1
|
Diverse Gene Expressions in the Prediction of Cuprizone-Induced Demyelination. Neurotox Res 2020; 37:732-742. [DOI: 10.1007/s12640-019-00154-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 12/13/2019] [Accepted: 12/19/2019] [Indexed: 10/25/2022]
|
2
|
Zhang Y, Zhou Y, Chen S, Hu Y, Zhu Z, Wang Y, Du N, Song T, Yang Y, Guo A, Wang Y. Macrophage migration inhibitory factor facilitates prostaglandin E 2 production of astrocytes to tune inflammatory milieu following spinal cord injury. J Neuroinflammation 2019; 16:85. [PMID: 30981278 PMCID: PMC6461812 DOI: 10.1186/s12974-019-1468-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
Background Astrocytes have been shown to produce several pro- and anti-inflammatory cytokines to maintain homeostasis of microenvironment in response to vast array of CNS insults. Some inflammation-related cytokines are responsible for regulating such cell events. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that can be inducibly expressed in the lesioned spinal cord. Unknown is whether MIF can facilitate the production of immunosuppressive factors from astrocytes to tune milieu following spinal cord injury. Methods Following establishment of contusion SCI rat model, correlation of PGE2 synthesis-related protein levels with that of MIF was assayed by Western blot. ELISA assay was used to detect production of PGE2, TNF-α, IL-1β, and IL-6. Immunohistochemistry was performed to observe colocalization of COX2 with GFAP- and S100β-positive astrocytes. The primary astrocytes were treated by various inhibitors to validate relevant signal pathway. Results The protein levels of MIF and COX2, but not of COX1, synchronously increased following spinal cord injury. Treatment of MIF inhibitor 4-IPP to the lesion sites significantly reduced the expression of COX2, mPGES-1, and as a consequence, the production of PGE2. Astrocytes responded robustly to the MIF interference, by which regulated MAPK/COX2/PGE2 signal pathway through coupling with the CD74 membrane receptor. MIF-induced production of PGE2 from astrocytes was able to suppress production of TNF-α, but boosted production of IL-1β and IL-6 in LPS-activated macrophages. Conclusion Collectively, these results reveal a novel function of MIF-mediated astrocytes, which fine-tune inflammatory microenvironment to maintain homeostasis. These suggest an alternative therapeutic strategy for CNS inflammation. Electronic supplementary material The online version of this article (10.1186/s12974-019-1468-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuxin Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.,Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Yue Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.,Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Shuxia Chen
- Department of Pediatrics, Yancheng City No.1 People's Hospital, Yancheng, 224005, People's Republic of China
| | - Yuming Hu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Zhenjie Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Nan Du
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Tiancheng Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China
| | - Aisong Guo
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China.
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, People's Republic of China.
| |
Collapse
|
3
|
Larson TA. Sex Steroids, Adult Neurogenesis, and Inflammation in CNS Homeostasis, Degeneration, and Repair. Front Endocrinol (Lausanne) 2018; 9:205. [PMID: 29760681 PMCID: PMC5936772 DOI: 10.3389/fendo.2018.00205] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/12/2018] [Indexed: 01/16/2023] Open
Abstract
Sex steroidal hormones coordinate the development and maintenance of tissue architecture in many organs, including the central nervous systems (CNS). Within the CNS, sex steroids regulate the morphology, physiology, and behavior of a wide variety of neural cells including, but not limited to, neurons, glia, endothelial cells, and immune cells. Sex steroids spatially and temporally control distinct molecular networks, that, in turn modulate neural activity, synaptic plasticity, growth factor expression and function, nutrient exchange, cellular proliferation, and apoptosis. Over the last several decades, it has become increasingly evident that sex steroids, often in conjunction with neuroinflammation, have profound impact on the occurrence and severity of neuropsychiatric and neurodegenerative disorders. Here, I review the foundational discoveries that established the regulatory role of sex steroids in the CNS and highlight recent advances toward elucidating the complex interaction between sex steroids, neuroinflammation, and CNS regeneration through adult neurogenesis. The majority of recent work has focused on neuroinflammatory responses following acute physical damage, chronic degeneration, or pharmacological insult. Few studies directly assess the role of immune cells in regulating adult neurogenesis under healthy, homeostatic conditions. As such, I also introduce tractable, non-traditional models for examining the role of neuroimmune cells in natural neuronal turnover, seasonal plasticity of neural circuits, and extreme CNS regeneration.
Collapse
Affiliation(s)
- Tracy A. Larson
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Ku SK, Bae JS. Inhibitory Effect of FXa on Secretory Group IIA Phospholipase A2. Inflammation 2016; 38:987-94. [PMID: 25399323 DOI: 10.1007/s10753-014-0062-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
It is well known that the expression level of secretory group IIA phospholipase A2 (sPLA2-IIA) is elevated in inflammatory diseases and lipopolysaccharide (LPS) upregulates the expression of sPLA2-IIA in human umbilical vein endothelial cells (HUVECs). Activated factor X (FXa) is an important enzyme in the coagulation cascade responsible for thrombin generation, and it influences cell signaling in various cell types by activating protease-activated receptors (PARs). Here, FX or FXa was examined for its effects on the expression and activity of sPLA2-IIA in HUVECs and mouse. Prior treatment of cells or mouse with FXa inhibited LPS-induced expression and activity of sPLA2-IIA via interacting with FXa receptor (effective cell protease receptor-1, EPR-1). And FXa suppressed the activation of cytosolic phospholipase A2 (cPLA2) and extracellular signal-regulated kinase (ERK) 1/2 by LPS. Therefore, these results suggest that FXa may inhibit LPS-mediated expression of sPLA2-IIA by suppression of cPLA2 and ERK 1/2.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan, 712-715, Republic of Korea
| | | |
Collapse
|
5
|
Ku SK, Yang EJ, Kang H, Jung B, Bae JS. Inhibitory effect of polyozellin on secretory group IIA phospholipase A2. Arch Pharm Res 2015; 39:271-278. [PMID: 26659873 DOI: 10.1007/s12272-015-0694-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022]
Abstract
The expression of secretory group IIA phospholipase A2 (sPLA2-IIA) is enhanced by development of inflammatory disorders. In this study, sPLA2-IIA expression was induced in the lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells and mice to evaluate the effect of polyozellin. Polyozellin, a major constituent of a Korea edible mushroom Polyozellus multiplex, has been known to exhibit the biological activities such as anti-oxidative and anti-inflammatory effects. Polyozellin remarkably suppressed the LPS-mediated protein expression and activity of sPLA2-IIA via inhibition of phosphorylation of cytosolic phospholipase A2 and extracellular signal-regulated kinase 1/2. These results demonstrated that polyozellin might play an important role in the modulation of sPLA2-IIA expression and activity in response to the inflammatory diseases.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Hanny University, Gyeongsan, 712-715, South Korea
| | - Eun-Ju Yang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | - Hyejin Kang
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Hanny University, Gyeongsan, 712-715, South Korea
| | - Byeongjin Jung
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Hanny University, Gyeongsan, 712-715, South Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea.
| |
Collapse
|
6
|
Sriramoju B, Kanwar RK, Kanwar JR. Nanoformulated Mutant SurR9-C84A: a Possible Key for Alzheimer’s and its Associated Inflammation. Pharm Res 2015; 32:2787-97. [DOI: 10.1007/s11095-015-1664-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 03/02/2015] [Indexed: 01/25/2023]
|
7
|
Clarner T, Janssen K, Nellessen L, Stangel M, Skripuletz T, Krauspe B, Hess FM, Denecke B, Beutner C, Linnartz-Gerlach B, Neumann H, Vallières L, Amor S, Ohl K, Tenbrock K, Beyer C, Kipp M. CXCL10 Triggers Early Microglial Activation in the Cuprizone Model. THE JOURNAL OF IMMUNOLOGY 2015; 194:3400-13. [DOI: 10.4049/jimmunol.1401459] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
8
|
|
9
|
Rehal S, von der Weid PY. Experimental ileitis alters prostaglandin biosynthesis in mesenteric lymphatic and blood vessels. Prostaglandins Other Lipid Mediat 2015; 116-117:37-48. [DOI: 10.1016/j.prostaglandins.2014.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/09/2014] [Accepted: 11/11/2014] [Indexed: 01/22/2023]
|
10
|
Identifying the role of microRNAs in spinal cord injury. Neurol Sci 2014; 35:1663-71. [PMID: 25231644 DOI: 10.1007/s10072-014-1940-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 08/06/2014] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is medically and socioeconomically debilitating, and effective treatments are lacking. The elucidation of the pathophysiological mechanisms underlying SCI is essential for developing effective treatments for SCI. MicroRNAs (miRNAs) are small non-coding RNA molecules (18-24 nucleotides long) that regulate gene expression by interacting with specific target sequences. Recent studies suggest that miRNAs can act as post-transcriptional regulators to inhibit mRNA translation. Bioinformatic analyses indicate that the altered expression of miRNAs has an effect on critical processes of SCI physiopathology, including astrogliosis, oxidative stress, inflammation, apoptosis, and neuroplasticity. Therefore, the study of miRNAs may provide new insights into the molecular mechanisms of SCI. Current studies have also provided potential therapeutic clinical applications that involve targeting mRNAs to treat SCI. This review summarizes the biogenesis and function of miRNAs and the roles of miRNAs in SCI. We also discuss the potential therapeutic applications of miRNA-based interventions for SCI.
Collapse
|
11
|
Krauspe BM, Dreher W, Beyer C, Baumgartner W, Denecke B, Janssen K, Langhans CD, Clarner T, Kipp M. Short-term cuprizone feeding verifies N-acetylaspartate quantification as a marker of neurodegeneration. J Mol Neurosci 2014; 55:733-48. [PMID: 25189319 DOI: 10.1007/s12031-014-0412-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 08/20/2014] [Indexed: 01/27/2023]
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) is a quantitative MR imaging technique often used to complement conventional MR imaging with specific metabolic information. A key metabolite is the amino acid derivative N-Acetylaspartate (NAA) which is an accepted marker to measure the extent of neurodegeneration in multiple sclerosis (MS) patients. NAA is catabolized by the enzyme aspartoacylase (ASPA) which is predominantly expressed in oligodendrocytes. Since the formation of MS lesions is paralleled by oligodendrocyte loss, NAA might accumulate in the brain, and therefore, the extent of neurodegeneration might be underestimated. In the present study, we used the well-characterized cuprizone model. There, the loss of oligodendrocytes is paralleled by a reduction in ASPA expression and activity as demonstrated by genome-wide gene expression analysis and enzymatic activity assays. Notably, brain levels of NAA were not increased as determined by gas chromatography-mass spectrometry and 1H-MRS. These important findings underpin the reliability of NAA quantification as a valid marker for the paraclinical determination of the extent of neurodegeneration, even under conditions of oligodendrocyte loss in which impaired metabolization of NAA is expected. Future studies have to reveal whether other enzymes are able to metabolize NAA or whether an excess of NAA is cleared by other mechanisms rather than enzymatic metabolism.
Collapse
Affiliation(s)
- Barbara Maria Krauspe
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhao H, Luo F, Li H, Zhang L, Yi Y, Wan J. Antinociceptive effect of tetrandrine on LPS-induced hyperalgesia via the inhibition of IKKβ phosphorylation and the COX-2/PGE₂ pathway in mice. PLoS One 2014; 9:e94586. [PMID: 24722146 PMCID: PMC3983227 DOI: 10.1371/journal.pone.0094586] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/17/2014] [Indexed: 11/19/2022] Open
Abstract
Tetrandrine (TET) is a bisbenzylisoquinoline alkaloid that is isolated from the Stephania Tetrandra. It is known to possess anti-inflammatory and immunomodulatory effects. We have shown that TET can effectively suppress the production of bacterial lipopolysaccharide (LPS)-induced inflammatory mediators, including cyclooxygenases (COXs), in macrophages. However, whether TET has an antinociceptive effect on LPS-induced hyperalgesia is unknown. In the present study, we investigated the potential antinociceptive effects of TET and the mechanisms by which it elicits its effects on LPS-induced hyperalgesia. LPS effectively evoked hyperalgesia and induced the production of PGE2 in the sera, brain tissues, and cultured astroglia. TET pretreatment attenuated all of these effects. LPS also activated inhibitor of κB (IκB) kinase β (IKKβ) and its downstream components in the IκB/nuclear factor (NF)-κB signaling pathway, including COX-2; the increase in expression levels of these components was significantly abolished by TET. Furthermore, in primary astroglia, knockdown of IKKβ, but not IKKα, reversed the effects of TET on the LPS-induced increase in IκB phosphorylation, P65 phosphorylation, and COX-2. Our results suggest that TET can effectively exert antinociceptive effects on LPS-induced hyperalgesia in mice by inhibiting IKKβ phosphorylation, which leads to the reduction in the production of important pain mediators, such as PGE2 and COX-2, via the IKKβ/IκB/NF-κB pathway.
Collapse
Affiliation(s)
- Hengguang Zhao
- Department of Dermatology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fuling Luo
- Department of Pharmacy, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhong Li
- Molecular oncology and epigenetics laboratory, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongfen Yi
- Department of Pathology, Molecular Medicine and Tumor Center, Chongqing Medical University, Chongqing, China
- * E-mail: (YFY); (JYW)
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
- * E-mail: (YFY); (JYW)
| |
Collapse
|
13
|
Sierra A, Beccari S, Diaz-Aparicio I, Encinas JM, Comeau S, Tremblay MÈ. Surveillance, phagocytosis, and inflammation: how never-resting microglia influence adult hippocampal neurogenesis. Neural Plast 2014; 2014:610343. [PMID: 24772353 PMCID: PMC3977558 DOI: 10.1155/2014/610343] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/11/2014] [Indexed: 12/27/2022] Open
Abstract
Microglia cells are the major orchestrator of the brain inflammatory response. As such, they are traditionally studied in various contexts of trauma, injury, and disease, where they are well-known for regulating a wide range of physiological processes by their release of proinflammatory cytokines, reactive oxygen species, and trophic factors, among other crucial mediators. In the last few years, however, this classical view of microglia was challenged by a series of discoveries showing their active and positive contribution to normal brain functions. In light of these discoveries, surveillant microglia are now emerging as an important effector of cellular plasticity in the healthy brain, alongside astrocytes and other types of inflammatory cells. Here, we will review the roles of microglia in adult hippocampal neurogenesis and their regulation by inflammation during chronic stress, aging, and neurodegenerative diseases, with a particular emphasis on their underlying molecular mechanisms and their functional consequences for learning and memory.
Collapse
Affiliation(s)
- Amanda Sierra
- Ikerbasque Foundation, 48011 Bilbao, Spain
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 48170 Zamudio, Spain
- Department of Neurosciences, University of the Basque Country, 48940 Leioa, Spain
| | - Sol Beccari
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 48170 Zamudio, Spain
- Department of Neurosciences, University of the Basque Country, 48940 Leioa, Spain
| | - Irune Diaz-Aparicio
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 48170 Zamudio, Spain
- Department of Neurosciences, University of the Basque Country, 48940 Leioa, Spain
| | - Juan M. Encinas
- Ikerbasque Foundation, 48011 Bilbao, Spain
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 48170 Zamudio, Spain
- Department of Neurosciences, University of the Basque Country, 48940 Leioa, Spain
| | - Samuel Comeau
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Canada G1P 4C7
- Département de Médecine Moléculaire, Université Laval, Canada G1V 4G2
| | - Marie-Ève Tremblay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Canada G1P 4C7
- Département de Médecine Moléculaire, Université Laval, Canada G1V 4G2
| |
Collapse
|
14
|
Jantas D, Krawczyk S, Lason W. The predominant protective effect of tianeptine over other antidepressants in models of neuronal apoptosis: the effect blocked by inhibitors of MAPK/ERK1/2 and PI3-K/Akt pathways. Neurotox Res 2013; 25:208-25. [PMID: 24105645 PMCID: PMC3889694 DOI: 10.1007/s12640-013-9430-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 08/24/2013] [Accepted: 09/25/2013] [Indexed: 11/25/2022]
Abstract
Tianeptine (Tian) possesses neuroprotective potential, however, little is known about the effect of this drug in models of neuronal apoptosis. In the present study, we aimed (1) to compare the neuroprotective capacities of some antidepressants (ADs) in the models of staurosporine (St)- and doxorubicin (Dox)-evoked cell death, activating the intracellular and the extracellular apoptotic pathway, respectively; (2) to identify the Tian-modulated steps underlying its neuroprotective action; (3) to test the effect of various ADs against Dox-evoked cell damage in glia cells. Primary neuronal and glia cell cultures and retinoic acid-differentiated human neuroblastoma SH-SY5Y (RA-SH-SY5Y) cells were co-treated with imipramine, fluoxetine, citalopram, reboxetine, mirtazapine or Tian and St or Dox. The data showed the predominant neuroprotective effect of Tian over other tested ADs against St- and Dox-induced cell damage in primary neurons and in RA-SH-SY5Y cells. This effect was shown to be caspase-3-independent but connected with attenuation of DNA fragmentation. Moreover, neuroprotection elicited by Tian was blocked by pharmacological inhibitors of MAPK/ERK1/2 and PI3-K/Akt signaling pathways as well by inhibitor of necroptosis, necrostatin-1. Interestingly, the protective effects of all tested ADs were demonstrated in primary glia cells against the Dox-evoked cell damage. The obtained data suggests the glial cells as a common target for protective action of various ADs whereas in relation to neuronal cells only Tian possesses such properties, at least against St- and Dox-induced cell damage. Moreover, this neuroprotective effect of Tian is caspase-3-independent and engages the regulation of survival pathways (MAPK/ERK1/2 and PI3-K/Akt).
Collapse
Affiliation(s)
- D Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343, Kraków, PL, Poland,
| | | | | |
Collapse
|
15
|
Xiang Y, Chen L, Liu H, Liu X, Wei X, Sun B, Wang T, Zhang X. Inhibition of sPLA₂-IIA prevents LPS-induced neuroinflammation by suppressing ERK1/2-cPLA₂α pathway in mice cerebral cortex. PLoS One 2013; 8:e77909. [PMID: 24130900 PMCID: PMC3793966 DOI: 10.1371/journal.pone.0077909] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 09/16/2013] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammation is involved in various central nervous system (CNS) disorders, including brain infections, ischemia, trauma, stroke, and degenerative CNS diseases. In the CNS inflammation, secretory phospholipase A₂-IIA (sPLA₂-IIA) acts as a mediator, resulting in the generation of the precursors of pro-inflammatory lipid mediators, such as prostaglandins (PGs) and leukotrienes (LTs). However, the role of sPLA₂-IIA in neuroinflammation is more complicated and remains unclear yet. In the present study, we investigated the effect of sPLA₂-IIA inhibition by specific inhibitor SC-215 on the inflammation in LPS-induced mice cerebral cortex and primary astrocytes. Our results showed that the inhibition of sPLA₂-IIA alleviated the release of PGE₂ by suppressing the activation of ERK1/2, cPLA₂α, COX-2 and mPGES-1. These findings demonstrated that sPLA₂-IIA showed the potential to regulate the neuroinflammation in vivo and in vitro, indicating that sPLA₂-IIA might be a novel target for the treatment of acute neuroinflammation.
Collapse
Affiliation(s)
- Yanxiao Xiang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Lin Chen
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Huiqing Liu
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Xiaoqian Liu
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Xinbing Wei
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Baozhu Sun
- Department of Anesthesiology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Tian Wang
- Department of Pharmacology, Yantai University School of Pharmacy, Yantai, Shandong, PR China
| | - Xiumei Zhang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
- * E-mail:
| |
Collapse
|
16
|
Azami Tameh A, Clarner T, Beyer C, Atlasi MA, Hassanzadeh G, Naderian H. Regional regulation of glutamate signaling during cuprizone-induced demyelination in the brain. Ann Anat 2013; 195:415-23. [PMID: 23711509 DOI: 10.1016/j.aanat.2013.03.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 02/25/2013] [Accepted: 03/18/2013] [Indexed: 12/14/2022]
Abstract
Glutamate excitotoxicity is associated with a wide range of neurodegenerative disorders and also seems to be involved in the pathology of demyelinating disorders such as multiple sclerosis (MS). Cuprizone-induced toxic demyelination shows clear characteristics of MS such as demyelination and axonal damage without the involvement of the innate immune system. In this study, we have evaluated glutamate signaling during cuprizone-induced demyelination in the white and gray matter of mouse brain by studying the expression of ionotropic and metabotropic glutamate-receptors and -transporters by Affymetrix gene array analysis, followed by real-time PCR and western blot analysis. Cellular localization of glutamate transporters was investigated by fluorescence double-labeling experiments. Comparing white and gray matter areas, the expression of glutamate receptors was region-specific. Among NMDA receptor subunits, NR2A was up-regulated in the demyelinated corpus callosum (CC), whereas the metabotropic glutamate receptor mGluR2 was down-regulated in demyelinated gray matter. Glutamate-aspartate transporter (GLAST) co-localizing with GFAP(+) astrocytes was increased in both demyelinated CC and telencephalic cortex, whereas Slc1a4 transporter was up-regulated only in CC. Our data indicate that cuprizone treatment affects glutamate-receptors and -transporters differently in gray and white matter brain areas revealing particularly regulation of GLAST and Slc1a4 compared with other genes. This might have an important influence on brain-region selective sensitivity to neurotoxic compounds and the progression of demyelination as has been reported for MS and other demyelinating neurological diseases.
Collapse
Affiliation(s)
- Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran; Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | | | | | | | | | | |
Collapse
|
17
|
Brisevac D, Bjelobaba I, Bajic A, Clarner T, Stojiljkovic M, Beyer C, Andjus P, Kipp M, Nedeljkovic N. Regulation of ecto-5′-nucleotidase (CD73) in cultured cortical astrocytes by different inflammatory factors. Neurochem Int 2012; 61:681-8. [DOI: 10.1016/j.neuint.2012.06.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Revised: 06/12/2012] [Accepted: 06/21/2012] [Indexed: 12/16/2022]
|
18
|
Font-Nieves M, Sans-Fons MG, Gorina R, Bonfill-Teixidor E, Salas-Pérdomo A, Márquez-Kisinousky L, Santalucia T, Planas AM. Induction of COX-2 enzyme and down-regulation of COX-1 expression by lipopolysaccharide (LPS) control prostaglandin E2 production in astrocytes. J Biol Chem 2012; 287:6454-68. [PMID: 22219191 DOI: 10.1074/jbc.m111.327874] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Pathological conditions and pro-inflammatory stimuli in the brain induce cyclooxygenase-2 (COX-2), a key enzyme in arachidonic acid metabolism mediating the production of prostanoids that, among other actions, have strong vasoactive properties. Although low basal cerebral COX-2 expression has been reported, COX-2 is strongly induced by pro-inflammatory challenges, whereas COX-1 is constitutively expressed. However, the contribution of these enzymes in prostanoid formation varies depending on the stimuli and cell type. Astrocyte feet surround cerebral microvessels and release molecules that can trigger vascular responses. Here, we investigate the regulation of COX-2 induction and its role in prostanoid generation after a pro-inflammatory challenge with the bacterial lipopolysaccharide (LPS) in astroglia. Intracerebral administration of LPS in rodents induced strong COX-2 expression mainly in astroglia and microglia, whereas COX-1 expression was predominant in microglia and did not increase. In cultured astrocytes, LPS strongly induced COX-2 and microsomal prostaglandin-E(2) (PGE(2)) synthase-1, mediated by the MyD88-dependent NFκB pathway and influenced by mitogen-activated protein kinase pathways. Studies in COX-deficient cells and using COX inhibitors demonstrated that COX-2 mediated the high production of PGE(2) and, to a lesser extent, other prostanoids after LPS. In contrast, LPS down-regulated COX-1 in an MyD88-dependent fashion, and COX-1 deficiency increased PGE(2) production after LPS. The results show that astrocytes respond to LPS by a COX-2-dependent production of prostanoids, mainly vasoactive PGE(2), and suggest that the coordinated down-regulation of COX-1 facilitates PGE(2) production after TLR-4 activation. These effects might induce cerebral blood flow responses to brain inflammation.
Collapse
Affiliation(s)
- Miriam Font-Nieves
- Department of Brain Ischemia and Neurodegeneration, Institute for Biomedical Research of Barcelona, Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
19
|
BLBP-expression in astrocytes during experimental demyelination and in human multiple sclerosis lesions. Brain Behav Immun 2011; 25:1554-68. [PMID: 21620951 DOI: 10.1016/j.bbi.2011.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/11/2011] [Accepted: 05/12/2011] [Indexed: 12/18/2022] Open
Abstract
Several lines of evidence indicate that remyelination represents one of the most effective mechanisms to achieve axonal protection. For reasons that are not yet understood, this process is often incomplete or fails in multiple sclerosis (MS). Activated astrocytes appear to be able to boost or inhibit endogenous repair processes. A better understanding of remyelination in MS and possible reasons for its failure is needed. Using the well-established toxic demyelination cuprizone model, we created lesions with either robust or impaired endogenous remyelination capacity. Lesions were analyzed for mRNA expression levels by Affymetrix GeneChip® arrays. One finding was the predominance of immune and stress response factors in the group of genes which were classified as remyelination-supporting factors. We further demonstrate that lesions with impaired remyelination capacity show weak expression of the radial-glia cell marker brain lipid binding protein (BLBP, also called B-FABP or FABP7). The expression of BLBP in activated astrocytes correlates with the presence of oligodendrocyte progenitor cells. BLBP-expressing astrocytes are also detected in experimental autoimmune encephalomyelitis during the remission phase. Furthermore, highest numbers of BLBP-expressing astrocytes were evident in lesions of early MS, whereas significantly less are present at the rim of (chronic)-active lesions from patients with long disease duration. Transfection experiments show that BLBP regulates growth factor expression in U87 astrocytoma cells. In conclusion, we provide evidence that expression of BLBP in activated astrocytes negatively correlates with disease duration and in parallel with remyelination failure.
Collapse
|
20
|
Kipp M, Clarner T, Gingele S, Pott F, Amor S, van der Valk P, Beyer C. Brain lipid binding protein (FABP7) as modulator of astrocyte function. Physiol Res 2011; 60:S49-60. [PMID: 21777034 DOI: 10.33549/physiolres.932168] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Over a century ago, hyperplasia and hypertrophy of astrocytes was noted as a histopathological hallmark of multiple sclerosis and was hypothesized to play an important role in the development and course of this disease. However until today, the factual contribution of astrocytes to multiple sclerosis is elusive. Astrocytes may play an active role during degeneration and demyelination by controlling local inflammation in the CNS, provoking damage of oligodendrocytes and axons, and glial scarring but might also be beneficial by creating a permissive environment for remyelination and oligodendrocyte precursor migration, proliferation, and differentiation. Recent findings from our lab suggest that brain lipid binding protein (FABP7) is implicated in the course of multiple sclerosis and the regulation of astrocyte function. The relevance of our findings and data from other groups are highlighted and discussed in this paper in the context of myelin repair.
Collapse
Affiliation(s)
- M Kipp
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | | | | | | | | | | | | |
Collapse
|
21
|
Huxtable AG, Vinit S, Windelborn JA, Crader SM, Guenther CH, Watters JJ, Mitchell GS. Systemic inflammation impairs respiratory chemoreflexes and plasticity. Respir Physiol Neurobiol 2011; 178:482-9. [PMID: 21729770 DOI: 10.1016/j.resp.2011.06.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 06/20/2011] [Accepted: 06/21/2011] [Indexed: 11/18/2022]
Abstract
Many lung and central nervous system disorders require robust and appropriate physiological responses to assure adequate breathing. Factors undermining the efficacy of ventilatory control will diminish the ability to compensate for pathology, threatening life itself. Although most of these same disorders are associated with systemic and/or neuroinflammation, and inflammation affects neural function, we are only beginning to understand interactions between inflammation and any aspect of ventilatory control (e.g. sensory receptors, rhythm generation, chemoreflexes, plasticity). Here we review available evidence, and present limited new data suggesting that systemic (or neural) inflammation impairs two key elements of ventilatory control: chemoreflexes and respiratory motor (versus sensory) plasticity. Achieving an understanding of mechanisms whereby inflammation undermines ventilatory control is fundamental since inflammation may diminish the capacity for natural, compensatory responses during pathological states, and the ability to harness respiratory plasticity as a therapeutic strategy in the treatment of devastating breathing disorders, such as during cervical spinal injury or motor neuron disease.
Collapse
Affiliation(s)
- A G Huxtable
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706, United States
| | | | | | | | | | | | | |
Collapse
|
22
|
Gonadal steroids prevent cell damage and stimulate behavioral recovery after transient middle cerebral artery occlusion in male and female rats. Brain Behav Immun 2011; 25:715-26. [PMID: 21277368 DOI: 10.1016/j.bbi.2011.01.013] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 01/12/2011] [Accepted: 01/21/2011] [Indexed: 12/14/2022] Open
Abstract
17β-estradiol (E) and progesterone (P) are neuroprotective factors in the brain preventing neuronal death under different injury paradigms. Our previous work demonstrates that both steroids compensate neuronal damage and activate distinct neuroprotective strategies such as improving local energy metabolism and abating pro-inflammatory responses. The current study explored steroid hormone-mediated protection from brain damage and restoration of behavioral function after 1h transient middle cerebral artery occlusion (tMCAO). Male and ovariectomized female rats were studied 24h after stroke. Both steroid hormones reduced the cortical infarct area in males and females to a similar extent. A maximum effect of ~60-70% reduction of the infarct size was evident after P and a combined treatment with both hormones. No infarct protection was seen in the basal ganglia. Testing of motor and sensory behavioral revealed an equal high degree of functional recovery in all three hormone groups. Gene expression studies in the delineated penumbra revealed that estrogen receptor (ER) alpha and beta are locally up-regulated. tMCAO-mediated induction of the pro-inflammatory chemokines CCL2, CCL5 and interleukin 6 was attenuated by E and P, whereas the expression of vascular endothelial growth factor (VEGF) was fortified. Local expression of microglia/macrophage/lymphocyte markers, i.e. Iba1, CD68 and CD3, were significantly reduced in the penumbra after hormone treatment suggesting attenuation of microglia and lymphocyte attraction. These results demonstrate the neuroprotective potency of a combined treatment with E and P under ischemic conditions in both sexes and point at the regulation of chemokine-microglia/lymphocyte interactions as a supposable mechanism implicated in cell protection.
Collapse
|
23
|
Mor E, Cabilly Y, Goldshmit Y, Zalts H, Modai S, Edry L, Elroy-Stein O, Shomron N. Species-specific microRNA roles elucidated following astrocyte activation. Nucleic Acids Res 2011; 39:3710-23. [PMID: 21247879 PMCID: PMC3089466 DOI: 10.1093/nar/gkq1325] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 11/14/2010] [Accepted: 11/14/2010] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that play a central role in regulation of gene expression by binding to target genes. Many miRNAs were associated with the function of the central nervous system (CNS) in health and disease. Astrocytes are the CNS most abundant glia cells, providing support by maintaining homeostasis and by regulating neuronal signaling, survival and synaptic plasticity. Astrocytes play a key role in repair of brain insults, as part of local immune reactivity triggered by inflammatory or pathological conditions. Thus, astrocyte activation, or astrogliosis, is an important outcome of the innate immune response, which can be elicited by endotoxins such as lipopolysaccharide (LPS) and cytokines such as interferon-gamma (IFN-γ). The involvement of miRNAs in inflammation and stress led us to hypothesize that astrogliosis is mediated by miRNA function. In this study, we compared the miRNA regulatory layer expressed in primary cultured astrocyte derived from rodents (mice) and primates (marmosets) brains upon exposure to LPS and IFN-γ. We identified subsets of differentially expressed miRNAs some of which are shared with other immunological related systems while others, surprisingly, are mouse and rat specific. Of interest, these specific miRNAs regulate genes involved in the tumor necrosis factor-alpha (TNF-α) signaling pathway, indicating a miRNA-based species-specific regulation. Our data suggests that miRNA function is more significant in the mechanisms governing astrocyte activation in rodents compared to primates.
Collapse
Affiliation(s)
- Eyal Mor
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Yuval Cabilly
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Yona Goldshmit
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Harel Zalts
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Shira Modai
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Liat Edry
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Orna Elroy-Stein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel and Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
24
|
Sergeeva MG, Aleshin SE, Grabeklis S, Reiser G. PPAR activation has dichotomous control on the expression levels of cytosolic and secretory phospholipase A2 in astrocytes; inhibition in naïve, untreated cells and enhancement in LPS-stimulated cells. J Neurochem 2010; 115:399-410. [PMID: 20670373 DOI: 10.1111/j.1471-4159.2010.06931.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the importance of cytosolic phospholipase A(2) type IVA (cPLA(2)) and secretory PLA(2) (sPLA(2)) in physiological and pathological responses of astrocytes in inflammatory conditions, the regulation of the expression of these genes is still unclear. Both genes have peroxisome proliferator-activated receptors (PPAR) binding sites in their promoters. The role of synthetic PPAR agonists in the regulation of gene expression in naïve and lipopolysaccharide (LPS)-stimulated rat astrocytes in culture was investigated. Exposure to LPS resulted in a time-dependent, fourfold transient increase of sPLA(2) expression, with maximum at 4 h; cPLA(2) expression was notably increased after 16-h LPS stimulation. Using selective PPARα, PPARβ/δ, and PPARγ agonists, we found that expression of both cPLA(2) and sPLA(2) is under PPAR control, but with different isotypes sensitivity. In naïve astrocytes, all three PPAR agonists significantly suppressed the expression of sPLA(2), while only PPARα and PPARγ activation suppressed cPLA(2) expression. Astonishingly, simultaneous addition of LPS with PPAR agonists evoked the opposite effect. All three PPAR agonists induced potentiation of cPLA(2) expression level. Potentiation of sPLA(2) expression was induced only by simultaneous addition of LPS with PPARγ agonist. By knockdown of PPARα, PPARβ/δ, and PPARγ, we confirmed the involvement of PPAR-dependent pathways. The important novelty of our findings is that both sPLA(2) and cPLA(2) are under dichotomous control of PPARs: suppression in naïve control cells, but induction in LPS-stimulated astrocytes.
Collapse
Affiliation(s)
- Marina G Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | | | | | | |
Collapse
|
25
|
Fuller S, Steele M, Münch G. Activated astroglia during chronic inflammation in Alzheimer's disease--do they neglect their neurosupportive roles? Mutat Res 2010; 690:40-49. [PMID: 19748514 DOI: 10.1016/j.mrfmmm.2009.08.016] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 08/08/2009] [Accepted: 08/15/2009] [Indexed: 05/28/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, characterized histopathologically by the extracellular deposition of beta-amyloid peptide in senile plaques, as well as intracellular neurofibrillary tangles (NFT) of hyperphosphorylated tau protein, extensive neuronal loss and synaptic changes in the hippocampus and cerebral cortex. In addition, the AD brain shows chronic inflammation characterized by an abundance of reactive astrocytes and activated microglia. In the healthy brain, astrocytes provide essential services for brain homeostasis and neuronal function, including metabolic support for neurons in the form of lactate, glutamate uptake and conversion into glutamine, and synthesis of glutathione and its precursors. In AD, a large body of evidence now suggests that by transforming from a basal to a reactive state, astrocytes neglect their neurosupportive functions, thus rendering neurons vulnerable to neurotoxins including pro-inflammatory cytokines and reactive oxygen species. This review will explain the normal functions of astrocytes, and how these cells might be activated to turn into inflammatory cells, actively contributing to neurodegeneration and neglecting their neurosupportive roles ("neuro-neglect hypothesis"). Furthermore, it is proposed that astrocytes might be promising target of therapeutic intervention for Alzheimer's disease, if these compromised functions can be normalized with pharmacological agents that are specifically designed to return astrocytes to a quiescent phenotype or supplement factors which activated astrocytes lack to produce.
Collapse
Affiliation(s)
- Stacey Fuller
- Department of Pharmacology, School of Medicine, University of Western Sydney, Campbelltown, Australia
| | | | | |
Collapse
|
26
|
van Neerven S, Regen T, Wolf D, Nemes A, Johann S, Beyer C, Hanisch UK, Mey J. Inflammatory chemokine release of astrocytes in vitro is reduced by all-trans retinoic acid. J Neurochem 2010; 114:1511-26. [PMID: 20557428 DOI: 10.1111/j.1471-4159.2010.06867.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The production of chemokines by astrocytes constitutes an important component of neuroinflammatory processes in the brain. As the transcriptional activator retinoic acid (RA), used for chemotherapy and dermatological applications, exerts anti-inflammatory effects on monocytes and lymphocytes, we have tested whether the physiologically occurring isomer, all-trans RA, affects chemokine expression by astrocytes. Under control conditions, primary cultures of murine cortical astrocytes expressed no or very low levels of CCL and CXCL chemokines. After treatment with bacterial lipopolysaccharides to simulate inflammation in vitro, we detected a strong increase in the release of CCL2 (to > 4 ng/mL in cell culture supernatant), CCL3 (> 20 ng/mL), CCL5 (> 25 ng/mL), CXCL1 (> 30 ng/mL) and CXCL2 (> 20 ng/mL). Although simultaneous exposure to RA did not significantly affect this response, 12 h pre-treatment with 0.1 microM all-trans RA strongly suppressed mRNA expression and protein release of all chemokines. The anti-inflammatory activity of RA engaged RA and retinoid X receptors and correlated with a decreased expression of the lipopolysaccharides co-receptor CD14. A minor reduction of nuclear NF-kappaB was observed but not significant, activation of Jun amino-terminal kinase, p38 and signal transducer and activator of transcription 3 were not altered by RA. The results suggest that retinoids should be further investigated as candidates for the treatment of neuroinflammation.
Collapse
|
27
|
Baertling F, Kokozidou M, Pufe T, Clarner T, Windoffer R, Wruck CJ, Brandenburg LO, Beyer C, Kipp M. ADAM12 is expressed by astrocytes during experimental demyelination. Brain Res 2010; 1326:1-14. [PMID: 20176000 DOI: 10.1016/j.brainres.2010.02.049] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 02/07/2010] [Accepted: 02/14/2010] [Indexed: 12/18/2022]
Abstract
A disintegrin and metalloproteinase (ADAM) 12 represents a member of a large family of similarly structured multi-domain proteins. In the central nervous system (CNS), ADAM12 has been suggested to play a role in brain development, glioblastoma cell proliferation, and in experimental autoimmune encephalomyelitis. Furthermore, ADAM12 was reported to be almost exclusively expressed by oligodendrocytes and could, therefore, be considered as suitable marker for this cell type. In the present study, we investigated ADAM12 expression in the healthy and pathologically altered murine CNS. As pathological paradigm, we used the cuprizone demyelination model in which myelin loss during multiple sclerosis is imitated. Besides APC(+) oligodendrocytes, SMI311(+) neurons and GFAP(+) astrocytes express ADAM12 in the adult mouse brain. ADAM12 expression was further analyzed in vitro. After the induction of demyelination, we observed that activated astrocytes are the main source of ADAM12 in brain regions affected by oligodendrocyte loss. Exposure of astrocytes in vitro to either lipopolysaccharides (LPS), tumor necrosis factor alpha (TNFalpha), glutamate, or hydrogen peroxide revealed a highly stimulus-specific regulation of ADAM12 expression which was not seen in microglial BV2 cells. It appears that LPS- and TNFalpha-induced ADAM12 expression is mediated via the classic NFkappaB pathway. In summary, we demonstrated that ADAM12 is not a suitable marker for oligodendrocytes. Our results further suggest that ADAM12 might be implicated in the course of distinct CNS diseases such as demyelinating disorders.
Collapse
Affiliation(s)
- Fabian Baertling
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Animal models of human cerebellar ataxias: a cornerstone for the therapies of the twenty-first century. THE CEREBELLUM 2009; 8:137-54. [PMID: 19669387 DOI: 10.1007/s12311-009-0127-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cerebellar ataxias represent a group of disabling neurological disorders. Our understanding of the pathogenesis of cerebellar ataxias is continuously expanding. A considerable number of laboratory animals with neurological mutations have been reported and numerous relevant animal models mimicking the phenotype of cerebellar ataxias are becoming available. These models greatly help dissecting the numerous mechanisms of cerebellar dysfunction, a major step for the assessment of therapeutics targeting a given deleterious pathway and for the screening of old or newly synthesized chemical compounds. Nevertheless, differences between animal models and human disorders should not be overlooked and difficulties in terms of characterization should not be occulted. The identification of the mutations of many hereditary ataxias, the development of valuable animal models, and the recent identifications of the molecular mechanisms underlying cerebellar disorders represent a combination of key factors for the development of anti-ataxic innovative therapies. It is anticipated that the twenty-first century will be the century of effective therapies in the field of cerebellar ataxias. The animal models are a cornerstone to reach this goal.
Collapse
|
29
|
Acs P, Kipp M, Norkute A, Johann S, Clarner T, Braun A, Berente Z, Komoly S, Beyer C. 17beta-estradiol and progesterone prevent cuprizone provoked demyelination of corpus callosum in male mice. Glia 2009; 57:807-14. [PMID: 19031445 DOI: 10.1002/glia.20806] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sex hormones, for example, estrogen and progesterone, are thought to affect and delay progression of multiple sclerosis (MS) in pregnant women. Although both steroid hormones are neuroprotective in the brain and elevated during pregnancy, only estrogen was tested in clinical trials. To evaluate the role of 17beta-estradiol (E) and progesterone (P) in prevention demyelination, young adult male mice were fed with cuprizone for a defined time interval and simultaneously treated with steroids by repeated injections into the neck region. The status of myelination was analyzed by magnetic resonance imaging and conventional histological staining. The individual application of E and P resulted only in a moderate prevention of demyelination in the corpus callosum (CC). The combined treatment with both steroid hormones counteracted the process of demyelination. Expression of the mature (PLP and MBP) and premature (PDGF-alpha-R) oligodendrocyte markers were significantly increased after hormone application in the affected CC. In addition, both hormones stimulated astrogliosis and the expression of IGF-1. Microglial invasion in demyelinated CC was pronounced and additionally localized in the midline of CC after hormone treatment. These data show that sex steroids can protect the brain from demyelination and stimulate remyelination. It appears that only the administration of both hormones is fully effective. The beneficial steroid effect requires interactions with oligodendrocytes possibly by preventing their degeneration or recruitment from precursor cells which are stimulated to remyelinated fibers. The positive hormonal influence on myelination in the CNS may be a future therapeutically strategy for the treatment of MS.
Collapse
Affiliation(s)
- Peter Acs
- Department of Neurology, Faculty of Medicine, University of Pécs, Rét u. 2, Pécs, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kipp M, Beyer C. Impact of sex steroids on neuroinflammatory processes and experimental multiple sclerosis. Front Neuroendocrinol 2009; 30:188-200. [PMID: 19393685 DOI: 10.1016/j.yfrne.2009.04.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 04/01/2009] [Accepted: 04/14/2009] [Indexed: 12/18/2022]
Abstract
Synthetic and natural estrogens as well as progestins modulate neuronal development and activity. Neurons and glia are endowed with high-affinity steroid receptors. Besides regulating brain physiology, both steroids conciliate neuroprotection against toxicity and neurodegeneration. The majority of data derive from in vitro studies, although more recently, animal models have proven the efficaciousness of steroids as neuroprotective factors. Indications for a safeguarding role also emerge from first clinical trials. Gender-specific prevalence of degenerative disorders might be associated with the loss of hormonal activity or steroid malfunctions. Our studies and evidence from the literature support the view that steroids attenuate neuroinflammation by reducing the pro-inflammatory property of astrocytes. This effect appears variable depending on the brain region and toxic condition. Both hormones can individually mediate protection, but they are more effective in cooperation. A second research line, using an animal model for multiple sclerosis, provides evidence that steroids achieve remyelination after demyelination. The underlying cellular mechanisms involve interactions with astroglia, insulin-like growth factor-1 responses, and the recruitment of oligodendrocytes.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
31
|
Palin K, Moreau ML, Sauvant J, Orcel H, Nadjar A, Duvoid-Guillou A, Dudit J, Rabié A, Moos F. Interleukin-6 activates arginine vasopressin neurons in the supraoptic nucleus during immune challenge in rats. Am J Physiol Endocrinol Metab 2009; 296:E1289-99. [PMID: 19258490 DOI: 10.1152/ajpendo.90489.2008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The increase of plasma arginin-vasopressin (AVP) release, which translates hypothalamic AVP neuron activation in response to immune challenge, appears to occur independently of plasma osmolality or blood pressure changes. Many studies have shown that major inflammatory mediators produced in response to peripheral inflammation, such as prostaglandin (PG)-E(2) and interleukin (IL)-1beta, excite AVP neurons. However, in vivo electrical activation of AVP neurons was still not assessed in relation to plasma AVP release, osmolality, or blood pressure or to the expression and role of inflammatory molecules like PG-E(2), IL-1beta, IL-6, and tumor necrosis factor-alpha (TNFalpha). This study aims at elucidating those factors that underlie the activation of AVP neurons in response to immune stimulation mimicked by an intraperitoneal injection of lipopolysaccharide (LPS) in male Wistar rats. LPS treatment concomittanlty decreased diuresis and increased plasma AVP as well as AVP neuron activity in vivo, and these effects occurred as early as 30 min. Activation was sustained for more than 6 h. Plasma osmolality did not change, whereas blood pressure only transiently increased during the first hour post-LPS. PG-E(2), IL-1beta, and TNFalpha mRNA expression were raised 3 h after LPS, whereas IL-6 mRNA level increased 30 min post-LPS. In vivo electrophysiological recordings showed that brain IL-6 injection increased AVP neuron activity similarly to peripheral LPS treatment. In contrast, brain injection of anti-IL-6 antibodies prevented the LPS induced-activation of AVP neurons. Taken together, these results suggest that the early activation of AVP neurons in response to LPS injection is induced by brain IL-6.
Collapse
Affiliation(s)
- Karine Palin
- Laboratoire PsyNuGen, Université de Bordeaux 2, CNRS UMR5226, INRA UMR1286, IFR8 Neurosciences, Bâtiment UFR de pharmacie, 146 rue Léo Saignat, Bordeaux, F-33076, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Trotter A, Hilgendorff A, Kipp M, Beyer C, Kueppers E, Kiossis E, Stuplich J, Pohlandt F, Thome U. Gender-related effects of prenatal administration of estrogen and progesterone receptor antagonists on VEGF and surfactant-proteins and on alveolarisation in the developing piglet lung. Early Hum Dev 2009; 85:353-9. [PMID: 19186013 DOI: 10.1016/j.earlhumdev.2008.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 11/16/2008] [Accepted: 12/21/2008] [Indexed: 01/09/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is essential for embryonic lung development and has been shown to be regulated by estradiol (E2) and progesterone (P). AIM To investigate the effects of prenatal E2 and P withdrawal by specific receptor antagonists on the mRNA expression of VEGF, surfactant proteins (SP-B and SP-C) and on alveolarisation in lung tissue of male and female pig fetuses. METHODS Fetuses from 10 sows were randomized to receive either both an intramuscular injection of the E2 receptor blocker ICI 182.780 and the P receptor blocker RTI 3021-022 (ICI+RTI, n=5) or a placebo injection (n=5) at 90 days of gestation (DOG, 115=term). After delivery by cesarean section on 114 DOG, tissue of the left lingula of the piglet's lung (28 placebo, 26 ICI+RTI) was obtained to determine the mRNA expression of VEGF, SP-B and SP-C. Lungs from 15 placebo and 14 ICI+RTI group piglets were removed and alveolar counts performed. RESULTS The ICI+RTI group showed significantly lower SP-C mRNA expression and alveolar counts compared to the placebo group (p=0.04 and 0.03, respectively). Diminished alveolarisation in the ICI+RTI group was mainly due to the reduction of alveolar counts in male piglets (p=0.02). Within the placebo group VEGF and SP-B mRNA expression in male piglets were significantly lower compared to female piglets (p=0.01 and 0.004, respectively). ICI+RTI treatment abolished this gender-related difference. CONCLUSION Estradiol and P antagonism affected gender-related differences of key proteins for pulmonary function and development and especially in males was associated with diminished alveolarisation.
Collapse
Affiliation(s)
- Andreas Trotter
- Section of Neonatology, Center for Perinatal Medicine, Children's Hospital, University of Bonn, Bonn, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Current World Literature. Curr Opin Neurol 2009; 22:321-9. [DOI: 10.1097/wco.0b013e32832cf9cb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Norkute A, Hieble A, Braun A, Johann S, Clarner T, Baumgartner W, Beyer C, Kipp M. Cuprizone treatment induces demyelination and astrocytosis in the mouse hippocampus. J Neurosci Res 2009; 87:1343-55. [PMID: 19021291 DOI: 10.1002/jnr.21946] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Memory impairment is outstanding within the spectrum of cognitive deficits in multiple sclerosis (MS) patients. Demyelination has been reported in the hippocampus formation of MS patients. The degree of hippocampus lesions in MS strongly correlates with progression of cognitive dysfunction. Because no appropriate animal model for the study of hippocampus demyelination has been established, we used the cuprizone mouse model to investigated demyelination in young adult and aged mice. The myelin status was analyzed by classical histological staining, immunocytochemistry for proteolipoprotein, and electron microscopy. Oligodendrocyte, astroglial, and microglia markers were studied. Cuprizone intoxication induced an almost complete demyelination of distinct hippocampus subregions to a similar extent in young adult and aged male mice. Demyelination was pronounced in a subset of white and gray matter areas, i.e., the stratum lacunosum moleculare containing the perforant path, medial alveus, stratum pyramidale in the cornu ammonis 2/3 region, and hilus region. Besides demyelination, affected areas displayed hypertrophic and hyperplastic astrocytosis. No significant effect on microglia invasion was detected at any investigated time point (0, 3, 5, and 7 weeks). We conclude that cuprizone-induced demyelination provides an adequate animal model to investigate appropriate therapy strategies for the prevention of hippocampus demyelination.
Collapse
Affiliation(s)
- Akvile Norkute
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Küppers E, Brito V, Wachter B, Wolburg H, Beyer C. Aquaporin-4 Isoform Expression in the Developing Mouse Nigro-striatal System. J Mol Neurosci 2009; 38:1. [DOI: 10.1007/s12031-008-9147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 08/27/2008] [Indexed: 11/29/2022]
|
36
|
Groebe A, Clarner T, Baumgartner W, Dang J, Beyer C, Kipp M. Cuprizone treatment induces distinct demyelination, astrocytosis, and microglia cell invasion or proliferation in the mouse cerebellum. THE CEREBELLUM 2009; 8:163-74. [PMID: 19259754 DOI: 10.1007/s12311-009-0099-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 02/11/2009] [Indexed: 10/21/2022]
Abstract
Demyelination of the cerebellum is a well-known phenomenon in human multiple sclerosis (MS). Concordantly, patients with MS frequently developed symptoms deriving from cerebellar lesions, i.e., dysmetria leading to hand dexterity impairment. Important advances in MS research have been made as a direct or indirect consequence of the establishment of adequate animal models. In this study, we used the cuprizone mouse model to investigate cerebellar demyelination in young adult male mice. The myelin status was analyzed by immunohistochemistry for proteolipoprotein and electron microscopy. The expression and presence of oligodendrocyte, astroglial, and microglia markers were supplementary studied. Cuprizone intoxication induced an almost complete demyelination of cerebellar nuclei. Cerebellar cortex regions were not (cortical gray matter) or only marginally (cortical white matter) affected. In addition, the affected areas displayed hypertrophic and hyperplastic astrocytosis accompanied by microglia or macrophage invasion. We conclude that cuprizone-induced demyelination pictures cerebellar deep gray matter involvement but not cerebellar cortex pathology as described for human MS. Behavioral changes after cuprizone described for this animal model may not only result from effects on commissural fiber tracts but also can arise from cerebellar demyelination.
Collapse
Affiliation(s)
- Angela Groebe
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Malaspina A, Jokic N, Huang WL, Priestley JV. Comparative analysis of the time-dependent functional and molecular changes in spinal cord degeneration induced by the G93A SOD1 gene mutation and by mechanical compression. BMC Genomics 2008; 9:500. [PMID: 18947433 PMCID: PMC2585103 DOI: 10.1186/1471-2164-9-500] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 10/23/2008] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Mutations of the superoxide dismutase 1 (SOD1) gene are linked to amyotrophic lateral sclerosis (ALS), an invariably fatal neurological condition involving cortico-spinal degeneration. Mechanical injury can also determine spinal cord degeneration and act as a risk factor for the development of ALS. RESULTS We have performed a comparative ontological analysis of the gene expression profiles of thoracic cord samples from rats carrying the G93A SOD1 gene mutation and from wild-type littermates subjected to mechanical compression of the spinal cord. Common molecular responses and gene expression changes unique to each experimental paradigm were evaluated against the functional development of each animal model. Gene Ontology categories crucial to protein folding, extracellular matrix and axonal formation underwent early activation in both experimental paradigms, but decreased significantly in the spinal cord from animals recovering from injury after 7 days and from the G93A SOD1 mutant rats at end-stage disease. Functional improvement after compression coincided with a massive up-regulation of growth-promoting gene categories including factors involved in angiogenesis and transcription, overcoming the more transitory surge of pro-apoptotic components and cell-cycle genes. The cord from G93A SOD1 mutants showed persistent over-expression of apoptotic and stress molecules with fewer neurorestorative signals, while functional deterioration was ongoing. CONCLUSION this study illustrates how cytoskeletal protein metabolism is central to trauma and genetically-induced spinal cord degeneration and elucidates the main molecular events accompanying functional recovery or decline in two different animal models of spinal cord degeneration.
Collapse
Affiliation(s)
- Andrea Malaspina
- Neuroscience Centre, Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, Whitechapel, London E1 2AT, UK.
| | | | | | | |
Collapse
|
38
|
Kampmann E, Johann S, van Neerven S, Beyer C, Mey J. Anti-inflammatory effect of retinoic acid on prostaglandin synthesis in cultured cortical astrocytes. J Neurochem 2008; 106:320-32. [PMID: 18394023 DOI: 10.1111/j.1471-4159.2008.05395.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prostanoids are important mediators of inflammation and pain signaling. Although it is now well accepted that astrocytes participate in inflammatory reactions in the CNS, the molecular regulation of this activity is still largely unknown. Specifically, the regulation of prostanoid synthesis by this type of glia remains to be resolved. Recent evidence suggests that the transcriptional regulator retinoic acid (RA) is involved in regulation of the immune response. We have investigated the expression pattern of the enzymes that catalyze prostanoid and leukotriene synthesis in cultured cortical astrocytes, their stimulation by lipopolysaccharides (LPS) and their regulation by RA. The data indicate that astrocytes are an important source of prostaglandins (PGs) and that RA reduces their inflammatory biosynthesis. LPS treatment induced the expression of enzymes for the production of arachidonic acid and PGs but caused down-regulation of a PG degrading enzyme and of leukotriene synthesizing enzymes that compete with PG synthesis. Consequently, the secretion of the PGE(2) was highly increased after LPS exposure. RA counteracted the inflammatory regulation of cyclooxygenase (COX)-2 mRNA and protein in astrocytes and thereby reduced the synthesis of PGE(2) by approximately 60%. In the absence of LPS, RA enhanced the expression of COX-1 mRNA. In conclusion, RA might be effective in suppressing inflammatory processes in the brain by inhibiting PG synthesis.
Collapse
|
39
|
Brain-region-specific astroglial responses in vitro after LPS exposure. J Mol Neurosci 2008; 35:235-43. [PMID: 18373222 DOI: 10.1007/s12031-008-9057-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2008] [Accepted: 02/20/2008] [Indexed: 12/31/2022]
Abstract
Astroglia is well-known to be integrated in the complex regulation of neuroinflammation in the central nervous system. Astrocytes become activated and synthesize cytokines, chemokines, and prostanoids during degenerative and vulnerable processes and interact with other immune-competent cells. Degenerative disorders often occur in a brain-region-specific fashion suggesting differences in the activity and reactivity of innate immune cells. We have investigated the potency of lipopolysaccharides (LPS) to differently stimulate astrocytes from the cortex and midbrain. Astroglial cultures were prepared from Bagg albino/c mice and exposed to LPS. Astrocytes from both brain areas already differed in their capacity and profile of cytokine expression under basal unstimulated conditions. In response to LPS, we observed both a region-specific pattern of up-regulation of distinct cytokines and differences in the extent and time-course of activation. Our data demonstrate that astrocytes reveal a region-specific basal profile of cytokine expression and a selective area-specific regulation of cytokines upon LPS-induced inflammation. This makes astrocytes likely candidates to be responsible for region-specific incidence rates of neurological and neurodegenerative disorders.
Collapse
|