
| | - Inge Huitinga
- Neuroimmunology Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences Amsterdam, Netherlands
| | - Paul J Lucassen
- Department Structural and Functional Plasticity of the Nervous System, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Anne-Marie van Dam
- Department Structural and Functional Plasticity of the Nervous System, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
360
|
Le OTT, Nguyen TTN, Lee SY. Phosphoinositide turnover in Toll-like receptor signaling and trafficking. BMB Rep 2015; 47:361-8. [PMID: 24856829 PMCID: PMC4163850 DOI: 10.5483/bmbrep.2014.47.7.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Indexed: 12/29/2022] Open
Abstract
Lipid components in biological membranes are essential for maintaining cellular function. Phosphoinositides, the phosphorylated derivatives of phosphatidylinositol (PI), regulate many critical cell processes involving membrane signaling, trafficking, and reorganization. Multiple metabolic pathways including phosphoinositide kinases and phosphatases and phospholipases tightly control spatio-temporal concentration of membrane phosphoinositides. Metabolizing enzymes responsible for PI 4,5-bisphosphate (PI(4,5)P2) production or degradation play a regulatory role in Toll-like receptor (TLR) signaling and trafficking. These enzymes include PI 4-phosphate 5-kinase, phosphatase and tensin homolog, PI 3-kinase, and phospholipase C. PI(4,5)P2 mediates the interaction with target cytosolic proteins to induce their membrane translocation, regulate vesicular trafficking, and serve as a precursor for other signaling lipids. TLR activation is important for the innate immune response and is implicated in diverse pathophysiological disorders. TLR signaling is controlled by specific interactions with distinct signaling and sorting adaptors. Importantly, TLR signaling machinery is differentially formed depending on a specific membrane compartment during signaling cascades. Although detailed mechanisms remain to be fully clarified, phosphoinositide metabolism is promising for a better understanding of such spatio-temporal regulation of TLR signaling and trafficking. [BMB Reports 2014; 47(7): 361-368]
Collapse
Affiliation(s)
- Oanh Thi Tu Le
- Neuroscience Graduate Program, Department of Biomedical Sciences, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi 443-721, Korea
| | - Tu Thi Ngoc Nguyen
- Neuroscience Graduate Program, Department of Biomedical Sciences, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi 443-721, Korea
| | - Sang Yoon Lee
- Neuroscience Graduate Program, Department of Biomedical Sciences, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi 443-721, Korea
| |
Collapse
|
361
|
Reduction of inflammatory responses by L-serine treatment leads to neuroprotection in mice after traumatic brain injury. Neuropharmacology 2015; 95:1-11. [PMID: 25747604 DOI: 10.1016/j.neuropharm.2015.02.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/15/2015] [Accepted: 02/23/2015] [Indexed: 11/22/2022]
Abstract
This study was designed to evaluate the neuroprotective effect of l-serine and the underlying mechanisms in mice after traumatic brain injury (TBI) induced using a weight drop model. The mice were intraperitoneally injected with l-serine 3 h after TBI and then injected twice each day for 7 days or until the end of the experiment. The neurological severity score, brain water content, lesion volume, and neurone loss were determined. The levels of TNF-α, IL-1β, IL-6, and IL-10 and the number of GFAP- and Iba-1-positive cells and activated caspase-3-positive neurones in the brain tissue ipsilateral to TBI were also measured. Simultaneously, the influences of l-serine on these variables were observed. In addition, the expression of glycine receptors and l-serine-induced currents were measured. We found l-serine treatment: 1) decreased the neurological deficit score, brain water content, lesion volume, and neurone loss; 2) inhibited activated caspase-3; and 3) reduced the levels of TNF-α, IL-1β and IL-6 and the number of GFAP- and Iba-1-positive cells. The effects of l-serine were antagonised by the administration of strychnine, an antagonist of glycine receptors. In addition, we found that glycine receptors were expressed mainly in the cortical neurones but less in the astrocytes or microglial cells, and l-serine activated these receptors and induced strychnine-sensitive currents in these neurones. In conclusion, l-serine induces the activation of glycine receptors, which alleviates neuronal excitotoxicity, a secondary brain injury process, thereby reduces the activation of astrocytes and microglial cells and secretion of proinflammatory cytokines and inhibits neuronal apoptosis. Thus, l-serine treatment leads to neuroprotection of brain tissue through reducing inflammatory responses and improves recovery of the neurological functions in mice after traumatic brain injury.
Collapse
|
362
|
Xiao Z, Ren P, Chao Y, Wang Q, Kuai J, Lv M, Chen L, Gao C, Sun X. Protective role of isoflurane pretreatment in rats with focal cerebral ischemia and the underlying molecular mechanism. Mol Med Rep 2015; 12:675-83. [PMID: 25738964 DOI: 10.3892/mmr.2015.3408] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 11/03/2014] [Indexed: 11/06/2022] Open
Abstract
Inflammation and immunity are important in the pathogenesis of cerebral ischemia. Toll-like receptor 4 (TLR4) is involved in the inflammatory responses of injured brain tissues. Emerging studies have focused on the effect of isoflurane (ISO) pretreatment on cerebral ischemia, however, the association between ISO pretreatment and TLR4 during cerebral ischemia remains to be elucidated. In the present study, the protective role of ISO pretreatment in rats with focal cerebral ischemia reperfusion was investigated and the molecular mechanism was discussed. Using a middle cerebral artery occlusion (MCAO) model, triphenyltetrazolium chloride staining was utilized to measure the infarct volume and brain edema and immunofluorescence staining was used to detect the MCAO-induced TLR4 expression and localization. Western blot analyses were conducted to quantify the protein expression levels of TLR4, myeloid differentiation primary response 88 (MyD88) and nuclear factor (NF)-κB in ischemic brain tissue at different time points. The results demonstrated that, following ISO pretreatment, the neurological deficits, brain edema and cerebral infarct size caused by ischemia/reperfusion were attenuated. The astrocyte and microglial activation in the brain tissue was decreased. In addition, the expression levels of TLR4, MyD88 and NF-κB were decreased. The present study indicated that ISO pretreatment may protect the brain from ischemic damage by downregulating the expression levels of TLR4, MyD88 and NF-κB.
Collapse
Affiliation(s)
- Zhibin Xiao
- Department of Anesthesiology, Tangdou Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Pengcheng Ren
- Department of Anesthesiology, Tangdou Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yang Chao
- Department of Stomatology, The 323 Hospital of People's Liberation Army, Xi'an, Shaanxi 710054, P.R. China
| | - Qianyun Wang
- Department of Medicine, The 323 Hospital of People's Liberation Army, Xi'an, Shaanxi 710054, P.R. China
| | - Jianke Kuai
- Department of Anesthesiology, Tangdou Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Miaomiao Lv
- Department of Anesthesiology, The 323 Hospital of People's Liberation Army, Xi'an, Shaanxi 710054, P.R. China
| | - Lei Chen
- Department of Obstetrics and Gynecology, The Navy General Hospital of People's Liberation Army, Beijing 100059, P.R. China
| | - Changjun Gao
- Department of Anesthesiology, Tangdou Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xude Sun
- Department of Anesthesiology, Tangdou Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
363
|
Su F, Yi H, Xu L, Zhang Z. Fluoxetine and S-citalopram inhibit M1 activation and promote M2 activation of microglia in vitro. Neuroscience 2015; 294:60-8. [PMID: 25711936 DOI: 10.1016/j.neuroscience.2015.02.028] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 12/27/2022]
Abstract
Increasing evidence has suggested that microglia dysfunction plays an important role in the pathogenesis of depression. Both classical activation (M1 activation) and alternative activation (M2 activation) may be involved in the process. M1-activated microglia secrete various pro-inflammatory cytokines and neurotoxic mediators, which may contribute to the development of depression, while M2-activated microglia promote tissue reconstruction by releasing anti-inflammatory cytokines involved in the process of depression. Selective serotonin reuptake inhibitors (SSRIs) are first-line treatments for depression, and their effects on immune system modulation have recently gained attention. Several studies have suggested that SSRIs affect the M1 activation of microglia, but results have varied. In addition, little is known about the effect of SSRIs on M2 activation in depression. The aim of this study was to investigate the effects of fluoxetine and S-citalopram, two widely used SSRIs in clinical, on both the M1 and M2 activation of microglia (the murine BV2 cell line and mouse primary microglia cell). The indexes of activation were measured by real-time polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA) and Western blot. The present results showed that both fluoxetine and S-citalopram significantly down-regulated the indexes of M1 activation and up-regulated the M2 activation indexes on mRNA and protein levels either in cell line or primary cells. Taken together, the results suggested that fluoxetine and S-citalopram modulated the immune system by inhibiting M1 activation and by improving M2 activation of microglia and that the immune system modulation may partially mediate the therapeutic effects of antidepressant drugs-SSRIs.
Collapse
Affiliation(s)
- F Su
- The Department of Neurology of Affiliated ZhongDa Hospital, The Institute of Neuropsychiatry and Medical School of Southeast University, Nanjing, China
| | - H Yi
- The Department of Neurology of Affiliated ZhongDa Hospital, The Institute of Neuropsychiatry and Medical School of Southeast University, Nanjing, China
| | - L Xu
- Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Z Zhang
- The Department of Neurology of Affiliated ZhongDa Hospital, The Institute of Neuropsychiatry and Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
364
|
Cianciulli A, Dragone T, Calvello R, Porro C, Trotta T, Lofrumento DD, Panaro MA. IL-10 plays a pivotal role in anti-inflammatory effects of resveratrol in activated microglia cells. Int Immunopharmacol 2015; 24:369-376. [DOI: 10.1016/j.intimp.2014.12.035] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/19/2014] [Accepted: 12/28/2014] [Indexed: 01/31/2023]
|
365
|
Ooi YY, Dheen ST, Tay SSW. Paracrine effects of mesenchymal stem cells-conditioned medium on microglial cytokines expression and nitric oxide production. Neuroimmunomodulation 2015; 22:233-42. [PMID: 25341618 DOI: 10.1159/000365483] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/19/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIM Microglia, the resident macrophages in the central nervous system, secrete various proinflammatory cytokines and undergo proliferation upon activation in various neurodegenerative diseases. Activation of microglia has been implicated in exacerbation of various neurodegenerative diseases. Recently, it has been proposed that mesenchymal stem cells (MSC) have immunosuppressive properties and the potential to moderate inflammation. This study aimed to elucidate the effects of MSC-conditioned medium (MSC-CM) in modulating microglial activation by analyzing microglial proinflammatory and anti-inflammatory factors [interleukin (IL)-6, tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS) and IL-10], signaling pathway molecules [NFκB, c-Jun N-terminal kinase (JNK) and MKP-1) and NO production. METHODS Immortalized murine microglia cell line, BV2 microglia and primary microglia isolated from C57BL/6 mouse pup brains were used in this study. Mouse MSC were isolated from the male C57BL/6 mouse tibia and fibula. The effects of MSC-CM on the expression of inflammatory cytokines and signaling molecules in microglia were elucidated using RT-PCR, immunofluorescence analysis and Western blot analysis. NO production in microglia was assessed using a Griess kit. RESULTS MSC-CM significantly reduced the mRNA and protein expression levels of proinflammatory cytokines (IL-6 and TNF-α) in microglia activated by lipopolysaccharide (LPS). In addition, MSC-CM significantly reduced the protein expression of NFκB, JNK and c-Jun, but increased the expression levels of IL-10 and MKP-1 in activated BV2 microglia. NO production and iNOS expression by BV2 microglia in MSC-CM were increased. CONCLUSIONS Overall, our findings suggest that MSC immunomodulate microglial activities through paracrine effects.
Collapse
Affiliation(s)
- Yin Yin Ooi
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
366
|
Argente-Arizón P, Freire-Regatillo A, Argente J, Chowen JA. Role of non-neuronal cells in body weight and appetite control. Front Endocrinol (Lausanne) 2015; 6:42. [PMID: 25859240 PMCID: PMC4374626 DOI: 10.3389/fendo.2015.00042] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022] Open
Abstract
The brain is composed of neurons and non-neuronal cells, with the latter encompassing glial, ependymal and endothelial cells, as well as pericytes and progenitor cells. Studies aimed at understanding how the brain operates have traditionally focused on neurons, but the importance of non-neuronal cells has become increasingly evident. Once relegated to supporting roles, it is now indubitable that these diverse cell types are fundamental for brain development and function, including that of metabolic circuits, and they may play a significant role in obesity onset and complications. They participate in processes of neurogenesis, synaptogenesis, and synaptic plasticity of metabolic circuits both during development and in adulthood. Some glial cells, such as tanycytes and astrocytes, transport circulating nutrients and metabolic factors that are fundamental for neuronal viability and activity into and within the hypothalamus. All of these cell types express receptors for a variety of metabolic factors and hormones, suggesting that they participate in metabolic function. They are the first line of defense against any assault to neurons. Indeed, microglia and astrocytes participate in the hypothalamic inflammatory response to high fat diet (HFD)-induced obesity, with this process contributing to inflammatory-related insulin and leptin resistance. Moreover, HFD-induced obesity and hyperleptinemia modify hypothalamic astroglial morphology, which is associated with changes in the synaptic inputs to neuronal metabolic circuits. Astrocytic contact with the microvasculature is increased by HFD intake and this could modify nutrient/hormonal uptake into the brain. In addition, progenitor cells in the hypothalamus are now known to have the capacity to renew metabolic circuits, and this can be affected by HFD intake and obesity. Here, we discuss our current understanding of how non-neuronal cells participate in physiological and physiopathological metabolic control.
Collapse
Affiliation(s)
- Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Julie A. Chowen, Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Avda. Menéndez Pelayo, 65, Madrid E-28009, Spain e-mail: ;
| |
Collapse
|
367
|
Winklewski PJ, Radkowski M, Demkow U. Cross-talk between the inflammatory response, sympathetic activation and pulmonary infection in the ischemic stroke. J Neuroinflammation 2014; 11:213. [PMID: 25539803 PMCID: PMC4297381 DOI: 10.1186/s12974-014-0213-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/02/2014] [Indexed: 01/29/2023] Open
Abstract
The immune system response and inflammation play a key role in brain injury during and after a stroke. The acute immune response is responsible for secondary brain tissue damage immediately after the stroke, followed by immunosuppression due to sympathetic nervous system activation. The latter increases risk of infection complications, such as pneumonia. The pneumonia-related inflammatory state can release a bystander autoimmune response against central nervous system antigens, thereby initiating a vicious circle. The aim of this review is to summarize the relationship between ischemic stroke, sympathetic nervous system activation and pulmonary infection.
Collapse
Affiliation(s)
- Pawel J Winklewski
- Institute of Human Physiology, Medical University of Gdansk, Tuwima Street 15, 80-210, Gdansk, Poland.
| | - Marek Radkowski
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, Warsaw, Poland.
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
368
|
Modulation properties of factors released by bone marrow stromal cells on activated microglia: an in vitro study. Sci Rep 2014; 4:7514. [PMID: 25524416 PMCID: PMC5378994 DOI: 10.1038/srep07514] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/24/2014] [Indexed: 12/14/2022] Open
Abstract
In the present paper we develop a new non-cell based (cell-free) therapeutic approach applied to BV2 microglial cells and spinal cord derived primary microglia (PM) using conditioned media from rat bone marrow stromal cells (BMSCs-CM). First we collected conditioned media (CM) from either naive or injured rat spinal cord tissue (SCI-CM, inflammatory stimulation agent) and from rat bone marrow stromal cells (BMSCs-CM, therapeutic immunomodulation agent). They were both subsequently checked for the presence of chemokines and growth, neurotrophic and neural migration factors using proteomics analysis. The data clearly showed that rat BMSCs-CM contain in vitro growth factors, neural migration factors, osteogenic factors, differentiating factors and immunomodulators, whereas SCI-CM contain chemokines, chemoattractant factors and neurotrophic factors. Afterwards we determined whether the BMSCs-CM affect chemotactic activity, NO production, morphological and pro-apoptotic changes of either BV2 or PM cells once activated with SCI-CM. Our results confirm the anti-migratory and NO-inhibitory effects of BMSCs-CM on SCI-CM-activated microglia with higher impact on primary microglia. The cytotoxic effect of BMSCs-CM occurred only on SCI-CM-stimulated BV2 cells and PM, not on naive BV2 cells, nor on PM. Taken together, the molecular cocktail found in BMSCs-CM is favorable for immunomodulatory properties.
Collapse
|
369
|
Brain APCs including microglia are only differential and positional polymorphs. Ann Neurosci 2014; 17:191-9. [PMID: 25205905 PMCID: PMC4117011 DOI: 10.5214/ans.0972.7531.1017410] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 09/22/2010] [Accepted: 10/04/2010] [Indexed: 12/23/2022] Open
Abstract
The antigen presentation to lymphocytes in brain occurs in two steps. Initially it happens at perivascular spaces by perivascular microglia/macrophage population and finally at the site of inflammation deep into brain parenchyma by the resident microglia. But recent evidence challanges the existing notion of involvement of distinct and different cells at these sites. Studies have shown that many of these microglial cells show dendritic cell phenotype in pathogenic and cytokine driven environment. Different subsets of the cell show wide range of myeloid lineage functions indicating a pre-differentiated status of the cell. Monocytic CD34(+)/B220(+) precursor cells have been transformed to microglial cells in vitro and transplantation of these cells show Iba-1 or F4/80 positivity with microglial phenotypes in vivo in adults. Even they can be converted into dendritic cell like forms. The interconvertability among macrophage-microglia-dendritic cells and final effector maturation according to the microenvironmental cues indicates existence of a pre-mature myeloid cell population concerned with antigen presentation and related functions in brain. With the substantial recent observation this article sketches the idea that brain APCs appearing as macrophage/microglia/DC like forms are derivatives of the same stock in response to their position and microenvironment. And also microglia is never any distinct cells, both in neonatal stage and adults.
Collapse
|
370
|
Costa RC, Orlando DR, Abreu CC, Nakagaki KYR, Mesquita LP, Nascimento LC, Silva AC, Maiorka PC, Peconick AP, Raymundo DL, Varaschin MS. Histological and immunohistochemical characterization of the inflammatory and glial cells in the central nervous system of goat fetuses and adult male goats naturally infected with Neospora caninum. BMC Vet Res 2014; 10:291. [PMID: 25495444 PMCID: PMC4270032 DOI: 10.1186/s12917-014-0291-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/25/2014] [Indexed: 12/03/2022] Open
Abstract
Background Neospora caninum is an apicomplexan protozoan that is considered one of the main agents responsible for abortion in ruminants. The lesions found in the central nervous system (CNS) of aborted fetuses show multifocal necrosis, gliosis, and perivascular cuffs of mononuclear cells, but the inflammatory and glial cells have not been immunophenotypically characterized. The lesions in the CNS of infected adult animals have rarely been described. Therefore, in this study, we characterized the lesions, the immunophenotypes of the inflammatory and glial cells and the expression of MHC-II and PCNA in the CNS of goats infected with N. caninum. The CNS of eight aborted fetuses and six adult male goats naturally infected with N. caninum were analyzed with lectin histochemistry (RCA1) and immunohistochemistry (with anti-CD3, −CD79α, −GFAP, −MHC-II, and -PCNA antibodies). All animals were the offspring of dams naturally infected with N. caninum. Results The microscopic lesions in the CNS of the aborted fetuses consisted of perivascular cuffs composed mainly of macrophages (RCA1+), rare T lymphocytes (CD3+), and rare B lymphocytes (CD79α+). Multifocal necrosis surrounded by astrocytes (GFAP+), gliosis composed predominantly of monocytic-lineage cells (macrophages and microglia, RCA1+), and the cysts of N. caninum, related (or not) to the lesions were present. Similar lesions were found in four of the six male goats, and multinucleate giant cells related to focal gliosis were also found in three adult goats. Anti-GFAP immunostaining showed astrocytes characterizing areas of glial scarring. Cysts of N. caninum were found in three adult male goats. The presence of N. caninum was evaluated with histopathology, immunohistochemistry, and PCR. Immunohistochemistry demonstrated anti-PCNA labeling of macrophages and microglia in the perivascular cuffs and the expression of MHC-II by microglia and endothelial cells in the CNS of the aborted fetuses and adult male goats. Conclusions Macrophages and microglia were the predominant inflammatory cells in the CNS of aborted fetuses and healthy adult male goats infected with N. caninum. Activated astrocytes were mainly associated with inflamed areas, suggesting that astrocytes were involved in the resolution of the lesions.
Collapse
Affiliation(s)
- Rafael Carneiro Costa
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | - Débora Ribeiro Orlando
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | - Camila Costa Abreu
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | | | - Leonardo Pereira Mesquita
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Av. Prof. Dr. Orlando Marques de Paiva, 87 - Cidade Universitária, São Paulo, SP, Brazil.
| | - Lismara Castro Nascimento
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | - Aline Costa Silva
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | - Paulo César Maiorka
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Av. Prof. Dr. Orlando Marques de Paiva, 87 - Cidade Universitária, São Paulo, SP, Brazil.
| | - Ana Paula Peconick
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | - Djeison Lutier Raymundo
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| | - Mary Suzan Varaschin
- Universidade Federal de Lavras, Setor de Patologia Veterinária, Caixa postal 3037, Lavras, MG, Brazil.
| |
Collapse
|
371
|
Gavgiotaki E, Filippidis G, Kalognomou M, Tsouko AA, Skordos I, Fotakis C, Athanassakis I. Third Harmonic Generation microscopy as a reliable diagnostic tool for evaluating lipid body modification during cell activation: the example of BV-2 microglia cells. J Struct Biol 2014; 189:105-13. [PMID: 25486610 DOI: 10.1016/j.jsb.2014.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/22/2014] [Accepted: 11/30/2014] [Indexed: 10/24/2022]
Abstract
Nonlinear optical processes have found widespread applications in fields ranging from fundamental physics to biomedicine. In this study, we attempted to evaluate cell activation by using the Third Harmonic Generation (THG) imaging microscopy as a new diagnostic tool. The BV-2 microglia cell line with or without activation by lipopolysaccharide was chosen as a representative biological model. The results showed that THG imaging could discriminate between the control versus activated state of BV-2 cells not only as to THG signal intensity but also as to THG signal area, while verifying once more that the majority of the intracellular detected signal corresponds to lipid bodies. Since THG imaging is a real time, non-destructive modality and does not require any prior cell processing and staining, the results presented here provide an important tool for normal versus activated cell discrimination, which could be proved very useful in the study of inflammation.
Collapse
Affiliation(s)
- E Gavgiotaki
- Institute of Electronic Structure and Laser, Foundation for Research and Technology Hellas, 71110 Heraklion, Greece; Department of Physics, University of Crete, Greece
| | - G Filippidis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology Hellas, 71110 Heraklion, Greece
| | - M Kalognomou
- Department of Biology, University of Crete, Heraklion 71409, Crete, Greece
| | - A A Tsouko
- Institute of Electronic Structure and Laser, Foundation for Research and Technology Hellas, 71110 Heraklion, Greece; Department of Physics, University of Crete, Greece
| | - I Skordos
- Department of Biology, University of Crete, Heraklion 71409, Crete, Greece
| | - C Fotakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology Hellas, 71110 Heraklion, Greece; Department of Physics, University of Crete, Greece
| | - I Athanassakis
- Department of Biology, University of Crete, Heraklion 71409, Crete, Greece.
| |
Collapse
|
372
|
Oligodendrocyte ablation triggers central pain independently of innate or adaptive immune responses in mice. Nat Commun 2014; 5:5472. [PMID: 25434649 PMCID: PMC4268702 DOI: 10.1038/ncomms6472] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023] Open
Abstract
Mechanisms underlying central neuropathic pain are poorly understood. Although glial dysfunction has been functionally linked with neuropathic pain, very little is known about modulation of pain by oligodendrocytes. Here we report that genetic ablation of oligodendrocytes rapidly triggers a pattern of sensory changes that closely resemble central neuropathic pain, which are manifest before overt demyelination. Primary oligodendrocyte loss is not associated with autoreactive T- and B-cell infiltration in the spinal cord and neither activation of microglia nor reactive astrogliosis contribute functionally to central pain evoked by ablation of oligodendrocytes. Instead, light and electron microscopic analyses reveal axonal pathology in the spinal dorsal horn and spinothalamic tract concurrent with the induction and maintenance of nociceptive hypersensitivity. These data reveal a role for oligodendrocytes in modulating pain and suggest that perturbation of oligodendrocyte functions that maintain axonal integrity can lead to central neuropathic pain independent of immune contributions. Whether oligodendrocytes have a role in the development of chronic pain is not clear. Here the authors show that oligodendrocyte depletion causes a neuropathic pain that sets in before demyelination and is independent of immune cell activation and infiltration.
Collapse
|
373
|
Yang JY, Xue X, Tian H, Wang XX, Dong YX, Wang F, Zhao YN, Yao XC, Cui W, Wu CF. Role of microglia in ethanol-induced neurodegenerative disease: Pathological and behavioral dysfunction at different developmental stages. Pharmacol Ther 2014; 144:321-37. [DOI: 10.1016/j.pharmthera.2014.07.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 07/03/2014] [Indexed: 01/04/2023]
|
374
|
Purpurogallin, a natural phenol, attenuates high-mobility group box 1 in subarachnoid hemorrhage induced vasospasm in a rat model. Int J Vasc Med 2014; 2014:254270. [PMID: 25485154 PMCID: PMC4251792 DOI: 10.1155/2014/254270] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/30/2014] [Accepted: 10/30/2014] [Indexed: 01/17/2023] Open
Abstract
High-mobility group box 1 (HMGB1) was shown to be an important extracellular mediator involved in vascular inflammation of animals following subarachnoid hemorrhage (SAH). This study is of interest to examine the efficacy of purpurogallin, a natural phenol, on the alternation of cytokines and HMGB1 in a SAH model. A rodent double hemorrhage SAH model was employed. Basilar arteries (BAs) were harvested to examine HMGB1 mRNA and protein expression (Western blot). CSF samples were to examine IL-1β, IL-6, IL-8, and TNF-α (rt-PCR). Deformed endothelial wall, tortuous elastic lamina, and necrotic smooth muscle were observed in the vessels of SAH groups but were absent in the purpurogallin group. IL-1β, IL-6, and TNF-α in the SAH only and SAH plus vehicle groups were significantly elevated (P < 0.01). Purpurgallin dose-dependently reduced HMGB1 protein expression. Likewise, high dose purpurogallin reduced TNF-α and HMGB1 mRNA levels. In conclusion, purpurogallin exerts its neuroinflammation effect through the dual effect of inhibiting IL-6 and TNF-α mRNA expression and reducing HMGB1 protein and mRNA expression. This study supports purpurogallin could attenuate both proinflammatory cytokines and late-onset inflammasome in SAH induced vasospasm.
Collapse
|
375
|
Huber AK, Duncker PC, Irani DN. Immune responses to non-tumor antigens in the central nervous system. Front Oncol 2014; 4:328. [PMID: 25431758 PMCID: PMC4230036 DOI: 10.3389/fonc.2014.00328] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/30/2014] [Indexed: 12/16/2022] Open
Abstract
The central nervous system (CNS), once viewed as an immune-privileged site protected by the blood-brain barrier (BBB), is now known to be a dynamic immunological environment through which immune cells migrate to prevent and respond to events such as localized infection. During these responses, endogenous glial cells, including astrocytes and microglia, become highly reactive and may secrete inflammatory mediators that regulate BBB permeability and recruit additional circulating immune cells. Here, we discuss the various roles played by astrocytes, microglia, and infiltrating immune cells during host immunity to non-tumor antigens in the CNS, focusing first on bacterial and viral infections, and then turning to responses directed against self-antigens in the setting of CNS autoimmunity.
Collapse
Affiliation(s)
- Amanda K Huber
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - Patrick C Duncker
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
376
|
Yoo Y, Byun K, Kang T, Bayarsaikhan D, Kim JY, Oh S, Kim YH, Kim SY, Chung WI, Kim SU, Lee B, Park YM. Amyloid-Beta-Activated Human Microglial Cells Through ER-Resident Proteins. J Proteome Res 2014; 14:214-23. [DOI: 10.1021/pr500926r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- YongCheol Yoo
- Department
of Biological Sciences, Korea Advanced Institute of Science and Technology, 373-1 Guseong-dong, Daejeon 305-701, Republic of Korea
- Center
for Cognition and Sociality, Institute for Basic Science (IBS), 5, Hwarang-ro 14-gil, Daejeon 305-811, Republic of Korea
- Mass
Spectrometer Research Center, Korea Basic Science Institute, 52
Eoeun-dong, Ochang, Chungcheongbuk-do 363-883, Republic of Korea
| | - Kyunghee Byun
- Center
for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45, Songdo-dong, Yeonsu-ku, Incheon 406-840, Republic of Korea
- Department
of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
| | - Taewook Kang
- Mass
Spectrometer Research Center, Korea Basic Science Institute, 52
Eoeun-dong, Ochang, Chungcheongbuk-do 363-883, Republic of Korea
| | - Delger Bayarsaikhan
- Center
for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45, Songdo-dong, Yeonsu-ku, Incheon 406-840, Republic of Korea
| | - Jin Young Kim
- Mass
Spectrometer Research Center, Korea Basic Science Institute, 52
Eoeun-dong, Ochang, Chungcheongbuk-do 363-883, Republic of Korea
| | - Seyeoun Oh
- Center
for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45, Songdo-dong, Yeonsu-ku, Incheon 406-840, Republic of Korea
| | - Young Hye Kim
- Mass
Spectrometer Research Center, Korea Basic Science Institute, 52
Eoeun-dong, Ochang, Chungcheongbuk-do 363-883, Republic of Korea
| | - Se-Young Kim
- Mass
Spectrometer Research Center, Korea Basic Science Institute, 52
Eoeun-dong, Ochang, Chungcheongbuk-do 363-883, Republic of Korea
| | - Won-Il Chung
- Department
of Biological Sciences, Korea Advanced Institute of Science and Technology, 373-1 Guseong-dong, Daejeon 305-701, Republic of Korea
| | - Seung U. Kim
- Department
of Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, British Columbia V5Z 1M9, Canada
| | - Bonghee Lee
- Center
for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45, Songdo-dong, Yeonsu-ku, Incheon 406-840, Republic of Korea
- Department
of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
| | - Young Mok Park
- Center
for Cognition and Sociality, Institute for Basic Science (IBS), 5, Hwarang-ro 14-gil, Daejeon 305-811, Republic of Korea
- Mass
Spectrometer Research Center, Korea Basic Science Institute, 52
Eoeun-dong, Ochang, Chungcheongbuk-do 363-883, Republic of Korea
| |
Collapse
|
377
|
Poddar NK, Zano S, Natarajan R, Yamamoto B, Viola RE. Enhanced brain distribution of modified aspartoacylase. Mol Genet Metab 2014; 113:219-24. [PMID: 25066302 PMCID: PMC4252805 DOI: 10.1016/j.ymgme.2014.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 10/25/2022]
Abstract
Canavan disease is a fatal neurological disorder caused by defects in the gene that produces the enzyme aspartoacylase. Enzyme replacement therapy can potentially be used to overcome these defects if a stable enzyme form that can gain access to the appropriate neural cells can be produced. Achieving the proper cellular targeting requires a modified form of aspartoacylase that can traverse the blood-brain barrier. A PEGylated form of aspartoacylase that shows dramatic enhancement in brain tissue access and distribution has been produced. While the mechanism of transport has not yet been established, this modified enzyme is significantly less immunogenic than unmodified aspartoacylase. These improved properties set the stage for more extensive enzyme replacement trials as a possible treatment strategy.
Collapse
Affiliation(s)
- Nitesh K Poddar
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Stephen Zano
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Reka Natarajan
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Bryan Yamamoto
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Ronald E Viola
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA.
| |
Collapse
|
378
|
Age-associated changes in rat immune system: Lessons learned from experimental autoimmune encephalomyelitis. Exp Gerontol 2014; 58:179-97. [DOI: 10.1016/j.exger.2014.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/24/2014] [Accepted: 08/12/2014] [Indexed: 01/15/2023]
|
379
|
Lehmann J, Härtig W, Seidel A, Füldner C, Hobohm C, Grosche J, Krueger M, Michalski D. Inflammatory cell recruitment after experimental thromboembolic stroke in rats. Neuroscience 2014; 279:139-54. [DOI: 10.1016/j.neuroscience.2014.08.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/28/2014] [Accepted: 08/17/2014] [Indexed: 10/24/2022]
|
380
|
Siebert JR, Conta Steencken A, Osterhout DJ. Chondroitin sulfate proteoglycans in the nervous system: inhibitors to repair. BIOMED RESEARCH INTERNATIONAL 2014; 2014:845323. [PMID: 25309928 PMCID: PMC4182688 DOI: 10.1155/2014/845323] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are widely expressed in the normal central nervous system, serving as guidance cues during development and modulating synaptic connections in the adult. With injury or disease, an increase in CSPG expression is commonly observed close to lesioned areas. However, these CSPG deposits form a substantial barrier to regeneration and are largely responsible for the inability to repair damage in the brain and spinal cord. This review discusses the role of CSPGs as inhibitors, the role of inflammation in stimulating CSPG expression near site of injury, and therapeutic strategies for overcoming the inhibitory effects of CSPGs and creating an environment conducive to nerve regeneration.
Collapse
Affiliation(s)
- Justin R. Siebert
- Lake Erie College of Osteopathic Medicine at Seton Hill, 20 Seton Hill Drive, Greensburg, PA 15601, USA
| | - Amanda Conta Steencken
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Donna J. Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
381
|
Luo D, Or TCT, Yang CLH, Lau ASY. Anti-inflammatory activity of iridoid and catechol derivatives from Eucommia ulmoides Oliver. ACS Chem Neurosci 2014; 5:855-66. [PMID: 25065689 DOI: 10.1021/cn5001205] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation and pro-inflammatory mediators play key roles in the pathogenesis of neurodegenerative diseases including stroke, which account for a significant burden of morbidity and mortality worldwide. Recently, the unsatisfactory pharmacotherapy and side effects of the drugs led to the development of alternative medicine for treating these diseases. Du Zhong (DZ), Eucommia ulmoides Oliver leaves, is a commonly used herb in the therapy of stroke in China. We hypothesize that the components from DZ inhibit neuroinflammation. In this study, DZ was extracted and the bioactive fractions with inhibitory effect on lipopolysaccharide (LPS)-stimulated nitric oxide (NO) production in BV-2 microglial cells were further separated using chromatography. Two purified bioactive compounds, genipin (compound C) and 4-(1,2-dimethoxyethyl)benzene-1,2-diol (compound F), were isolated and identified after spectroscopic analysis. The results showed that they inhibited LPS-stimulated NO and tumor necrosis factor-alpha (TNF-α) production. Genipin exerted its anti-inflammatory effects through PI3K/Akt signaling pathway, whereas compound F inhibited phosphorylation of p38 mitogen-activated protein kinase (MAPK). In conclusion, genipin and compound F have potential for developing into new drugs for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Dan Luo
- Molecular
Chinese Medicine Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Terry C. T. Or
- Molecular
Chinese Medicine Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Cindy L. H. Yang
- Molecular
Chinese Medicine Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Allan S. Y. Lau
- Molecular
Chinese Medicine Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Cytokine
Biology Group, Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
382
|
Popiolek-Barczyk K, Rojewska E, Jurga AM, Makuch W, Zador F, Borsodi A, Piotrowska A, Przewlocka B, Mika J. Minocycline enhances the effectiveness of nociceptin/orphanin FQ during neuropathic pain. BIOMED RESEARCH INTERNATIONAL 2014; 2014:762930. [PMID: 25276817 PMCID: PMC4168034 DOI: 10.1155/2014/762930] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/01/2014] [Indexed: 12/27/2022]
Abstract
Nociceptin/orphanin FQ (N/OFQ) antinociception, which is mediated selectively by the N/OFQ peptide receptor (NOP), was demonstrated in pain models. In this study, we determine the role of activated microglia on the analgesic effects of N/OFQ in a rat model of neuropathic pain induced by chronic constriction injury (CCI) to the sciatic nerve. Repeated 7-day administration of minocycline (30 mg/kg i.p.), a drug that affects microglial activation, significantly reduced pain in CCI-exposed rats and it potentiates the analgesic effects of administered N/OFQ (2.5-5 μg i.t.). Minocycline also downregulates the nerve injury-induced upregulation of NOP protein in the dorsal lumbar spinal cord. Our in vitro study showed that minocycline reduced NOP mRNA, but not protein, level in rat primary microglial cell cultures. In [(35)S]GTPγS binding assays we have shown that minocycline increases the spinal N/OFQ-stimulated NOP signaling. We suggest that the modulation of the N/OFQ system by minocycline is due to the potentiation of its neuronal antinociceptive activity and weakening of the microglial cell activation. This effect is beneficial for pain relief, and these results suggest new targets for the development of drugs that are effective against neuropathic pain.
Collapse
Affiliation(s)
- Katarzyna Popiolek-Barczyk
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| | - Agnieszka M. Jurga
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| | - Ferenz Zador
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt 62 Street, Szeged 6726, Hungary
| | - Anna Borsodi
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt 62 Street, Szeged 6726, Hungary
| | - Anna Piotrowska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Cracow, Poland
| |
Collapse
|
383
|
Legido A, Katsetos CD. Experimental studies in epilepsy: immunologic and inflammatory mechanisms. Semin Pediatr Neurol 2014; 21:197-206. [PMID: 25510941 DOI: 10.1016/j.spen.2014.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this article, we review the literature based on experimental studies lending credence to a relationship between epilepsy and immune-mediated mechanisms linked to central nervous system innate immunity. The brain innate immunity responses to neuronal injury or excessive neuronal activity are mediated by resident microglia and astroglia, but also neurons play an immunomodulatory role. Antigens or antibodies applied to the brain trigger an epileptogenic and inflammatory response. Furthermore, seizure activity and status epilepticus elicit the production and release of proinflammatory cytokines and chemokines. The immune pathogenesis of epilepsy involves complex cell-to-cell interactions including a cross talk between astrocytes and neurons, between astrocytes and brain microvascular endothelial cells, as well as reciprocal leukocyte-endothelial interactions in the context of disruption of the blood-brain barrier. There is a large body of literature from experimental studies showing that seizures can initiate a cascade of innate and adaptive immune responses from various cellular sources and perpetuate neuroinflammation through mechanisms involving transcription of inflammatory genes or posttranslational changes in cytokine release machinery. These inflammatory processes could also possibly contribute to the pathogenesis of comorbidities often associated with epilepsy. This opens exciting possibilities for the development of disease-modifying drugs aimed at mitigating neuroinflammation as a means of ameliorating epileptogenesis and lessening or preventing postictal brain injury.
Collapse
Affiliation(s)
- Agustín Legido
- Section of Neurology, St. Christopher's Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA.
| | - Christos D Katsetos
- Section of Neurology, St. Christopher's Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA
| |
Collapse
|
384
|
Zhang YH, Chen H, Chen Y, Wang L, Cai YH, Li M, Wen HQ, Du J, An R, Luo QL, Wang XL, Lun ZR, Xu YH, Shen JL. Activated microglia contribute to neuronal apoptosis in Toxoplasmic encephalitis. Parasit Vectors 2014; 7:372. [PMID: 25128410 PMCID: PMC4143554 DOI: 10.1186/1756-3305-7-372] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/01/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A plethora of evidence shows that activated microglia play a critical role in the pathogenesis of the central nervous system (CNS). Toxoplasmic encephalitis (TE) frequently occurs in HIV/AIDS patients. However, knowledge remains limited on the contributions of activated microglia to the pathogenesis of TE. METHODS A murine model of reactivated encephalitis was generated in a latent infection with Toxoplasma gondii induced by cyclophosphamide. The neuronal apoptosis in the CNS and the profile of pro-inflammatory cytokines were assayed in both in vitro and in vivo experiments. RESULTS Microglial cells were found to be activated in the cortex and hippocampus in the brain tissues of mice. The in vivo expression of interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS) were up-regulated in TE mice, and accordingly, the neuronal apoptosis was significantly increased. The results were positively correlated with those of the in vitro experiments. Additionally,apoptosis of the mouse neuroblastoma type Neuro2a (N2a) remarkably increased when the N2a was co-cultured in transwell with microglial cells and Toxoplasma tachyzoites. Both in vivo and in vitro experiments showed that minocycline (a microglia inhibitor) treatment notably reduced microglial activation and neuronal apoptosis. CONCLUSIONS Activated microglia contribute to neuronal apoptosis in TE and inhibition of microglia activation might represent a novel therapeutic strategy of TE.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yuan-hong Xu
- The Key Laboratory of Zoonoses and Pathogen Biology Anhui, and Department of Parasitology, Anhui Medical University, Hefei, China.
| | | |
Collapse
|
385
|
Jansen AHP, Reits EAJ, Hol EM. The ubiquitin proteasome system in glia and its role in neurodegenerative diseases. Front Mol Neurosci 2014; 7:73. [PMID: 25152710 PMCID: PMC4126450 DOI: 10.3389/fnmol.2014.00073] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/10/2014] [Indexed: 12/12/2022] Open
Abstract
The ubiquitin proteasome system (UPS) is crucial for intracellular protein homeostasis and for degradation of aberrant and damaged proteins. The accumulation of ubiquitinated proteins is a hallmark of many neurodegenerative diseases, including amyotrophic lateral sclerosis, Alzheimer’s, Parkinson’s, and Huntington’s disease, leading to the hypothesis that proteasomal impairment is contributing to these diseases. So far, most research related to the UPS in neurodegenerative diseases has been focused on neurons, while glial cells have been largely disregarded in this respect. However, glial cells are essential for proper neuronal function and adopt a reactive phenotype in neurodegenerative diseases, thereby contributing to an inflammatory response. This process is called reactive gliosis, which in turn affects UPS function in glial cells. In many neurodegenerative diseases, mostly neurons show accumulation and aggregation of ubiquitinated proteins, suggesting that glial cells may be better equipped to maintain proper protein homeostasis. During an inflammatory reaction, the immunoproteasome is induced in glia, which may contribute to a more efficient degradation of disease-related proteins. Here we review the role of the UPS in glial cells in various neurodegenerative diseases, and we discuss how studying glial cell function might provide essential information in unraveling mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Anne H P Jansen
- Department of Cell Biology and Histology, Academic Medical Center Amsterdam, Netherlands
| | - Eric A J Reits
- Department of Cell Biology and Histology, Academic Medical Center Amsterdam, Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands ; Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences Amsterdam, Netherlands ; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Netherlands
| |
Collapse
|
386
|
Liu Y, Zhang R, Yan K, Chen F, Huang W, Lv B, Sun C, Xu L, Li F, Jiang X. Mesenchymal stem cells inhibit lipopolysaccharide-induced inflammatory responses of BV2 microglial cells through TSG-6. J Neuroinflammation 2014; 11:135. [PMID: 25088370 PMCID: PMC4128538 DOI: 10.1186/1742-2094-11-135] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/21/2014] [Indexed: 12/20/2022] Open
Abstract
Microglia are the primary immunocompetent cells in brain tissue and microglia-mediated inflammation is associated with the pathogenesis of various neuronal disorders. Recently, many studies have shown that mesenchymal stem cells (MSCs) display a remarkable ability to modulate inflammatory and immune responses through the release of a variety of bioactive molecules, thereby protecting the central nervous system. Previously, we reported that MSCs have the ability to modulate inflammatory responses in a traumatic brain injury model and that the potential mechanisms may be partially attributed to upregulated TNF-α stimulated gene/protein 6 (TSG-6) expression. However, whether TSG-6 exerts an anti-inflammatory effect by affecting microglia is not fully understood. In this study, we investigated the anti-inflammatory effects of MSCs and TSG-6 in an in vitro lipopolysaccharide (LPS)-induced BV2 microglial activation model. We found that MSCs and TSG-6 significantly inhibited the expression of pro-inflammatory mediators in activated microglia. However, MSC effects on microglia were attenuated when TSG-6 expression was silenced. In addition, we found that the activation of nuclear factor (NF)-κB and mitogen-activated protein kinase (MAPK) pathways in LPS-stimulated BV2 microglial cells was significantly inhibited by TSG-6. Furthermore, we found that the presence of CD44 in BV2 microglial cells was essential for MSC- and TSG-6-mediated inhibition of pro-inflammatory gene expression and of NF-κB and MAPK activation in BV2 microglial cells. The results of this study suggest that MSCs can modulate microglia activation through TSG-6 and that TSG-6 attenuates the inflammatory cascade in activated microglia. Our study indicates that novel mechanisms are responsible for the immunomodulatory effect of MSCs on microglia and that MSCs, as well as TSG-6, might be promising therapeutic agents for the treatment of neurotraumatic injuries or neuroinflammatory diseases associated with microglial activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiaodan Jiang
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Gongye Road, Guangzhou 510282, China.
| |
Collapse
|
387
|
García-Cáceres C, Fuente-Martín E, Díaz F, Granado M, Argente-Arizón P, Frago LM, Freire-Regatillo A, Barrios V, Argente J, Chowen JA. The opposing effects of ghrelin on hypothalamic and systemic inflammatory processes are modulated by its acylation status and food intake in male rats. Endocrinology 2014; 155:2868-80. [PMID: 24848869 DOI: 10.1210/en.2014-1074] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ghrelin is an endogenous hormone that stimulates appetite and adipose tissue accrual. Both the acylated (AG) and non-acylated (DAG) isoforms of this hormone are also reported to exert anti-inflammatory and protective effects systemically and in the central nervous system. As inflammatory processes have been implicated in obesity-associated secondary complications, we hypothesized that this natural appetite stimulator may protect against negative consequences resulting from excessive food intake. Adult male Wistar rats were treated icv (5 μg/day) with AG, DAG, the ghrelin mimetic GH-releasing peptide (GHRP)-6, AG, and pair-fed with controls (AG-pf) or saline for 14 days. Regardless of food intake AG increased visceral adipose tissue (VAT) and decreased circulating cytokine levels. However, AG reduced cytokine production in VAT only in rats fed ad libitum. Hypothalamic cytokine production was increased in AG-treated rats fed ad libitum and by DAG, but intracellular inflammatory signaling pathways associated with insulin and leptin resistance were unaffected. Gliosis was not observed in response to any treatment as glial markers were either reduced or unaffected. AG, DAG, and GHRP-6 stimulated production of hypothalamic insulin like-growth factor I that is involved in cell protective mechanisms. In hypothalamic astrocyte cell cultures AG decreased tumor necrosis factorα and DAG decreased interleukin-1β mRNA levels, suggesting direct anti-inflammatory effects on astrocytes. Thus, whereas ghrelin stimulates food intake and weight gain, it may also induce mechanisms of cell protection that help to detour or delay systemic inflammatory responses and hypothalamic gliosis due to excess weight gain, as well as its associated pathologies.
Collapse
Affiliation(s)
- Cristina García-Cáceres
- Hospital Infantil Universitario Niño Jesús, Department of Endocrinology, Instituto de Investigación La Princesa, Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28009 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
388
|
Schwab JM, Zhang Y, Kopp MA, Brommer B, Popovich PG. The paradox of chronic neuroinflammation, systemic immune suppression, autoimmunity after traumatic chronic spinal cord injury. Exp Neurol 2014; 258:121-129. [PMID: 25017893 PMCID: PMC4099970 DOI: 10.1016/j.expneurol.2014.04.023] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/19/2014] [Accepted: 04/21/2014] [Indexed: 02/06/2023]
Abstract
During the transition from acute to chronic stages of recovery after spinal cord injury (SCI), there is an evolving state of immunologic dysfunction that exacerbates the problems associated with the more clinically obvious neurologic deficits. Since injury directly affects cells embedded within the "immune privileged/specialized" milieu of the spinal cord, maladaptive or inefficient responses are likely to occur. Collectively, these responses qualify as part of the continuum of "SCI disease" and are important therapeutic targets to improve neural repair and neurological outcome. Generic immune suppressive therapies have been largely unsuccessful, mostly because inflammation and immunity exert both beneficial (plasticity enhancing) and detrimental (e.g. glia- and neurodegenerative; secondary damage) effects and these functions change over time. Moreover, "compartimentalized" investigations, limited to only intraspinal inflammation and associated cellular or molecular changes in the spinal cord, neglect the reality that the structure and function of the CNS are influenced by systemic immune challenges and that the immune system is 'hardwired' into the nervous system. Here, we consider this interplay during the progression from acute to chronic SCI. Specifically, we survey impaired/non-resolving intraspinal inflammation and the paradox of systemic inflammatory responses in the context of ongoing chronic immune suppression and autoimmunity. The concepts of systemic inflammatory response syndrome (SIRS), compensatory anti-inflammatory response syndrome (CARS) and "neurogenic" spinal cord injury-induced immune depression syndrome (SCI-IDS) are discussed as determinants of impaired "host-defense" and trauma-induced autoimmunity.
Collapse
Affiliation(s)
- Jan M. Schwab
- Department of Neurology and Experimental Neurology, Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charite - Universitatsmedizin Berlin, D-10117 Berlin, Germany
- Spinal Cord Injury Center, Trauma Hospital Berlin, D-12683 Berlin, Germany
| | - Yi Zhang
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Marcel A. Kopp
- Department of Neurology and Experimental Neurology, Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charite - Universitatsmedizin Berlin, D-10117 Berlin, Germany
| | - Benedikt Brommer
- Department of Neurology and Experimental Neurology, Clinical and Experimental Spinal Cord Injury Research (Neuroparaplegiology), Charite - Universitatsmedizin Berlin, D-10117 Berlin, Germany
| | - Phillip G. Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center, The Ohio State University Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
389
|
CHENG WENJING, LI YUNHONG, HOU XIAOLIN, ZHANG NAN, MA JIAO, DING FEIJIA, LI FAN, MIAO ZHENHUA, ZHANG YANLI, QI QI, LI GUANGHUA, SHEN YING, LIU JUAN, HUANG WEIDONG, WANG YIN. HSP60 is involved in the neuroprotective effects of naloxone. Mol Med Rep 2014; 10:2172-6. [DOI: 10.3892/mmr.2014.2411] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 05/09/2014] [Indexed: 11/05/2022] Open
|
390
|
Roles of TLR3 and RIG-I in mediating the inflammatory response in mouse microglia following Japanese encephalitis virus infection. J Immunol Res 2014; 2014:787023. [PMID: 25101306 PMCID: PMC4101954 DOI: 10.1155/2014/787023] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/03/2014] [Indexed: 12/28/2022] Open
Abstract
Japanese encephalitis virus (JEV) infection can cause central nervous system disease with irreversible neurological damage in humans and animals. Evidence suggests that overactivation of microglia leads to greatly increased neuronal damage during JEV infection. However, the mechanism by which JEV induces the activation of microglia remains unclear. Toll-like receptor 3 (TLR3) and retinoic acid-inducible gene I (RIG-I) can recognize double-stranded RNA, and their downstream signaling results in production of proinflammatory mediators. In this study, we investigated the roles of TLR3 and RIG-I in the inflammatory response caused by JEV infection in the mouse microglial cell line. JEV infection induced the expression of TLR3 and RIG-I and the activation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (p38MAPK). Knockdown of TLR3 and RIG-I attenuated activation of ERK, p38MAPK, activator protein 1 (AP-1), and nuclear factor κB (NF-κB). Secretion of TNF-α, IL-6, and CCL-2, which was induced by JEV, was reduced by TLR3 and RIG-I knockdown and inhibitors of phosphorylated ERK and p38MAPK. Furthermore, viral proliferation was increased following knockdown of TLR3 and RIG-I. Our findings suggest that the signaling pathways of TLR3 and RIG-I play important roles in the JEV-induced inflammatory response of microglia.
Collapse
|
391
|
Barreto GE, Santos-Galindo M, Garcia-Segura LM. Selective estrogen receptor modulators regulate reactive microglia after penetrating brain injury. Front Aging Neurosci 2014; 6:132. [PMID: 24999330 PMCID: PMC4064706 DOI: 10.3389/fnagi.2014.00132] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/05/2014] [Indexed: 11/13/2022] Open
Abstract
Following brain injury, microglia assume a reactive-like state and secrete pro-inflammatory molecules that can potentiate damage. A therapeutic strategy that may limit microgliosis is of potential interest. In this context, selective estrogen receptor modulators, such as raloxifene and tamoxifen, are known to reduce microglia activation induced by neuroinflammatory stimuli in young animals. In the present study, we have assessed whether raloxifene and tamoxifen are able to affect microglia activation after brain injury in young and aged animals in time points relevant to clinics, which is hours after brain trauma. Volume fraction of MHC-II(+) microglia was estimated according to the point-counting method of Weibel within a distance of 350 μm from the lateral border of the wound, and cellular morphology was measured by fractal analysis. Two groups of animals were studied: (1) young rats, ovariectomized at 2 months of age; and (2) aged rats, ovariectomized at 18 months of age. Fifteen days after ovariectomy animals received a stab wound brain injury and the treatment with estrogenic compounds. Our findings indicate that raloxifene and tamoxifen reduced microglia activation in both young and aged animals. Although the volume fraction of reactive microglia was found lower in aged animals, this was accompanied by important changes in cell morphology, where aged microglia assume a bushier and hyperplasic aspect when compared to young microglia. These data suggest that early regulation of microglia activation provides a mechanism by which selective estrogen receptors modulators (SERMs) may exert a neuroprotective effect in the setting of a brain trauma.
Collapse
Affiliation(s)
- George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | | | | |
Collapse
|
392
|
Sweet TB, Panda N, Hein AM, Das SL, Hurley SD, Olschowka JA, Williams JP, O'Banion MK. Central Nervous System Effects of Whole-Body Proton Irradiation. Radiat Res 2014; 182:18-34. [DOI: 10.1667/rr13699.1] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Tara Beth Sweet
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Nirlipta Panda
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Amy M. Hein
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Shoshana L. Das
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Sean D. Hurley
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - John A. Olschowka
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - M. Kerry O'Banion
- Department of Neurobiology and Anatomy, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
393
|
Krauthausen M, Saxe S, Zimmermann J, Emrich M, Heneka MT, Müller M. CXCR3 modulates glial accumulation and activation in cuprizone-induced demyelination of the central nervous system. J Neuroinflammation 2014; 11:109. [PMID: 24930935 PMCID: PMC4096537 DOI: 10.1186/1742-2094-11-109] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 05/15/2014] [Indexed: 12/18/2022] Open
Abstract
Background The functional state of glial cells, like astrocytes and microglia, critically modulates the course of neuroinflammatory and neurodegenerative diseases and can have both detrimental and beneficial effects. Glial cell function is tightly controlled by cellular interactions in which cytokines are important messengers. Recent studies provide evidence that in particular chemokines are important modulators of glial cell function. During the course of CNS diseases like multiple sclerosis or Alzheimer’s disease, and in the corresponding animal models, the chemokines CXCL9 and CXCL10 are abundantly expressed at sites of glial activation, arguing for an important role of these chemokines and their corresponding receptor CXCR3 in glial activation. To clarify the role of this chemokine system in glial cell activation, we characterized the impact of CXCR3 on glial activation in a model of toxic demyelination in which glial activation without a prominent influx of hematogenous cells is prototypical. Methods We investigated the impact of CXCR3 on cuprizone-induced demyelination, comparing CXCR3-deficient mice with wild type controls. The clinical course during cuprizone feeding was documented for five weeks and for the subsequent four days withdrawal of the cuprizone diet (5.5 weeks). Glial activation was characterized using histological, histomorphometric and phenotypic analysis. Molecular analysis for (de)myelination and neuroinflammation was applied to characterize the effect of cuprizone on CXCR3-deficient mice and control animals. Results CXCR3-deficient mice displayed a milder clinical course during cuprizone feeding and a more rapid body weight recovery after offset of diet. In the CNS, CXCR3 deficiency significantly attenuated the accumulation and activation of microglia and astrocytes. Moreover, a deficiency of CXCR3 reduced the expression of the microglial activation markers CD45 and CD11b. Compared to controls, we observed a vast reduction of RNA levels for proinflammatory cytokines and chemokines like Ccl2, Cxcl10, Tnf and Il6 within the CNS of cuprizone-treated mice. Lastly, CXCR3 deficiency had no major effects on the course of demyelination during cuprizone feeding. Conclusions The CXCR3 chemokine system is critically involved in the intrinsic glial activation during cuprizone-induced demyelination, which significantly modulates the distribution of glial cells and the local cytokine milieu.
Collapse
Affiliation(s)
- Marius Krauthausen
- Department of Neurology, Universitätsklinikum Bonn, Sigmund-Freud-Str, 25, D-53105 Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
394
|
Reduced inflammatory phenotype in microglia derived from neonatal rat spinal cord versus brain. PLoS One 2014; 9:e99443. [PMID: 24914808 PMCID: PMC4051776 DOI: 10.1371/journal.pone.0099443] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 05/15/2014] [Indexed: 12/22/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system (CNS). Membrane bound sensors on their processes monitor the extracellular environment and respond to perturbations of the CNS such as injury or infection. Once activated, microglia play a crucial role in determining neuronal survival. Recent studies suggest that microglial functional response properties vary across different regions of the CNS. However, the activation profiles of microglia derived from the spinal cord have not been evaluated against brain microglia in vitro. Here, we studied the morphological properties and secretion of inflammatory and trophic effectors by microglia derived from the brain or spinal cord of neonatal rats under basal culture conditions and after activation with lipopolysaccharide (LPS). Our results demonstrate that spinal microglia assume a less inflammatory phenotype after LPS activation, with reduced release of the inflammatory effectors tumor necrosis factor alpha, interleukin-1 beta, and nitric oxide, a less amoeboid morphology, and reduced phagocytosis relative to brain-derived microglia. Phenotypic differences between brain and spinal microglia are an important consideration when evaluating anti-inflammatory or immunomodulatory therapies for brain versus spinal injury.
Collapse
|
395
|
Barr GA, Hunter DA. Interactions between glia, the immune system and pain processes during early development. Dev Psychobiol 2014; 56:1698-710. [PMID: 24910104 DOI: 10.1002/dev.21229] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/15/2014] [Indexed: 01/10/2023]
Abstract
Pain is a serious problem for infants and children and treatment options are limited. Moreover, infants born prematurely or hospitalized for illness likely have concurrent infection that activates the immune system. It is now recognized that the immune system in general and glia in particular influence neurotransmission and that the neural bases of pain are intimately connected to immune function. We know that injuries that induce pain activate immune function and suppressing the immune system alleviates pain. Despite this advance in our understanding, virtually nothing is known of the role that the immune system plays in pain processing in infants and children, even though pain is a serious clinical issue in pediatric medicine. This brief review summarizes the existing data on immune-neural interactions in infants, providing evidence for the immaturity of these interactions.
Collapse
Affiliation(s)
- Gordon A Barr
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104.
| | | |
Collapse
|
396
|
Schachtele SJ, Hu S, Sheng WS, Mutnal MB, Lokensgard JR. Glial cells suppress postencephalitic CD8+ T lymphocytes through PD-L1. Glia 2014; 62:1582-94. [PMID: 24890099 PMCID: PMC4141010 DOI: 10.1002/glia.22701] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/30/2014] [Accepted: 05/15/2014] [Indexed: 01/06/2023]
Abstract
Engagement of the programmed death (PD)−1 receptor on activated cells by its ligand (PD‐L1) is a mechanism for suppression of activated T‐lymphocytes. Microglia, the resident inflammatory cells of the brain, are important for pathogen detection and initiation of innate immunity, however, a novel role for these cells as immune regulators has also emerged. PD‐L1 on microglia has been shown to negatively regulate T‐cell activation in models of multiple sclerosis and acute viral encephalitis. In this study, we investigated the role of glial cell PD‐L1 in controlling encephalitogenic CD8+ T‐lymphocytes, which infiltrate the brain to manage viral infection, but remain to produce chronic neuroinflammation. Using a model of chronic neuroinflammation following murine cytomegalovirus (MCMV)‐induced encephalitis, we found that CD8+ T‐cells persisting within the brain expressed PD‐1. Conversely, activated microglia expressed PD‐L1. In vitro, primary murine microglia, which express low basal levels of PD‐L1, upregulated the co‐inhibitory ligand on IFN‐γ‐treatment. Blockade of the PD‐1: PD‐L1 pathway in microglial: CD8+ T‐cell co‐cultures increased T‐cell IFN‐γ and interleukin (IL)−2 production. We observed a similar phenomenon following blockade of this co‐inhibitory pathway in astrocyte: CD8+ T‐cell co‐cultures. Using ex vivo cultures of brain leukocytes, including microglia and CD8+ T‐cells, obtained from mice with MCMV‐induced chronic neuroinflammation, we found that neutralization of either PD‐1 or PD‐L1 increased IFN‐γ production from virus‐specific CD8+ T‐cells stimulated with MCMV IE1168–176 peptide. These data demonstrate that microglia and astrocytes control antiviral T‐cell responses and suggest a therapeutic potential of PD1: PD‐L1 modulation to manage the deleterious consequences of uncontrolled neuroinflammation. GLIA 2014;62:1582–1594 Microglia and astrocytes exert regulatory control over T‐cells during chronic neuroinflammation following viral brain infection. Post-encephalitic glial cells express PD‐L1 and suppress persistent CD8 T‐cells via the PD‐1: PD‐L1 inhibitory pathway.
Collapse
Affiliation(s)
- Scott J Schachtele
- Department of Medicine, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota, McGuire Translational Research Facility, Minneapolis, Minnesota
| | | | | | | | | |
Collapse
|
397
|
Li R, Xu W, Chen Y, Qiu W, Shu Y, Wu A, Dai Y, Bao J, Lu Z, Hu X. Raloxifene suppresses experimental autoimmune encephalomyelitis and NF-κB-dependent CCL20 expression in reactive astrocytes. PLoS One 2014; 9:e94320. [PMID: 24722370 PMCID: PMC3983123 DOI: 10.1371/journal.pone.0094320] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/14/2014] [Indexed: 12/19/2022] Open
Abstract
Recent clinical data have led to the consideration of sexual steroids as new potential therapeutic tools for multiple sclerosis. Selective estrogen receptor modulators can exhibit neuroprotective effects like estrogen, with fewer systemic estrogen side effects than estrogen, offering a more promising therapeutic modality for multiple sclerosis. The important role of astrocytes in a proinflammatory effect mediated by CCL20 signaling on inflammatory cells has been documented. Their potential contribution to selective estrogen receptor modulator-mediated protection is still unknown. Using a mouse model of chronic neuroinflammation, we report that raloxifene, a selective estrogen receptor modulator, alleviated experimental autoimmune encephalomyelitis–an animal model of multiple sclerosis–and decreased astrocytic production of CCL20. Enzyme-linked immunosorbent assay, immunohistochemistry imaging and transwell migration assays revealed that reactive astrocytes express CCL20, which promotes Th17 cell migration. In cultured rodent astrocytes, raloxifene inhibited IL-1β-induced CCL20 expression and chemotaxis ability for Th17 migration, whereas the estrogen receptor antagonist ICI 182,780 blocked this effect. Western blotting further indicated that raloxifene suppresses IL-1β-induced NF-κB activation (phosphorylation of p65) and translocation but does not affect phosphorylation of IκB. In conclusion, these data demonstrate that raloxifene provides robust neuroprotection against experimental autoimmune encephalomyelitis, partially via an inhibitory action on CCL20 expression and NF-κB pathways in reactive astrocytes. Our results contribute to a better understanding of the critical roles of raloxifene in treating experimental autoimmune encephalomyelitis and uncover reactive astrocytes as a new target for the inhibitory action of estrogen receptors on chemokine CCL20 expression.
Collapse
MESH Headings
- Animals
- Astrocytes/drug effects
- Astrocytes/pathology
- Cell Movement/drug effects
- Cells, Cultured
- Chemokine CCL20/antagonists & inhibitors
- Chemokine CCL20/genetics
- Chemokine CCL20/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Female
- Fulvestrant
- Gene Expression Regulation
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/genetics
- Multiple Sclerosis/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Raloxifene Hydrochloride/pharmacology
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/genetics
- Receptors, Estrogen/immunology
- Selective Estrogen Receptor Modulators/pharmacology
- Signal Transduction
- Th17 Cells/drug effects
- Th17 Cells/pathology
Collapse
Affiliation(s)
- Rui Li
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wen Xu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Ying Chen
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wei Qiu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yaqing Shu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Aimin Wu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yongqiang Dai
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Jian Bao
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhengqi Lu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xueqiang Hu
- Multiple Sclerosis Center, Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- * E-mail:
| |
Collapse
|
398
|
Shpak M, Hall AW, Goldberg MM, Derryberry DZ, Ni Y, Iyer VR, Cowperthwaite MC. An eQTL analysis of the human glioblastoma multiforme genome. Genomics 2014; 103:252-63. [DOI: 10.1016/j.ygeno.2014.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/11/2014] [Accepted: 02/24/2014] [Indexed: 12/20/2022]
|
399
|
RETINAL MORPHOLOGIC CHANGES AND CONCENTRATIONS OF CYTOKINES IN EYES WITH DIABETIC MACULAR EDEMA. Retina 2014; 34:741-8. [DOI: 10.1097/iae.0b013e3182a48917] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
400
|
Morphological Analyses on Retinal Glial Responses to Glaucomatous Injury Evoked by Venous Cauterization. Appl Microsc 2014. [DOI: 10.9729/am.2014.44.1.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|