1
|
Saenz‐Antoñanzas A, Muñoz‐Culla M, Rigo P, Ruiz‐Barreiro L, Moreno‐Valladares M, Alberro A, Cruces‐Salguero S, Arroyo‐Izaga M, Arranz AM, Otaegui D, Guillemot F, Matheu A. Centenarian hippocampus displays high levels of astrocytic metallothioneins. Aging Cell 2024; 23:e14201. [PMID: 38769809 PMCID: PMC11320342 DOI: 10.1111/acel.14201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/10/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024] Open
Abstract
The hippocampus is a brain area linked to cognition. The mechanisms that maintain cognitive activity in humans are poorly understood. Centenarians display extreme longevity which is generally accompanied by better quality of life, lower cognitive impairment, and reduced incidence of pathologies including neurodegenerative diseases. We performed transcriptomic studies in hippocampus samples from individuals of different ages (centenarians [≥97 years], old, and young) and identified a differential gene expression pattern in centenarians compared to the other two groups. In particular, several isoforms of metallothioneins (MTs) were highly expressed in centenarians. Moreover, we identified that MTs were mainly expressed in astrocytes. Functional studies in human primary astrocytes revealed that MT1 and MT3 are necessary for their homeostasis maintenance. Overall, these results indicate that the expression of MTs specifically in astrocytes is a mechanism for protection during aging.
Collapse
Affiliation(s)
| | - Maider Muñoz‐Culla
- Multiple Sclerosis GroupBiodonostia Health Research InstituteSan SebastianSpain
- CIBERNED, ISCIIIMadridSpain
- Department of Basic Psychological Processes and their DevelopmentUniversity of the Basque Country (UPV/EHU)San SebastianSpain
| | - Piero Rigo
- Neural Stem Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Leire Ruiz‐Barreiro
- Laboratory of Humanized Models of Disease, Achucarro Basque Center for NeuroscienceLeioaSpain
| | | | - Ainhoa Alberro
- Multiple Sclerosis GroupBiodonostia Health Research InstituteSan SebastianSpain
- CIBERNED, ISCIIIMadridSpain
| | | | - Marta Arroyo‐Izaga
- BIOMICs Research Group, Microfluidics & BIOMICs, Department of Pharmacy and Food Sciences, Lascaray Research CenterUniversity of the Basque Country (UPV/EHU), BioarabaVitoriaSpain
| | - Amaia M. Arranz
- Laboratory of Humanized Models of Disease, Achucarro Basque Center for NeuroscienceLeioaSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - David Otaegui
- Multiple Sclerosis GroupBiodonostia Health Research InstituteSan SebastianSpain
- CIBERNED, ISCIIIMadridSpain
| | | | - Ander Matheu
- Cellular Oncology GroupBiodonostia Health Research InstituteSan SebastianSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
- CIBERFES, ISCIIIMadridSpain
| |
Collapse
|
2
|
Sanchis P, Fernández‐Gayol O, Vizueta J, Comes G, Canal C, Escrig A, Molinero A, Giralt M, Hidalgo J. Microglial cell‐derived interleukin‐6 influences behavior and inflammatory response in the brain following traumatic brain injury. Glia 2019; 68:999-1016. [DOI: 10.1002/glia.23758] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Paula Sanchis
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Olaya Fernández‐Gayol
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Joel Vizueta
- Departament de Genètica, Microbiologia i Estadística and Institut de Recerca de la Biodiversitat (IRBio), Facultat de BiologiaUniversitat de Barcelona Barcelona Spain
| | - Gemma Comes
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Carla Canal
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Anna Escrig
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Amalia Molinero
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Mercedes Giralt
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| | - Juan Hidalgo
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of BiosciencesUniversitat Autònoma de Barcelona Barcelona Spain
| |
Collapse
|
3
|
Izzy S, Liu Q, Fang Z, Lule S, Wu L, Chung JY, Sarro-Schwartz A, Brown-Whalen A, Perner C, Hickman SE, Kaplan DL, Patsopoulos NA, El Khoury J, Whalen MJ. Time-Dependent Changes in Microglia Transcriptional Networks Following Traumatic Brain Injury. Front Cell Neurosci 2019; 13:307. [PMID: 31440141 PMCID: PMC6694299 DOI: 10.3389/fncel.2019.00307] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
The neuroinflammatory response to traumatic brain injury (TBI) is critical to both neurotoxicity and neuroprotection, and has been proposed as a potentially modifiable driver of secondary injury in animal and human studies. Attempts to broadly target immune activation have been unsuccessful in improving outcomes, in part because the precise cellular and molecular mechanisms driving injury and outcome at acute, subacute, and chronic time points after TBI remain poorly defined. Microglia play a critical role in neuroinflammation and their persistent activation may contribute to long-term functional deficits. Activated microglia are characterized by morphological transformation and transcriptomic changes associated with specific inflammatory states. We analyzed the temporal course of changes in inflammatory genes of microglia isolated from injured brains at 2, 14, and 60 days after controlled cortical impact (CCI) in mice, a well-established model of focal cerebral contusion. We identified a time dependent, injury-associated change in the microglial gene expression profile toward a reduced ability to sense tissue damage, perform housekeeping, and maintain homeostasis in the early stages following CCI, with recovery and transition to a specialized inflammatory state over time. This later state starts at 14 days post-injury and is characterized by a biphasic pattern of IFNγ, IL-4, and IL-10 gene expression changes, with concurrent proinflammatory and anti-inflammatory gene changes. Our transcriptomic data sets are an important step to understand microglial role in TBI pathogenesis at the molecular level and identify common pathways that affect outcome. More studies to evaluate gene expression at the single cell level and focusing on subacute and chronic timepoint are warranted.
Collapse
Affiliation(s)
- Saef Izzy
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Qiong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai, China
| | - Zhou Fang
- Harvard Medical School, Boston, MA, United States.,Systems Biology and Computer Science Program, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sevda Lule
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Limin Wu
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Joon Yong Chung
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Aliyah Sarro-Schwartz
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Alexander Brown-Whalen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Caroline Perner
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Suzanne E Hickman
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Nikolaos A Patsopoulos
- Harvard Medical School, Boston, MA, United States.,Systems Biology and Computer Science Program, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joseph El Khoury
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Michael J Whalen
- Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Harvard Medical School, Boston, MA, United States.,Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Fernández-Gayol O, Sanchis P, Aguilar K, Navarro-Sempere A, Comes G, Molinero A, Giralt M, Hidalgo J. Different Responses to a High-Fat Diet in IL-6 Conditional Knockout Mice Driven by Constitutive GFAP-Cre and Synapsin 1-Cre Expression. Neuroendocrinology 2019; 109:113-130. [PMID: 30636247 DOI: 10.1159/000496845] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/12/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Interleukin-6 (IL-6) is a major cytokine controlling body weight and metabolism, at least in part through actions in the central nervous system (CNS) from local sources. METHODS We herewith report results obtained in conditional IL-6 KO mice for brain cells (Il6ΔGfap and Il6ΔSyn). RESULTS The reporter RiboTag mouse line demonstrated specific astrocytic expression of GFAP-dependent Cre in the hypothalamus but not in other brain areas, whereas that of synapsin 1-dependent Cre was specific for neurons. Feeding a high-fat diet (HFD) or a control diet showed that Il6ΔGfap and Il6ΔSyn mice were more prone and resistant, respectively, to HFD-induced obesity. Energy intake was not altered in HFD experiments, but it was reduced in Il6ΔSyn male mice following a 24-h fast. HFD increased circulating insulin, leptin, and cholesterol levels, decreased triglycerides, and caused impaired responses to the insulin and glucose tolerance tests. In Il6ΔGfap mice, the only significant difference observed was an increase in insulin levels of females, whereas in Il6ΔSyn mice the effects of HFD were decreased. Hypothalamic Agrp expression was significantly decreased by HFD, further decreased in Il6ΔGfap, and increased in Il6ΔSyn female mice. Hypothalamic Il-6 mRNA levels were not decreased in Il6ΔSyn mice and even increased in Il6ΔGfapmale mice. Microarray analysis of hypothalamic RNA showed that female Il6ΔGfap mice had increased interferon-related pathways and affected processes in behavior, modulation of chemical synaptic transmission, learning, and memory. CONCLUSION The present results demonstrate that brain production of IL-6 regulates body weight in the context of caloric excess and that the cellular source is critical.
Collapse
Affiliation(s)
- Olaya Fernández-Gayol
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Paula Sanchis
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kevin Aguilar
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alicia Navarro-Sempere
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Comes
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Amalia Molinero
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mercedes Giralt
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Hidalgo
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain,
| |
Collapse
|
5
|
Effect of Metallothionein-III on Mercury-Induced Chemokine Gene Expression. TOXICS 2018; 6:toxics6030048. [PMID: 30103553 PMCID: PMC6161308 DOI: 10.3390/toxics6030048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 11/16/2022]
Abstract
Mercury compounds are known to cause central nervous system disorders; however the detailed molecular mechanisms of their actions remain unclear. Methylmercury increases the expression of several chemokine genes, specifically in the brain, while metallothionein-III (MT-III) has a protective role against various brain diseases. In this study, we investigated the involvement of MT-III in chemokine gene expression changes in response to methylmercury and mercury vapor in the cerebrum and cerebellum of wild-type mice and MT-III null mice. No difference in mercury concentration was observed between the wild-type mice and MT-III null mice in any brain tissue examined. The expression of Ccl3 in the cerebrum and of Cxcl10 in the cerebellum was increased by methylmercury in the MT-III null but not the wild-type mice. The expression of Ccl7 in the cerebellum was increased by mercury vapor in the MT-III null mice but not the wild-type mice. However, the expression of Ccl12 and Cxcl12 was increased in the cerebrum by methylmercury only in the wild-type mice and the expression of Ccl3 in the cerebellum was increased by mercury vapor only in the wild-type mice. These results indicate that MT-III does not affect mercury accumulation in the brain, but that it affects the expression of some chemokine genes in response to mercury compounds.
Collapse
|
6
|
Baek A, Cho SR, Kim SH. Elucidation of Gene Expression Patterns in the Brain after Spinal Cord Injury. Cell Transplant 2018; 26:1286-1300. [PMID: 28933220 PMCID: PMC5657738 DOI: 10.1177/0963689717715822] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological disease. The pathophysiological mechanisms of SCI have been reported to be relevant to central nervous system injury such as brain injury. In this study, gene expression of the brain after SCI was elucidated using transcriptome analysis to characterize the temporal changes in global gene expression patterns in a SCI mouse model. Subjects were randomly classified into 3 groups: sham control, acute (3 h post-injury), and subacute (2 wk post-injury) groups. We sought to confirm the genes differentially expressed between post-injured groups and sham control group. Therefore, we performed transcriptome analysis to investigate the enriched pathways associated with pathophysiology of the brain after SCI using Database for Annotation Visualization, and Integrated Discovery (DAVID), which yielded Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. Following enriched pathways were found in the brain: oxidative phosphorylation pathway; inflammatory response pathways—cytokine–cytokine receptor interaction and chemokine signaling pathway; and endoplasmic reticulum (ER) stress-related pathways—antigen processing and presentation and mitogen-activated protein kinase signaling pathway. Oxidative phosphorylation pathway was identified at acute phase, while inflammation response and ER stress-related pathways were identified at subacute phase. Since the following pathways—oxidative phosphorylation pathway, inflammatory response pathways, and ER stress-related pathways—have been well known in the SCI, we suggested a link between SCI and brain injury. These mechanisms provide valuable reference data for better understanding pathophysiological processes in the brain after SCI.
Collapse
Affiliation(s)
- Ahreum Baek
- 1 Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea.,2 Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Rae Cho
- 2 Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,5 Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Hoon Kim
- 1 Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| |
Collapse
|
7
|
Nielsen AE, Bohr A, Penkowa M. The Balance between Life and Death of Cells: Roles of Metallothioneins. Biomark Insights 2017. [DOI: 10.1177/117727190600100016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Metallothionein (MT) is a highly conserved, low-molecular-weight, cysteine-rich protein that occurs in 4 isoforms (MT-I to MT-IV), of which MT-I+II are the major and best characterized proteins. This review will focus on mammalian MT-I+II and their functional impact upon cellular survival and death, as seen in two rather contrasting pathological conditions: Neurodegeneration and neoplasms. MT-I+II have analogous functions including: 1) Antioxidant scavenging of reactive oxygen species (ROS); 2) Cytoprotection against degeneration and apoptosis; 3) Stimulation of cell growth and repair including angiogenesis/revascularization, activation of stem/progenitor cells, and neuroregeneration. Thereby, MT-I+II mediate neuroprotection, CNS restoration and clinical recovery during neurodegenerative disorders. Due to the promotion of cell survival, increased MT-I+II levels have been associated with poor tumor prognosis, although the data are less clear and direct causative roles of MT-I+II in oncogenesis remain to be identified. The MT-I+II molecular mechanisms of actions are not fully elucidated. However, their role in metal ion homeostasis might be fundamental in controlling Zn-dependent transcription factors, protein synthesis, cellular energy levels/metabolism and cell redox state. Here, the neuroprotective and regenerative functions of MT-I+II are reviewed, and the presumed link to oncogenesis is critically perused.
Collapse
Affiliation(s)
- Allan Evald Nielsen
- Section of Neuroprotection, Centre of Inflammation and Metabolism
- The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Adam Bohr
- Section of Neuroprotection, Centre of Inflammation and Metabolism
- The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Milena Penkowa
- Section of Neuroprotection, Centre of Inflammation and Metabolism
- The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark
| |
Collapse
|
8
|
Metallothionein in Brain Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5828056. [PMID: 29085556 PMCID: PMC5632493 DOI: 10.1155/2017/5828056] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/17/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
Metallothioneins are a family of proteins which are able to bind metals intracellularly, so their main function is to regulate the cellular metabolism of essential metals. There are 4 major isoforms of MTs (I-IV), three of which have been localized in the central nervous system. MT-I and MT-II have been localized in the spinal cord and brain, mainly in astrocytes, whereas MT-III has been found mainly in neurons. MT-I and MT-II have been considered polyvalent proteins whose main function is to maintain cellular homeostasis of essential metals such as zinc and copper, but other functions have also been considered: detoxification of heavy metals, regulation of gene expression, processes of inflammation, and protection against free radicals generated by oxidative stress. On the other hand, the MT-III has been related in events of pathogenesis of neurodegenerative diseases such as Parkinson and Alzheimer. Likewise, the participation of MTs in other neurological disorders has also been reported. This review shows recent evidence about the role of MT in the central nervous system and its possible role in neurodegenerative diseases as well as in brain disorders.
Collapse
|
9
|
Jiang L, Hu Y, He X, Lv Q, Wang TH, Xia QJ. Breviscapine reduces neuronal injury caused by traumatic brain injury insult: partly associated with suppression of interleukin-6 expression. Neural Regen Res 2017; 12:90-95. [PMID: 28250753 PMCID: PMC5319248 DOI: 10.4103/1673-5374.198990] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Breviscapine, extracted from the herb Erigeron breviscapus, is widely used for the treatment of cardiovascular diseases, cerebral infarct, and stroke, but its mechanism of action remains unclear. This study established a rat model of traumatic brain injury induced by controlled cortical impact, and injected 75 μg breviscapine via the right lateral ventricle. We found that breviscapine significantly improved neurobehavioral dysfunction at 6 and 9 days after injection. Meanwhile, interleukin-6 expression was markedly down-regulated following breviscapine treatment. Our results suggest that breviscapine is effective in promoting neurological behavior after traumatic brain injury and the underlying molecular mechanism may be associated with the suppression of interleukin-6.
Collapse
Affiliation(s)
- Ling Jiang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yue Hu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiang He
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Lv
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qing-Jie Xia
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
10
|
Gupta M, Mishra SK, Kumar BSH, Khushu S, Rana P. Early detection of whole body radiation induced microstructural and neuroinflammatory changes in hippocampus: A diffusion tensor imaging and gene expression study. J Neurosci Res 2016; 95:1067-1078. [PMID: 27436454 DOI: 10.1002/jnr.23833] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/21/2016] [Accepted: 06/21/2016] [Indexed: 11/09/2022]
Abstract
Ionizing radiation is known to a cause systemic inflammatory response within hours of exposure that may affect the central nervous system (CNS). The present study was carried out to look upon the influence of radiation induced systemic inflammatory response in hippocampus within 24 hr of whole body radiation exposure. A Diffusion Tensor Imaging (DTI) study was conducted in mice exposed to a 5-Gy radiation dose through a 60 Co source operating at 2.496 Gy/min at 3 hr and 24 hr post irradiation and in sham-irradiated controls using 7 T animal MRI system. The results showed a significant decrease in Mean Diffusivity (MD), Radial Diffusivity (RD), and Axial Diffusivity (AD) in hippocampus at 24 hr compared with controls. Additionally, marked change in RD was observed at 3 hr. Increased serum C-Reactive Protein (CRP) level depicted an increased systemic/peripheral inflammation. The neuroinflammatory response in hippocampus was characterized by increased mRNA expression of IL-1β, IL-6, and Cox-2 at the 24 hr time point. Additionally, in the irradiated group, reactive astrogliosis was illustrated, with noticeable changes in GFAP expression at 24 hr. Altered diffusivity and enhanced neuroinflammatory expression in the hippocampal region showed peripheral inflammation induced changes in brain. Moreover, a negative correlation between gene expression and DTI parameters depicted a neuroinflammation induced altered microenvironment that might affect water diffusivity. The study showed that there was an influence of whole body radiation exposure on hippocampus even during the early acute phase that could be reflected in terms of neuroinflammatory response as well as microstructural changes. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mamta Gupta
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Sushanta Kumar Mishra
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - B S Hemanth Kumar
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Poonam Rana
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
11
|
Lamprecht MR, Elkin BS, Kesavabhotla K, Crary JF, Hammers JL, Huh JW, Raghupathi R, Morrison B. Strong Correlation of Genome-Wide Expression after Traumatic Brain Injury In Vitro and In Vivo Implicates a Role for SORLA. J Neurotrauma 2016; 34:97-108. [PMID: 26919808 DOI: 10.1089/neu.2015.4306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The utility of in vitro models of traumatic brain injury (TBI) depends on their ability to recapitulate the in vivo TBI cascade. In this study, we used a genome-wide approach to compare changes in gene expression at several time points post-injury in both an in vitro model and an in vivo model of TBI. We found a total of 2073 differentially expressed genes in our in vitro model and 877 differentially expressed genes in our in vivo model when compared to noninjured controls. We found a strong correlation in gene expression changes between the two models (r = 0.69), providing confidence that the in vitro model represented at least part of the in vivo injury cascade. From these data, we searched for genes with significant changes in expression over time (analysis of covariance) and identified sorting protein-related receptor with A-type repeats (SORLA). SORLA directs amyloid precursor protein to the recycling pathway by direct binding and away from amyloid-beta producing enzymes. Mutations of SORLA have been linked to Alzheimer's disease (AD). We confirmed downregulation of SORLA expression in organotypic hippocampal slice cultures by immunohistochemistry and Western blotting and present preliminary data from human tissue that is consistent with these experimental results. Together, these data suggest that the in vitro model of TBI used in this study strongly recapitulates the in vivo TBI pathobiology and is well suited for future mechanistic or therapeutic studies. The data also suggest the possible involvement of SORLA in the post-traumatic cascade linking TBI to AD.
Collapse
Affiliation(s)
- Michael R Lamprecht
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Benjamin S Elkin
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,2 MEA Forensic Engineers & Scientists , Mississauga, Ontario, Canada
| | - Kartik Kesavabhotla
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - John F Crary
- 3 Department of Pathology, Fishberg Department of Neuroscience, Friedman Brain Institute , and the Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jennifer L Hammers
- 4 Office of Chief Medical Examiner , City of New York, New York, New York
| | - Jimmy W Huh
- 5 Department of Anesthesia and Critical Care, Children's Hospital of Philadelphia , Philadelphia, Pennsylvania
| | - Ramesh Raghupathi
- 6 Department of Neurobiology and Anatomy, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Barclay Morrison
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| |
Collapse
|
12
|
White TE, Surles-Zeigler MC, Ford GD, Gates AS, Davids B, Distel T, LaPlaca MC, Ford BD. Bilateral gene interaction hierarchy analysis of the cell death gene response emphasizes the significance of cell cycle genes following unilateral traumatic brain injury. BMC Genomics 2016; 17:130. [PMID: 26912237 PMCID: PMC4765060 DOI: 10.1186/s12864-016-2412-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 01/26/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Delayed or secondary cell death that is caused by a cascade of cellular and molecular processes initiated by traumatic brain injury (TBI) may be reduced or prevented if an effective neuroprotective strategy is employed. Microarray and subsequent bioinformatic analyses were used to determine which genes, pathways and networks were significantly altered 24 h after unilateral TBI in the rat. Ipsilateral hemi-brain, the corresponding contralateral hemi-brain, and naïve (control) brain tissue were used for microarray analysis. RESULTS Ingenuity Pathway Analysis showed cell death and survival (CD) to be a top molecular and cellular function associated with TBI on both sides of the brain. One major finding was that the overall gene expression pattern suggested an increase in CD genes in ipsilateral brain tissue and suppression of CD genes contralateral to the injury which may indicate an endogenous protective mechanism. We created networks of genes of interest (GOI) and ranked the genes by the number of direct connections each had in the GOI networks, creating gene interaction hierarchies (GIHs). Cell cycle was determined from the resultant GIHs to be a significant molecular and cellular function in post-TBI CD gene response. CONCLUSIONS Cell cycle and apoptosis signalling genes that were highly ranked in the GIHs and exhibited either the inverse ipsilateral/contralateral expression pattern or contralateral suppression were identified and included STAT3, CCND1, CCND2, and BAX. Additional exploration into the remote suppression of CD genes may provide insight into neuroprotective mechanisms that could be used to develop therapies to prevent cell death following TBI.
Collapse
Affiliation(s)
- Todd E White
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Monique C Surles-Zeigler
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Gregory D Ford
- Division of Natural Sciences and Physical Education, Georgia Highlands College, 5441 Highway 20, NE, Cartersville, GA, 30121, USA.
| | - Alicia S Gates
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Benem Davids
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Timothy Distel
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
- University of California-Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA.
| | - Michelle C LaPlaca
- Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Byron D Ford
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
- University of California-Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA.
| |
Collapse
|
13
|
Pardo L, Schlüter A, Valor LM, Barco A, Giralt M, Golbano A, Hidalgo J, Jia P, Zhao Z, Jové M, Portero-Otin M, Ruiz M, Giménez-Llort L, Masgrau R, Pujol A, Galea E. Targeted activation of CREB in reactive astrocytes is neuroprotective in focal acute cortical injury. Glia 2016; 64:853-74. [PMID: 26880229 DOI: 10.1002/glia.22969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 01/07/2023]
Abstract
The clinical challenge in acute injury as in traumatic brain injury (TBI) is to halt the delayed neuronal loss that occurs hours and days after the insult. Here we report that the activation of CREB-dependent transcription in reactive astrocytes prevents secondary injury in cerebral cortex after experimental TBI. The study was performed in a novel bitransgenic mouse in which a constitutively active CREB, VP16-CREB, was targeted to astrocytes with the Tet-Off system. Using histochemistry, qPCR, and gene profiling we found less neuronal death and damage, reduced macrophage infiltration, preserved mitochondria, and rescued expression of genes related to mitochondrial metabolism in bitransgenic mice as compared to wild type littermates. Finally, with meta-analyses using publicly available databases we identified a core set of VP16-CREB candidate target genes that may account for the neuroprotective effect. Enhancing CREB activity in astrocytes thus emerges as a novel avenue in acute brain post-injury therapeutics.
Collapse
Affiliation(s)
- Luis Pardo
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, L'hospitalet De Llobregat, Barcelona, 08907, Spain
| | - Luis M Valor
- Instituto De Neurociencias De Alicante, Universidad Miguel Hernández/Consejo Superior De Investigaciones Científicas, Sant Joan D'alacant, Alicante, 03550, Spain
| | - Angel Barco
- Instituto De Neurociencias De Alicante, Universidad Miguel Hernández/Consejo Superior De Investigaciones Científicas, Sant Joan D'alacant, Alicante, 03550, Spain
| | - Mercedes Giralt
- Institut De Neurociències and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma, Barcelona, 08193, Spain
| | - Arantxa Golbano
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Juan Hidalgo
- Institut De Neurociències and Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma, Barcelona, 08193, Spain
| | - Peilin Jia
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, 25198, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, 25198, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, L'hospitalet De Llobregat, Barcelona, 08907, Spain
| | - Lydia Giménez-Llort
- Institut De Neurociènces and Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma De Barcelona, Bellaterra, 08193, Spain
| | - Roser Masgrau
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, L'hospitalet De Llobregat, Barcelona, 08907, Spain.,Institució Catalana De Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Elena Galea
- Institut De Neurociències and Unitat De Bioquímica, Facultat De Medicina, Universitat Autònoma De Barcelona, Bellaterra, Barcelona, 08193, Spain.,Institució Catalana De Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
14
|
Lynes MA, Hidalgo J, Manso Y, Devisscher L, Laukens D, Lawrence DA. Metallothionein and stress combine to affect multiple organ systems. Cell Stress Chaperones 2014; 19:605-11. [PMID: 24584987 PMCID: PMC4147071 DOI: 10.1007/s12192-014-0501-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 01/23/2014] [Accepted: 01/23/2014] [Indexed: 12/16/2022] Open
Abstract
Metallothioneins (MTs) are a family of low molecular weight, cysteine-rich, metal-binding proteins that have a wide range of functions in cellular homeostasis and immunity. MTs can be induced by a variety of conditions including metals, glucocorticoids, endotoxin, acute phase cytokines, stress, and irradiation. In addition to their important immunomodulatory functions, MTs can protect essential cellular compartments from toxicants, serve as a reservoir of essential heavy metals, and regulate cellular redox potential. Many of the roles of MTs in the neuroinflammation, intestinal inflammation, and stress response have been investigated and were the subject of a session at the 6th International Congress on Stress Proteins in Biology and Medicine in Sheffield, UK. Like the rest of the cell stress response, there are therapeutic opportunities that arise from an understanding of MTs, and these proteins also provide potential insights into the world of the heat shock protein.
Collapse
Affiliation(s)
- Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, 06269, USA,
| | | | | | | | | | | |
Collapse
|
15
|
Xu B, Yu DM, Liu FS. Effect of siRNA‑induced inhibition of IL‑6 expression in rat cerebral gliocytes on cerebral edema following traumatic brain injury. Mol Med Rep 2014; 10:1863-8. [PMID: 25109513 DOI: 10.3892/mmr.2014.2462] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 04/25/2014] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effect of RNA interference (RNAi) on the inhibition of interleukin (IL)‑6 expression in rat cerebral gliocytes in vitro and rat cerebral traumatic tissues in vivo, as well as the effect of RNAi on cerebral edema. pSUPER vectors containing IL‑6 small hairpin RNA (pSUPER‑IL‑6 1‑5) were designed, constructed and transfected into C6 rat glioma cells using cationic liposomes. ELISA was used to select the plasmid with the strongest interference effect. A freefall method was used to generate a rat brain injury model and rats were randomly divided into treatment, empty plasmid and control groups (n=14/group). IL‑6 levels, water content and sodium content were determined in the brain tissues at 24 and 72 h post‑injury. pSUPER‑IL‑6 was effectively transfected into C6 cells and was found to inhibit the expression of IL‑6 rather than IL‑8. The pSUPER‑IL‑6 1 vector was most effective in inducing RNAi. In vivo, IL‑6 levels were observed to be lowest in the interference group and there were statistically significant differences in water and sodium content among the experimental groups (P<0.05). RNAi was found to inhibit IL‑6 expression in vivo and in vitro in rat cerebral gliocytes, and the reduction of the IL‑6 levels was found to reduce post‑traumatic cerebral edema.
Collapse
Affiliation(s)
- Bin Xu
- Department of Emergency, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Dong-Ming Yu
- Department of Emergency, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Fu-Sheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
16
|
Histone deacetylase inhibitors are neuroprotective and preserve NGF-mediated cell survival following traumatic brain injury. Proc Natl Acad Sci U S A 2013; 110:10747-52. [PMID: 23754423 DOI: 10.1073/pnas.1308950110] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acute traumatic brain injury (TBI) is associated with long-term cognitive and behavioral dysfunction. In vivo studies have shown histone deacetylase inhibitors (HDACis) to be neuroprotective following TBI in rodent models. HDACis are intriguing candidates because they are capable of provoking widespread genetic changes and modulation of protein function. By using known HDACis and a unique small-molecule pan-HDACi (LB-205), we investigated the effects and mechanisms associated with HDACi-induced neuroprotection following CNS injury in an astrocyte scratch assay in vitro and a rat TBI model in vivo. We demonstrate the preservation of sufficient expression of nerve growth factor (NGF) and activation of the neurotrophic tyrosine kinase receptor type 1 (TrkA) pathway following HDACi treatment to be crucial in stimulating the survival of CNS cells after TBI. HDACi treatment up-regulated the expression of NGF, phospho-TrkA, phospho-protein kinase B (p-AKT), NF-κB, and B-cell lymphoma 2 (Bcl-2) cell survival factors while down-regulating the expression of p75 neurotrophin receptor (NTR), phospho-JNK, and Bcl-2-associated X protein apoptosis factors. HDACi treatment also increased the expression of the stem cell biomarker nestin, and decreased the expression of reactive astrocyte biomarker GFAP within damaged tissue following TBI. These findings provide further insight into the mechanisms by which HDACi treatment after TBI is neuroprotective and support the continued study of HDACis following acute TBI.
Collapse
|
17
|
Gene expression patterns following unilateral traumatic brain injury reveals a local pro-inflammatory and remote anti-inflammatory response. BMC Genomics 2013; 14:282. [PMID: 23617241 PMCID: PMC3669032 DOI: 10.1186/1471-2164-14-282] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 04/15/2013] [Indexed: 01/13/2023] Open
Abstract
Background Traumatic brain injury (TBI) results in irreversible damage at the site of impact and initiates cellular and molecular processes that lead to secondary neural injury in the surrounding tissue. We used microarray analysis to determine which genes, pathways and networks were significantly altered using a rat model of TBI. Adult rats received a unilateral controlled cortical impact (CCI) and were sacrificed 24 h post-injury. The ipsilateral hemi-brain tissue at the site of the injury, the corresponding contralateral hemi-brain tissue, and naïve (control) brain tissue were used for microarray analysis. Ingenuity Pathway Analysis (IPA) software was used to identify molecular pathways and networks that were associated with the altered gene expression in brain tissues following TBI. Results Inspection of the top fifteen biological functions in IPA associated with TBI in the ipsilateral tissues revealed that all had an inflammatory component. IPA analysis also indicated that inflammatory genes were altered on the contralateral side, but many of the genes were inversely expressed compared to the ipsilateral side. The contralateral gene expression pattern suggests a remote anti-inflammatory molecular response. We created a network of the inversely expressed common (i.e., same gene changed on both sides of the brain) inflammatory response (IR) genes and those IR genes included in pathways and networks identified by IPA that changed on only one side. We ranked the genes by the number of direct connections each had in the network, creating a gene interaction hierarchy (GIH). Two well characterized signaling pathways, toll-like receptor/NF-kappaB signaling and JAK/STAT signaling, were prominent in our GIH. Conclusions Bioinformatic analysis of microarray data following TBI identified key molecular pathways and networks associated with neural injury following TBI. The GIH created here provides a starting point for investigating therapeutic targets in a ranked order that is somewhat different than what has been presented previously. In addition to being a vehicle for identifying potential targets for post-TBI therapeutic strategies, our findings can also provide a context for evaluating the potential of therapeutic agents currently in development.
Collapse
|
18
|
Quintana A, Erta M, Ferrer B, Comes G, Giralt M, Hidalgo J. Astrocyte-specific deficiency of interleukin-6 and its receptor reveal specific roles in survival, body weight and behavior. Brain Behav Immun 2013; 27:162-73. [PMID: 23085146 DOI: 10.1016/j.bbi.2012.10.011] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 10/01/2012] [Accepted: 10/10/2012] [Indexed: 12/22/2022] Open
Abstract
Interleukin-6 (IL-6) is a major cytokine which controls not only the immune system but also exhibits many other functions including effects in the central nervous system (CNS). IL-6 is known to be produced by different cells in the CNS, and all the major CNS do respond to IL-6, which makes it difficult to dissect the specific roles of each cell type when assessing the role of IL-6 in the brain. We have produced for the first time floxed mice for IL-6 and have crossed them with GFAP-Cre mice to delete IL-6 in astrocytes (Ast-IL-6 KO mice), and have compared their phenotype with that of mice with deletion of IL-6 receptor in astrocytes (Ast-IL6R KO mice). Our results indicate a major prosurvival role of the astrocyte IL-6 system at early ages (intrauterine life), which was also involved to various degrees in the control of adult body weight, locomotor activity, anxiety and exploratory behaviors. In some occasions deleting IL-6R in astrocytes mimicked the phenotype of Ast-IL-6 KO mice (i.e. activity), while in others the opposite was observed (i.e. exploration), suggesting autocrine and paracrine (presumably on neurons) roles of astrocyte IL-6. Our results suggest important roles of the astrocyte IL-6 system on normal brain physiology, in some cases totally unexpected from previous results with total IL-6 KO mice.
Collapse
Affiliation(s)
- Albert Quintana
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
19
|
Erta M, Quintana A, Hidalgo J. Interleukin-6, a major cytokine in the central nervous system. Int J Biol Sci 2012; 8:1254-66. [PMID: 23136554 PMCID: PMC3491449 DOI: 10.7150/ijbs.4679] [Citation(s) in RCA: 778] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/19/2012] [Indexed: 12/21/2022] Open
Abstract
Interleukin-6 (IL-6) is a cytokine originally identified almost 30 years ago as a B-cell differentiation factor, capable of inducing the maturation of B cells into antibody-producing cells. As with many other cytokines, it was soon realized that IL-6 was not a factor only involved in the immune response, but with many critical roles in major physiological systems including the nervous system. IL-6 is now known to participate in neurogenesis (influencing both neurons and glial cells), and in the response of mature neurons and glial cells in normal conditions and following a wide arrange of injury models. In many respects, IL-6 behaves in a neurotrophin-like fashion, and seemingly makes understandable why the cytokine family that it belongs to is known as neuropoietins. Its expression is affected in several of the main brain diseases, and animal models strongly suggest that IL-6 could have a role in the observed neuropathology and that therefore it is a clear target of strategic therapies.
Collapse
Affiliation(s)
- María Erta
- Instituto de Neurociencias y Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Biociencias, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
20
|
Ley EJ, Clond MA, Singer MB, Shouhed D, Salim A. IL6 Deficiency Affects Function After Traumatic Brain Injury. J Surg Res 2011; 170:253-6. [DOI: 10.1016/j.jss.2011.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/07/2011] [Accepted: 03/02/2011] [Indexed: 11/16/2022]
|
21
|
Kruse F, Bosse F, Vogelaar CF, Brazda N, Küry P, Gasis M, Müller HW. Cortical gene expression in spinal cord injury and repair: insight into the functional complexity of the neural regeneration program. Front Mol Neurosci 2011; 4:26. [PMID: 21994489 PMCID: PMC3182759 DOI: 10.3389/fnmol.2011.00026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/05/2011] [Indexed: 11/29/2022] Open
Abstract
Traumatic spinal cord injury (SCI) results in the formation of a fibrous scar acting as a growth barrier for regenerating axons at the lesion site. We have previously shown (Klapka et al., 2005) that transient suppression of the inhibitory lesion scar in rat spinal cord leads to long distance axon regeneration, retrograde rescue of axotomized cortical motoneurons, and improvement of locomotor function. Here we applied a systemic approach to investigate for the first time specific and dynamic alterations in the cortical gene expression profile following both thoracic SCI and regeneration-promoting anti-scarring treatment (AST). In order to monitor cortical gene expression we carried out microarray analyses using total RNA isolated from layer V/VI of rat sensorimotor cortex at 1–60 days post-operation (dpo). We demonstrate that cortical neurons respond to injury by massive changes in gene expression, starting as early as 1 dpo. AST, in turn, results in profound modifications of the lesion-induced expression profile. The treatment attenuates SCI-triggered transcriptional changes of genes related to inhibition of axon growth and impairment of cell survival, while upregulating the expression of genes associated with axon outgrowth, cell protection, and neural development. Thus, AST not only modifies the local environment impeding spinal cord regeneration by reduction of fibrous scarring in the injured spinal cord, but, in addition, strikingly changes the intrinsic capacity of cortical pyramidal neurons toward enhanced cell maintenance and axonal regeneration.
Collapse
Affiliation(s)
- Fabian Kruse
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
22
|
Metallothionein and brain inflammation. J Biol Inorg Chem 2011; 16:1103-13. [PMID: 21678079 DOI: 10.1007/s00775-011-0802-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/02/2011] [Indexed: 10/18/2022]
Abstract
Since the seminal discoveries of Bert Vallee regarding zinc and metallothioneins (MTs) more than 50 years ago, thousands of studies have been published concerning this fascinating story. One of the most active areas of research is the involvement of these proteins in the inflammatory response in general, and in neuroinflammation in particular. We describe the general aspects of the inflammatory response, highlighting the essential role of the major cytokine interleukin-6, and review briefly the expression and function of MTs in the central nervous system in the context of neuroinflammation. Particular attention is paid to the Tg2576 Alzheimer disease mouse model and the preliminary results obtained in mice into which human Zn(7)MT-2A was injected, which suggest a reversal of the behavioral deficits while enhancing amyloid plaque load and gliosis.
Collapse
|
23
|
Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S. Interleukin-6, a mental cytokine. ACTA ACUST UNITED AC 2011; 67:157-83. [PMID: 21238488 DOI: 10.1016/j.brainresrev.2011.01.002] [Citation(s) in RCA: 278] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 12/21/2010] [Accepted: 01/08/2011] [Indexed: 12/18/2022]
Abstract
Almost a quarter of a century ago, interleukin-6 (IL-6) was discovered as an inflammatory cytokine involved in B cell differentiation. Today, IL-6 is recognized to be a highly versatile cytokine, with pleiotropic actions not only in immune cells, but also in other cell types, such as cells of the central nervous system (CNS). The first evidence implicating IL-6 in brain-related processes originated from its dysregulated expression in several neurological disorders such as multiple sclerosis, Alzheimer's disease and Parkinson's disease. In addition, IL-6 was shown to be involved in multiple physiological CNS processes such as neuron homeostasis, astrogliogenesis and neuronal differentiation. The molecular mechanisms underlying IL-6 functions in the brain have only recently started to emerge. In this review, an overview of the latest discoveries concerning the actions of IL-6 in the nervous system is provided. The central position of IL-6 in the neuroinflammatory reaction pattern, and more specifically, the role of IL-6 in specific neurodegenerative processes, which accompany Alzheimer's disease, multiple sclerosis and excitotoxicity, are discussed. It is evident that IL-6 has a dichotomic action in the CNS, displaying neurotrophic properties on the one hand, and detrimental actions on the other. This is in agreement with its central role in neuroinflammation, which evolved as a beneficial process, aimed at maintaining tissue homeostasis, but which can become malignant when exaggerated. In this perspective, it is not surprising that 'well-meant' actions of IL-6 are often causing harm instead of leading to recovery.
Collapse
Affiliation(s)
- Anneleen Spooren
- Laboratory of Eukaryotic Signal Transduction and Gene Expression, University of Ghent, K.L. Ledeganckstraat 35, 9000 Gent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
24
|
Barr TL, Alexander S, Conley Y. Gene expression profiling for discovery of novel targets in human traumatic brain injury. Biol Res Nurs 2010; 13:140-53. [PMID: 21112922 DOI: 10.1177/1099800410385671] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Several clinical trials have failed to demonstrate a significant effect on outcome following human traumatic brain injury (TBI) despite promising results obtained in preclinical animal studies. These failures may be due in part to a misinterpretation of the findings obtained in preclinical animal models of TBI, a misunderstanding of the complexity of the human response to TBI, limited knowledge about the biological pathways that interact to contribute to good and bad outcomes after brain injury, and the effects of genomic variability and environment on individual recovery. Recent publications suggest that data obtained from gene expression profiling studies of complex neurological diseases such as stroke, multiple sclerosis (MS), Alzheimer's and Parkinson's may contribute to a more informed understanding of what affects outcome following TBI. These data may help to bridge the gap between successful preclinical studies and negative clinical trials in humans to reveal novel targets for therapy. Gene expression profiling has the capability to identify biomarkers associated with response to TBI, elucidate complex genetic interactions that may play a role in outcome following TBI, and reveal biological pathways related to brain health. This review highlights the current state of the literature on gene expression profiling for neurological disease and discusses its ability to aid in unraveling the variable human response to TBI and the potential for it to offer treatment strategies in an area where we currently have limited therapeutic options primarily based on supportive care.
Collapse
Affiliation(s)
- Taura L Barr
- West Virginia University School of Nursing & Center for Neuroscience, Morgantown, WV, USA.
| | | | | |
Collapse
|
25
|
Israelsson C, Wang Y, Kylberg A, Pick CG, Hoffer BJ, Ebendal T. Closed head injury in a mouse model results in molecular changes indicating inflammatory responses. J Neurotrauma 2010; 26:1307-14. [PMID: 19317611 DOI: 10.1089/neu.2008.0676] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cerebral gene expression changes in response to traumatic brain injury will provide useful information in the search for future trauma treatment. In order to characterize the outcome of mild brain injury, we studied C57BL/6J mice in a weight-drop, closed head injury model. At various times post-injury, mRNA was isolated from neocortex and hippocampus and transcriptional alterations were studied using quantitative reverse transcriptase PCR and gene array analysis. At three days post-injury, the results showed unilateral injury responses, both in neocortex and hippocampus, with the main effect seen on the side of the skull hit by the dropping weight. Upregulated transcripts encoded products characterizing reactive astrocytes, phagocytes, microglia, and immune-reactive cells. Markers for oligodendrocytes and T-cells were not altered. Notably, strong differences in the responses among individual mice were seen (e.g., for the Gfap transcript expressed by reactive astrocytes and the chemokine Ccl3 transcript expressed by activated microglial cells). In conclusion, mild TBI chiefly activates transcripts leading to tissue signaling, inflammatory processes, and chemokine signaling, as in focal brain injury, suggesting putative targets for drug development.
Collapse
Affiliation(s)
- Charlotte Israelsson
- Developmental Neuroscience, Department of Neuroscience, Biomedical Center, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
26
|
Manso Y, Serra M, Comes G, Giralt M, Carrasco J, Cols N, Vasák M, González-Duarte P, Hidalgo J. The comparison of mouse full metallothionein-1 versus alpha and beta domains and metallothionein-1-to-3 mutation following traumatic brain injury reveals different biological motifs. J Neurosci Res 2010; 88:1708-18. [PMID: 20127815 DOI: 10.1002/jnr.22342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Traumatic injury to the brain is one of the leading causes of injury-related death or disability, but current therapies are limited. Previously it has been shown that the antioxidant proteins metallothioneins (MTs) are potent neuroprotective factors in animal models of brain injury. The exogenous administration of MTs causes effects consistent with the roles proposed from studies in knock-out mice. We herewith report the results comparing full mouse MT-1 with the independent alpha and beta domains, alone or together, in a cryoinjury model. The lesion of the cortex caused the mice to perform worse in the horizontal ladder beam and the rota-rod tests; all the proteins showed a modest effect in the former test, while only full MT-1 improved the performance of animals in the rota-rod, and the alpha domain showed a rather detrimental effect. Gene expression analysis by RNA protection assay demonstrated that all proteins may alter the expression of host-response genes such as GFAP, Mac1 and ICAM, in some cases being the beta domain more effective than the alpha domain or even the full MT-1. A MT-1-to-MT-3 mutation blunted some but not all the effects caused by the normal MT-1, and in some cases increased its potency. Thus, splitting the two MT-1 domains do not seem to eliminate all MT functions but certainly modifies them, and different motifs seem to be present in the protein underlying such functions.
Collapse
Affiliation(s)
- Yasmina Manso
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology Unit, Faculty of Biosciences, Autonomous University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Israelsson C, Bengtsson H, Kylberg A, Kullander K, Lewén A, Hillered L, Ebendal T. Distinct cellular patterns of upregulated chemokine expression supporting a prominent inflammatory role in traumatic brain injury. J Neurotrauma 2008; 25:959-74. [PMID: 18665806 DOI: 10.1089/neu.2008.0562] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cerebral gene expressions change in response to traumatic brain injury (TBI), and future trauma treatment may improve with increased knowledge about these regulations. We subjected C57BL/6J mice to injury by controlled cortical impact (CCI). At various time points post-injury, mRNA from neocortex and hippocampus was isolated, and transcriptional alterations studied using quantitative real-time polymerase chain reaction (PCR) and gene array analysis. Spatial distribution of enhanced expression was characterized by in situ hybridization. Products of the upregulated transcripts serve functions in a range of cellular mechanisms, including stress, inflammation and immune responses, and tissue remodeling. We also identified increased transcript levels characterizing reactive astrocytes, oligodendrocytes, and microglia, and furthermore, we demonstrated a novel pattern of scattered cell clusters expressing the chemokine Cxcl10. Notably, a sustained increase in integrin alpha X (Itgax), characterizing antigen-presenting dendritic cells, was found with the transcript located to similar cell clusters. In contrast, T-cell receptor alpha transcript showed only a modest increase. The induced P-selectin (Selp) expression level in endothelial cells, and chemokines from microglia, may guide perivascular accumulation of extravasating inflammatory monocytes differentiating into dendritic cells. In conclusion, our study shows that following TBI, secondary injury chiefly involves inflammatory processes and chemokine signaling, which comprise putative targets for pharmaceutical neuroprotection.
Collapse
Affiliation(s)
- Charlotte Israelsson
- Department of Neuroscience, Developmental Neuroscience, Biomedical Center, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
28
|
Quintana A, Molinero A, Borup R, Nielsen FC, Campbell IL, Penkowa M, Hidalgo J. Effect of astrocyte-targeted production of IL-6 on traumatic brain injury and its impact on the cortical transcriptome. Dev Neurobiol 2008; 68:195-208. [PMID: 18000830 DOI: 10.1002/dneu.20584] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Interleukin-6 (IL-6) is one of the key players in the response of the brain cortex to injury. We have described previously that astrocyte-driven production of IL-6 (GFAP-IL6) in transgenic mice, although causing spontaneous neuroinflammation and long term damage, is beneficial after an acute (freeze) injury in the cortex, increasing healing and decreasing oxidative stress and apoptosis. To determine the transcriptional basis for these responses here we analyzed the global gene expression profile of the cortex, at 0 (unlesioned), 1 or 4 days post lesion (dpl), in both GFAP-IL6 mice and their control littermates. GFAP-IL6 mice showed an increase in genes associated with the inflammatory response both at 1 dpl (Iftm1, Endod1) and 4 dpl (Gfap, C4b), decreased expression of proapoptotic genes (i.e. Gadd45b, Clic4, p21) as well as reduced expression of genes involved in the control of oxidative stress (Atf4). Furthermore, the presence of IL-6 altered the expression of genes involved in hemostasis (Vwf), cell migration and proliferation (Cap2), and synaptic activity (Vamp2). All these changes in gene expression could underlie the phenotype of the GFAP-IL6 mice after injury, but many other possible factors were also identified in this study, highlighting the utility of this approach for deciphering new pathways orchestrated by IL-6.
Collapse
Affiliation(s)
- Albert Quintana
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology Unit, Faculty of Sciences, Autonomous University of Barcelona, Bellaterra, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
29
|
Pedersen MV, Helweg-Larsen RB, Nielsen FC, Berezin V, Bock E, Penkowa M. The synthetic NCAM-derived peptide, FGL, modulates the transcriptional response to traumatic brain injury. Neurosci Lett 2008; 437:148-53. [PMID: 18436381 DOI: 10.1016/j.neulet.2008.03.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 03/13/2008] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
Cerebral responses to traumatic brain injury (TBI) include up- and downregulation of a vast number of proteins involved in endogenous inflammatory responses and defense mechanisms developing postinjury. The present study analyzed the global gene expression profile in response to cryo-induced TBI by means of microarray analysis. Adolescent rats were subjected to TBI and treated with either placebo or a neural cell adhesion molecule (NCAM)-derived fibroblast growth factor receptor (FGFR) agonist, FGL peptide, which has been demonstrated to have neuroprotective effects. mRNA levels were measured at various time-points postlesion (6 h, 1 day and 4 days). The effects of injury, treatment, and injury-treatment interaction were observed. TBI alone rendered a large number of genes affected. Analysis of lesion and treatment interactions resulted in a clear effect of the interaction between injury and FGL-treatment compared to injury and placebo-treatment. Genes affected by TBI alone included inflammation markers, protein kinases, ion channel members and growth factors. Genes encoding regulators of apoptosis, signal transduction and metabolism were altered by the interaction between FGL-treatment and TBI. FGL-treatment in non-injured animals rendered genes regulating signaling, transport and cytoskeleton maintenance significantly increased. Thus, the hypothesis of a putative neuroprotective role of FGL was supported by our findings.
Collapse
Affiliation(s)
- Martin Volmer Pedersen
- Protein Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
30
|
Metallothionein in the central nervous system: Roles in protection, regeneration and cognition. Neurotoxicology 2008; 29:489-503. [PMID: 18313142 DOI: 10.1016/j.neuro.2007.12.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Accepted: 12/22/2007] [Indexed: 12/23/2022]
Abstract
Metallothionein (MT) is an enigmatic protein, and its physiological role remains a matter of intense study and debate 50 years after its discovery. This is particularly true of its function in the central nervous system (CNS), where the challenge remains to link its known biochemical properties of metal binding and free radical scavenging to the intricate workings of brain. In this compilation of four reports, first delivered at the 11th International Neurotoxicology Association (INA-11) Meeting, June 2007, the authors present the work of their laboratories, each of which gives an important insight into the actions of MT in the brain. What emerges is that MT has the potential to contribute to a variety of processes, including neuroprotection, regeneration, and even cognitive functions. In this article, the properties and CNS expression of MT are briefly reviewed before Dr Hidalgo describes his pioneering work using transgenic models of MT expression to demonstrate how this protein plays a major role in the defence of the CNS against neurodegenerative disorders and other CNS injuries. His group's work leads to two further questions, what are the mechanisms at the cellular level by which MT acts, and does this protein influence higher order issues of architecture and cognition? These topics are addressed in the second and third sections of this review by Dr West, and Dr Levin and Dr Eddins, respectively. Finally, Dr Aschner examines the ability of MT to protect against a specific toxicant, methylmercury, in the CNS.
Collapse
|
31
|
Quintana A, Molinero A, Florit S, Manso Y, Comes G, Carrasco J, Giralt M, Borup R, Nielsen FC, Campbell IL, Penkowa M, Hidalgo J. Diverging mechanisms for TNF-alpha receptors in normal mouse brains and in functional recovery after injury: From gene to behavior. J Neurosci Res 2008; 85:2668-85. [PMID: 17131423 DOI: 10.1002/jnr.21126] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cytokines, such as tumour necrosis factor (TNF)-alpha and lymphotoxin-alpha, have been described widely to play important roles in the brain in physiologic conditions and after traumatic injury. However, the exact mechanisms involved in their function have not been fully elucidated. We give some insight on their role by using animals lacking either Type 1 receptor (TNFR1KO) or Type 2 (TNFR2KO) and their controls (C57Bl/6). Both TNFR1KO and to a greater extent TNFR2KO mice showed increased exploration/activity neurobehavioral traits in the hole board test, such as rearings, head dippings, and ambulations, compared with wild-type mice, suggesting an inhibitory role of TNFR1/TNFR2 signaling. In contrast, no significant differences were observed in the elevated plus maze test, ruling out a major role of these receptors in the control of anxiety. We next evaluated the response to a freeze injury to the somatosensorial cortex. The effect of the cryolesion on motor function was evaluated with the horizontal ladder beam test, and the results showed that both TNFR1KO and TNFR2KO mice made fewer errors, suggesting a detrimental role for TNFR1/TNFR2 signaling for coping with brain damage. Expression of approximately 22600 genes was analyzed using an Affymetrix chip (MOE430A) at 0 (unlesioned), 1, or 4 days post-lesion in the three strains. The results show a unique and major role of both TNF receptors on the pattern of gene expression elicited by the injury but also in normal conditions, and suggest that blocking of TNFR1/TNFR2 receptors may be beneficial after a traumatic brain injury.
Collapse
Affiliation(s)
- Albert Quintana
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology Unit, Faculty of Sciences, Autonomous University of Barcelona, Bellaterra, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chung RS, Hidalgo J, West AK. New insight into the molecular pathways of metallothionein-mediated neuroprotection and regeneration. J Neurochem 2007; 104:14-20. [PMID: 17986229 DOI: 10.1111/j.1471-4159.2007.05026.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
There is a large body of evidence demonstrating that metallothioneins (MTs) expressed in astrocytes following CNS injury, exhibit both neuroprotective and neuroregenerative properties and are critical for recovery outcomes. As these proteins lack signal peptides, and have well characterized free radical scavenging and heavy metal binding properties, the neuroprotective functions of MTs have been attributed to these intracellular roles. However, there is an increasing realization that the neuroprotective functions of MTs may also involve an extracellular component. In this issue of Journal of Neurochemistry, Ambjørn et al. reveal considerable insight into this novel function of MTs. In this review, we examine the seminal work of Ambjørn et al. in the context of our current understanding of the role of MT in astrocyte-neuron interactions in the injured brain, and also discuss the significant therapeutic potential of their work.
Collapse
Affiliation(s)
- R S Chung
- NeuroRepair Group, Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia.
| | | | | |
Collapse
|
33
|
Post hoc pattern matching: assigning significance to statistically defined expression patterns in single channel microarray data. BMC Bioinformatics 2007; 8:240. [PMID: 17615071 PMCID: PMC1934919 DOI: 10.1186/1471-2105-8-240] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 07/05/2007] [Indexed: 11/28/2022] Open
Abstract
Background Researchers using RNA expression microarrays in experimental designs with more than two treatment groups often identify statistically significant genes with ANOVA approaches. However, the ANOVA test does not discriminate which of the multiple treatment groups differ from one another. Thus, post hoc tests, such as linear contrasts, template correlations, and pairwise comparisons are used. Linear contrasts and template correlations work extremely well, especially when the researcher has a priori information pointing to a particular pattern/template among the different treatment groups. Further, all pairwise comparisons can be used to identify particular, treatment group-dependent patterns of gene expression. However, these approaches are biased by the researcher's assumptions, and some treatment-based patterns may fail to be detected using these approaches. Finally, different patterns may have different probabilities of occurring by chance, importantly influencing researchers' conclusions about a pattern and its constituent genes. Results We developed a four step, post hoc pattern matching (PPM) algorithm to automate single channel gene expression pattern identification/significance. First, 1-Way Analysis of Variance (ANOVA), coupled with post hoc 'all pairwise' comparisons are calculated for all genes. Second, for each ANOVA-significant gene, all pairwise contrast results are encoded to create unique pattern ID numbers. The # genes found in each pattern in the data is identified as that pattern's 'actual' frequency. Third, using Monte Carlo simulations, those patterns' frequencies are estimated in random data ('random' gene pattern frequency). Fourth, a Z-score for overrepresentation of the pattern is calculated ('actual' against 'random' gene pattern frequencies). We wrote a Visual Basic program (StatiGen) that automates PPM procedure, constructs an Excel workbook with standardized graphs of overrepresented patterns, and lists of the genes comprising each pattern. The visual basic code, installation files for StatiGen, and sample data are available as supplementary material. Conclusion The PPM procedure is designed to augment current microarray analysis procedures by allowing researchers to incorporate all of the information from post hoc tests to establish unique, overarching gene expression patterns in which there is no overlap in gene membership. In our hands, PPM works well for studies using from three to six treatment groups in which the researcher is interested in treatment-related patterns of gene expression. Hardware/software limitations and extreme number of theoretical expression patterns limit utility for larger numbers of treatment groups. Applied to a published microarray experiment, the StatiGen program successfully flagged patterns that had been manually assigned in prior work, and further identified other gene expression patterns that may be of interest. Thus, over a moderate range of treatment groups, PPM appears to work well. It allows researchers to assign statistical probabilities to patterns of gene expression that fit a priori expectations/hypotheses, it preserves the data's ability to show the researcher interesting, yet unanticipated gene expression patterns, and assigns the majority of ANOVA-significant genes to non-overlapping patterns.
Collapse
|
34
|
Quintana A, Giralt M, Molinero A, Campbell IL, Penkowa M, Hidalgo J. Analysis of the cerebral transcriptome in mice subjected to traumatic brain injury: importance of IL-6. Neuroimmunomodulation 2007; 14:139-43. [PMID: 18073505 DOI: 10.1159/000110637] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury is one of the leading causes of incapacity and death among young people. Injury to the brain elicits a potent inflammatory response, comprising recruitment of inflammatory cells, reactive astrogliosis and activation of brain macrophages. Under the influence of presumably several cytokines and growth factors, a cascade of events is activated that result ultimately in increased oxidative stress and tissue damage, but also in activation of counterregulatory factors and tissue regeneration. The complexity of this response is being unraveled by high-throughput methodologies such as microarrays. The combination of these modern techniques with the comparison of normal and genetically modified mice boosts the significance of the results obtained. With this approach, we have demonstrated that a cytokine such as interleukin-6 is one of the key players in the response of the brain to injury.
Collapse
Affiliation(s)
- Albert Quintana
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology Unit, Faculty of Sciences, Autonomous University of Barcelona, Bellaterra, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Penkowa M, Cáceres M, Borup R, Nielsen FC, Poulsen CB, Quintana A, Molinero A, Carrasco J, Florit S, Giralt M, Hidalgo J. Novel roles for metallothionein-I + II (MT-I + II) in defense responses, neurogenesis, and tissue restoration after traumatic brain injury: Insights from global gene expression profiling in wild-type and MT-I + II knockout mice. J Neurosci Res 2006; 84:1452-74. [PMID: 16941634 DOI: 10.1002/jnr.21043] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Traumatic injury to the brain is one of the leading causes of injury-related death or disability, especially among young people. Inflammatory processes and oxidative stress likely underlie much of the damage elicited by injury, but the full repertoire of responses involved is not well known. A genomic approach, such as the use of microarrays, provides much insight in this regard, especially if combined with the use of gene-targeted animals. We report here the results of one of these studies comparing wild-type and metallothionein-I + II knockout mice subjected to a cryolesion of the somatosensorial cortex and killed at 0, 1, 4, 8, and 16 days postlesion (dpl) using Affymetrix genechips/oligonucleotide arrays interrogating approximately 10,000 different murine genes (MG_U74Av2). Hierarchical clustering analysis of these genes readily shows an orderly pattern of gene responses at specific times consistent with the processes involved in the initial tissue injury and later regeneration of the parenchyma, as well as a prominent effect of MT-I + II deficiency. The results thoroughly confirmed the importance of the antioxidant proteins MT-I + II in the response of the brain to injury and opened new avenues that were confirmed by immunohistochemistry. Data in KO, MT-I-overexpressing, and MT-II-injected mice strongly suggest a role of these proteins in postlesional activation of neural stem cells.
Collapse
Affiliation(s)
- Milena Penkowa
- Section of Neuroprotection, Centre of Inflammation and Metabolism, The Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Novak JP, Kim SY, Xu J, Modlich O, Volsky DJ, Honys D, Slonczewski JL, Bell DA, Blattner FR, Blumwald E, Boerma M, Cosio M, Gatalica Z, Hajduch M, Hidalgo J, McInnes RR, Miller III MC, Penkowa M, Rolph MS, Sottosanto J, St-Arnaud R, Szego MJ, Twell D, Wang C. Generalization of DNA microarray dispersion properties: microarray equivalent of t-distribution. Biol Direct 2006; 1:27. [PMID: 16959036 PMCID: PMC1586001 DOI: 10.1186/1745-6150-1-27] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Accepted: 09/07/2006] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND DNA microarrays are a powerful technology that can provide a wealth of gene expression data for disease studies, drug development, and a wide scope of other investigations. Because of the large volume and inherent variability of DNA microarray data, many new statistical methods have been developed for evaluating the significance of the observed differences in gene expression. However, until now little attention has been given to the characterization of dispersion of DNA microarray data. RESULTS Here we examine the expression data obtained from 682 Affymetrix GeneChips with 22 different types and we demonstrate that the Gaussian (normal) frequency distribution is characteristic for the variability of gene expression values. However, typically 5 to 15% of the samples deviate from normality. Furthermore, it is shown that the frequency distributions of the difference of expression in subsets of ordered, consecutive pairs of genes (consecutive samples) in pair-wise comparisons of replicate experiments are also normal. We describe a consecutive sampling method, which is employed to calculate the characteristic function approximating standard deviation and show that the standard deviation derived from the consecutive samples is equivalent to the standard deviation obtained from individual genes. Finally, we determine the boundaries of probability intervals and demonstrate that the coefficients defining the intervals are independent of sample characteristics, variability of data, laboratory conditions and type of chips. These coefficients are very closely correlated with Student's t-distribution. CONCLUSION In this study we ascertained that the non-systematic variations possess Gaussian distribution, determined the probability intervals and demonstrated that the K(alpha) coefficients defining these intervals are invariant; these coefficients offer a convenient universal measure of dispersion of data. The fact that the K(alpha) distributions are so close to t-distribution and independent of conditions and type of arrays suggests that the quantitative data provided by Affymetrix technology give "true" representation of physical processes, involved in measurement of RNA abundance. REVIEWERS This article was reviewed by Yoav Gilad (nominated by Doron Lancet), Sach Mukherjee (nominated by Sandrine Dudoit) and Amir Niknejad and Shmuel Friedland (nominated by Neil Smalheiser).
Collapse
Affiliation(s)
- Jaroslav P Novak
- McGill University and Genome Québec Innovation Centre, 740 Docteur Penfield Avenue, Montreal, Québec, H3A 1A4, Canada
| | - Seon-Young Kim
- Human Genomics Laboratory, Genome Research Center, 52 Eoeun-dong, Yuseong-gu, Daejon, 305-333, Korea
| | - Jun Xu
- Transcriptional Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Olga Modlich
- Institut fur Onkologische Chemie, Heinrich Heine Universitat Dusseldorf, Moorenstr. 5, D-40225 Dusseldorf, Germany
| | - David J Volsky
- St. Luke's-Roosevelt Hospital Center and Columbia University, Molecular Virology Division, 432 West 58th Street, Antenucci Building, Room 709, New York, NY 10019, USA
| | - David Honys
- Institute of Experimental Botany AS CR, Rozvojová 135, CZ-165 02, Praha 6, Czech Republic and Charles University in Prague, Department of Plant Physiology, Viničná 5, 12844, Praha 2, Czech Republic
| | - Joan L Slonczewski
- Department of Biology, Higley Hall, 202 N. College Dr., Kenyon College, Gambier, OH 43022, USA
| | - Douglas A Bell
- Environmental Genomics Section, C3-03, PO Box 12233, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Fred R Blattner
- Department of Genetics, 425 Henry Mall, University of Wisconsin, Madison, WI 53706, USA
| | - Eduardo Blumwald
- Department of Plant Sciences, University of California, One Shields Ave, Davis, CA 95616, USA
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Slot 522-3, Little Rock AR 72205, USA
| | - Manuel Cosio
- Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Zoran Gatalica
- Department of Pathology, Creighton University School of Medicine, 601 North 30th Street, Omaha, NE, 68131-2197, USA
| | - Marian Hajduch
- Laboratory of Experimental Medicine, Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Puskinova 6, 775 20 Olomouc, Czech Republic
| | - Juan Hidalgo
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology unit, Faculty of Sciences, Autonomous University of Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Roderick R McInnes
- Programs in Genetics and Developmental Biology, The Research Institute, The Hospital for Sick Children, Toronto, Canada M5G 1X8; Departments of Molecular and Medical Genetics and Pediatrics, University of Toronto, Toronto, M5S 1A1, Canada
| | - Merrill C Miller III
- Environmental Genomics Section, C3-03, PO Box 12233, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Milena Penkowa
- Section of Neuroprotection, Centre of Inflammation and Metabolism, The Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen Denmark
| | - Michael S Rolph
- Arthritis and Inflammation Research Program, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst NSW 2010, Australia
| | - Jordan Sottosanto
- Department of Plant Sciences, University of California, One Shields Ave, Davis, CA 95616, USA
| | - Rene St-Arnaud
- Genetics Unit, Shriners Hospital for Children and Departments of Surgery and Human Genetics, McGill University, Montréal H3A 2T5, Québec, Canada
| | - Michael J Szego
- Programs in Genetics and Developmental Biology, The Research Institute, The Hospital for Sick Children, Toronto, Canada M5G 1X8; Departments of Molecular and Medical Genetics, University of Toronto, Toronto, M5S 1A1, Canada
| | - David Twell
- Department of Biology, University of Leicester, LE1 7RH Leicester, UK
| | - Charles Wang
- Transcriptional Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, Cedars-Sinai Medical Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90048, USA
| |
Collapse
|
37
|
Abstract
Metallothioneins (MTs) constitute a family of cysteine-rich metalloproteins involved in cytoprotection during pathology. In mammals there are four isoforms (MT-I - IV), of which MT-I and -II (MT-I + II) are the best characterized MT proteins in the brain. Accumulating studies have demonstrated MT-I + II as multipurpose factors important for host defense responses, immunoregulation, cell survival and brain repair. This review will focus on expression and roles of MT-I + II in the disordered brain. Initially, studies of genetically modified mice with MT-I + II deficiency or endogenous MT-I overexpression demonstrated the importance of MT-I + II for coping with brain pathology. In addition, exogenous MT-I or MT-II injected intraperitoneally is able to promote similar effects as those of endogenous MT-I + II, which indicates that MT-I + II have both extra- and intracellular actions. In injured brain, MT-I + II inhibit macrophages, T lymphocytes and their formation of interleukins, tumor necrosis factor-alpha, matrix metalloproteinases, and reactive oxygen species. In addition, MT-I + II enhance cell cycle progression, mitosis and cell survival, while neuronal apoptosis is inhibited. The precise mechanisms downstream of MT-I + II have not been fully established, but convincing data show that MT-I + II are essential for coping with neuropathology and for brain recovery. As MT-I and/or MT-II compounds are well tolerated, they may provide a potential therapy for a range of brain disorders.
Collapse
Affiliation(s)
- Milena Penkowa
- Section of Neuroprotection, Centre of Inflammation and Metabolism at The Faculty of Health Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
38
|
Abstract
1. Microarrays, a recent development, provide a revolutionary platform to analyse thousands of genes at once. They have enormous potential in the study of biological processes in health and disease and, perhaps, microarrays have become crucial tools in diagnostic applications and drug discovery. 2. Microarray based studies have provided the essential impetus for biomedical experiments, such as identification of disease-causing genes in malignancies and regulatory genes in the cell cycle mechanism. Microarrays can identify genes for new and unique potential drug targets, predict drug responsiveness for individual patients and, finally, initiate gene therapy and prevention strategies. 3. The present article reviews the principles and technological concerns, as well as the steps involved in obtaining and analysing of data. Furthermore, applications of microarray based experiments in drug target identifications and validation strategies are discussed. 4. To exemplify how this tool can be useful, in the present review we provide an overview of some of the past and potential future aspects of microarray technology and present a broad overview of this rapidly growing field.
Collapse
Affiliation(s)
- Manikandan Jayapal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
39
|
Quintana A, Giralt M, Rojas S, Penkowa M, Campbell IL, Hidalgo J, Molinero A. Differential role of tumor necrosis factor receptors in mouse brain inflammatory responses in cryolesion brain injury. J Neurosci Res 2006; 82:701-16. [PMID: 16267827 DOI: 10.1002/jnr.20680] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) is one of the mediators dramatically increased after traumatic brain injury that leads to the activation, proliferation, and hypertrophy of mononuclear, phagocytic cells and gliosis. Eventually, TNF-alpha can induce both apoptosis and necrosis via intracellular signaling. This cytokine exerts its functions via interaction with two receptors: type-1 receptor (TNFR1) and type-2 receptor (TNFR2). In this work, the inflammatory response after a freeze injury (cryolesion) in the cortex was studied in wild-type (WT) animals and in mice lacking TNFR1 (TNFR1 KO) or TNFR2 (TNFR2 KO). Lack of TNFR1, but not of TNFR2, significantly decreased the inflammatory response and tissue damage elicited by the cryolesion at both 3 and 7 days postlesion, with decreased gliosis, lower IL-1beta immunostaining, and a reduction of apoptosis markers. Cryolesion produced a clear induction of the proinflammatory cytokines interleukin (IL)-1alpha, IL-1beta, IL-6, and TNF-alpha; this induction was significantly lower in the TNFR1 KO mice. Host response genes (ICAM-1, A20, EB22/5, and GFAP) were also induced by the cryolesion, but to a lesser extent in TNFR1 KO mice. Lack of TNFR1 signaling also affected the expression of apoptosis/cell death-related genes (Fas, Rip, p53), matrix metalloproteinases (MMP3, MMP9, MMP12), and their inhibitors (TIMP1), suggesting a role of TNFR1 in extracellular matrix remodeling after injury. However, GDNF, NGF, and BDNF expression were not affected by TNFR1 deficiency. Overall, these results suggest that TNFR1 is involved in the early establishment of the inflammatory response and that its deficiency causes a decreased inflammatory response and tissue damage following brain injury.
Collapse
Affiliation(s)
- Albert Quintana
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology Unit, Faculty of Sciences, Autonomous University of Barcelona, Bellaterra, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Alesci S, Rodak M, Ilias I, Zhou R, Manji HK. The genomics of mood disorders. PROGRESS IN BRAIN RESEARCH 2006; 158:129-39. [PMID: 17027694 DOI: 10.1016/s0079-6123(06)58006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Salvatore Alesci
- Clinical Neuroendocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
41
|
Lindberg C, Hjorth E, Post C, Winblad B, Schultzberg M. Cytokine production by a human microglial cell line: effects of beta-amyloid and alpha-melanocyte-stimulating hormone. Neurotox Res 2005; 8:267-76. [PMID: 16371321 DOI: 10.1007/bf03033980] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Senile plaques in the Alzheimer's disease (AD) are formed by aggregation of beta-amyloid (Abeta) peptide. Abeta peptide has been shown to activate microglia and stimulate their production of inflammatory factors, such as cytokines. In the AD brain, the continued presence of amyloid plaques may keep microglia persistently activated, leading to chronic inflammation in the CNS. It is well established that alpha-melanocyte-stimulating hormone (alpha-MSH) gives rise to anti-inflammatory and anti-pyretic effects. The biological activities of alpha-MSH are mediated by one or more of the melanocortin receptor (MCR) subtypes, i.e. MCR1 - MCR5. The aim of the present study was to determine the effect of alpha-MSH alone and on Abeta-activated microglial cells with regard to the secretion of inflammatory cytokines, such as interleukin-6 (IL-6), and to determine which receptor subtype mediates the effects of alpha-MSH. The human microglial cell line, CHME3, was incubated for 24 h with freshly dissolved Abeta(1-40), interferon-gamma (IFN-gamma) and/or alpha-MSH. Freshly dissolved Abeta(1-40) (5-60 microM) resulted in a dose-dependent decrease in cell viability, along with a dose-dependent increase in IL-6 release. Neither IFN-gamma nor alpha-MSH affected the Abeta-induced secretion of IL-6, but resulted in a dose-dependent increase in basal IL-6 release. Agouti, the endogenous antagonist of MCR1 and 4, further increased the alpha-MSH-induced secretion of IL-6. RT-PCR showed the expression of MCR1, MCR3, MCR4 and MCR5 mRNA. The combined data suggest that the effect of alpha-MSH in increasing IL-6 release from the human microglial cell line is mediated by MCR3 or MCR5.
Collapse
Affiliation(s)
- Catharina Lindberg
- Karolinska Institutet, Neurotec Department, Division of Experimental Geriatrics, Karolinska University Hospital Huddinge, Novum, SE-141 86 Stockholm, Sweden
| | | | | | | | | |
Collapse
|