1
|
Oasa S, Chen G, Schultzberg M, Terenius L. Small Molecule Decoy of Amyloid-β Aggregation Blocks Activation of Microglia-Like Cells. J Alzheimers Dis 2024; 101:787-796. [PMID: 39240634 PMCID: PMC11492064 DOI: 10.3233/jad-231399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2024] [Indexed: 09/07/2024]
Abstract
Background Aggregated forms of the amyloid-β (Aβ) peptides which form protofibrils and fibrils in the brain are signatures of Alzheimer's disease (AD). Aggregates are also recognized by microglia, which in early phases may be protective and in later phases contribute to the pathology. We have identified several small molecules, decoys which interfere with Aβ oligomerization and induce other aggregation trajectories leading to aggregated macrostructures which are non-toxic. Objective This study investigates whether the small-molecule decoys affect microglial activation in terms of cytokine secretion and phagocytosis of Aβ peptide. Methods The effects of the decoys (NSC 69318, NSC 100873, NSC 16224) were analyzed in a model of human THP-1 monocytes differentiated to microglia-like cells. The cells were activated by Aβ40 and Aβ42 peptides, respectively, and after treatment with each decoy the secreted levels of pro-inflammatory cytokines and the Aβ phagocytosis were analyzed. Results NSC16224, which generates a double-stranded aggregate of thin protofibrils, was found to block Aβ40- and Aβ42-induced increase in microglial secretion of pro-inflammatory cytokines. NSC 69318, selective for neurotoxicity of Aβ42, and NSC 100873 did not significantly reduce the microglial activation in terms of cytokine secretion. The uptake of Aβ42 was not affected by anyone of the decoys. Conclusions Our findings open the possibility that the molecular decoys of Aβ aggregation may block microglial activation by Aβ40 and Aβ42 in addition to blocking neurotoxicity as shown previously.
Collapse
Affiliation(s)
- Sho Oasa
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gefei Chen
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Marianne Schultzberg
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Bioclinicum J10 : 30, Karolinska Institutet, Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Jahandideh A, Yarizadeh M, Noei-Khesht Masjedi M, Fatehnejad M, Jahandideh R, Soheili R, Eslami Y, Zokaei M, Ahmadvand A, Ghalamkarpour N, Kumar Pandey R, Nabi Afjadi M, Payandeh Z. Macrophage's role in solid tumors: two edges of a sword. Cancer Cell Int 2023; 23:150. [PMID: 37525217 PMCID: PMC10391843 DOI: 10.1186/s12935-023-02999-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023] Open
Abstract
The tumor microenvironment is overwhelmingly dictated by macrophages, intimately affiliated with tumors, exercising pivotal roles in multiple processes, including angiogenesis, extracellular matrix reconfiguration, cellular proliferation, metastasis, and immunosuppression. They further exhibit resilience to chemotherapy and immunotherapy via meticulous checkpoint blockades. When appropriately stimulated, macrophages can morph into a potent bidirectional component of the immune system, engulfing malignant cells and annihilating them with cytotoxic substances, thus rendering them intriguing candidates for therapeutic targets. As myelomonocytic cells relentlessly amass within tumor tissues, macrophages rise as prime contenders for cell therapy upon the development of chimeric antigen receptor effector cells. Given the significant incidence of macrophage infiltration correlated with an unfavorable prognosis and heightened resistance to chemotherapy in solid tumors, we delve into the intricate role of macrophages in cancer propagation and their promising potential in confronting four formidable cancer variants-namely, melanoma, colon, glioma, and breast cancers.
Collapse
Affiliation(s)
- Arian Jahandideh
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- Usern Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahsa Yarizadeh
- Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Maryam Noei-Khesht Masjedi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Fatehnejad
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Romina Jahandideh
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roben Soheili
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Yeganeh Eslami
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ardavan Ahmadvand
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nogol Ghalamkarpour
- Department of Clinical Laboratory Sciences, School of Allied Medicine, Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Rajan Kumar Pandey
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Zahra Payandeh
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
3
|
A Review on Phyto-Therapeutic Approaches in Alzheimer's Disease. J Funct Biomater 2023; 14:jfb14010050. [PMID: 36662097 PMCID: PMC9861153 DOI: 10.3390/jfb14010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/29/2022] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Neurodegenerative diseases occur due to progressive and sometimes irreversible loss of function and death of nerve cells. A great deal of effort is being made to understand the pathogenesis of neurodegenerative diseases. In particular, the prevalence of Alzheimer's disease (AD) is quite high, and only symptomatic therapy is available due to the absence of radical treatment. The aim of this review is to try to elucidate the general pathogenesis of AD, to provide information about the limit points of symptomatic treatment approaches, and to emphasize the potential neurologic effects of phytocompounds as new tools as therapeutic agents for disease prevention, retardation, and therapy. This survey also covers the notable properties of herbal compounds such as their effects on the inhibition of an enzyme called acetylcholinesterase, which has significant value in the treatment of AD. It has been proven that phytopharmaceuticals have long-term effects that could protect nervous system health, eliminate inflammatory responses, improve cognitive damage, provide anti-aging effects in the natural aging process, and alleviate dementia sequelae. Herbal-based therapeutic agents can afford many advantages and can be used as potentially as new-generation therapeutics or complementary agents with high compliance, fewer adverse effects, and lower cost in comparison to the traditional pharmaceutical agents in the fight against AD.
Collapse
|
4
|
The Pivotal Immunoregulatory Functions of Microglia and Macrophages in Glioma Pathogenesis and Therapy. JOURNAL OF ONCOLOGY 2022; 2022:8903482. [PMID: 35419058 PMCID: PMC9001141 DOI: 10.1155/2022/8903482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/24/2022] [Indexed: 12/16/2022]
Abstract
Gliomas are mixed solid tumors composed of both neoplastic and nonneoplastic cells. In glioma microenvironment, the most common nonneoplastic and infiltrating cells are macrophages and microglia. Microglia are the exact phagocytes of the central nervous system, whereas macrophages are myeloid immune cells that are depicted with ardent phagocytosis. Microglia are heterogeneously located in almost all nonoverlapping sections of the brain as well as the spinal cord, while macrophages are derived from circulating monocytes. Microglia and macrophages utilize a variety of receptors for the detection of molecules, particles, and cells that they engulf. Both microglia and peripheral macrophages interact directly with vessels both in the periphery of and within the tumor. In glioma milieu, normal human astrocytes, glioma cells, and microglia all exhibited the ability of phagocytosing glioma cells and precisely apoptotic tumor cells. Also, microglia and macrophages are robustly triggered by the glioma via the expression of chemoattractants such as monocyte chemoattractant protein, stromal-derived factor-1, and macrophage-colony stimulating factor. Glioma-associated microglia and/or macrophages positively correlated with glioma invasiveness, immunosuppression, and patients' poor outcome, making these cells a suitable target for immunotherapeutic schemes.
Collapse
|
5
|
Lee S, Choi WS. Protective Role of Microglia on Neuronal Survival after Exposure to Amyloid Beta. Chonnam Med J 2022; 58:13-17. [PMID: 35169554 PMCID: PMC8813657 DOI: 10.4068/cmj.2022.58.1.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 11/06/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of neurodegeneration. It is characterized by deposits of amyloid beta (Aβ) plaques and impaired memory. Microglia are associated with AD. They are activated in the AD brain and AD models. However, the exact role of microglia has not been established. We thus investigated the role of microglia in AD models using a primary culture and an ex-vivo assay. We showed that oligomerized Aβ is toxic to neurons in the primary culture. In the ex-vivo assay, a microglial cell line removed amyloid plaques in the brain of 5XFAD (AD model) mice. To verify if microglia can be protective for the neuron, we co-cultured neurons with primary microglia and treated them with Aβ. The loss of neurons, induced by amyloid toxicity, was attenuated by co-cultured microglia. Taken together, our data suggest that microglia promote neuronal survival by phagocytic clearance of Aβ in AD models.
Collapse
Affiliation(s)
- Sunjun Lee
- School of Biological Sciences and Technology, College of Natural Sciences, Chonnam National University, Gwangju, Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, College of Natural Sciences, Chonnam National University, Gwangju, Korea
| |
Collapse
|
6
|
Energy homeostasis deregulation is attenuated by TUDCA treatment in streptozotocin-induced Alzheimer's disease mice model. Sci Rep 2021; 11:18114. [PMID: 34518585 PMCID: PMC8437965 DOI: 10.1038/s41598-021-97624-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/27/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. While cognitive deficits remain the major manifestation of AD, metabolic and non-cognitive abnormalities, such as alterations in food intake, body weight and energy balance are also present, both in AD patients and animal models. In this sense, the tauroursodeoxycholic acid (TUDCA) has shown beneficial effects both in reducing the central and cognitive markers of AD, as well as in attenuating the metabolic disorders associated with it. We previously demonstrated that TUDCA improves glucose homeostasis and decreases the main AD neuromarkers in the streptozotocin-induced AD mouse model (Stz). Besides that, TUDCA-treated Stz mice showed lower body weight and adiposity. Here, we investigated the actions of TUDCA involved in the regulation of body weight and adiposity in Stz mice, since the effects of TUDCA in hypothalamic appetite control and energy homeostasis have not yet been explored in an AD mice model. The TUDCA-treated mice (Stz + TUDCA) displayed lower food intake, higher energy expenditure (EE) and respiratory quotient. In addition, we observed in the hypothalamus of the Stz + TUDCA mice reduced fluorescence and gene expression of inflammatory markers, as well as normalization of the orexigenic neuropeptides AgRP and NPY expression. Moreover, leptin-induced p-JAK2 and p-STAT3 signaling in the hypothalamus of Stz + TUDCA mice was improved, accompanied by reduced acute food intake after leptin stimulation. Taken together, we demonstrate that TUDCA treatment restores energy metabolism in Stz mice, a phenomenon that is associated with reduced food intake, increased EE and improved hypothalamic leptin signaling. These findings suggest treatment with TUDCA as a promising therapeutic intervention for the control of energy homeostasis in AD individuals.
Collapse
|
7
|
Ye JY, Hao Q, Zong Y, Shen Y, Zhang Z, Ma C. Sophocarpine Attenuates Cognitive Impairment and Promotes Neurogenesis in a Mouse Model of Alzheimer's Disease. Neuroimmunomodulation 2021; 28:166-177. [PMID: 34320497 DOI: 10.1159/000508655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/08/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Alzheimer's disease (AD), which is characterized by abnormal deposition of amyloid-β (Aβ) plaques and impaired neurogenesis and cognition, still lacks an optimally effective therapeutic agent for its management, and mounting evidence has shown that inflammatory processes are implicated in AD. Sophocarpine has been reported to exert inflammation-regulating effects in various diseases. However, whether sophocarpine can exert anti-neuroinflammatory and neuroprotective effects in AD remains unclear. This study investigated whether sophocarpine could ameliorate the pathological features and potential mechanisms in a mouse AD model. METHODS APP/PS1 mice were treated with sophocarpine for 8 weeks. We quantified the effects of sophocarpine treatment on cognitive performance using a behavioral test. Brain Aβ deposits and neurogenesis were evaluated using immunofluorescence staining. We also assessed the morphology and inflammatory changes induced by sophocarpine administration and its expression in the hippocampus. RESULTS Administration of sophocarpine significantly alleviated cognitive impairment and reduced neural loss. APP/PS1 mice treated with sophocarpine showed reduced Aβ plaque deposits and enhanced neurogenesis. Sophocarpine markedly decreased the expression of inflammation markers and inhibited microglial activation. CONCLUSIONS Sophocarpine could potentially alleviate cognitive impairment and brain damage in APP/PS1 mice with its neuroprotective effects via modulation of the inflammatory pathway.
Collapse
Affiliation(s)
- Jian-Ya Ye
- Department of Nursing, Hebei University of Chinese Medicine, Shijiazhang, China
| | - Qingmao Hao
- Department of Nursing, Hebei University of Chinese Medicine, Shijiazhang, China
| | - Yijun Zong
- Department of Nursing, Hebei University of Chinese Medicine, Shijiazhang, China
| | - Yongqing Shen
- Department of Nursing, Hebei University of Chinese Medicine, Shijiazhang, China
| | - Zhiqin Zhang
- Department of Nursing, Hebei University of Chinese Medicine, Shijiazhang, China
| | - Changsheng Ma
- Neurobiology Laboratory, Institute of Basic Medicine, Hebei Medical University, Shijiazhang, China
| |
Collapse
|
8
|
Kwon S, Moreno-Gonzalez I, Taylor-Presse K, Edwards Iii G, Gamez N, Calderon O, Zhu B, Velasquez FC, Soto C, Sevick-Muraca EM. Impaired Peripheral Lymphatic Function and Cerebrospinal Fluid Outflow in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 69:585-593. [PMID: 31104026 DOI: 10.3233/jad-190013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cerebrospinal fluid (CSF) outflow from the brain occurs through absorption into the arachnoid villi and, more predominantly, through meningeal and olfactory lymphatics that ultimately drain into the peripheral lymphatics. Impaired CSF outflow has been postulated as a contributing mechanism in Alzheimer's disease (AD). Herein we conducted near-infrared fluorescence imaging of CSF outflow into the peripheral lymph nodes (LNs) and of peripheral lymphatic function in a transgenic mouse model of AD (5XFAD) and wild-type (WT) littermates. CSF outflow was assessed from change in fluorescence intensity in the submandibular LNs as a function of time following bolus, an intrathecal injection of indocyanine green (ICG). Peripheral lymphatic function was measured by assessing lymphangion contractile function in lymphatics draining into the popliteal LN following intradermal ICG injection in the dorsal aspect of the hind paw. The results show 1) significantly impaired CSF outflow into the submandibular LNs of 5XFAD mice and 2) reduced contractile frequency in the peripheral lymphatics as compared to WT mice. Impaired CSF clearance was also evidenced by reduction of fluorescence on ventral surfaces of extracted brains of 5XFAD mice at euthanasia. These results support the hypothesis that lymphatic congestion caused by reduced peripheral lymphatic function could limit CSF outflow and may contribute to the cause and/or progression of AD.
Collapse
Affiliation(s)
- Sunkuk Kwon
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Ines Moreno-Gonzalez
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Kathleen Taylor-Presse
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - George Edwards Iii
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Nazaret Gamez
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Olivia Calderon
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Banghe Zhu
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Fred Christian Velasquez
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Claudio Soto
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Eva M Sevick-Muraca
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| |
Collapse
|
9
|
Simvastatin Prevents Long-Term Cognitive Deficits in Sepsis Survivor Rats by Reducing Neuroinflammation and Neurodegeneration. Neurotox Res 2020; 38:871-886. [PMID: 32524380 DOI: 10.1007/s12640-020-00222-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023]
Abstract
Sepsis-associated encephalopathy causes brain dysfunction that can result in cognitive impairments in sepsis survivor patients. In previous work, we showed that simvastatin attenuated oxidative stress in brain structures related to memory in septic rats. However, there is still a need to evaluate the long-term impact of simvastatin administration on brain neurodegenerative processes and cognitive damage in sepsis survivors. Here, we investigated the possible neuroprotective role of simvastatin in neuroinflammation, and neurodegeneration conditions of brain structures related to memory in rats at 10 days after sepsis survival. Male Wistar rats (250-300 g) were submitted to cecal ligation and puncture (CLP, n = 42) or remained as non-manipulated (naïve, n = 30). Both groups were treated (before and after the surgery) by gavage with simvastatin (20 mg/kg) or an equivalent volume of saline and observed for 10 days. Simvastatin-treated rats that survived to sepsis showed a reduction in the levels of nitrate, IL1-β, and IL-6 and an increase in Bcl-2 protein expression in the prefrontal cortex and hippocampus, and synaptophysin only in the hippocampus. Immunofluorescence revealed a reduction of glial activation, neurodegeneration, apoptosis, and amyloid aggregates confirmed by quantification of GFAP, Iba-1, phospho Ser396-tau, total tau, cleaved caspase-3, and thioflavin-S in the prefrontal cortex and hippocampus. In addition, treated animals presented better performance in tasks involving habituation memory, discriminative, and aversive memory. These results suggest that statins exert a neuroprotective role by upregulation of the Bcl-2 and gliosis reduction, which may prevent the cognitive deficit observed in sepsis survivor animals.
Collapse
|
10
|
Zhou Z, Hou J, Mo Y, Ren M, Yang G, Qu Z, Hu Y. Geniposidic acid ameliorates spatial learning and memory deficits and alleviates neuroinflammation via inhibiting HMGB-1 and downregulating TLR4/2 signaling pathway in APP/PS1 mice. Eur J Pharmacol 2020; 869:172857. [DOI: 10.1016/j.ejphar.2019.172857] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022]
|
11
|
Uyaniker S, van der Spek SJF, Reinders NR, Xiong H, Li KW, Bossers K, Smit AB, Verhaagen J, Kessels HW. The Effects of Sindbis Viral Vectors on Neuronal Function. Front Cell Neurosci 2019; 13:362. [PMID: 31440143 PMCID: PMC6694438 DOI: 10.3389/fncel.2019.00362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/25/2019] [Indexed: 11/16/2022] Open
Abstract
Viral vectors are attractive tools to express genes in neurons. Transduction of neurons with a recombinant, replication-deficient Sindbis viral vector is a method of choice for studying the effects of short-term protein overexpression on neuronal function. However, to which extent Sindbis by itself may affect neurons is not fully understood. We assessed effects of neuronal transduction with a Sindbis viral vector on the transcriptome and proteome in organotypic hippocampal slice cultures, and analyzed the electrophysiological properties of individual CA1 neurons, at 24 h and 72 h after viral vector injection. Whereas Sindbis caused substantial gene expression alterations, changes at the protein level were less pronounced. Alterations in transcriptome and proteome were predominantly limited to proteins involved in mediating anti-viral innate immune responses. Sindbis transduction did not affect the intrinsic electrophysiological properties of individual neurons: the membrane potential and neuronal excitability were similar between transduced and non-transduced CA1 neurons up to 72 h after Sindbis injection. Synaptic currents also remained unchanged upon Sindbis transduction, unless slices were massively infected for 72 h. We conclude that Sindbis viral vectors at low transduction rates are suitable for studying short-term effects of a protein of interest on electrophysiological properties of neurons, but not for studies on the regulation of gene expression.
Collapse
Affiliation(s)
- Seçil Uyaniker
- Laboratory for Neuroregeneration, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Sophie J F van der Spek
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, Netherlands
| | - Niels R Reinders
- Laboratory for Neuroregeneration, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Cellular and Computational Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Hui Xiong
- Laboratory for Neuroregeneration, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Cellular and Computational Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, Netherlands
| | - Koen Bossers
- Laboratory for Neuroregeneration, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, Netherlands
| | - Helmut W Kessels
- Laboratory for Neuroregeneration, The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Cellular and Computational Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
Rustenhoven J, Smith AM, Smyth LC, Jansson D, Scotter EL, Swanson MEV, Aalderink M, Coppieters N, Narayan P, Handley R, Overall C, Park TIH, Schweder P, Heppner P, Curtis MA, Faull RLM, Dragunow M. PU.1 regulates Alzheimer's disease-associated genes in primary human microglia. Mol Neurodegener 2018; 13:44. [PMID: 30124174 PMCID: PMC6102813 DOI: 10.1186/s13024-018-0277-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/10/2018] [Indexed: 12/16/2022] Open
Abstract
Background Microglia play critical roles in the brain during homeostasis and pathological conditions. Understanding the molecular events underpinning microglial functions and activation states will further enable us to target these cells for the treatment of neurological disorders. The transcription factor PU.1 is critical in the development of myeloid cells and a major regulator of microglial gene expression. In the brain, PU.1 is specifically expressed in microglia and recent evidence from genome-wide association studies suggests that reductions in PU.1 contribute to a delayed onset of Alzheimer’s disease (AD), possibly through limiting neuroinflammatory responses. Methods To investigate how PU.1 contributes to immune activation in human microglia, microarray analysis was performed on primary human mixed glial cultures subjected to siRNA-mediated knockdown of PU.1. Microarray hits were confirmed by qRT-PCR and immunocytochemistry in both mixed glial cultures and isolated microglia following PU.1 knockdown. To identify attenuators of PU.1 expression in microglia, high throughput drug screening was undertaken using a compound library containing FDA-approved drugs. NanoString and immunohistochemistry was utilised to investigate the expression of PU.1 itself and PU.1-regulated mediators in primary human brain tissue derived from neurologically normal and clinically and pathologically confirmed cases of AD. Results Bioinformatic analysis of gene expression upon PU.1 silencing in mixed glial cultures revealed a network of modified AD-associated microglial genes involved in the innate and adaptive immune systems, particularly those involved in antigen presentation and phagocytosis. These gene changes were confirmed using isolated microglial cultures. Utilising high throughput screening of FDA-approved compounds in mixed glial cultures we identified the histone deacetylase inhibitor vorinostat as an effective attenuator of PU.1 expression in human microglia. Further characterisation of vorinostat in isolated microglial cultures revealed gene and protein changes partially recapitulating those seen following siRNA-mediated PU.1 knockdown. Lastly, we demonstrate that several of these PU.1-regulated genes are expressed by microglia in the human AD brain in situ. Conclusions Collectively, these results suggest that attenuating PU.1 may be a valid therapeutic approach to limit microglial-mediated inflammatory responses in AD and demonstrate utility of vorinostat for this purpose. Electronic supplementary material The online version of this article (10.1186/s13024-018-0277-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Amy M Smith
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Leon C Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Molly E V Swanson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, New Zealand
| | - Miranda Aalderink
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Natacha Coppieters
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, New Zealand
| | - Pritika Narayan
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand.,School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Renee Handley
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand.,School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Chris Overall
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, USA.,Departmemt of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas I H Park
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, New Zealand
| | | | | | - Maurice A Curtis
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand. .,Centre for Brain Research, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
13
|
Cumaoğlu A, Yerer MB. The Effects of Aldose Reductase Inhibitor Quercetin and Monochloropivaloylquercetin in Amyloid β Peptide (1–42) Induced Neuroinflammation in Microglial Cells. Nat Prod Commun 2018. [DOI: 10.1177/1934578x1801300611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Microglial over-activation plays a crucial roles during neuroinflammation. Aldose reductase (AR) is one of the enzymes that has been linked to inflammatory processes in several diseases. Therefore, inhibition of AR is considered as an important strategy to reduce inflammation. In the present study, Quercetin (Q) and monochloropivaloylquercetin (MCPQ) showed potent inhibition on AR expression and anti-neuroinflammatory effects in Amyloid β (Aβ) peptide (1–42) induced inflammatory process by inhibiting expression of inflammatory mediators from microglial cells. Furthermore, ablation of AR caused a significant reduction on COX2 expression in Aβ-induced neuroinflammation. Q and MCPQ suppressed COX2 mRNA and protein expression, which further resulted in downstream inhibition of prostaglandin E2 (PGE2) release in Aβ-induced neuroinflammatory process. Additionally, Aβ treatment resulted in activation of Mitogen Activated Protein Kinase (MAPK) and increased translocation of Nuclear Factor Kappa B (NFκB). Q and Sorbinil significantly reduced the activation of MAPK, at the same time Q, MCPQ and sorbinil decreased nuclear translocation of NFκB and diminished tumor necrosis factor (TNF)-α release in Aβ-induced neuroinflammation. The results suggested that AR is a probable target for treatment of neuroinflammation as well as Q and MCPQ could be effective agents for treating or preventing inflammation-related neurodegenerative diseases by AR inhibition.
Collapse
Affiliation(s)
- Ahmet Cumaoğlu
- Department of Biochemistry, Faculty of Pharmacy, Erciyes University, Kayseri, 38039, Turkey
| | - Mükerrem Betül Yerer
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, 38039, Turkey
| |
Collapse
|
14
|
Kim J, Kwon YH, Kim CS, Tu TH, Kim BS, Joe Y, Chung HT, Goto T, Kawada T, Park T, Choi MS, Kim MS, Yu R. The involvement of 4-1BB/4-1BBL signaling in glial cell-mediated hypothalamic inflammation in obesity. FEBS Open Bio 2018; 8:843-853. [PMID: 29744298 PMCID: PMC5929936 DOI: 10.1002/2211-5463.12426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/09/2018] [Accepted: 03/29/2018] [Indexed: 01/12/2023] Open
Abstract
Obesity‐induced inflammation occurs not only in peripheral tissues but also in areas of the central nervous system. Glial cells such as astrocytes and microglia play crucial roles in obesity‐related hypothalamic inflammation, leading to the derangement of energy metabolism and neurodegenerative pathologies. Here, we show that the interaction of 4‐1BB/4‐1BBL between lipid‐laden astrocytes/microglia promotes hypothalamic inflammation in obesity. Stimulation of 4‐1BB, a member of the TNF receptor superfamily, and/or its ligand 4‐1BBL on astrocytes and/or microglia with a specific agonist resulted in activation of the inflammatory signaling pathway and enhanced production of inflammatory mediators. Contact coculture of lipid‐laden astrocytes and microglia increased the production of inflammatory mediators, and blockade of the 4‐1BB/4‐1BBL interaction reduced the inflammatory response. Moreover, deficiency of 4‐1BB reduced hypothalamic inflammation in obese mice fed an high‐fat diet. These findings suggest that 4‐1BBL/4‐1BB signaling enhances the glial cell‐mediated inflammatory cross talk and participates in obesity‐induced hypothalamic inflammation.
Collapse
Affiliation(s)
- Jiye Kim
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Yoon-Hee Kwon
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Chu-Sook Kim
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Thai H Tu
- Department of Food Science and Nutrition University of Ulsan South Korea
| | - Byung-Sam Kim
- Department of Biological Science University of Ulsan South Korea
| | - Yeonsoo Joe
- Department of Biological Science University of Ulsan South Korea
| | - Hun T Chung
- Department of Biological Science University of Ulsan South Korea
| | - Tsuyoshi Goto
- Graduate School of Agriculture Kyoto University Uji Japan
| | - Teruo Kawada
- Graduate School of Agriculture Kyoto University Uji Japan
| | - Taesun Park
- Department of Food and Nutrition Yonsei University Seoul South Korea
| | - Myung-Sook Choi
- Department of Food Science and Nutrition Center for Food and Nutritional Genomics Research Kyungpook National University Daegu South Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism University of Ulsan College of Medicine Seoul South Korea
| | - Rina Yu
- Department of Food Science and Nutrition University of Ulsan South Korea
| |
Collapse
|
15
|
Schultz R, Krug M, Precht M, Wohl SG, Witte OW, Schmeer C. Frataxin overexpression in Müller cells protects retinal ganglion cells in a mouse model of ischemia/reperfusion injury in vivo. Sci Rep 2018; 8:4846. [PMID: 29555919 PMCID: PMC5859167 DOI: 10.1038/s41598-018-22887-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/02/2018] [Indexed: 01/28/2023] Open
Abstract
Müller cells are critical for retinal function and neuronal survival but can become detrimental in response to retinal ischemia and increased oxidative stress. Elevated oxidative stress increases expression of the mitochondrial enzyme frataxin in the retina, and its overexpression is neuroprotective after ischemia. Whether frataxin expression in Müller cells might improve their function and protect neurons after ischemia is unknown. The aim of this study was to evaluate the effect of frataxin overexpression in Müller cells on neuronal survival after retinal ischemia/reperfusion in the mouse in vivo. Retinal ischemia/reperfusion was induced in mice overexpressing frataxin in Müller cells by transient elevation of intraocular pressure. Retinal ganglion cells survival was determined 14 days after lesion. Expression of frataxin, antioxidant enzymes, growth factors and inflammation markers was determined with qRT-PCR, Western blotting and immunohistochemistry 24 hours after lesion. Following lesion, there was a 65% increase in the number of surviving RGCs in frataxin overexpressing mice. Improved survival was associated with increased expression of the antioxidant enzymes Gpx1 and Sod1 as well as the growth factors Cntf and Lif. Additionally, microglial activation was decreased in these mice. Therefore, support of Müller cell function constitutes a feasible approach to reduce neuronal degeneration after ischemia.
Collapse
Affiliation(s)
- Rowena Schultz
- Department of Ophthalmology, Jena University Hospital, Jena, Germany
| | - Melanie Krug
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Michel Precht
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Stefanie G Wohl
- Department of Biological Structure, University of Washington Seattle, Seattle, United States
| | - Otto W Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christian Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
16
|
The Effect of Osteopontin on Microglia. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1879437. [PMID: 28698867 PMCID: PMC5494082 DOI: 10.1155/2017/1879437] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/29/2017] [Accepted: 05/24/2017] [Indexed: 12/16/2022]
Abstract
Osteopontin (OPN) is a proinflammatory cytokine that can be secreted from many cells, including activated macrophages and T-lymphocytes, and is widely distributed in many tissues and cells. OPN, a key factor in tissue repairing and extracellular matrix remodeling after injury, is a constituent of the extracellular matrix of the central nervous system (CNS). Recently, the role of OPN in neurodegenerative diseases has gradually caused widespread concern. Microglia are resident macrophage-like immune cells in CNS and play a vital role in both physiological and pathological conditions, including restoring the integrity of the CNS and promoting the progression of neurodegenerative disorders. Microglia's major function is to maintain homeostasis and the normal function of the CNS, both during development and in response to CNS injury. Although the functional mechanism of OPN in CNS neurodegenerative diseases has yet to be fully elucidated, most studies suggest that OPN play a role in pathogenesis of neurodegenerative diseases or in neuroprotection by regulating the activation and function of microglia. Here, we summarize the functions of OPN on microglia in response to various stimulations in vitro and in vivo.
Collapse
|
17
|
Kata D, Földesi I, Feher LZ, Hackler L, Puskas LG, Gulya K. A novel pleiotropic effect of aspirin: Beneficial regulation of pro- and anti-inflammatory mechanisms in microglial cells. Brain Res Bull 2017; 132:61-74. [PMID: 28528204 DOI: 10.1016/j.brainresbull.2017.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 04/27/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Aspirin, one of the most widely used non-steroidal anti-inflammatory drugs, has extensively studied effects on the cardiovascular system. To reveal further pleiotropic, beneficial effects of aspirin on a number of pro- and anti-inflammatory microglial mechanisms, we performed morphometric and functional studies relating to phagocytosis, pro- and anti-inflammatory cytokine production (IL-1β, tumor necrosis factor-α (TNF-α) and IL-10, respectively) and analyzed the expression of a number of inflammation-related genes, including those related to the above functions, in pure microglial cells. We examined the effects of aspirin (0.1mM and 1mM) in unchallenged (control) and bacterial lipopolysaccharide (LPS)-challenged secondary microglial cultures. Aspirin affected microglial morphology and functions in a dose-dependent manner as it inhibited LPS-elicited microglial activation by promoting ramification and the inhibition of phagocytosis in both concentrations. Remarkably, aspirin strongly reduced the pro-inflammatory IL-1β and TNF-α production, while it increased the anti-inflammatory IL-10 level in LPS-challenged cells. Moreover, aspirin differentially regulated the expression of a number of inflammation-related genes as it downregulated such pro-inflammatory genes as Nos2, Kng1, IL1β, Ptgs2 or Ccr1, while it upregulated some anti-inflammatory genes such as IL10, Csf2, Cxcl1, Ccl5 or Tgfb1. Thus, the use of aspirin could be beneficial for the prophylaxis of certain neurodegenerative disorders as it effectively ameliorates inflammation in the brain.
Collapse
Affiliation(s)
- Diana Kata
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary.
| | - Imre Földesi
- Department of Laboratory Medicine, University of Szeged, Szeged, Hungary.
| | | | | | | | - Karoly Gulya
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary.
| |
Collapse
|
18
|
Brain Endothelial- and Epithelial-Specific Interferon Receptor Chain 1 Drives Virus-Induced Sickness Behavior and Cognitive Impairment. Immunity 2016; 44:901-12. [PMID: 27096319 DOI: 10.1016/j.immuni.2016.04.005] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 10/19/2015] [Accepted: 01/05/2016] [Indexed: 12/22/2022]
Abstract
Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.
Collapse
|
19
|
Daulatzai MA. Dysfunctional Sensory Modalities, Locus Coeruleus, and Basal Forebrain: Early Determinants that Promote Neuropathogenesis of Cognitive and Memory Decline and Alzheimer’s Disease. Neurotox Res 2016; 30:295-337. [DOI: 10.1007/s12640-016-9643-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
|
20
|
Jamalidoust M, Ravanshad M, Namayandeh M, Zare M, Asaei S, Ziyaeyan M. Construction of AAV-rat-IL4 and Evaluation of its Modulating Effect on Aβ (1-42)-Induced Proinflammatory Cytokines in Primary Microglia and the B92 Cell Line by Quantitative PCR Assay. Jundishapur J Microbiol 2016; 9:e30444. [PMID: 27217922 PMCID: PMC4870549 DOI: 10.5812/jjm.30444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/27/2015] [Accepted: 09/29/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Interleukin-4 (IL-4), as the most prominent anti-inflammatory cytokine, plays an important role in modulating microglial activation and inflammatory responses in Alzheimer's disease (AD), a chronic inflammatory disorder. OBJECTIVES The current study aimed to develop a new recombinant Adeno-associated viral (rAAV) vector that delivers IL-4 and then assess the counterbalancing effect of the new construct along with recombinant IL-4 (rIL-4) protein in in-vitro models of AD. MATERIALS AND METHODS The rAAV-IL4 was originally prepared and then employed along with rIL-4 protein to counter Amyloid β (1-42)-induced proinflammatory cytokines in a primary microglia cell culture and the B92 rat microglia continuous cell line, using relative Real-Time PCR assay. RESULTS Aβ (1-42) stimulated the production of the proinflammatory cytokines IL6, IL1β, TNFα, and IL18 in both the primary microglia cell culture and the B92 cell line. Both the rAAV-IL4 construct and the rIL-4 protein were found to inhibit production of the most important Aβ (1-42)-induced proinflammatory cytokine mRNAs in the two types of cells with different patterns. CONCLUSIONS It seems that the new construct can serve as an appropriate option in the modulation of Aβ-induced proinflammatory cytokine gene expression and microglia activation in patients affected by AD.
Collapse
Affiliation(s)
- Marzieh Jamalidoust
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Mehrdad Ravanshad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
- Corresponding author: Mehrdad Ravanshad, Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran. Tel: +98-2182883836, Fax: +98-2188013030, E-mail:
| | - Mandana Namayandeh
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Maryam Zare
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Sadaf Asaei
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Mazyar Ziyaeyan
- Alborzi Clinical Microbiology Research Center, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| |
Collapse
|
21
|
Montesinos J, Pascual M, Rodríguez-Arias M, Miñarro J, Guerri C. Involvement of TLR4 in the long-term epigenetic changes, rewarding and anxiety effects induced by intermittent ethanol treatment in adolescence. Brain Behav Immun 2016; 53:159-171. [PMID: 26686767 DOI: 10.1016/j.bbi.2015.12.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 12/11/2022] Open
Abstract
Studies in humans and experimental animals have demonstrated the vulnerability of the adolescent brain to actions of ethanol and the long-term consequences of binge drinking, including the behavioral and cognitive deficits that result from alcohol neurotoxicity, and increased risk to alcohol abuse and dependence. Although the mechanisms that participate in these effects are largely unknown, we have shown that ethanol by activating innate immune receptors, toll-like receptor 4 (TLR4), induces neuroinflammation, impairs myelin proteins and causes cognitive dysfunctions in adolescent mice. Since neuroimmune signaling is also involved in alcohol abuse, the aim of this study was to assess whether ethanol treatment in adolescence promotes the long-term synaptic and molecular events associated with alcohol abuse and addiction. Using wild-type (WT) and TLR4-deficient (TLR4-KO) adolescent mice treated intermittently with ethanol (3g/kg) for 2 weeks, we showed that binge-like ethanol treatment in adolescent mice promotes short- and long-term alterations in synaptic plasticity and epigenetic changes in the promoter region of bdnf and fosb, which increased their expression in the mPFC of young adult animals. These molecular events were associated with long-term rewarding and anxiogenic-related behavioral effects, along with increased alcohol preference. Our results further showed the participation of neuroimmune system activation and the TLR4 signaling response since deficient mice in TLR4 (TLR4-KO) are protected against molecular and behavioral alterations of ethanol in the adolescent brain. Our results highlight a new role of the neuroimmune function and open up new avenues to develop pharmacological treatments that can normalize the immune signaling responsible for long-term effects in adolescence, including alcohol abuse and related disorders.
Collapse
Affiliation(s)
- Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Jose Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012 Valencia, Spain.
| |
Collapse
|
22
|
von Bernhardi R, Cornejo F, Parada GE, Eugenín J. Role of TGFβ signaling in the pathogenesis of Alzheimer's disease. Front Cell Neurosci 2015; 9:426. [PMID: 26578886 PMCID: PMC4623426 DOI: 10.3389/fncel.2015.00426] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/09/2015] [Indexed: 12/19/2022] Open
Abstract
Aging is the main risk factor for Alzheimer’s disease (AD); being associated with conspicuous changes on microglia activation. Aged microglia exhibit an increased expression of cytokines, exacerbated reactivity to various stimuli, oxidative stress, and reduced phagocytosis of β-amyloid (Aβ). Whereas normal inflammation is protective, it becomes dysregulated in the presence of a persistent stimulus, or in the context of an inflammatory environment, as observed in aging. Thus, neuroinflammation can be a self-perpetuating deleterious response, becoming a source of additional injury to host cells in neurodegenerative diseases. In aged individuals, although transforming growth factor β (TGFβ) is upregulated, its canonical Smad3 signaling is greatly reduced and neuroinflammation persists. This age-related Smad3 impairment reduces protective activation while facilitating cytotoxic activation of microglia through several cellular mechanisms, potentiating microglia-mediated neurodegeneration. Here, we critically discuss the role of TGFβ-Smad signaling on the cytotoxic activation of microglia and its relevance in the pathogenesis of AD. Other protective functions, such as phagocytosis, although observed in aged animals, are not further induced by inflammatory stimuli and TGFβ1. Analysis in silico revealed that increased expression of receptor scavenger receptor (SR)-A, involved in Aβ uptake and cell activation, by microglia exposed to TGFβ, through a Smad3-dependent mechanism could be mediated by transcriptional co-factors Smad2/3 over the MSR1 gene. We discuss that changes of TGFβ-mediated regulation could at least partially mediate age-associated microglia changes, and, together with other changes on inflammatory response, could result in the reduction of protective activation and the potentiation of cytotoxicity of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Francisca Cornejo
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Guillermo E Parada
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Jaime Eugenín
- Laboratory of Neural Systems, Faculty of Chemistry and Biology, Department of Biology, Universidad de Santiago de Chile Santiago, Chile
| |
Collapse
|
23
|
Majd S, Power JH, Grantham HJM. Neuronal response in Alzheimer's and Parkinson's disease: the effect of toxic proteins on intracellular pathways. BMC Neurosci 2015; 16:69. [PMID: 26499115 PMCID: PMC4619058 DOI: 10.1186/s12868-015-0211-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 10/13/2015] [Indexed: 01/09/2023] Open
Abstract
Accumulation of protein aggregates is the leading cause of cellular dysfunction in neurodegenerative disorders. Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease, Prion disease and motor disorders such as amyotrophic lateral sclerosis, present with a similar pattern of progressive neuronal death, nervous system deterioration and cognitive impairment. The common characteristic is an unusual misfolding of proteins which is believed to cause protein deposition and trigger degenerative signals in the neurons. A similar clinical presentation seen in many neurodegenerative disorders suggests the possibility of shared neuronal responses in different disorders. Despite the difference in core elements of deposits in each neurodegenerative disorder, the cascade of neuronal reactions such as activation of glycogen synthase kinase-3 beta, mitogen-activated protein kinases, cell cycle re-entry and oxidative stress leading to a progressive neurodegeneration are surprisingly similar. This review focuses on protein toxicity in two neurodegenerative diseases, AD and PD. We reviewed the activated mechanisms of neurotoxicity in response to misfolded beta-amyloid and α-synuclein, two major toxic proteins in AD and PD, leading to neuronal apoptosis. The interaction between the proteins in producing an overlapping pathological pattern will be also discussed.
Collapse
Affiliation(s)
- Shohreh Majd
- Centre for Neuroscience and Paramedic Unit, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| | - John H Power
- Department of Human Physiology, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| | - Hugh J M Grantham
- Centre for Neuroscience and Paramedic Unit, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| |
Collapse
|
24
|
Meadowcroft MD, Connor JR, Yang QX. Cortical iron regulation and inflammatory response in Alzheimer's disease and APPSWE/PS1ΔE9 mice: a histological perspective. Front Neurosci 2015; 9:255. [PMID: 26257600 PMCID: PMC4511841 DOI: 10.3389/fnins.2015.00255] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/10/2015] [Indexed: 12/25/2022] Open
Abstract
Disruption of iron homeostasis and increased glial response are known to occur in brains afflicted by Alzheimer's disease (AD). While the APP/PS1 transgenic mouse model recapitulates the hallmark amyloid-beta plaque pathology of AD, it does so in a different neuronal mileu than humans. Understanding the iron characteristics and glial response of the APP/PS1 model is important when testing new treatment procedures and translating these results. Brain tissue from AD patients, APP/PS1 mice, and controls were stained for iron, H- and L-ferritin, microglia, astrocytes, Aβ40∕42, and degenerating neurons. The histological data demonstrate differences in ferritin, iron distribution, gliosis, and Aβ plaque composition between APP/PS1 and AD tissue. Specifically, an association between focal iron deposition and Aβ plaques is found ubiquitously throughout the AD tissue and is not observed in the APP/PS1 mouse model. Ferritin, microglia, and astrocyte staining show differential response patterns to amyloid plaques in AD and the APP/PS1 tissue. Aβ 40 and 42 antibody and thioflavin staining demonstrate morphological differences in plaque composition. The histological data support the hypothesis that iron distribution, iron management, and glial response histologically differ between the APP/PS1 and AD brain. Acknowledging the caveat that there are distinct plaque, iron, and glial contrasts between the AD brain and the APP/PS1 mouse is crucial when utilizing this model.
Collapse
Affiliation(s)
- Mark D Meadowcroft
- Department of Neurosurgery, Milton S. Hershey Medical Center, The Pennsylvania State University - College of Medicine Hershey, PA, USA ; Department of Radiology (The Center for NMR Research), Milton S. Hershey Medical Center, The Pennsylvania State University - College of Medicine Hershey, PA, USA
| | - James R Connor
- Department of Neurosurgery, Milton S. Hershey Medical Center, The Pennsylvania State University - College of Medicine Hershey, PA, USA
| | - Qing X Yang
- Department of Neurosurgery, Milton S. Hershey Medical Center, The Pennsylvania State University - College of Medicine Hershey, PA, USA ; Department of Radiology (The Center for NMR Research), Milton S. Hershey Medical Center, The Pennsylvania State University - College of Medicine Hershey, PA, USA
| |
Collapse
|
25
|
Bishnoi RJ, Palmer RF, Royall DR. Serum interleukin (IL)-15 as a biomarker of Alzheimer's disease. PLoS One 2015; 10:e0117282. [PMID: 25710473 PMCID: PMC4339977 DOI: 10.1371/journal.pone.0117282] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 12/19/2014] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL-15), a pro-inflammatory cytokine has been studied as a possible marker of Alzheimer’s disease (AD); however its exact role in neuro-inflammation or the pathogenesis AD is not well understood yet. A Multiple Indicators Multiple Causes (MIMIC) approach was used to examine the relationship between serum IL-15 levels and AD in a well characterized AD cohort, the Texas Alzheimer's Research and Care Consortium (TARCC). Instead of categorical diagnoses, we used two latent construct d (for dementia) and g’ (for cognitive impairments not contributing to functional impairments) in our analysis. The results showed that the serum IL-15 level has significant effects on cognition, exclusively mediated by latent construct d and g’. Contrasting directions of association lead us to speculate that IL-15’s effects in AD are mediated through functional networks as d scores have been previously found to be specifically related to default mode network (DMN). Our finding warrants the need for further research to determine the changes in structural and functional networks corresponding to serum based biomarkers levels.
Collapse
Affiliation(s)
- Ram J. Bishnoi
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail:
| | - Raymond F. Palmer
- Department of Family and Community Medicine, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Donald R. Royall
- Department of Psychiatry, Family and Community Medicine, and Medicine, University of Texas Health Science Center, South Texas Veterans’ Health System Audie L. Murphy Division, Geriatric Research Education and Clinical Centers, San Antonio, Texas, United States of America
| |
Collapse
|
26
|
Lim C, Hammond CJ, Hingley ST, Balin BJ. Chlamydia pneumoniae infection of monocytes in vitro stimulates innate and adaptive immune responses relevant to those in Alzheimer's disease. J Neuroinflammation 2014; 11:217. [PMID: 25540075 PMCID: PMC4295513 DOI: 10.1186/s12974-014-0217-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/07/2014] [Indexed: 11/24/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a progressive neurodegenerative disorder in which infection with Chlamydia pneumoniae (Cpn) has been associated. Cpn is an obligate intracellular respiratory pathogen that may enter the central nervous system (CNS) following infection and trafficking of monocytes through the blood-brain barrier. Following this entry, these cells may secrete pro-inflammatory cytokines and chemokines that have been identified in the AD brain, which have been thought to contribute to AD neurodegeneration. The objectives of this work were: (i) to determine if Cpn infection influences monocyte gene transcript expression at 48 hours post-infection and (ii) to analyze whether pro-inflammatory cytokines are produced and secreted from these cells over 24 to 120 hours post-infection. Methods Gene transcription was analyzed by RT-PCR using an innate and adaptive immunity microarray with 84 genes organized into 5 functional categories: inflammatory response, host defense against bacteria, antibacterial humoral response, septic shock, and cytokines, chemokines and their receptors. Statistical analysis of the results was performed using the Student's t-test. P-values ≤ 0.05 were considered to be significant. ELISA was performed on supernatants from uninfected and Cpn-infected THP1 monocytes followed by statistical analysis with ANOVA. Results When Cpn-infected THP1 human monocytes were compared to control uninfected monocytes at 48 hours post-infection, 17 genes were found to have a significant 4-fold or greater expression, and no gene expression was found to be down-regulated. Furthermore, cytokine secretion (IL-1β, IL-6, IL-8) appears to be maintained for an extended period of infection. Conclusions Utilizing RT-PCR and ELISA techniques, our data demonstrate that Cpn infection of THP1 human monocytes promotes an innate immune response and suggests a potential role in the initiation of inflammation in sporadic/late-onset Alzheimer’s disease.
Collapse
Affiliation(s)
- Charles Lim
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| | - Christine J Hammond
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| | - Susan T Hingley
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| | - Brian J Balin
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA.
| |
Collapse
|
27
|
Qu Z, Meng F, Bomgarden RD, Viner RI, Li J, Rogers JC, Cheng J, Greenlief CM, Cui J, Lubahn DB, Sun GY, Gu Z. Proteomic quantification and site-mapping of S-nitrosylated proteins using isobaric iodoTMT reagents. J Proteome Res 2014; 13:3200-11. [PMID: 24926564 PMCID: PMC4084841 DOI: 10.1021/pr401179v] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
S-Nitrosylation is a redox-based protein post-translational
modification in response to nitric oxide signaling and is involved
in a wide range of biological processes. Detection and quantification
of protein S-nitrosylation have been challenging
tasks due to instability and low abundance of the modification. Many
studies have used mass spectrometry (MS)-based methods with different
thiol-reactive reagents to label and identify proteins with S-nitrosylated cysteine (SNO-Cys). In this study, we developed
a novel iodoTMT switch assay (ISA) using an isobaric set of thiol-reactive
iodoTMTsixplex reagents to specifically detect and quantify protein S-nitrosylation. Irreversible labeling of SNO-Cys with the
iodoTMTsixplex reagents enables immune-affinity detection of S-nitrosylated proteins, enrichment of iodoTMT-labeled peptides
by anti-TMT resin, and importantly, unambiguous modification site-mapping
and multiplex quantification by liquid chromatography–tandem
MS. Additionally, we significantly improved anti-TMT peptide enrichment
efficiency by competitive elution. Using ISA, we identified a set
of SNO-Cys sites responding to lipopolysaccharide (LPS) stimulation
in murine BV-2 microglial cells and revealed effects of S-allyl cysteine from garlic on LPS-induced protein S-nitrosylation in antioxidative signaling and mitochondrial metabolic
pathways. ISA proved to be an effective proteomic approach for quantitative
analysis of S-nitrosylation in complex samples and
will facilitate the elucidation of molecular mechanisms of nitrosative
stress in disease.
Collapse
Affiliation(s)
- Zhe Qu
- Department of Pathology and Anatomical Sciences, ‡Department of Biochemistry, and §Center for Translational Neuroscience, University of Missouri School of Medicine , Columbia, Missouri, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Raj DDA, Jaarsma D, Holtman IR, Olah M, Ferreira FM, Schaafsma W, Brouwer N, Meijer MM, de Waard MC, van der Pluijm I, Brandt R, Kreft KL, Laman JD, de Haan G, Biber KPH, Hoeijmakers JHJ, Eggen BJL, Boddeke HWGM. Priming of microglia in a DNA-repair deficient model of accelerated aging. Neurobiol Aging 2014; 35:2147-60. [PMID: 24799273 DOI: 10.1016/j.neurobiolaging.2014.03.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/21/2014] [Accepted: 03/23/2014] [Indexed: 12/16/2022]
Abstract
Aging is associated with reduced function, degenerative changes, and increased neuroinflammation of the central nervous system (CNS). Increasing evidence suggests that changes in microglia cells contribute to the age-related deterioration of the CNS. The most prominent age-related change of microglia is enhanced sensitivity to inflammatory stimuli, referred to as priming. It is unclear if priming is due to intrinsic microglia ageing or induced by the ageing neural environment. We have studied this in Ercc1 mutant mice, a DNA repair-deficient mouse model that displays features of accelerated aging in multiple tissues including the CNS. In Ercc1 mutant mice, microglia showed hallmark features of priming such as an exaggerated response to peripheral lipopolysaccharide exposure in terms of cytokine expression and phagocytosis. Specific targeting of the Ercc1 deletion to forebrain neurons resulted in a progressive priming response in microglia exemplified by phenotypic alterations. Summarizing, these data show that neuronal genotoxic stress is sufficient to switch microglia from a resting to a primed state.
Collapse
Affiliation(s)
- Divya D A Raj
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Dick Jaarsma
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Inge R Holtman
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marta Olah
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Filipa M Ferreira
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wandert Schaafsma
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michel M Meijer
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Monique C de Waard
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ingrid van der Pluijm
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Renata Brandt
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Karim L Kreft
- Department of Immunology, Erasmus University Medical Center and MS Center ErasMS, Rotterdam, the Netherlands; Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jon D Laman
- Department of Immunology, Erasmus University Medical Center and MS Center ErasMS, Rotterdam, the Netherlands
| | - Gerald de Haan
- Department of Cell Biology, European Research Institute on the Biology of Aging, University of Groningen, University Medical Center, Groningen, the Netherlands
| | - Knut P H Biber
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Psychiatry and Psychotherapy, University Medical Center, Freiburg, Germany
| | - Jan H J Hoeijmakers
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hendrikus W G M Boddeke
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
29
|
Qu Z, Meng F, Zhou H, Li J, Wang Q, Wei F, Cheng J, Greenlief CM, Lubahn DB, Sun GY, Liu S, Gu Z. NitroDIGE analysis reveals inhibition of protein S-nitrosylation by epigallocatechin gallates in lipopolysaccharide-stimulated microglial cells. J Neuroinflammation 2014; 11:17. [PMID: 24472655 PMCID: PMC3922161 DOI: 10.1186/1742-2094-11-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 01/20/2014] [Indexed: 12/28/2022] Open
Abstract
Background Nitric oxide (NO) is a signaling molecule regulating numerous cellular functions in development and disease. In the brain, neuronal injury or neuroinflammation can lead to microglial activation, which induces NO production. NO can react with critical cysteine thiols of target proteins forming S-nitroso-proteins. This modification, known as S-nitrosylation, is an evolutionarily conserved redox-based post-translational modification (PTM) of specific proteins analogous to phosphorylation. In this study, we describe a protocol for analyzing S-nitrosylation of proteins using a gel-based proteomic approach and use it to investigate the modes of action of a botanical compound found in green tea, epigallocatechin-3-gallate (EGCG), on protein S-nitrosylation after microglial activation. Methods/Results To globally and quantitatively analyze NO-induced protein S-nitrosylation, the sensitive gel-based proteomic method, termed NitroDIGE, was developed by combining two-dimensional differential in-gel electrophoresis (2-D DIGE) with the modified biotin switch technique (BST) using fluorescence-tagged CyDye™ thiol reactive agents to label S-nitrosothiols. The NitroDIGE method showed high specificity and sensitivity in detecting S-nitrosylated proteins (SNO-proteins). Using this approach, we identified a subset of SNO-proteins ex vivo by exposing immortalized murine BV-2 microglial cells to a physiological NO donor, or in vivo by exposing BV-2 cells to endotoxin lipopolysaccharides (LPS) to induce a proinflammatory response. Moreover, EGCG was shown to attenuate S-nitrosylation of proteins after LPS-induced activation of microglial cells primarily by modulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated oxidative stress response. Conclusions These results demonstrate that NitroDIGE is an effective proteomic strategy for “top-down” quantitative analysis of protein S-nitrosylation in multi-group samples in response to nitrosative stress due to excessive generation of NO in cells. Using this approach, we have revealed the ability of EGCG to down-regulate protein S-nitrosylation in LPS-stimulated BV-2 microglial cells, consistent with its known antioxidant effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Zezong Gu
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
30
|
Yao ZA, Xu L, Wu HG. Immunomodulatory function of κ-carrageenan oligosaccharides acting on LPS-activated microglial cells. Neurochem Res 2013; 39:333-43. [PMID: 24357352 DOI: 10.1007/s11064-013-1228-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 10/22/2013] [Accepted: 12/11/2013] [Indexed: 01/08/2023]
Abstract
The major neurodegenerative diseases are characterized by increasing of activated-microglial cells and inflammatory cytokines in the central nervous system. Carrageenan extracted from red algae is a kind of polysaccharide with sulfate groups. The oligosaccharides were obtained from carrageenan by enzymatic degradation. To detect the immunomodulatory activity of κ-carrageenan oligosaccharides (KOS) on microglial cells and the relationship to the sulfate group content, the desulfated derivatives of KOS (DSK) were obtained by dimethyl sulfoxide-methanol-pyridine method. KOS was labeled with fluorescein isothiocyanate. The effect of KOS and DSK on lipopolysaccharide (LPS)-activated microglial cells was detected. Hematoxylin-eosin staining and flow cytometric were used to detect the cell viability. The "scratch" migration assay, ornithine analysis and RT-PCR were used to determine the cell migration, arginase and TNF-α released by microglial cells, respectively. The effect of LPS and KOS on microglial cells was determined by flow cytometry and laser scanning confocal microscopy. The results showed that KOS and DSK could inhibit the cell viability, arginase and TNF-α released by LPS-activated microglia cell with concentration dependent manner. But the effect of DSK was weaker than that of KOS. KOS aggregated on the cell surface firstly, and then they enter into the cell to the nucleus, spread over the entire cell finally. But the exist of LPS could prevent the entrance of KOS. It could be concluded that KOS could protect microglial cells from being activated by LPS, and its inhibition function had relationship to the sulfate group content of KOS, while there were competition between LPS and KOS.
Collapse
Affiliation(s)
- Zi-Ang Yao
- College of Life Science and Technology, Dalian University, Dalian, 116622, China
| | | | | |
Collapse
|
31
|
Molecular Dissection of Cyclosporin A's Neuroprotective Effect Reveals Potential Therapeutics for Ischemic Brain Injury. Brain Sci 2013; 3:1325-56. [PMID: 24961531 PMCID: PMC4061870 DOI: 10.3390/brainsci3031325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 07/30/2013] [Accepted: 08/14/2013] [Indexed: 12/02/2022] Open
Abstract
After the onset of brain ischemia, a series of events leads ultimately to the death of neurons. Many molecules can be pharmacologically targeted to protect neurons during these events, which include glutamate release, glutamate receptor activation, excitotoxicity, Ca2+ influx into cells, mitochondrial dysfunction, activation of intracellular enzymes, free radical production, nitric oxide production, and inflammation. There have been a number of attempts to develop neuroprotectants for brain ischemia, but many of these attempts have failed. It was reported that cyclosporin A (CsA) dramatically ameliorates neuronal cell damage during ischemia. Some researchers consider ischemic cell death as a unique process that is distinct from both apoptosis and necrosis, and suggested that mitochondrial dysfunction and Δψ collapse are key steps for ischemic cell death. It was also suggested that CsA has a unique neuroprotective effect that is related to mitochondrial dysfunction. Here, I will exhibit examples of neuroprotectants that are now being developed or in clinical trials, and will discuss previous researches about the mechanism underlying the unique CsA action. I will then introduce the results of our cDNA subtraction experiment with or without CsA administration in the rat brain, along with our hypothesis about the mechanism underlying CsA’s effect on transcriptional regulation.
Collapse
|
32
|
Yoo SW, Chang DY, Lee HS, Kim GH, Park JS, Ryu BY, Joe EH, Lee YD, Kim SS, Suh-Kim H. Immune following suppression mesenchymal stem cell transplantation in the ischemic brain is mediated by TGF-β. Neurobiol Dis 2013; 58:249-57. [PMID: 23759293 DOI: 10.1016/j.nbd.2013.06.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 05/22/2013] [Accepted: 06/01/2013] [Indexed: 12/16/2022] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) has been shown to enhance the recovery of brain functions following ischemic injury. Although immune modulation has been suggested to be one of the mechanisms, the molecular mechanisms underlying improved recovery has not been clearly identified. Here, we report that MSCs secrete transforming growth factor-beta (TGF-β) to suppress immune propagation in the ischemic rat brain. Ischemic stroke caused global death of resident cells in the infarcted area, elevated the monocyte chemoattractant protein-1 (MCP-1) level, and evoked massive infiltration of circulating CD68+ immune cells through the impaired blood-brain barrier. Transplantation of MSCs at day 3 post-ischemia blocked the subsequent upregulation of MCP-1 in the ischemic area and the infiltration of additional CD68+ immune cells. MSC-conditioned media decreased the migration and MCP-1 production of freshly isolated immune cells in vitro, and this effect was blocked by an inhibitor of TGF-β signaling or an anti-TGF-β neutralizing antibody. Finally, transplantation of TGF-β1-silenced MSCs failed to attenuate the infiltration of CD68+ cells into the ischemic brain, and was associated with only minor improvements in motor function. These results indicate that TGF-β is key to the ability of MSCs to beneficially attenuate immune reactions in the ischemic brain. Our findings offer insight into the interactions between allogeneic MSCs and the host immune system, reinforcing the prospective clinical value of using MSCs in the treatment of neurological disorders involving inflammation-mediated secondary damage.
Collapse
Affiliation(s)
- Seung-Wan Yoo
- Department of Anatomy, Ajou University School of Medicine, Suwon, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Browne TC, McQuillan K, McManus RM, O’Reilly JA, Mills KHG, Lynch MA. IFN-γ Production by Amyloid β–Specific Th1 Cells Promotes Microglial Activation and Increases Plaque Burden in a Mouse Model of Alzheimer’s Disease. THE JOURNAL OF IMMUNOLOGY 2013; 190:2241-51. [DOI: 10.4049/jimmunol.1200947] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
34
|
Harati R, Villégier AS, Banks WA, Mabondzo A. Susceptibility of juvenile and adult blood-brain barrier to endothelin-1: regulation of P-glycoprotein and breast cancer resistance protein expression and transport activity. J Neuroinflammation 2012; 9:273. [PMID: 23253775 PMCID: PMC3547749 DOI: 10.1186/1742-2094-9-273] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 11/07/2012] [Indexed: 12/12/2022] Open
Abstract
Background P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) play a critical role in keeping neurotoxic substances from entering the brain. We and others have previously reported an impact of inflammation on the regulation of adult blood–brain barrier (BBB) efflux transporters. However, studies in children have not been done. From the pediatric clinical perspective, it is important to understand how the central nervous system (CNS) and BBB drug efflux transporters differ in childhood from those of adults under normal and inflammatory conditions. Therefore, we examined and compared the regulation of P-gp and BCRP expression and transport activity in young and adult BBB and investigated the molecular mechanisms underlying inflammatory responses. Methods Rats at postnatal day (P) P21 and P84, corresponding to the juvenile and adult stages of human brain maturation, respectively, were treated with endothelin-1 (ET-1) given by the intracerebroventricular (icv) route. Twenty-four hours later, we measured P-gp and BCRP protein expression in isolated brain capillary by immunoblotting as well as by transport activity in vivo by measuring the unbound drug partitioning coefficient of the brain (Kp,uu,brain) of known efflux transporter substrates administered intravenously. Glial activation was measured by immunohistochemistry. The release of cytokines/chemokines (interleukins-1α, 1-β (IL-1β), -6 (IL-6), -10 (IL-10), monocyte chemoattractant protein (MCP-1/CCL2), fractalkine and tissue inhibitor of metalloproteinases-1 (TIMP-1)) were simultaneously measured in brain and serum samples using the Agilent Technology cytokine microarray. Results We found that juvenile and adult BBBs exhibited similar P-gp and BCRP transport activities in the normal physiological conditions. However, long-term exposure of the juvenile brain to low-dose of ET-1 did not change BBB P-gp transport activity but tended to decrease BCRP transport activity in the juvenile brain, while a significant increase of the activity of both transporters was evidenced at the BBB in the adult brain. Moreover, juvenile and adult brain showed differences in their expression profiles of cytokines and chemokines mediated by ET-1. Conclusions BBB transporter activity during neuroinflammation differs between the juvenile and adult brains. These findings emphasize the importance of considering differential P-gp and BCRP transport regulation mechanisms between adult and juvenile BBB in the context of neuroinflammation.
Collapse
Affiliation(s)
- Rania Harati
- CEA, Direction des Sciences du Vivant, iBiTec-S, Service de Pharmacologie et d'Immunoanalyse, Gif-sur-Yvette, France
| | | | | | | |
Collapse
|
35
|
Cao L, Beaulac H, Eurich A. Differential lumbar spinal cord responses among wild type, CD4 knockout, and CD40 knockout mice in spinal nerve L5 transection-induced neuropathic pain. Mol Pain 2012; 8:88. [PMID: 23249743 PMCID: PMC3545955 DOI: 10.1186/1744-8069-8-88] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 12/14/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Our previous studies have indicated that both lumbar spinal cord-infiltrating CD4+ T cells and microglial CD40 contribute to the maintenance of mechanical hypersensitivity in a murine model of neuropathic pain spinal nerve L5 transection (L5Tx). To further delineate the CD4 and CD40-mediated mechanisms involved in the development of L5Tx-induced neuropathic pain behaviors, we examined the lumbar spinal cord mononuclear cells of wild type (WT) BALB/c, BALB/c-CD4 knockout (KO), and BALB/c-CD40 KO mice via flow cytometry. RESULTS In WT mice, L5Tx induced significant but transient (at day 3 and/or day 7) increases of the total numbers of mononuclear cells, microglial cells (CD45loCD11b+), and infiltrating leukocytes (CD45hi) in the ipsilateral side of the spinal cord. In CD4 KO mice, significant elevation of microglia was detected only on day 7 post-L5Tx, while no significant increase in infiltrating leukocytes post-L5Tx was observed. CD40 KO mice did not exhibit any of the changes observed in WT mice. Furthermore, neutralizing CD40 antibody treatment indicated an early involvement of CD40 signaling in the development of L5Tx-induced mechanical hypersensitivity. CONCLUSIONS Altogether, data indicate that both CD4 and CD40 play a role in L5Tx-induced leukocyte infiltration into the lumbar spinal cord but have differential contributions to spinal cord microglial activation following peripheral nerve injury.
Collapse
Affiliation(s)
- Ling Cao
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA.
| | | | | |
Collapse
|
36
|
Jones RS, Minogue AM, Connor TJ, Lynch MA. Amyloid-β-induced astrocytic phagocytosis is mediated by CD36, CD47 and RAGE. J Neuroimmune Pharmacol 2012; 8:301-11. [PMID: 23238794 DOI: 10.1007/s11481-012-9427-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/02/2012] [Indexed: 11/26/2022]
Abstract
Astrocytes, the most numerous glial cell in the brain, have multiple functions and are key to maintenance of homeostasis in the central nervous system. Microglia are the resident immunocompetent cells in the brain and share several functions with macrophages, including their phagocytic ability. Indeed microglia are the resident phagocytes in the brain and express numerous cell surface proteins which act to enable receptor-mediated phagocytosis. However recent evidence suggests that astrocytes express some genes which permit phagocytosis of phosphatidylserine-decorated cells and this probably explains sporadic reports in the literature which suggest that astrocytes become phagocytic following brain trauma. Here we examined the potential of astrocytes to phagocytose fluorescently-labelled latex beads and amyloid-β (Aβ) and report that they competently engulf both in a manner that relies on actin polymerization since it was inhibited by cytochalasin D. The data indicate that incubation of cultured astrocytes or microglia with Aβ increased phagocytosis and markers of activation of both cell types. Aβ was found to markedly increase expression of the putative Aβ-binding receptors CD36 and CD47 in astrocytes, while it decreased expression of the receptor for advanced glycation endproducts (RAGE). It is demonstrated that blocking these receptors using a neutralizing antibody attenuated Aβ-induced phagocytosis of latex beads by astrocytes. Interestingly blocking these receptors also decreased uptake of beads even in the absence of Aβ. Here we demonstrate that astrocytes are competent phagocytes and are capable of engulfing Aβ.
Collapse
Affiliation(s)
- Raasay S Jones
- Trinity College Institute of Neuroscience, Department of Physiology, Lloyd Building, Trinity College, Dublin 2, Ireland
| | | | | | | |
Collapse
|
37
|
Hematopoietic prostaglandin D synthase inhibitors. PROGRESS IN MEDICINAL CHEMISTRY 2012; 51:97-133. [PMID: 22520473 DOI: 10.1016/b978-0-12-396493-9.00004-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
38
|
von Bernhardi R, Eugenín J. Alzheimer's disease: redox dysregulation as a common denominator for diverse pathogenic mechanisms. Antioxid Redox Signal 2012; 16:974-1031. [PMID: 22122400 DOI: 10.1089/ars.2011.4082] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and a progressive neurodegeneration that appears to result from multiple pathogenic mechanisms (including protein misfolding/aggregation, involved in both amyloid β-dependent senile plaques and tau-dependent neurofibrillary tangles), metabolic and mitochondrial dysfunction, excitoxicity, calcium handling impairment, glial cell dysfunction, neuroinflammation, and oxidative stress. Oxidative stress, which could be secondary to several of the other pathophysiological mechanisms, appears to be a major determinant of the pathogenesis and progression of AD. The identification of oxidized proteins common for mild cognitive impairment and AD suggests that key oxidation pathways are triggered early and are involved in the initial progression of the neurodegenerative process. Abundant data support that oxidative stress, also considered as a main factor for aging, the major risk factor for AD, can be a common key element capable of articulating the divergent nature of the proposed pathogenic factors. Pathogenic mechanisms influence each other at different levels. Evidence suggests that it will be difficult to define a single-target therapy resulting in the arrest of progression or the improvement of AD deterioration. Since oxidative stress is present from early stages of disease, it appears as one of the main targets to be included in a clinical trial. Exploring the articulation of AD pathogenic mechanisms by oxidative stress will provide clues for better understanding the pathogenesis and progression of this dementing disorder and for the development of effective therapies to treat this disease.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | |
Collapse
|
39
|
Involvement of calcitonin gene-related peptide and CCL2 production in CD40-mediated behavioral hypersensitivity in a model of neuropathic pain. ACTA ACUST UNITED AC 2012; 7:117-28. [PMID: 22377050 DOI: 10.1017/s1740925x12000026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The neuropeptide calcitonin gene-related peptide (CGRP) is known to play a pro-nociceptive role after peripheral nerve injury upon its release from primary afferent neurons in preclinical models of neuropathic pain. We previously demonstrated a critical role for spinal cord microglial CD40 in the development of spinal nerve L5 transection (L5Tx)-induced mechanical hypersensitivity. Herein, we investigated whether CGRP is involved in the CD40-mediated behavioral hypersensitivity. First, L5Tx was found to significantly induce CGRP expression in wild-type (WT) mice up to 14 days post-L5Tx. This increase in CGRP expression was reduced in CD40 knockout (KO) mice at day 14 post-L5Tx. Intrathecal injection of the CGRP antagonist CGRP8-37 significantly blocked L5Tx-induced mechanical hypersensitivity. In vitro, CGRP induced glial IL-6 and CCL2 production, and CD40 stimulation added to the effects of CGRP in neonatal glia. Further, there was decreased CCL2 production in CD40 KO mice compared to WT mice 21 days post-L5Tx. However, CGRP8-37 did not significantly affect spinal cord CCL2 production following L5Tx in WT mice. Altogether, these data suggest that CD40 contributes to the maintenance of behavioral hypersensitivity following peripheral nerve injury in part through two distinct pathways, the enhancement of CGRP expression and spinal cord CCL2 production.
Collapse
|
40
|
Abstract
Parkinson's disease (PD) is the most common age-related motoric neurodegenerative disease initially described in the 1800's by James Parkinson as the 'Shaking Palsy'. Loss of the neurotransmitter dopamine was recognized as underlying the pathophysiology of the motor dysfunction; subsequently discovery of dopamine replacement therapies brought substantial symptomatic benefit to PD patients. However, these therapies do not fully treat the clinical syndrome nor do they alter the natural history of this disorder motivating clinicians and researchers to further investigate the clinical phenotype, pathophysiology/pathobiology and etiology of this devastating disease. Although the exact cause of sporadic PD remains enigmatic studies of familial and rare toxicant forms of this disorder have laid the foundation for genome wide explorations and environmental studies. The combination of methodical clinical evaluation, systematic pathological studies and detailed genetic analyses have revealed that PD is a multifaceted disorder with a wide-range of clinical symptoms and pathology that include regions outside the dopamine system. One common thread in PD is the presence of intracytoplasmic inclusions that contain the protein, α-synuclein. The presence of toxic aggregated forms of α-synuclein (e.g., amyloid structures) are purported to be a harbinger of subsequent pathology. In fact, PD is both a cerebral amyloid disease and the most common synucleinopathy, that is, diseases that display accumulations of α-synuclein. Here we present our current understanding of PD etiology, pathology, clinical symptoms and therapeutic approaches with an emphasis on misfolded α-synuclein.
Collapse
Affiliation(s)
- Timothy R. Mhyre
- Department of Neuroscience, Georgetown University Medical Center, NRB EP08, 3970 Reservoir Road NW, 20057, Washington, DC, USA,
| | - James T. Boyd
- University of Vermont College of Medicine, 1 South Prospect Street, DU-Arnold 4416-UHC, 05401, Burlington, VT, USA,
| | - Robert W. Hamill
- Department of Neurology, University of Vermont College of Medicine, 89 Beaumont Avenue, Given Hall Room C225, 05405, Burlington, VT, USA,
| | - Kathleen A. Maguire-Zeiss
- Department of Neuroscience, Center for Neural Injury and RecoveryGeorgetown University Medical Center, 3970 Reservoir Road, NW NRB EP08, 20057, Washington, DC, USA,
| |
Collapse
|
41
|
Sheng W, Zong Y, Mohammad A, Ajit D, Cui J, Han D, Hamilton JL, Simonyi A, Sun AY, Gu Z, Hong JS, Weisman GA, Sun GY. Pro-inflammatory cytokines and lipopolysaccharide induce changes in cell morphology, and upregulation of ERK1/2, iNOS and sPLA₂-IIA expression in astrocytes and microglia. J Neuroinflammation 2011; 8:121. [PMID: 21943492 PMCID: PMC3206447 DOI: 10.1186/1742-2094-8-121] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/24/2011] [Indexed: 11/29/2022] Open
Abstract
Background Activation of glial cells, including astrocytes and microglia, has been implicated in the inflammatory responses underlying brain injury and neurodegenerative diseases including Alzheimer's and Parkinson's diseases. Although cultured astrocytes and microglia are capable of responding to pro-inflammatory cytokines and lipopolysaccharide (LPS) in the induction and release of inflammatory factors, no detailed analysis has been carried out to compare the induction of iNOS and sPLA2-IIA. In this study, we investigated the effects of cytokines (TNF-alpha, IL-1beta, and IFN-gamma) and LPS + IFN-gamma to induce temporal changes in cell morphology and induction of p-ERK1/2, iNOS and sPLA2-IIA expression in immortalized rat (HAPI) and mouse (BV-2) microglial cells, immortalized rat astrocytes (DITNC), and primary microglia and astrocytes. Methods/Results Cytokines (TNF-alpha, IL-1beta, and IFN-gamma) and LPS + IFN-gamma induced a time-dependent increase in fine processes (filopodia) in microglial cells but not in astrocytes. Filopodia production was attributed to IFN-gamma and was dependent on ERK1/2 activation. Cytokines induced an early (15 min) and a delayed phase (1 ~ 4 h) increase in p-ERK1/2 expression in microglial cells, and the delayed phase increase corresponded to the increase in filopodia production. In general, microglial cells are more active in responding to cytokines and LPS than astrocytes in the induction of NO. Although IFN-gamma and LPS could individually induce NO, additive production was observed when IFN-gamma was added together with LPS. On the other hand, while TNF-alpha, IL-1beta, and LPS could individually induce sPLA2-IIA mRNA and protein expression, this induction process does not require IFN-gamma. Interestingly, neither rat immortalized nor primary microglial cells were capable of responding to cytokines and LPS in the induction of sPLA2-IIA expression. Conclusion These results demonstrated the utility of BV-2 and HAPI cells as models for investigation on cytokine and LPS induction of iNOS, and DITNC astrocytes for induction of sPLA2-IIA. In addition, results further demonstrated that cytokine-induced sPLA2-IIA is attributed mainly to astrocytes and not microglial cells.
Collapse
Affiliation(s)
- Wenwen Sheng
- Department of Biochemistry, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kawakami M, Yoshimoto T, Nakagata N, Yamamura KI, Siesjo BK. Effects of cyclosporin A administration on gene expression in rat brain. Brain Inj 2011; 25:614-23. [PMID: 21534739 DOI: 10.3109/02699052.2011.571229] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PRIMARY OBJECTIVE The immunosuppressant cyclosporin A (CsA) is reported to have a strong anti-ischemic effect. Although this neuroprotective effect is speculated to be related to the blockade of a mitochondrial permeability transition pore (mPTP), the underlying molecular mechanism remains to be elucidated. This study focused on the effect of CsA on transcriptional regulation in brain cells. METHODS CsA and a control substance were injected into rat brains and purified extracted mRNA. Both mRNAs were compared using a cDNA subtraction technique. RESULTS Nine significantly up-regulated genes and seven significantly down-regulated genes were detected following CsA administration. All of the up-regulated genes are neurotrophic or reported to have roles in regeneration of brain tissue. Among the down-regulated genes, three are known to be detrimental to neuronal cells and are also reported to facilitate the pathology of Alzheimer's disease (AD) and four genes are related to oxidative metabolism. CONCLUSIONS Strong immunosuppression would present as a side-effect during CsA use as a neuroprotectant. The results of this study will help to discriminate between the CsA immunosuppressive effect and the neuroprotective effect at the molecular level and may lead to the development of new conceptual and pharmacological tools.
Collapse
Affiliation(s)
- Minoru Kawakami
- Laboratory of Phylogeny, Institute of Molecular Embryology and Genetics, Kumamoto University, Japan.
| | | | | | | | | |
Collapse
|
43
|
Koistinaho J, Malm T, Goldsteins G. Glycogen synthase kinase-3β: a mediator of inflammation in Alzheimer's disease? Int J Alzheimers Dis 2011; 2011:129753. [PMID: 21629736 PMCID: PMC3100542 DOI: 10.4061/2011/129753] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 03/04/2011] [Indexed: 02/03/2023] Open
Abstract
Proliferation and activation of microglial cells is a neuropathological characteristic of brain injury and neurodegeneration, including Alzheimer's disease. Microglia act as the first and main form of immune defense in the nervous system. While the primary function of microglia is to survey and maintain the cellular environment optimal for neurons in the brain parenchyma by actively scavenging the brain for damaged brain cells and foreign proteins or particles, sustained activation of microglia may result in high production of proinflammatory mediators that disturb normal brain functions and even cause neuronal injury. Glycogen synthase kinase-3β has been recently identified as a major regulator of immune system and mediates inflammatory responses in microglia. Glycogen synthase kinase-3β has been extensively investigated in connection to tau and amyloid β toxicity, whereas reports on the role of this enzyme in neuroinflammation in Alzheimer's disease are negligible. Here we review and discuss the role of glycogen synthase-3β in immune cells in the context of Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Jari Koistinaho
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | | | | |
Collapse
|
44
|
Yang G, Meng Y, Li W, Yong Y, Fan Z, Ding H, Wei Y, Luo J, Ke ZJ. Neuronal MCP-1 mediates microglia recruitment and neurodegeneration induced by the mild impairment of oxidative metabolism. Brain Pathol 2010; 21:279-97. [PMID: 21029241 DOI: 10.1111/j.1750-3639.2010.00445.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Chemokines are implicated in the neuroinflammation of several chronic neurodegenerative disorders. However, the precise role of chemokines in neurodegeneration is unknown. Thiamine deficiency (TD) causes abnormal oxidative metabolism in the brain as well as a well-defined microglia activation and neurodegeneration in the submedial thalamus nucleus (SmTN), which are common features of neurodegenerative diseases. We evaluated the role of chemokines in neurodegeneration and the underlying mechanism in a TD model. Among the chemokines examined, TD selectively induced neuronal expression of monocyte chemoattractant protein-1 (MCP-1) in the SmTN prior to microglia activation and neurodegeneration. The conditioned medium collected from TD-induced neurons caused microglia activation. With a neuron/microglia co-culture system, we showed that MCP-1-induced neurotoxicity required the presence of microglia, and exogenous MCP-1 was able to activate microglia and stimulated microglia to produce cytokines. A MCP-1 neutralizing antibody inhibited MCP-1-induced microglia activation and neuronal death in culture and in the thalamus. MCP-1 knockout mice were resistant to TD-induced neuronal death in SmTN. TD selectively induced the accumulation of reactive oxygen species in neurons, and antioxidants blocked TD-induced MCP-1 expression. Together, our results indicated an induction of neuronal MCP-1 during mild impairment of oxidative metabolism caused by microglia recruitment/activation, which exacerbated neurodegeneration.
Collapse
Affiliation(s)
- Guang Yang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Heneka MT, O'Banion MK, Terwel D, Kummer MP. Neuroinflammatory processes in Alzheimer's disease. J Neural Transm (Vienna) 2010; 117:919-47. [PMID: 20632195 DOI: 10.1007/s00702-010-0438-z] [Citation(s) in RCA: 330] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 06/16/2010] [Indexed: 12/12/2022]
Abstract
Generation of neurotoxic amyloid beta peptides and their deposition along with neurofibrillary tangle formation represent key pathological hallmarks in Alzheimer's disease (AD). Recent evidence suggests that inflammation may be a third important component which, once initiated in response to neurodegeneration or dysfunction, may actively contribute to disease progression and chronicity. Various neuroinflammatory mediators including complement activators and inhibitors, chemokines, cytokines, radical oxygen species and inflammatory enzyme systems are expressed and released by microglia, astrocytes and neurons in the AD brain. Degeneration of aminergic brain stem nuclei including the locus ceruleus and the nucleus basalis of Meynert may facilitate the occurrence of inflammation in their projection areas given the antiinflammatory and neuroprotective action of their key transmitters norepinephrine and acetylcholine. While inflammation has been thought to arise secondary to degeneration, recent experiments demonstrated that inflammatory mediators may stimulate amyloid precursor protein processing by various means and therefore can establish a vicious cycle. Despite the fact that some aspects of inflammation may even be protective for bystander neurons, antiinflammatory treatment strategies should therefore be considered. Non-steroidal anti-inflammatory drugs have been shown to reduce the risk and delay the onset to develop AD. While, the precise molecular mechanism underlying this effect is still unknown, a number of possible mechanisms including cyclooxygenase 2 or gamma-secretase inhibition and activation of the peroxisome proliferator activated receptor gamma may alone or, more likely, in concert account for the epidemiologically observed protection.
Collapse
Affiliation(s)
- Michael T Heneka
- Department of Neurology, Clinical Neurosciences, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany.
| | | | | | | |
Collapse
|
46
|
Hjorth E, Frenkel D, Weiner H, Schultzberg M. Effects of immunomodulatory substances on phagocytosis of abeta(1-42) by human microglia. Int J Alzheimers Dis 2010; 2010. [PMID: 20798889 PMCID: PMC2925296 DOI: 10.4061/2010/798424] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/24/2010] [Indexed: 01/24/2023] Open
Abstract
Glial activation and increased inflammation characterize neuropathology in Alzheimer's disease (AD). The aim was to develop a model for studying phagocytosis of beta-amyloid (Abeta) peptide by human microglia and to test effects thereupon by immunomodulatory substances. Human CHME3 microglia showed intracellular Abeta(1-42) colocalized with lysosome-associated membrane protein-2, indicating phagocytosis. This was increased by interferon-gamma, and to a lesser degree with Protollin, a proteosome-based adjuvant. Secretion of brain-derived neurotrophic factor (BDNF) was decreased by Abeta(1-42) and by interferon-gamma and interleukin-1beta. These cytokines, but not Abeta(1-42), stimulated interleukin-6 release. Microglia which phagocytosed Abeta(1-42) exhibited a higher degree of expression of interleukin-1 receptor type I and inducible nitric oxide synthase. In conclusion, we show that human microglia are able to phagocytose Abeta(1-42) and that this is associated with expression of inflammatory markers. Abeta(1-42) and interferon-gamma decreased BDNF secretion suggesting a new neuropathological role for Abeta(1-42) and the inflammation accompanying AD.
Collapse
Affiliation(s)
- Erik Hjorth
- Division of Neurodegeneration, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 86 Stockholm, Sweden
| | | | | | | |
Collapse
|
47
|
TGF-beta1 blockade of microglial chemotaxis toward Abeta aggregates involves SMAD signaling and down-regulation of CCL5. J Neuroinflammation 2010; 7:28. [PMID: 20429874 PMCID: PMC2878297 DOI: 10.1186/1742-2094-7-28] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 04/29/2010] [Indexed: 12/30/2022] Open
Abstract
Background Overactivated microglia that cluster at neuritic plaques constantly release neurotoxins, which actively contribute to progressive neurodegeneration in Alzheimer's disease (AD). Therefore, attenuating microglial clustering can reduce focal neuroinflammation at neuritic plaques. Previously, we identified CCL5 and CCL2 as prominent chemokines that mediate the chemotaxis of microglia toward beta-amyloid (Aβ)aggregates. Although transforming growth factor-β1 (TGF-β1) has been shown to down-regulate the expression of chemokines in activated microglia, whether TGF-β1 can reduce the chemotaxis of microglia toward neuritic plaques in AD remains unclear. Methods In the present study, we investigated the effects of TGF-β1 on Aβ-induced chemotactic migration of BV-2 microglia using time-lapse recording, transwell assay, real-time PCR, ELISA, and western blotting. Results The cell tracing results suggest that the morphological characteristics and migratory patterns of BV-2 microglia resemble those of microglia in slice cultures. Using this model system, we discovered that TGF-β1 reduces Aβ-induced BV-2 microglial clustering in a dose-dependent manner. Chemotactic migration of these microglial cells toward Aβ aggregates was significantly attenuated by TGF-β1. However, these microglia remained actively moving without any reduction in migration speed. Pharmacological blockade of TGF-β1 receptor I (ALK5) by SB431542 treatment reduced the inhibitory effects of TGF-β1 on Aβ-induced BV-2 microglial clustering, while preventing TGF-β1-mediated cellular events, including SMAD2 phosphorylation and CCL5 down-regulation. Conclusions Our results suggest that TGF-β1 reduces Aβ-induced microglial chemotaxis via the SMAD2 pathway. The down-regulation of CCL5 by TGF-β1 at least partially contributes to the clustering of microglia at Aβ aggregates. The attenuating effects of SB431542 upon TGF-β1-suppressed microglial clustering may be mediated by restoration of CCL5 to normal levels. TGF-β1 may ameliorate microglia-mediated neuroinflammation in AD by preventing activated microglial clustering at neuritic plaques.
Collapse
|
48
|
Blalock EM, Grondin R, Chen KC, Thibault O, Thibault V, Pandya JD, Dowling A, Zhang Z, Sullivan P, Porter NM, Landfield PW. Aging-related gene expression in hippocampus proper compared with dentate gyrus is selectively associated with metabolic syndrome variables in rhesus monkeys. J Neurosci 2010; 30:6058-71. [PMID: 20427664 PMCID: PMC3155249 DOI: 10.1523/jneurosci.3956-09.2010] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 02/15/2010] [Accepted: 03/21/2010] [Indexed: 11/21/2022] Open
Abstract
Age-dependent metabolic syndrome (MetS) is a well established risk factor for cardiovascular disease, but it also confers major risk for impaired cognition in normal aging or Alzheimer's disease (AD). However, little is known about the specific pathways mediating MetS-brain interactions. Here, we performed the first studies quantitatively linking MetS variables to aging changes in brain genome-wide expression and mitochondrial function. In six young adult and six aging female rhesus monkeys, we analyzed gene expression in two major hippocampal subdivisions critical for memory/cognitive function [hippocampus proper, or cornu ammonis (CA), and dentate gyrus (DG)]. Genes that changed with aging [aging-related genes (ARGs)] were identified in each region. Serum variables reflecting insulin resistance and dyslipidemia were used to construct a quantitative MetS index (MSI). This MSI increased with age and correlated negatively with hippocampal mitochondrial function (state III oxidation). More than 2000 ARGs were identified in CA and/or DG, in approximately equal numbers, but substantially more ARGs in CA than in DG were correlated selectively with the MSI. Pathways represented by MSI-correlated ARGs were determined from the Gene Ontology Database and literature. In particular, upregulated CA ARGs representing glucocorticoid receptor (GR), chromatin assembly/histone acetyltransferase, and inflammatory/immune pathways were closely associated with the MSI. These results suggest a novel model in which MetS is associated with upregulation of hippocampal GR-dependent transcription and epigenetic coactivators, contributing to decreased mitochondrial function and brain energetic dysregulation. In turn, these MSI-associated neuroenergetic changes may promote inflammation, neuronal vulnerability, and risk of cognitive impairment/AD.
Collapse
Affiliation(s)
- Eric M Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Pistell PJ, Morrison CD, Gupta S, Knight AG, Keller JN, Ingram DK, Bruce-Keller AJ. Cognitive impairment following high fat diet consumption is associated with brain inflammation. J Neuroimmunol 2010; 219:25-32. [PMID: 20004026 PMCID: PMC2823983 DOI: 10.1016/j.jneuroim.2009.11.010] [Citation(s) in RCA: 449] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 11/11/2009] [Accepted: 11/11/2009] [Indexed: 02/07/2023]
Abstract
C57Bl/6 mice were administered a high fat, Western diet (WD, 41% fat) or a very high fat lard diet (HFL, 60% fat), and evaluated for cognitive ability using the Stone T-maze and for biochemical markers of brain inflammation. WD consumption resulted in significantly increased body weight and astrocyte reactivity, but not impaired cognition, microglial reactivity, or heightened cytokine levels. HFL increased body weight, and impaired cognition, increased brain inflammation, and decreased BDNF. Collectively, these data suggest that while different diet formulations can increase body weight, the ability of high fat diets to disrupt cognition is linked to brain inflammation.
Collapse
Affiliation(s)
- Paul J. Pistell
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Christopher D. Morrison
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Sunita Gupta
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Alecia G. Knight
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Jeffrey N. Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | - Donald K. Ingram
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808
| | | |
Collapse
|
50
|
Amyloid β-peptide directly induces spontaneous calcium transients, delayed intercellular calcium waves and gliosis in rat cortical astrocytes. ASN Neuro 2010; 2:e00026. [PMID: 20001968 PMCID: PMC2810812 DOI: 10.1042/an20090035] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 11/23/2009] [Accepted: 12/08/2009] [Indexed: 12/14/2022] Open
Abstract
The contribution of astrocytes to the pathophysiology of AD (Alzheimer's disease) and the molecular and signalling mechanisms that potentially underlie them are still very poorly understood. However, there is mounting evidence that calcium dysregulation in astrocytes may be playing a key role. Intercellular calcium waves in astrocyte networks in vitro can be mechanically induced after Aβ (amyloid β-peptide) treatment, and spontaneously forming intercellular calcium waves have recently been shown in vivo in an APP (amyloid precursor protein)/PS1 (presenilin 1) Alzheimer's transgenic mouse model. However, spontaneous intercellular calcium transients and waves have not been observed in vitro in isolated astrocyte cultures in response to direct Aβ stimulation in the absence of potentially confounding signalling from other cell types. Here, we show that Aβ alone at relatively low concentrations is directly able to induce intracellular calcium transients and spontaneous intercellular calcium waves in isolated astrocytes in purified cultures, raising the possibility of a potential direct effect of Aβ exposure on astrocytes in vivo in the Alzheimer's brain. Waves did not occur immediately after Aβ treatment, but were delayed by many minutes before spontaneously forming, suggesting that intracellular signalling mechanisms required sufficient time to activate before intercellular effects at the network level become evident. Furthermore, the dynamics of intercellular calcium waves were heterogeneous, with distinct radial or longitudinal propagation orientations. Lastly, we also show that changes in the expression levels of the intermediate filament proteins GFAP (glial fibrillary acidic protein) and S100B are affected by Aβ-induced calcium changes differently, with GFAP being more dependent on calcium levels than S100B.
Collapse
|