51
|
Spanos F, Liddelow SA. An Overview of Astrocyte Responses in Genetically Induced Alzheimer's Disease Mouse Models. Cells 2020; 9:E2415. [PMID: 33158189 PMCID: PMC7694249 DOI: 10.3390/cells9112415] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Despite many years of intense research, there is currently still no effective treatment. Multiple cell types contribute to disease pathogenesis, with an increasing body of data pointing to the active participation of astrocytes. Astrocytes play a pivotal role in the physiology and metabolic functions of neurons and other cells in the central nervous system. Because of their interactions with other cell types, astrocyte functions must be understood in their biologic context, thus many studies have used mouse models, of which there are over 190 available for AD research. However, none appear able to fully recapitulate the many functional changes in astrocytes reported in human AD brains. Our review summarizes the observations of astrocyte biology noted in mouse models of familial and sporadic AD. The limitations of AD mouse models will be discussed and current attempts to overcome these disadvantages will be described. With increasing understanding of the non-neuronal contributions to disease, the development of new methods and models will provide further insights and address important questions regarding the roles of astrocytes and other non-neuronal cells in AD pathophysiology. The next decade will prove to be full of exciting opportunities to address this devastating disease.
Collapse
Affiliation(s)
- Fokion Spanos
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA;
| | - Shane A. Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA;
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
52
|
Prantner D, Nallar S, Vogel SN. The role of RAGE in host pathology and crosstalk between RAGE and TLR4 in innate immune signal transduction pathways. FASEB J 2020; 34:15659-15674. [PMID: 33131091 DOI: 10.1096/fj.202002136r] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
Although the innate immune receptor protein, Receptor for Advanced Glycation End products (RAGE), has been extensively studied, there has been renewed interest in RAGE for its potential role in sepsis, along with a host of other inflammatory diseases of chronic, noninfectious origin. In contrast to other innate immune receptors, for example, Toll-like receptors (TLRs), that recognize ligands derived from pathogenic organisms that are collectively known as "pathogen-associated molecular patterns" (PAMPs) or host-derived "damage-associated molecular patterns" (DAMPs), RAGE has been shown to recognize a broad collection of DAMPs exclusively. Historically, these DAMPs have been shown to be pro-inflammatory in nature. Early studies indicated that the adaptor molecule, MyD88, might be important for this change. More recent studies have explored further the mechanisms underlying this inflammatory change. Overall, the newer results have shown that there is extensive crosstalk between RAGE and TLRs. The three canonical RAGE ligands, Advanced Glycation End products (AGEs), HMGB1, and S100 proteins, have all been shown to activate both TLRs and RAGE to varying degrees in order to induce inflammation in in vitro models. As with any field that delves deeply into innate signaling, obstacles of reagent purity may be a cause of some of the discrepancies in the literature, and we have found that commercial antibodies that have been widely used exhibit a high degree of nonspecificity. Nonetheless, the weight of published evidence has led us to speculate that RAGE may be physically interacting with TLRs on the cell surface to elicit inflammation via MyD88-dependent signaling.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Shreeram Nallar
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
53
|
Trujillo-Estrada L, Gomez-Arboledas A, Forner S, Martini AC, Gutierrez A, Baglietto-Vargas D, LaFerla FM. Astrocytes: From the Physiology to the Disease. Curr Alzheimer Res 2020; 16:675-698. [PMID: 31470787 DOI: 10.2174/1567205016666190830110152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
Abstract
Astrocytes are key cells for adequate brain formation and regulation of cerebral blood flow as well as for the maintenance of neuronal metabolism, neurotransmitter synthesis and exocytosis, and synaptic transmission. Many of these functions are intrinsically related to neurodegeneration, allowing refocusing on the role of astrocytes in physiological and neurodegenerative states. Indeed, emerging evidence in the field indicates that abnormalities in the astrocytic function are involved in the pathogenesis of multiple neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD) and Amyotrophic Lateral Sclerosis (ALS). In the present review, we highlight the physiological role of astrocytes in the CNS, including their communication with other cells in the brain. Furthermore, we discuss exciting findings and novel experimental approaches that elucidate the role of astrocytes in multiple neurological disorders.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Stefânia Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Antonia Gutierrez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| |
Collapse
|
54
|
Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol Neurodegener 2020; 15:40. [PMID: 32677986 PMCID: PMC7364557 DOI: 10.1186/s13024-020-00391-7] [Citation(s) in RCA: 541] [Impact Index Per Article: 108.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder seen in age-dependent dementia. There is currently no effective treatment for AD, which may be attributed in part to lack of a clear underlying mechanism. Studies within the last few decades provide growing evidence for a central role of amyloid β (Aβ) and tau, as well as glial contributions to various molecular and cellular pathways in AD pathogenesis. Herein, we review recent progress with respect to Aβ- and tau-associated mechanisms, and discuss glial dysfunction in AD with emphasis on neuronal and glial receptors that mediate Aβ-induced toxicity. We also discuss other critical factors that may affect AD pathogenesis, including genetics, aging, variables related to environment, lifestyle habits, and describe the potential role of apolipoprotein E (APOE), viral and bacterial infection, sleep, and microbiota. Although we have gained much towards understanding various aspects underlying this devastating neurodegenerative disorder, greater commitment towards research in molecular mechanism, diagnostics and treatment will be needed in future AD research.
Collapse
Affiliation(s)
- Tiantian Guo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Denghong Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Yuzhe Zeng
- Department of Orthopaedics, Orthopaedic Center of People's Liberation Army, The Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Timothy Y Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Yingjun Zhao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
55
|
A Role of Low-Density Lipoprotein Receptor-Related Protein 4 (LRP4) in Astrocytic Aβ Clearance. J Neurosci 2020; 40:5347-5361. [PMID: 32457076 DOI: 10.1523/jneurosci.0250-20.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/10/2020] [Accepted: 05/16/2020] [Indexed: 01/28/2023] Open
Abstract
Amyloid-β (Aβ) deposition occurs years before cognitive symptoms appear and is considered a cause of Alzheimer's disease (AD). The imbalance of Aβ production and clearance leads to Aβ accumulation and Aβ deposition. Increasing evidence indicates an important role of astrocytes, the most abundant cell type among glial cells in the brain, in Aβ clearance. We explored the role of low-density lipoprotein receptor-related protein 4 (LRP4), a member of the LDLR family, in AD pathology. We show that Lrp4 is specifically expressed in astrocytes and its levels in astrocytes were higher than those of Ldlr and Lrp1, both of which have been implicated in Aβ uptake. LRP4 was reduced in postmortem brain tissues of AD patients. Genetic deletion of the Lrp4 gene augmented Aβ plaques in 5xFAD male mice, an AD mouse model, and exacerbated the deficits in neurotransmission, synchrony between the hippocampus and PFC, and cognition. Mechanistically, LRP4 promotes Aβ uptake by astrocytes likely by interacting with ApoE. Together, our study demonstrates that astrocytic LRP4 plays an important role in Aβ pathology and cognitive function.SIGNIFICANCE STATEMENT This study investigates how astrocytes, a type of non-nerve cells in the brain, may contribute to Alzheimer's disease (AD) development. We demonstrate that the low-density lipoprotein receptor-related protein 4 (LRP4) is reduced in the brain of AD patients. Mimicking the reduced levels in an AD mouse model exacerbates cognitive impairment and increases amyloid aggregates that are known to damage the brain. We show that LRP4 could promote the clearance of amyloid protein by astrocytes. Our results reveal a previously unappreciated role of LRP4 in AD development.
Collapse
|
56
|
Isenberg JS, Roberts DD. Thrombospondin-1 in maladaptive aging responses: a concept whose time has come. Am J Physiol Cell Physiol 2020; 319:C45-C63. [PMID: 32374675 DOI: 10.1152/ajpcell.00089.2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous age-dependent alterations at the molecular, cellular, tissue and organ systems levels underlie the pathophysiology of aging. Herein, the focus is upon the secreted protein thrombospondin-1 (TSP1) as a promoter of aging and age-related diseases. TSP1 has several physiological functions in youth, including promoting neural synapse formation, mediating responses to ischemic and genotoxic stress, minimizing hemorrhage, limiting angiogenesis, and supporting wound healing. These acute functions of TSP1 generally require only transient expression of the protein. However, accumulating basic and clinical data reinforce the view that chronic diseases of aging are associated with accumulation of TSP1 in the extracellular matrix, which is a significant maladaptive contributor to the aging process. Identification of the relevant cell types that chronically produce and respond to TSP1 and the molecular mechanisms that mediate the resulting maladaptive responses could direct the development of therapeutic agents to delay or revert age-associated maladies.
Collapse
Affiliation(s)
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
57
|
Microglial and Astrocytic Function in Physiological and Pathological Conditions: Estrogenic Modulation. Int J Mol Sci 2020; 21:ijms21093219. [PMID: 32370112 PMCID: PMC7247358 DOI: 10.3390/ijms21093219] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
There are sexual differences in the onset, prevalence, and outcome of numerous neurological diseases. Thus, in Alzheimer’s disease, multiple sclerosis, and major depression disorder, the incidence in women is higher than in men. In contrast, men are more likely to present other pathologies, such as amyotrophic lateral sclerosis, Parkinson’s disease, and autism spectrum. Although the neurological contribution to these diseases has classically always been studied, the truth is that neurons are not the only cells to be affected, and there are other cells, such as glial cells, that are also involved and could be key to understanding the development of these pathologies. Sexual differences exist not only in pathology but also in physiological processes, which shows how cells are differentially regulated in males and females. One of the reasons these sexual differences may occur could be due to the different action of sex hormones. Many studies have shown an increase in aromatase levels in the brain, which could indicate the main role of estrogens in modulating proinflammatory processes. This review will highlight data about sex differences in glial physiology and how estrogenic compounds, such as estradiol and tibolone, could be used as treatment in neurological diseases due to their anti-inflammatory effects and the ability to modulate glial cell functions.
Collapse
|
58
|
Upadhya R, Zingg W, Shetty S, Shetty AK. Astrocyte-derived extracellular vesicles: Neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J Control Release 2020; 323:225-239. [PMID: 32289328 DOI: 10.1016/j.jconrel.2020.04.017] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) released by neural cells play an essential role in brain homeostasis and the crosstalk between neural cells and the periphery. EVs are diverse, nano-sized vesicles, which transport proteins, nucleic acids, and lipids between cells over short and long expanses and hence are proficient for modulating the target cells. EVs released from neural cells are implicated in synaptic plasticity, neuron-glia interface, neuroprotection, neuroregeneration, and the dissemination of neuropathological molecules. This review confers the various properties of EVs secreted by astrocytes and their potential role in health and disease with a focus on evolving concepts. Naïve astrocytes shed EVs containing a host of neuroprotective compounds, which include fibroblast growth factor-2, vascular endothelial growth factor, and apolipoprotein-D. Stimulated astrocytes secrete EVs with neuroprotective molecules including heat shock proteins, synapsin 1, unique microRNAs, and glutamate transporters. Well-characterized astrocyte-derived EVs (ADEVs) generated in specific culture conditions and ADEVs that are engineered to carry the desired miRNAs or proteins are likely useful for treating brain injury and neurogenerative diseases. On the other hand, in conditions such as Alzheimer's disease (AD), stroke, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), and other neuroinflammatory conditions, EVs released by activated astrocytes appear to mediate or exacerbate the pathological processes. The examples include ADEVs spreading the dysregulated complement system in AD, mediating motoneuron toxicity in ALS, and stimulating peripheral leukocyte migration into the brain in inflammatory conditions. Strategies restraining the release of EVs by activated astrocytes or modulating the composition of ADEVs are likely beneficial for treating neurodegenerative diseases. Also, periodic analyses of ADEVs in the blood is useful for detecting astrocyte-specific biomarkers in different neurological conditions and for monitoring disease progression and remission with distinct therapeutic approaches.
Collapse
Affiliation(s)
- Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Winston Zingg
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Siddhant Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA.
| |
Collapse
|
59
|
Xie Z, Zhao J, Wang H, Jiang Y, Yang Q, Fu Y, Zeng H, Hölscher C, Xu J, Zhang Z. Magnolol alleviates Alzheimer's disease-like pathology in transgenic C. elegans by promoting microglia phagocytosis and the degradation of beta-amyloid through activation of PPAR-γ. Biomed Pharmacother 2020; 124:109886. [PMID: 32000045 DOI: 10.1016/j.biopha.2020.109886] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/25/2019] [Accepted: 12/29/2019] [Indexed: 12/28/2022] Open
Abstract
This study aims to investigate whether magnolol (MG), a natural neolignane compound, can prevent AD induced by beta-amyloid (Aβ) and the possible mechanisms involved. MG dose-dependently reduces Aβ deposition, toxicity and memory impairment caused by Aβ in transgenic C. elegans. More importantly, these effects are reversed by GW9662, a selective peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist. MG is more effective in enhancing PPAR-γ luciferase levels than honokiol (HK). Meanwhile, MG has the potential to bind with the ligand binding domain of PPAR-γ (PPAR-γ-LBD). As expected, MG inhibited the luciferase activity of NF-κB and its target genes of inflammatory cytokines, and this effect was blocked by GW9662. The luciferase activity of Nrf2-ARE expression can be activated by MG and decreased Aβ-induced reactive oxygen species (ROS). The target gene LXR of PPAR-γ is activated by MG, which upregulates ApoE and promotes microglia phagocytosis and the degradation of Aβ, and these effects were also reversed by GW9662. In summary, MG can attenuate Aβ-induced AD and the underlying mechanism is the reduction of inflammation and promotion of phagocytosis and degradation of Aβ, which is dependent on PPAR-γ.
Collapse
Affiliation(s)
- Zhishen Xie
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jianping Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Hui Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yali Jiang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Qiaoling Yang
- Department of Pharmacy, Children's Hospital of Shanghai, Children's Hospital Affiliate to Shanghai Jiao Tong University, Shanghai 200040, China
| | - Yu Fu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Huahui Zeng
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiangyan Xu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
60
|
Crespo-Castrillo A, Garcia-Segura LM, Arevalo MA. The synthetic steroid tibolone exerts sex-specific regulation of astrocyte phagocytosis under basal conditions and after an inflammatory challenge. J Neuroinflammation 2020; 17:37. [PMID: 31992325 PMCID: PMC6986022 DOI: 10.1186/s12974-020-1719-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/19/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Tibolone is a synthetic steroid used in clinical practice for the treatment of climacteric symptoms and osteoporosis. Active metabolites of tibolone, generated in target tissues, have an affinity for estrogen and androgen receptors. Astrocytes are direct targets for estrogenic compounds and previous studies have shown that tibolone protects brain cortical neurons in association with a reduction in reactive astrogliosis in a mouse model of traumatic brain injury. Since phagocytosis is a crucial component of the neuroprotective function exerted by astrocytes, in the present study, we have assessed whether tibolone regulates phagocytosis in primary astrocytes incubated with brain-derived cellular debris. METHODS Male and female astrocyte cell cultures were obtained from newborn (P0-P2) female and male Wistar rats. Astrocytic phagocytosis was first characterized using carboxylate beads, Escherichia coli particles, or brain-derived cellular debris. Then, the effect of tibolone on the phagocytosis of Cy3-conjugated cellular debris was quantified by measuring the intensity of Cy3 dye-emitted fluorescence in a given GFAP immunoreactive area. Before the phagocytosis assays, astrocytes were incubated with tibolone in the presence or absence of estrogen or androgen receptor antagonists or an inhibitor of the enzyme that synthesizes estradiol. The effect of tibolone on phagocytosis was analyzed under basal conditions and after inflammatory stimulation with lipopolysaccharide. RESULTS Tibolone stimulated phagocytosis of brain-derived cellular debris by male and female astrocytes, with the effect being more pronounced in females. The effect of tibolone in female astrocytes was blocked by a selective estrogen receptor β antagonist and by an androgen receptor antagonist. None of these antagonists affected tibolone-induced phagocytosis in male astrocytes. In addition, the inhibition of estradiol synthesis in the cultures enhanced the stimulatory effect of tibolone on phagocytosis in male astrocytes but blocked the effect of the steroid in female cells under basal conditions. However, after inflammatory stimulation, the inhibition of estradiol synthesis highly potentiated the stimulation of phagocytosis by tibolone, particularly in female astrocytes. CONCLUSIONS Tibolone exerts sex-specific regulation of phagocytosis in astrocytes of both sexes, both under basal conditions and after inflammatory stimulation.
Collapse
Affiliation(s)
| | - Luis-Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria-Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain. .,Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
61
|
Cheloha RW, Li Z, Bousbaine D, Woodham AW, Perrin P, Volarić J, Ploegh HL. Internalization of Influenza Virus and Cell Surface Proteins Monitored by Site-Specific Conjugation of Protease-Sensitive Probes. ACS Chem Biol 2019; 14:1836-1844. [PMID: 31348637 DOI: 10.1021/acschembio.9b00493] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Commonly used methods to monitor internalization of cell surface structures involve application of fluorescently or otherwise labeled antibodies against the target of interest. Genetic modification of the protein of interest, for example through creation of fusions with fluorescent or enzymatically active protein domains, is another approach to follow trafficking behavior. The former approach requires indirect methods, such as multiple rounds of cell staining, to distinguish between a target that remains surface-disposed and an internalized and/or recycled species. The latter approach necessitates the creation of fusions whose behavior may not accurately reflect that of their unmodified counterparts. Here, we report a method for the characterization of protein internalization in real time through sortase-mediated, site-specific labeling of single-domain antibodies or viral proteins with a newly developed, cathepsin-sensitive quenched-fluorophore probe. Quenched probes of this type have been used to measure enzyme activity in complex environments and for different cell types, but not as a sensor of protein movement into living cells. This approach allows a quantitative assessment of the movement of proteins into protease-containing endosomes in real time in living cells. We demonstrate considerable variation in the rate of endosomal delivery for different cell surface receptors. We were also able to characterize the kinetics of influenza virus delivery to cathepsin-positive compartments, showing highly coordinated arrival in endosomal compartments. This approach should be useful for identifying proteins expressed on cells of interest for targeted endosomal delivery of payloads, such as antibody-drug conjugates or antigens that require processing.
Collapse
Affiliation(s)
- Ross W. Cheloha
- Boston Children’s Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, Massachusetts 02115, United States
| | - Zeyang Li
- Boston Children’s Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, Massachusetts 02115, United States
- Massachusetts Institute of Technology, 455 Main St, Cambridge, Massachusetts 02142, United States
| | - Djenet Bousbaine
- Boston Children’s Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, Massachusetts 02115, United States
- Massachusetts Institute of Technology, 455 Main St, Cambridge, Massachusetts 02142, United States
| | - Andrew W. Woodham
- Boston Children’s Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, Massachusetts 02115, United States
| | - Priscillia Perrin
- Massachusetts Institute of Technology, 455 Main St, Cambridge, Massachusetts 02142, United States
| | - Jana Volarić
- Boston Children’s Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, Massachusetts 02115, United States
| | - Hidde L. Ploegh
- Boston Children’s Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, Massachusetts 02115, United States
| |
Collapse
|
62
|
Wang Z, Zhao Y, Xu N, Zhang S, Wang S, Mao Y, Zhu Y, Li B, Jiang Y, Tan Y, Xie W, Yang BB, Zhang Y. NEAT1 regulates neuroglial cell mediating Aβ clearance via the epigenetic regulation of endocytosis-related genes expression. Cell Mol Life Sci 2019; 76:3005-3018. [PMID: 31006037 PMCID: PMC6647258 DOI: 10.1007/s00018-019-03074-9] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/22/2019] [Accepted: 03/18/2019] [Indexed: 12/17/2022]
Abstract
The accumulation of intracellular β-amyloid peptide (Aβ) is important pathological characteristic of Alzheimer's disease (AD). However, the exact underlying molecular mechanism remains to be elucidated. Here, we reported that Nuclear Paraspeckle Assembly Transcript 1 (NEAT1), a long n on-coding RNA, exhibits repressed expression in the early stage of AD and its down-regulation declines neuroglial cell mediating Aβ clearance via inhibiting expression of endocytosis-related genes. We find that NEAT1 is associated with P300/CBP complex and its inhibition affects H3K27 acetylation (H3K27Ac) and H3K27 crotonylation (H3K27Cro) located nearby to the transcription start site of many genes, including endocytosis-related genes. Interestingly, NEAT1 inhibition down-regulates H3K27Ac but up-regulates H3K27Cro through repression of acetyl-CoA generation. NEAT1 also mediates the binding between STAT3 and H3K27Ac but not H3K27Cro. Therefore, the decrease of H3K27Ac and/or the increase of H3K27Cro declines expression of multiple related genes. Collectively, this study first reveals the different roles of H3K27Ac and H3K27Cro in regulation of gene expression and provides the insight of the epigenetic regulatory mechanism of NEAT1 in gene expression and AD pathology.
Collapse
MESH Headings
- Acetyl Coenzyme A/metabolism
- Acetylation/drug effects
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Amyloid beta-Peptides/metabolism
- Amyloid beta-Peptides/pharmacology
- Animals
- Caveolin 2/antagonists & inhibitors
- Caveolin 2/genetics
- Caveolin 2/metabolism
- Disease Models, Animal
- Epigenesis, Genetic
- Gene Expression/drug effects
- Histones/metabolism
- Mice
- Mice, Transgenic
- Neuroglia/cytology
- Neuroglia/metabolism
- Peptide Fragments/metabolism
- Peptide Fragments/pharmacology
- RNA Interference
- RNA, Long Noncoding/antagonists & inhibitors
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/metabolism
- Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- STAT3 Transcription Factor/metabolism
- Transforming Growth Factor beta2/antagonists & inhibitors
- Transforming Growth Factor beta2/genetics
- Transforming Growth Factor beta2/metabolism
- p300-CBP Transcription Factors/metabolism
Collapse
Affiliation(s)
- Ziqiang Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yiwan Zhao
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Naihan Xu
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- Open FIESTA Center, Tsinghua University, Shenzhen, 518055, China
| | - Shikuan Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Songmao Wang
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Yunhao Mao
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- Open FIESTA Center, Tsinghua University, Shenzhen, 518055, China
| | - Yuanchang Zhu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Bing Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Ying Tan
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- Open FIESTA Center, Tsinghua University, Shenzhen, 518055, China
| | - Weidong Xie
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
- Open FIESTA Center, Tsinghua University, Shenzhen, 518055, China
| | - Burton B Yang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Department of Laboratory Medicine and Pathobiology, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.
| | - Yaou Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Key Laboratory in Health Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China.
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China.
- Open FIESTA Center, Tsinghua University, Shenzhen, 518055, China.
| |
Collapse
|
63
|
de Retana SF, Marazuela P, Solé M, Colell G, Bonaterra A, Sánchez-Quesada JL, Montaner J, Maspoch D, Cano-Sarabia M, Hernández-Guillamon M. Peripheral administration of human recombinant ApoJ/clusterin modulates brain beta-amyloid levels in APP23 mice. ALZHEIMERS RESEARCH & THERAPY 2019; 11:42. [PMID: 31077261 PMCID: PMC6511153 DOI: 10.1186/s13195-019-0498-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Background ApoJ/clusterin is a multifunctional protein highly expressed in the brain. The implication of ApoJ in β-amyloid (Aβ) fibrillization and clearance in the context of Alzheimer’s disease has been widely studied, although the source and concentration of ApoJ that promotes or inhibits Aβ cerebral accumulation is not clear yet. ApoJ is abundant in plasma and approximately 20% can appear bound to HDL-particles. In this regard, the impact of plasmatic ApoJ and its lipidation status on cerebral β-amyloidosis is still not known. Hence, our main objective was to study the effect of a peripheral increase of free ApoJ or reconstituted HDL particles containing ApoJ in an experimental model of cerebral β-amyloidosis. Methods Fourteen-month-old APP23 transgenic mice were subjected to subchronic intravenous treatment with rHDL-rApoJ nanodiscs or free rApoJ for 1 month. Aβ concentration and distribution in the brain, as well as Aβ levels in plasma and CSF, were determined after treatments. Other features associated to AD pathology, such as neuronal loss and neuroinflammation, were also evaluated. Results Both ApoJ-based treatments prevented the Aβ accumulation in cerebral arteries and induced a decrease in total brain insoluble Aβ42 levels. The peripheral treatment with rApoJ also induced an increase in the Aβ40 levels in CSF, whereas the concentration remained unaltered in plasma. At all the endpoints studied, the lipidation of rApoJ did not enhance the protective properties of free rApoJ. The effects obtained after subchronic treatment with free rApoJ were accompanied by a reduction in hippocampal neuronal loss and an enhancement of the expression of a phagocytic marker in microglial cells surrounding Aβ deposits. Finally, despite the activation of this phagocytic phenotype, treatments did not induce a global neuroinflammatory status. In fact, free rApoJ treatment was able to reduce the levels of interleukin-17 (IL17) and keratinocyte chemoattractant (KC) chemokine in the brain. Conclusions Our results demonstrate that an increase in circulating human rApoJ induces a reduction of insoluble Aβ and CAA load in the brain of APP23 mice. Thus, our study suggests that peripheral interventions, based on treatments with multifunctional physiological chaperones, offer therapeutic opportunities to regulate the cerebral Aβ load. Electronic supplementary material The online version of this article (10.1186/s13195-019-0498-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofía Fernández de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Guillem Colell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Anna Bonaterra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,CIBER of Diabetes and Metabolism (CIBERDEM), ISCIII, Madrid, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, , Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, , Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain.
| |
Collapse
|
64
|
Gabandé‐Rodríguez E, Keane L, Capasso M. Microglial phagocytosis in aging and Alzheimer's disease. J Neurosci Res 2019; 98:284-298. [DOI: 10.1002/jnr.24419] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/20/2019] [Accepted: 03/08/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Enrique Gabandé‐Rodríguez
- Department of Molecular Neuropathology Centro de Biología Molecular “Severo Ochoa” (CSIC‐UAM) Madrid Spain
| | - Lily Keane
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| | - Melania Capasso
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| |
Collapse
|
65
|
Byun YG, Chung WS. In Vitro Engulfment Assay to Measure Phagocytic Activity of Astrocytes Using Synaptosomes. Methods Mol Biol 2019; 1938:155-168. [PMID: 30617979 DOI: 10.1007/978-1-4939-9068-9_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Astrocytes eliminate unnecessary synapses, neural debris, and pathogenic proteins such as amyloid β plaque. Although the emerging evidences suggest that the phagocytic roles of astrocytes are critical in maintaining brain homeostasis during development as well as pathogenic conditions, the efficient assay for measuring phagocytic capacity and kinetics of astrocytes has been lacking. Here we present in vitro engulfment assay using purified astrocytes and synaptosomes. Based on imaging methods, either fluorescent or pH indicator-conjugated synaptosomes can be used in this assay.
Collapse
Affiliation(s)
- Youkyeong Gloria Byun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
66
|
Cunningham C, Dunne A, Lopez-Rodriguez AB. Astrocytes: Heterogeneous and Dynamic Phenotypes in Neurodegeneration and Innate Immunity. Neuroscientist 2018; 25:455-474. [PMID: 30451065 DOI: 10.1177/1073858418809941] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Astrocytes are the most numerous cell type in the brain and perform several essential functions in supporting neuronal metabolism and actively participating in neural circuit and behavioral function. They also have essential roles as innate immune cells in responding to local neuropathology, and the manner in which they respond to brain injury and degeneration is the subject of increasing attention in neuroscience. Although activated astrocytes have long been thought of as a relatively homogenous population, which alter their phenotype in a relatively stereotyped way upon central nervous system injury, the last decade has revealed substantial heterogeneity in the basal state and significant heterogeneity of phenotype during reactive astrocytosis. Thus, phenotypic diversity occurs at two distinct levels: that determined by regionality and development and that determined by temporally dynamic changes to the environment of astrocytes during pathology. These inflammatory and pathological states shape the phenotype of these cells, with different consequences for destruction or recovery of the local tissue, and thus elucidating these phenotypic changes has significant therapeutic implications. In this review, we will focus on the phenotypic heterogeneity of astrocytes in health and disease and their propensity to change that phenotype upon subsequent stimuli.
Collapse
Affiliation(s)
- Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland.,School of Medicine, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Ana Belen Lopez-Rodriguez
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| |
Collapse
|
67
|
Jung YJ, Chung WS. Phagocytic Roles of Glial Cells in Healthy and Diseased Brains. Biomol Ther (Seoul) 2018; 26:350-357. [PMID: 29316776 PMCID: PMC6029679 DOI: 10.4062/biomolther.2017.133] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/12/2017] [Accepted: 09/26/2017] [Indexed: 11/10/2022] Open
Abstract
Glial cells are receiving much attention since they have been recognized as important regulators of many aspects of brain function and disease. Recent evidence has revealed that two different glial cells, astrocytes and microglia, control synapse elimination under normal and pathological conditions via phagocytosis. Astrocytes use the MEGF10 and MERTK phagocytic pathways, and microglia use the classical complement pathway to recognize and eliminate unwanted synapses. Notably, glial phagocytosis also contributes to the clearance of disease-specific protein aggregates, such as β-amyloid, huntingtin, and α-synuclein. Here we reivew recent findings showing that glial cells are active regulators in brain functions through phagocytosis and that changes in glial phagocytosis contribute to the pathogenesis of various neurodegenerative diseases. A better understanding of the cellular and molecular mechanisms of glial phagocytosis in healthy and diseased brains will greatly improve our current approach in treating these diseases.
Collapse
Affiliation(s)
- Yeon-Joo Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
68
|
Sankar SB, Donegan RK, Shah KJ, Reddi AR, Wood LB. Heme and hemoglobin suppress amyloid β-mediated inflammatory activation of mouse astrocytes. J Biol Chem 2018; 293:11358-11373. [PMID: 29871926 DOI: 10.1074/jbc.ra117.001050] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/24/2018] [Indexed: 11/06/2022] Open
Abstract
Glial immune activity is a key feature of Alzheimer's disease (AD). Given that the blood factors heme and hemoglobin (Hb) are both elevated in AD tissues and have immunomodulatory roles, here we sought to interrogate their roles in modulating β-amyloid (Aβ)-mediated inflammatory activation of astrocytes. We discovered that heme and Hb suppress immune activity of primary mouse astrocytes by reducing expression of several proinflammatory cytokines (e.g. RANTES (regulated on activation normal T cell expressed and secreted)) and the scavenger receptor CD36 and reducing internalization of Aβ(1-42) by astrocytes. Moreover, we found that certain soluble (>75-kDa) Aβ(1-42) oligomers are primarily responsible for astrocyte activation and that heme or Hb association with these oligomers reverses inflammation. We further found that heme up-regulates phosphoprotein signaling in the phosphoinositide 3-kinase (PI3K)/Akt pathway, which regulates a number of immune functions, including cytokine expression and phagocytosis. The findings in this work suggest that dysregulation of Hb and heme levels in AD brains may contribute to impaired amyloid clearance and that targeting heme homeostasis may reduce amyloid pathogenesis. Altogether, we propose heme as a critical molecular link between amyloid pathology and AD risk factors, such as aging, brain injury, and stroke, which increase Hb and heme levels in the brain.
Collapse
Affiliation(s)
- Sitara B Sankar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Rebecca K Donegan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Kajol J Shah
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Amit R Reddi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332.
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332.
| |
Collapse
|
69
|
Söllvander S, Nikitidou E, Gallasch L, Zyśk M, Söderberg L, Sehlin D, Lannfelt L, Erlandsson A. The Aβ protofibril selective antibody mAb158 prevents accumulation of Aβ in astrocytes and rescues neurons from Aβ-induced cell death. J Neuroinflammation 2018; 15:98. [PMID: 29592816 PMCID: PMC5875007 DOI: 10.1186/s12974-018-1134-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/19/2018] [Indexed: 11/10/2022] Open
Abstract
Background Currently, several amyloid beta (Aβ) antibodies, including the protofibril selective antibody BAN2401, are in clinical trials. The murine version of BAN2401, mAb158, has previously been shown to lower the levels of pathogenic Aβ and prevent Aβ deposition in animal models of Alzheimer’s disease (AD). However, the cellular mechanisms of the antibody’s action remain unknown. We have recently shown that astrocytes effectively engulf Aβ42 protofibrils, but store rather than degrade the ingested Aβ aggregates. In a co-culture set-up, the incomplete degradation of Aβ42 protofibrils by astrocytes results in increased neuronal cell death, due to the release of extracellular vesicles, containing N-truncated, neurotoxic Aβ. Methods The aim of the present study was to investigate if the accumulation of Aβ in astrocytes can be affected by the Aβ protofibril selective antibody mAb158. Co-cultures of astrocytes, neurons, and oligodendrocytes, derived from embryonic mouse cortex, were exposed to Aβ42 protofibrils in the presence or absence of mAb158. Results Our results demonstrate that the presence of mAb158 almost abolished Aβ accumulation in astrocytes. Consequently, mAb158 treatment rescued neurons from Aβ-induced cell death. Conclusion Based on these findings, we conclude that astrocytes may play a central mechanistic role in anti-Aβ immunotherapy. Electronic supplementary material The online version of this article (10.1186/s12974-018-1134-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofia Söllvander
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Elisabeth Nikitidou
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Linn Gallasch
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Marlena Zyśk
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Linda Söderberg
- BioArctic AB, Warfvinges väg 35, SE-112 51, Stockholm, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Erlandsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
70
|
White CS, Lawrence CB, Brough D, Rivers-Auty J. Inflammasomes as therapeutic targets for Alzheimer's disease. Brain Pathol 2018; 27:223-234. [PMID: 28009077 DOI: 10.1111/bpa.12478] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease is the most common form of progressive dementia, typified initially by short term memory deficits which develop into a dramatic global cognitive decline. The classical hall marks of Alzheimer's disease include the accumulation of amyloid oligomers and fibrils, and the intracellular formation of neurofibrillary tangles of hyperphosphorylated tau. It is now clear that inflammation also plays a central role in the pathogenesis of the disease through a number of neurotoxic mechanisms. Microglia are the key immune regulators of the CNS which detect amyloidopathy through cell surface and cytosolic pattern recognition receptors (PRRs) and respond by initiating inflammation through the secretion of cytokines such as interleukin-1β (IL-1β). Inflammasomes, which regulate IL-1β release, are formed following activation of cytosolic PRRs, and using genetic and pharmacological approaches, NLRP3 and NLRP1 inflammasomes have been found to be integral in pathogenic neuroinflammation in animal models of Alzheimer's disease. Therefore, the inflammasomes are very promising novel pharmacological targets which merit further research in the continued endeavor for efficacious therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Claire S White
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - Catherine B Lawrence
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - David Brough
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jack Rivers-Auty
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
71
|
Byun YG, Chung WS. A Novel In Vitro Live-imaging Assay of Astrocyte-mediated Phagocytosis Using pH Indicator-conjugated Synaptosomes. J Vis Exp 2018. [PMID: 29443098 DOI: 10.3791/56647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Astrocytes are the major cell type in the brain and directly contact synapses and blood vessels. Although microglial cells have been considered the major immune cells and only phagocytes in the brain, recent studies have shown that astrocytes also participate in various phagocytic processes, such as developmental synapse elimination and clearance of amyloid beta plaques in Alzheimer's disease (AD). Despite these findings, the efficiency of astrocyte engulfment and degradation of their targets is unclear compared with that of microglia. This lack of information is mostly due to the lack of an assay system in which the kinetics of astrocyte- and microglia-mediated phagocytosis are easily comparable. To achieve this goal, we have developed a long-term live-imaging in vitro phagocytosis assay to evaluate the phagocytic capacity of purified astrocytes and microglia. In this assay, real-time detection of engulfment and degradation is possible using pH indicator-conjugated synaptosomes, which emit bright red fluorescence in acidic organelles, such as lysosomes. Our novel assay provides simple and effective detection of phagocytosis through live-imaging. In addition, this in vitro phagocytosis assay can be used as a screening platform to identify chemicals and compounds that can enhance or inhibit the phagocytic capacity of astrocytes. As synaptic pruning malfunction and pathogenic protein accumulation have been shown to cause mental disorders or neurodegenerative diseases, chemicals and compounds that modulate the phagocytic capacity of glial cells should be helpful in treating various neurological disorders.
Collapse
Affiliation(s)
- Youkyeong Gloria Byun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology;
| |
Collapse
|
72
|
Astroglial Responses to Amyloid-Beta Progression in a Mouse Model of Alzheimer’s Disease. Mol Imaging Biol 2018; 20:605-614. [DOI: 10.1007/s11307-017-1153-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
73
|
Li MZ, Zheng LJ, Shen J, Li XY, Zhang Q, Bai X, Wang QS, Ji JG. SIRT1 facilitates amyloid beta peptide degradation by upregulating lysosome number in primary astrocytes. Neural Regen Res 2018; 13:2005-2013. [PMID: 30233076 PMCID: PMC6183050 DOI: 10.4103/1673-5374.239449] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previous studies have shown that sirtuin 1 (SIRT1) reduces the production of neuronal amyloid beta (Aβ) and inhibits the inflammatory response of glial cells, thereby generating a neuroprotective effect against Aβ neurotoxicity in animal models of Alzheimer's disease. However, the protective effect of SIRT1 on astrocytes is still under investigation. This study established a time point model for the clearance of Aβ in primary astrocytes. Results showed that 12 hours of culture was sufficient for endocytosis of oligomeric Aβ, and 36 hours sufficient for effective degradation. Immunofluorescence demonstrated that Aβ degradation in primary astrocytes relies on lysosome function. Enzymatic agonists or SIRT1 inhibitors were used to stimulate cells over a concentration gradient. Aβ was co-cultured for 36 hours in medium. Western blot assay results under different conditions revealed that SIRT1 relies on its deacetylase activity to promote intracellular Aβ degradation. The experiment further screened SIRT1 using quantitative proteomics to investigate downstream, differentially expressed proteins in the Aβ degradation pathway and selected the ones related to enzyme activity of SIRT1. Most of the differentially expressed proteins detected are close to the primary astrocyte lysosomal pathway. Immunofluorescence staining demonstrated that SIRT1 relies on its deacetylase activity to upregulate lysosome number in primary astrocytes. Taken together, these findings confirm that SIRT1 relies on its deacetylase activity to upregulate lysosome number, thereby facilitating oligomeric Aβ degradation in primary astrocytes.
Collapse
Affiliation(s)
- Min-Zhe Li
- General Surgery Department, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Liang-Jun Zheng
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Jian Shen
- General Surgery Department, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xin-Ya Li
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Qi Zhang
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Xue Bai
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Qing-Song Wang
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Jian-Guo Ji
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
74
|
de Godoy MA, Saraiva LM, de Carvalho LRP, Vasconcelos-Dos-Santos A, Beiral HJV, Ramos AB, Silva LRDP, Leal RB, Monteiro VHS, Braga CV, de Araujo-Silva CA, Sinis LC, Bodart-Santos V, Kasai-Brunswick TH, Alcantara CDL, Lima APCA, da Cunha-E Silva NL, Galina A, Vieyra A, De Felice FG, Mendez-Otero R, Ferreira ST. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. J Biol Chem 2017; 293:1957-1975. [PMID: 29284679 DOI: 10.1074/jbc.m117.807180] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a disabling and highly prevalent neurodegenerative condition, for which there are no effective therapies. Soluble oligomers of the amyloid-β peptide (AβOs) are thought to be proximal neurotoxins involved in early neuronal oxidative stress and synapse damage, ultimately leading to neurodegeneration and memory impairment in AD. The aim of the current study was to evaluate the neuroprotective potential of mesenchymal stem cells (MSCs) against the deleterious impact of AβOs on hippocampal neurons. To this end, we established transwell cocultures of rat hippocampal neurons and MSCs. We show that MSCs and MSC-derived extracellular vesicles protect neurons against AβO-induced oxidative stress and synapse damage, revealed by loss of pre- and postsynaptic markers. Protection by MSCs entails three complementary mechanisms: 1) internalization and degradation of AβOs; 2) release of extracellular vesicles containing active catalase; and 3) selective secretion of interleukin-6, interleukin-10, and vascular endothelial growth factor to the medium. Results support the notion that MSCs may represent a promising alternative for cell-based therapies in AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Renata B Leal
- From the Institute of Biophysics Carlos Chagas Filho
| | | | | | | | | | | | | | | | | | | | - Antonio Galina
- the Institute of Medical Biochemistry Leopoldo de Meis, and
| | - Adalberto Vieyra
- From the Institute of Biophysics Carlos Chagas Filho.,the National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | | | | | - Sergio T Ferreira
- From the Institute of Biophysics Carlos Chagas Filho, .,the Institute of Medical Biochemistry Leopoldo de Meis, and
| |
Collapse
|
75
|
González-Reyes RE, Nava-Mesa MO, Vargas-Sánchez K, Ariza-Salamanca D, Mora-Muñoz L. Involvement of Astrocytes in Alzheimer's Disease from a Neuroinflammatory and Oxidative Stress Perspective. Front Mol Neurosci 2017; 10:427. [PMID: 29311817 PMCID: PMC5742194 DOI: 10.3389/fnmol.2017.00427] [Citation(s) in RCA: 352] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer disease (AD) is a frequent and devastating neurodegenerative disease in humans, but still no curative treatment has been developed. Although many explicative theories have been proposed, precise pathophysiological mechanisms are unknown. Due to the importance of astrocytes in brain homeostasis they have become interesting targets for the study of AD. Changes in astrocyte function have been observed in brains from individuals with AD, as well as in AD in vitro and in vivo animal models. The presence of amyloid beta (Aβ) has been shown to disrupt gliotransmission, neurotransmitter uptake, and alter calcium signaling in astrocytes. Furthermore, astrocytes express apolipoprotein E and are involved in the production, degradation and removal of Aβ. As well, changes in astrocytes that precede other pathological characteristics observed in AD, point to an early contribution of astroglia in this disease. Astrocytes participate in the inflammatory/immune responses of the central nervous system. The presence of Aβ activates different cell receptors and intracellular signaling pathways, mainly the advanced glycation end products receptor/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, responsible for the transcription of pro-inflammatory cytokines and chemokines in astrocytes. The release of these pro-inflammatory agents may induce cellular damage or even stimulate the production of Aβ in astrocytes. Additionally, Aβ induces the appearance of oxidative stress (OS) and production of reactive oxygen species and reactive nitrogen species in astrocytes, affecting among others, intracellular calcium levels, NADPH oxidase (NOX), NF-κB signaling, glutamate uptake (increasing the risk of excitotoxicity) and mitochondrial function. Excessive neuroinflammation and OS are observed in AD, and astrocytes seem to be involved in both. The Aβ/NF-κB interaction in astrocytes may play a central role in these inflammatory and OS changes present in AD. In this paper, we also discuss therapeutic measures highlighting the importance of astrocytes in AD pathology. Several new therapeutic approaches involving phenols (curcumin), phytoestrogens (genistein), neuroesteroids and other natural phytochemicals have been explored in astrocytes, obtaining some promising results regarding cognitive improvements and attenuation of neuroinflammation. Novel strategies comprising astrocytes and aimed to reduce OS in AD have also been proposed. These include estrogen receptor agonists (pelargonidin), Bambusae concretio Salicea, Monascin, and various antioxidatives such as resveratrol, tocotrienol, anthocyanins, and epicatechin, showing beneficial effects in AD models.
Collapse
Affiliation(s)
- Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - Daniel Ariza-Salamanca
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Laura Mora-Muñoz
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
76
|
Gustafsson G, Lindström V, Rostami J, Nordström E, Lannfelt L, Bergström J, Ingelsson M, Erlandsson A. Alpha-synuclein oligomer-selective antibodies reduce intracellular accumulation and mitochondrial impairment in alpha-synuclein exposed astrocytes. J Neuroinflammation 2017; 14:241. [PMID: 29228971 PMCID: PMC5725978 DOI: 10.1186/s12974-017-1018-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/01/2017] [Indexed: 11/29/2022] Open
Abstract
Background Due to its neurotoxic properties, oligomeric alpha-synuclein (α-syn) has been suggested as an attractive target for passive immunization against Parkinson’s disease (PD). In mouse models of PD, antibody treatment has been shown to lower the levels of pathogenic α-syn species, including oligomers, although the mechanisms of action remain unknown. We have previously shown that astrocytes rapidly engulf α-syn oligomers that are intracellularly stored, rather than degraded, resulting in impaired mitochondria. Methods The aim of the present study was to investigate if the accumulation of α-syn in astrocytes can be affected by α-syn oligomer-selective antibodies. Co-cultures of astrocytes, neurons, and oligodendrocytes were derived from embryonic mouse cortex and exposed to α-syn oligomers or oligomers pre-incubated with oligomer-selective antibodies. Results In the presence of antibodies, the astrocytes displayed an increased clearance of the exogenously added α-syn, and consequently, the α-syn accumulation in the culture was markedly reduced. Moreover, the addition of antibodies rescued the astrocytes from the oligomer-induced mitochondrial impairment. Conclusions Our results demonstrate that oligomer-selective antibodies can prevent α-syn accumulation and mitochondrial dysfunction in cultured astrocytes.
Collapse
Affiliation(s)
- Gabriel Gustafsson
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Veronica Lindström
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Jinar Rostami
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Eva Nordström
- BioArctic AB, Warfvinges väg 35, 112 51, Stockholm, Sweden
| | - Lars Lannfelt
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Joakim Bergström
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Martin Ingelsson
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Anna Erlandsson
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
77
|
Gomez‐Arboledas A, Davila JC, Sanchez‐Mejias E, Navarro V, Nuñez‐Diaz C, Sanchez‐Varo R, Sanchez‐Mico MV, Trujillo‐Estrada L, Fernandez‐Valenzuela JJ, Vizuete M, Comella JX, Galea E, Vitorica J, Gutierrez A. Phagocytic clearance of presynaptic dystrophies by reactive astrocytes in Alzheimer's disease. Glia 2017; 66:637-653. [PMID: 29178139 PMCID: PMC5814816 DOI: 10.1002/glia.23270] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 01/01/2023]
Abstract
Reactive astrogliosis, a complex process characterized by cell hypertrophy and upregulation of components of intermediate filaments, is a common feature in brains of Alzheimer's patients. Reactive astrocytes are found in close association with neuritic plaques; however, the precise role of these glial cells in disease pathogenesis is unknown. In this study, using immunohistochemical techniques and light and electron microscopy, we report that plaque-associated reactive astrocytes enwrap, engulf and may digest presynaptic dystrophies in the hippocampus of amyloid precursor protein/presenilin-1 (APP/PS1) mice. Microglia, the brain phagocytic population, was apparently not engaged in this clearance. Phagocytic reactive astrocytes were present in 35% and 67% of amyloid plaques at 6 and 12 months of age, respectively. The proportion of engulfed dystrophic neurites was low, around 7% of total dystrophies around plaques at both ages. This fact, along with the accumulation of dystrophic neurites during disease course, suggests that the efficiency of the astrocyte phagocytic process might be limited or impaired. Reactive astrocytes surrounding and engulfing dystrophic neurites were also detected in the hippocampus of Alzheimer's patients by confocal and ultrastructural analysis. We posit that the phagocytic activity of reactive astrocytes might contribute to clear dysfunctional synapses or synaptic debris, thereby restoring impaired neural circuits and reducing the inflammatory impact of damaged neuronal parts and/or limiting the amyloid pathology. Therefore, potentiation of the phagocytic properties of reactive astrocytes may represent a potential therapy in Alzheimer's disease.
Collapse
Affiliation(s)
- Angela Gomez‐Arboledas
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Jose C. Davila
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Elisabeth Sanchez‐Mejias
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Victoria Navarro
- Dpto. Bioquimica y Biologia Molecular, Facultad de FarmaciaUniversidad de SevillaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Instituto de Biomedicina de Sevilla (IBiS)‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSpain
| | - Cristina Nuñez‐Diaz
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Raquel Sanchez‐Varo
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Maria Virtudes Sanchez‐Mico
- Dpto. Bioquimica y Biologia Molecular, Facultad de FarmaciaUniversidad de SevillaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Instituto de Biomedicina de Sevilla (IBiS)‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSpain
| | - Laura Trujillo‐Estrada
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Juan Jose Fernandez‐Valenzuela
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Marisa Vizuete
- Dpto. Bioquimica y Biologia Molecular, Facultad de FarmaciaUniversidad de SevillaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Instituto de Biomedicina de Sevilla (IBiS)‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSpain
| | - Joan X. Comella
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de BarcelonaSpain
- Institut de Recerca de l'Hopital Univesitary de la Vall d'Hebron (VHIR)BarcelonaSpain
| | - Elena Galea
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de BarcelonaSpain
- ICREA, Pg. Lluís Companys 23Barcelona08010Spain
| | - Javier Vitorica
- Dpto. Bioquimica y Biologia Molecular, Facultad de FarmaciaUniversidad de SevillaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
- Instituto de Biomedicina de Sevilla (IBiS)‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSpain
| | - Antonia Gutierrez
- Dpto. Biologia Celular, Genetica y Fisiologia. Facultad de CienciasInstituto de Biomedicina de Malaga (IBIMA), Universidad de MalagaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| |
Collapse
|
78
|
Human Astrocytes Transfer Aggregated Alpha-Synuclein via Tunneling Nanotubes. J Neurosci 2017; 37:11835-11853. [PMID: 29089438 PMCID: PMC5719970 DOI: 10.1523/jneurosci.0983-17.2017] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/20/2017] [Accepted: 10/11/2017] [Indexed: 12/30/2022] Open
Abstract
Many lines of evidence suggest that the Parkinson's disease (PD)-related protein α-synuclein (α-SYN) can propagate from cell to cell in a prion-like manner. However, the cellular mechanisms behind the spreading remain elusive. Here, we show that human astrocytes derived from embryonic stem cells actively transfer aggregated α-SYN to nearby astrocytes via direct contact and tunneling nanotubes (TNTs). Failure in the astrocytes' lysosomal digestion of excess α-SYN oligomers results in α-SYN deposits in the trans-Golgi network followed by endoplasmic reticulum swelling and mitochondrial disturbances. The stressed astrocytes respond by conspicuously sending out TNTs, enabling intercellular transfer of α-SYN to healthy astrocytes, which in return deliver mitochondria, indicating a TNT-mediated rescue mechanism. Using a pharmacological approach to inhibit TNT formation, we abolished the transfer of both α-SYN and mitochondria. Together, our results highlight the role of astrocytes in α-SYN cell-to-cell transfer, identifying possible pathophysiological events in the PD brain that could be of therapeutic relevance. SIGNIFICANCE STATEMENT Astrocytes are the major cell type in the brain, yet their role in Parkinson's disease progression remains elusive. Here, we show that human astrocytes actively transfer aggregated α-synuclein (α-SYN) to healthy astrocytes via direct contact and tunneling nanotubes (TNTs), rather than degrade it. The astrocytes engulf large amounts of oligomeric α-SYN that are subsequently stored in the trans-Golgi network region. The accumulation of α-SYN in the astrocytes affects their lysosomal machinery and induces mitochondrial damage. The stressed astrocytes respond by sending out TNTs, enabling intercellular transfer of α-SYN to healthy astrocytes. Our findings highlight an unexpected role of astrocytes in the propagation of α-SYN pathology via TNTs, revealing astrocytes as a potential target for therapeutic intervention.
Collapse
|
79
|
Hopley RT, Haller E, Rojiani MV, Rojiani AM. Morphologic and Elemental Analysis of Primary Melanosis of the Dentate Nucleus: Review and Correlation With Neuromelanin. J Neuropathol Exp Neurol 2017; 76:949-956. [PMID: 29044415 DOI: 10.1093/jnen/nlx084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Primary melanosis of the dentate nucleus is a rarely described entity with neither known cause nor definitive clinicopathologic correlation. We revisit this previously reported phenomenon by presenting one such case with a review of the pathology as well as additional investigations including elemental analysis by energy-dispersive X-ray, immunohistochemistry and electron microscopy. The lesion presented macroscopically as a sharply defined, black pigmentation that was restricted to the dentate nucleus of the cerebellum. Other deep nuclei were uninvolved. Similarly, other areas of the cerebellum, brainstem, and supratentorial regions were macroscopically free of pigment. Microscopically, however, the pigment was noted to be present, albeit in microscopic deposits, within layers of the cerebellar cortex. Additionally, immunohistochemistry and electron microscopy defined an intracellular component within astrocytes. X-ray analysis of the pigment showed it to consist almost entirely of sulfur, an element known to be prominent in neuromelanin. This report also describes an association of the pigment with astrocytes by ultrastructural examination. We discuss the results of our findings in the context of etiopathogenetic considerations, seeking to gain a better understanding of this abnormal pigmentation and its relationship to neuromelanin.
Collapse
Affiliation(s)
- Richard T Hopley
- Department of Pathology, School of Medicine, University of South Florida-Morsani, Tampa, Florida; Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Edward Haller
- Department of Pathology, School of Medicine, University of South Florida-Morsani, Tampa, Florida; Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Mumtaz V Rojiani
- Department of Pathology, School of Medicine, University of South Florida-Morsani, Tampa, Florida; Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Amyn M Rojiani
- Department of Pathology, School of Medicine, University of South Florida-Morsani, Tampa, Florida; Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
80
|
Amyloid-beta neurotoxicity and clearance are both regulated by glial group II metabotropic glutamate receptors. Neuropharmacology 2017; 123:274-286. [DOI: 10.1016/j.neuropharm.2017.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/05/2017] [Accepted: 05/07/2017] [Indexed: 11/20/2022]
|
81
|
FTY720 Attenuates Infection-Induced Enhancement of Aβ Accumulation in APP/PS1 Mice by Modulating Astrocytic Activation. J Neuroimmune Pharmacol 2017. [PMID: 28620801 DOI: 10.1007/s11481-017-9753-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
It is well established that infection has a significant detrimental effect on patients with Alzheimer's disease (AD), accelerating cognitive decline and, even in healthy ageing individuals, increasing amyloid-β (Aβ) accumulation in the brain. In animal models of AD infection can also cause damage, with evidence of increased neuroinflammation, amyloid pathology and deterioration of cognitive function. These changes are against a backdrop of an age- and AD-related increase in susceptibility to infection. Here we set out to determine whether FTY720, a molecule that binds sphingosine-1-phosphate (S1P) receptors and with known immunosuppressant effects mediating its therapeutic action in multiple sclerosis (MS), might modulate the impact of infection in a mouse model of AD. Transgenic mice that overexpress amyloid precursor protein (APP) and presenilin 1 (PS1; APP/PS1 mice) and their littermates were/were not infected with Bordetella pertussis and were treated orally with FTY720 or vehicle beginning 3 days before infection. Infection increased astrocytic activation and enhanced blood brain barrier (BBB) permeability and these changes were attenuated in FTY720-treated B. pertussis-infected mice. Significantly, infection increased Aβ containing plaques and soluble Aβ and these infection-related changes were also attenuated in FTY720-treated B. pertussis-infected mice. The data suggest that this effect results from an FTY720-induced increase in Aβ phagocytosis by astrocytes. FTY720 did not impact on genotype-related changes in the absence of an infection indicating that its potential usefulness is restricted to reducing the impact of acute inflammatory stimuli in AD.
Collapse
|
82
|
Acosta C, Anderson HD, Anderson CM. Astrocyte dysfunction in Alzheimer disease. J Neurosci Res 2017; 95:2430-2447. [PMID: 28467650 DOI: 10.1002/jnr.24075] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022]
Abstract
Astrocytes are glial cells that are distributed throughout the central nervous system in an arrangement optimal for chemical and physical interaction with neuronal synapses and brain blood supply vessels. Neurotransmission modulates astrocytic excitability by activating an array of cell surface receptors and transporter proteins, resulting in dynamic changes in intracellular Ca2+ or Na+ . Ionic and electrogenic astrocytic changes, in turn, drive vital cell nonautonomous effects supporting brain function, including regulation of synaptic activity, neuronal metabolism, and regional blood supply. Alzheimer disease (AD) is associated with aberrant oligomeric amyloid β generation, which leads to extensive proliferation of astrocytes with a reactive phenotype and abnormal regulation of these processes. Astrocytic morphology, Ca2+ responses, extracellular K+ removal, glutamate transport, amyloid clearance, and energy metabolism are all affected in AD, resulting in a deleterious set of effects that includes glutamate excitotoxicity, impaired synaptic plasticity, reduced carbon delivery to neurons for oxidative phosphorylation, and dysregulated linkages between neuronal energy demand and regional blood supply. This review summarizes how astrocytes are affected in AD and describes how these changes are likely to influence brain function. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Crystal Acosta
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Canadian Centre for Agri-food Research in Health and Medicine, St. Boniface Hospital Research, Winnipeg, Manitoba, Canada
| | - Hope D Anderson
- Canadian Centre for Agri-food Research in Health and Medicine, St. Boniface Hospital Research, Winnipeg, Manitoba, Canada.,College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Christopher M Anderson
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
83
|
Kawabe K, Takano K, Moriyama M, Nakamura Y. Transglutaminases Derived from Astrocytes Accelerate Amyloid β Aggregation. Neurochem Res 2017; 42:2384-2391. [DOI: 10.1007/s11064-017-2258-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
|
84
|
Campos-Peña V, Toral-Rios D, Becerril-Pérez F, Sánchez-Torres C, Delgado-Namorado Y, Torres-Ossorio E, Franco-Bocanegra D, Carvajal K. Metabolic Syndrome as a Risk Factor for Alzheimer's Disease: Is Aβ a Crucial Factor in Both Pathologies? Antioxid Redox Signal 2017; 26:542-560. [PMID: 27368351 DOI: 10.1089/ars.2016.6768] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Recently, chronic degenerative diseases have become one of the main health problems worldwide. That is the case of Alzheimer's disease (AD) and metabolic syndrome (MetS), whose expression can be influenced by different risk factors. Recent Advances: In recent decades, it has been widely described that MetS increases the risk of cognitive impairment and dementia. MetS pathogenesis involves several vascular risk factors such as diabetes, dyslipidemia, hypertension, and insulin resistance (I/R). CRITICAL ISSUES Reported evidence shows that vascular risk factors are associated with AD, particularly in the development of protein aggregation, inflammation, oxidative stress, neuronal dysfunction, and disturbances in signaling pathways, with insulin receptor signaling being a common alteration between MetS and AD. FUTURE DIRECTIONS Insulin signaling has been involved in tau phosphorylation and amyloid β (Aβ) metabolism. However, it has also been demonstrated that Aβ oligomers can bind to insulin receptors, triggering their internalization, decreasing neuron responsiveness to insulin, and promoting insulin I/R. Thus, it could be argued that Aβ could be a convergent factor in the development of both pathologies. Antioxid. Redox Signal. 26, 542-560.
Collapse
Affiliation(s)
| | - Danira Toral-Rios
- 2 Departamento de Fisiología Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Mexico City, Mexico
| | | | - Carmen Sánchez-Torres
- 4 Departamento of Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Mexico City, Mexico
| | | | - Elimar Torres-Ossorio
- 6 Facultad de Química, Universidad Nacional Autónoma de México , Mexico City, Mexico
| | | | - Karla Carvajal
- 7 Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría , Mexico City, Mexico
| |
Collapse
|
85
|
Eltony SA, Abdelhameed SY. Effect of chronic administration of sildenafil citrate (Viagra) on the histology of the retina and optic nerve of adult male rat. Tissue Cell 2017; 49:323-335. [PMID: 28237322 DOI: 10.1016/j.tice.2017.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Abnormal vision has been reported by 3% of patients treated with sildenafil citrate (Viagra). Although many men use Viagra for an extended period for treatment of erectile dysfunction, the implications of the long term-daily use of it on the retina and optic nerve are unclear. AIM OF THE WORK To investigate the effect of chronic daily use of sildenafil citrate in a dose equivalent to men preferred therapeutic dose on the histology of the retina and optic nerve of adult male rat. MATERIAL & METHODS Eighteen adult male Wistar rats were equally divided into three groups. Group I: control. Group II: treated with sildenafil citrate orally (10mg/kg/day) for 8 weeks. Group III (withdrawal): treated as group II and then left for 4 weeks without treatment. Specimens from the retina and optic nerve were processed for light and electron microscopy. RESULTS In sildenafil citrate treated group, the retina and optic nerve revealed vacuolations and congested blood capillaries with apoptotic endothelial and pericytic cells, and thickened basal lamina. Caspase-3 (apoptotic marker) and CD31 (endothelial marker) expression increased. Glial cells revealed morphological changes: Müller cells lost their processes, activated microglia, astrocytic clasmatodendrosis, degenerated oligodendrocytes surrounded by disintegrated myelin sheathes of the optic nerve fibers. The retina and optic nerve of the withdrawal group revealed less vacuolations and congestion, and partial recovery of the glial cells. CONCLUSION Chronic treatment with sildenafil citrate (Viagra) caused toxic effect on the structure of the retina and optic nerve of the rat. Partial recovery was observed after drug withdrawal.
Collapse
Affiliation(s)
- Sohair A Eltony
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Egypt.
| | - Sally Y Abdelhameed
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Assiut University, Egypt
| |
Collapse
|
86
|
Schubert M, Volckaert K, Karl M, Morton A, Liehm P, Miles GB, Powis SJ, Gather MC. Lasing in Live Mitotic and Non-Phagocytic Cells by Efficient Delivery of Microresonators. Sci Rep 2017; 7:40877. [PMID: 28102341 PMCID: PMC5244359 DOI: 10.1038/srep40877] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/13/2016] [Indexed: 01/03/2023] Open
Abstract
Reliable methods to individually track large numbers of cells in real time are urgently needed to advance our understanding of important biological processes like cancer metastasis, neuronal network development and wound healing. It has recently been suggested to introduce microscopic whispering gallery mode lasers into the cytoplasm of cells and to use their characteristic, size-dependent emission spectrum as optical barcode but so far there is no evidence that this approach is generally applicable. Here, we describe a method that drastically improves intracellular delivery of resonators for several cell types, including mitotic and non-phagocytic cells. In addition, we characterize the influence of resonator size on the spectral characteristics of the emitted laser light and identify an optimum size range that facilitates tagging and tracking of thousands of cells simultaneously. Finally, we observe that the microresonators remain internalized by cells during cell division, which enables tagging several generations of cells.
Collapse
Affiliation(s)
- Marcel Schubert
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Klara Volckaert
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Markus Karl
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Andrew Morton
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Philipp Liehm
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Simon J Powis
- School of Medicine, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Malte C Gather
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| |
Collapse
|
87
|
Abstract
Following cellular engulfment, nanoparticles end up in the lysosomes, making them an ideal tool for modifying the lysosomal environment. Here, we describe how acidic nanoparticles can be used to lower the pH of lysosomes in cultured, primary astrocytes and thereby increase their degradation capacity. To guarantee that the cell culture is completely devoid of professional phagocytes, we isolate, expand, and differentiate neural stem cells from embryonic mouse cortex to achieve astrocytes for these experiments. Immunostainings with LAMP2-specific antibodies can be performed to verify the lysosomal localization of the nanoparticles, and the effect on lysosomal acidification can easily be followed with LysoTracker dye.
Collapse
Affiliation(s)
- Camilla Lööv
- MassGeneral Institute for Neurodegeneration, Massachusetts General Hospital, Harvard Medical School, MA, 02129, Charlestown, USA
| | - Anna Erlandsson
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden.
| |
Collapse
|
88
|
Zhao S, Yu Z, Liu Y, Bai Y, Jiang Y, van Leyen K, Yang YG, Lok JM, Whalen MJ, Lo EH, Wang X. CD47 deficiency improves neurological outcomes of traumatic brain injury in mice. Neurosci Lett 2016; 643:125-130. [PMID: 27931776 DOI: 10.1016/j.neulet.2016.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/02/2016] [Accepted: 12/03/2016] [Indexed: 10/20/2022]
Abstract
CD47 is a receptor for signal-regulatory protein alpha (SIRPα) in self-recognition by the innate immune system, and a receptor of thrombospondin-1 (TSP-1) contributing to vascular impairment in response to stress. However, the roles of CD47 in traumatic brain injury (TBI) have not been investigated. In this study we aimed to test our hypothesis that CD47 mediates early neutrophil brain infiltration and late brain vascular remodeling after TBI. Mice were subjected to TBI using a controlled cortical impact (CCI) device. We examined early phase neutrophil infiltration, and late phase brain vessel density, pro-angiogenic markers VEGF and Ang-1 protein expression, neurological function deficits and lesion volumes for up to three weeks after TBI. Our results show that mice deficient in CD47 (CD47 Knockout) had significantly less brain neutrophil infiltration at 24h, upregulated VEGF expression in peri-lesion cortex at 7 and 14days, and increased blood vessel density at 21days after TBI, compared to wild type (WT) mice. CD47 knockout also significantly decreased sensorimotor function deficits and reduced brain lesion volume at 21days after TBI. We conclude that CD47 may play pathological roles in brain neutrophil infiltration, progression of brain tissue damage, impairment of cerebrovascular remodeling and functional recovery after TBI.
Collapse
Affiliation(s)
- Song Zhao
- Departments of Orthopedic and Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, China; Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA.
| | - Zhanyang Yu
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA.
| | - Yu Liu
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA; Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Yang Bai
- Departments of Orthopedic and Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, China.
| | - Yinghua Jiang
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA.
| | - Klaus van Leyen
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA.
| | - Yong-Guang Yang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| | - Josephine M Lok
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA; Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Michael J Whalen
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA.
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Room 2401, Charlestown, MA 02129, USA.
| |
Collapse
|
89
|
Tondera C, Laube M, Pietzsch J. Insights into binding of S100 proteins to scavenger receptors: class B scavenger receptor CD36 binds S100A12 with high affinity. Amino Acids 2016; 49:183-191. [PMID: 27734162 PMCID: PMC5241339 DOI: 10.1007/s00726-016-2349-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/06/2016] [Indexed: 11/29/2022]
Abstract
The EF-hand type calcium-binding protein S100A12 exerts numerous intra- and extracellular functions of (patho)physiological relevance. Therefore, receptors of S100A12 are of high interest for research and clinical applications. Beside the extensively studied receptor for advanced glycation endproducts (RAGE), G-protein coupled receptors and more recently, scavenger receptors are suggested to be putative S100A12 receptors. Own findings and further information from the literature predestined CD36, a class B scavenger receptor, as promising candidate. To substantiate or prove against this hypothesis, this study aimed at investigation of interaction of S100A12 and CD36 on molecular and cellular level by the use of surface plasmon resonance (SPR), radio- and fluorescence-tracer-based cell binding, and cell activation experiments. S100A12 revealed binding affinity to CD36 in the low nanomolar range, essentially, at the CD36 thrombospondin-1 binding site. Additionally, S100A12-mediated translocation of CD36 to the membrane and elevation of both CD36 and peroxisome proliferator-activated receptor γ (PPARγ) expression was observed, which suggest a potential regulatory function of S100A12–CD36 interaction.
Collapse
Affiliation(s)
- Christoph Tondera
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany. .,Department of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
90
|
Lasič E, Galland F, Vardjan N, Šribar J, Križaj I, Leite MC, Zorec R, Stenovec M. Time-dependent uptake and trafficking of vesicles capturing extracellular S100B in cultured rat astrocytes. J Neurochem 2016; 139:309-323. [PMID: 27488079 DOI: 10.1111/jnc.13754] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/21/2016] [Accepted: 07/26/2016] [Indexed: 01/16/2023]
Abstract
Astrocytes, the most heterogeneous glial cells in the central nervous system, contribute to brain homeostasis, by regulating a myriad of functions, including the clearance of extracellular debris. When cells are damaged, cytoplasmic proteins may exit into the extracellular space. One such protein is S100B, which may exert toxic effects on neighboring cells unless it is removed from the extracellular space, but the mechanisms of this clearance are poorly understood. By using time-lapse confocal microscopy and fluorescently labeled S100B (S100B-Alexa488 ) and fluorescent dextran (Dextran546 ), a fluid phase uptake marker, we examined the uptake of fluorescently labeled S100B-Alexa488 from extracellular space and monitored trafficking of vesicles that internalized S100B-Alexa488 . Initially, S100B-Alexa488 and Dextran546 internalized with distinct rates into different endocytotic vesicles; S100B-Alexa488 internalized into smaller vesicles than Dextran546 . At a later stage, S100B-Alexa488 -positive vesicles substantially co-localized with Dextran546 -positive endolysosomes and with acidic LysoTracker-positive vesicles. Cell treatment with anti-receptor for advanced glycation end products (RAGE) antibody, which binds to RAGE, a 'scavenger receptor', partially inhibited uptake of S100B-Alexa488 , but not of Dextran546 . The dynamin inhibitor dynole 34-2 inhibited internalization of both fluorescent probes. Directional mobility of S100B-Alexa488 -positive vesicles increased over time and was inhibited by ATP stimulation, an agent that increases cytosolic free calcium concentration ([Ca2+ ]i ). We conclude that astrocytes exhibit RAGE- and dynamin-dependent vesicular mechanism to efficiently remove S100B from the extracellular space. If a similar process occurs in vivo, astroglia may mitigate the toxic effects of extracellular S100B by this process under pathophysiologic conditions. This study reveals the vesicular clearance mechanism of extracellular S100B in astrocytes. Initially, fluorescent S100B internalizes into smaller endocytotic vesicles than dextran molecules. At a later stage, both probes co-localize within endolysosomes. S100B internalization is both dynamin- and RAGE-dependent, whereas dextran internalization is dependent on dynamin. Vesicle internalization likely mitigates the toxic effects of extracellular S100B and other waste products.
Collapse
Affiliation(s)
- Eva Lasič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fabiana Galland
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Celica Biomedical, Ljubljana, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia. .,Celica Biomedical, Ljubljana, Slovenia.
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia. .,Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
91
|
Ries M, Sastre M. Mechanisms of Aβ Clearance and Degradation by Glial Cells. Front Aging Neurosci 2016; 8:160. [PMID: 27458370 PMCID: PMC4932097 DOI: 10.3389/fnagi.2016.00160] [Citation(s) in RCA: 393] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/17/2016] [Indexed: 12/24/2022] Open
Abstract
Glial cells have a variety of functions in the brain, ranging from immune defense against external and endogenous hazardous stimuli, regulation of synaptic formation, calcium homeostasis, and metabolic support for neurons. Their dysregulation can contribute to the development of neurodegenerative disorders, including Alzheimer’s disease (AD). One of the most important functions of glial cells in AD is the regulation of Amyloid-β (Aβ) levels in the brain. Microglia and astrocytes have been reported to play a central role as moderators of Aβ clearance and degradation. The mechanisms of Aβ degradation by glial cells include the production of proteases, including neprilysin, the insulin degrading enzyme, and the endothelin-converting enzymes, able to hydrolyse Aβ at different cleavage sites. Besides these enzymes, other proteases have been described to have some role in Aβ elimination, such as plasminogen activators, angiotensin-converting enzyme, and matrix metalloproteinases. Other relevant mediators that are released by glial cells are extracellular chaperones, involved in the clearance of Aβ alone or in association with receptors/transporters that facilitate their exit to the blood circulation. These include apolipoproteins, α2macroglobulin, and α1-antichymotrypsin. Finally, astrocytes and microglia have an essential role in phagocytosing Aβ, in many cases via a number of receptors that are expressed on their surface. In this review, we examine all of these mechanisms, providing an update on the latest research in this field.
Collapse
Affiliation(s)
- Miriam Ries
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital London, UK
| | - Magdalena Sastre
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital London, UK
| |
Collapse
|
92
|
Astrocyte-Microglia Cross Talk through Complement Activation Modulates Amyloid Pathology in Mouse Models of Alzheimer's Disease. J Neurosci 2016; 36:577-89. [PMID: 26758846 DOI: 10.1523/jneurosci.2117-15.2016] [Citation(s) in RCA: 409] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Increasing evidence supports a role of neuroinflammation in the pathogenesis of Alzheimer's disease (AD). Previously, we identified a neuron-glia signaling pathway whereby Aβ acts as an upstream activator of astroglial nuclear factor kappa B (NF-κB), leading to the release of complement C3, which acts on the neuronal C3a receptor (C3aR) to influence dendritic morphology and cognitive function. Here we report that astrocytic complement activation also regulates Aβ dynamics in vitro and amyloid pathology in AD mouse models through microglial C3aR. We show that in primary microglial cultures, acute C3 or C3a activation promotes, whereas chronic C3/C3a treatment attenuates, microglial phagocytosis and that the effect of chronic C3 exposure can be blocked by cotreatment with a C3aR antagonist and by genetic deletion of C3aR. We further demonstrate that Aβ pathology and neuroinflammation in amyloid precursor protein (APP) transgenic mice are worsened by astroglial NF-κB hyperactivation and resulting C3 elevation, whereas treatment with the C3aR antagonist (C3aRA) ameliorates plaque load and microgliosis. Our studies define a complement-dependent intercellular cross talk in which neuronal overproduction of Aβ activates astroglial NF-κB to elicit extracellular release of C3. This promotes a pathogenic cycle by which C3 in turn interacts with neuronal and microglial C3aR to alter cognitive function and impair Aβ phagocytosis. This feedforward loop can be effectively blocked by C3aR inhibition, supporting the therapeutic potential of C3aR antagonists under chronic neuroinflammation conditions. SIGNIFICANCE STATEMENT The complement pathway is activated in Alzheimer's disease. Here we show that the central complement factor C3 secreted from astrocytes interacts with microglial C3a receptor (C3aR) to mediate β-amyloid pathology and neuroinflammation in AD mouse models. Our study provides support for targeting C3aR as a potential therapy for Alzheimer's disease.
Collapse
|
93
|
Söllvander S, Nikitidou E, Brolin R, Söderberg L, Sehlin D, Lannfelt L, Erlandsson A. Accumulation of amyloid-β by astrocytes result in enlarged endosomes and microvesicle-induced apoptosis of neurons. Mol Neurodegener 2016; 11:38. [PMID: 27176225 PMCID: PMC4865996 DOI: 10.1186/s13024-016-0098-z] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 04/15/2016] [Indexed: 01/22/2023] Open
Abstract
Background Despite the clear physical association between activated astrocytes and amyloid-β (Aβ) plaques, the importance of astrocytes and their therapeutic potential in Alzheimer’s disease remain elusive. Soluble Aβ aggregates, such as protofibrils, have been suggested to be responsible for the widespread neuronal cell death in Alzheimer’s disease, but the mechanisms behind this remain unclear. Moreover, ineffective degradation is of great interest when it comes to the development and progression of neurodegeneration. Based on our previous results that astrocytes are extremely slow in degrading phagocytosed material, we hypothesized that astrocytes may be an important player in these processes. Hence, the aim of this study was to clarify the role of astrocytes in clearance, spreading and neuronal toxicity of Aβ. Results To examine the role of astrocytes in Aβ pathology, we added Aβ protofibrils to a co-culture system of primary neurons and glia. Our data demonstrates that astrocytes rapidly engulf large amounts of Aβ protofibrils, but then store, rather than degrade the ingested material. The incomplete digestion results in a high intracellular load of toxic, partly N-terminally truncated Aβ and severe lysosomal dysfunction. Moreover, secretion of microvesicles containing N-terminally truncated Aβ, induce apoptosis of cortical neurons. Conclusions Taken together, our results suggest that astrocytes play a central role in the progression of Alzheimer’s disease, by accumulating and spreading toxic Aβ species. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0098-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofia Söllvander
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Elisabeth Nikitidou
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Robin Brolin
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Linda Söderberg
- BioArctic Neuroscience AB, Warfvinges väg 35, SE-112 51, Stockholm, Sweden
| | - Dag Sehlin
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Lars Lannfelt
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Anna Erlandsson
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
94
|
Daschil N, Humpel C. Green-Fluorescent Protein(+) Astrocytes Attach to Beta-Amyloid Plaques in an Alzheimer Mouse Model and Are Sensitive for Clasmatodendrosis. Front Aging Neurosci 2016; 8:75. [PMID: 27092076 PMCID: PMC4825400 DOI: 10.3389/fnagi.2016.00075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/28/2016] [Indexed: 01/14/2023] Open
Abstract
Alzheimer's disease (AD) is pathologically characterized by beta-amyloid (Aβ) plaques and Tau pathology. It is well-established that Aβ plaques are surrounded by reactive astrocytes, highly expressing glial fibrillary acidic protein (GFAP). In order to study the cellular interaction of reactive astrocytes with Aβ plaques, we crossbred mice overexpressing amyloid precursor protein (APP) with the Swedish-Dutch-Iowa mutations (APP-SweDI) with mice expressing green fluorescent protein (GFP) under the GFAP-promotor. Three-dimensional confocal microscopy revealed a tight association and intense sprouting of astrocytic finely branched processes towards Aβ plaques in 12 month old mice. In order to study phagocytosis, 110 μm thick brain slices from 12 month old crossbred mice were cultured overnight, however, we found that the GFP fluorescence faded, distal processes degenerated and a complete loss of astrocytic morphology was seen (clasmatodendrosis). In summary, our data show that GFP(+) reactive astrocytes make intense contact with Aβ plaques but these cells are highly vulnerable for degeneration.
Collapse
Affiliation(s)
- Nina Daschil
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Department of Psychiatry, Psychosomatics and Psychotherapy, Medical University of Innsbruck Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Department of Psychiatry, Psychosomatics and Psychotherapy, Medical University of Innsbruck Innsbruck, Austria
| |
Collapse
|
95
|
Wang X, Zhang XG, Zhou TT, Li N, Jang CY, Xiao ZC, Ma QH, Li S. Elevated Neuronal Excitability Due to Modulation of the Voltage-Gated Sodium Channel Nav1.6 by Aβ1-42. Front Neurosci 2016; 10:94. [PMID: 27013956 PMCID: PMC4783403 DOI: 10.3389/fnins.2016.00094] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022] Open
Abstract
Aberrant increases in neuronal network excitability may contribute to the cognitive deficits in Alzheimer's disease (AD). However, the mechanisms underlying hyperexcitability are not fully understood. Such overexcitation of neuronal networks has been detected in the brains of APP/PS1 mice. In the present study, using current-clamp recording techniques, we observed that 12 days in vitro (DIV) primary cultured pyramidal neurons from P0 APP/PS1 mice exhibited a more prominent action potential burst and a lower threshold than WT littermates. Moreover, after treatment with Aβ1−42 peptide, 12 DIV primary cultured neurons showed similar changes, to a greater degree than in controls. Voltage-clamp recordings revealed that the voltage-dependent sodium current density of neurons incubated with Aβ1−42 was significantly increased, without change in the voltage-dependent sodium channel kinetic characteristics. Immunohistochemistry and western blot results showed that, after treatment with Aβ1−42, expressions of Nav and Nav1.6 subtype increased in cultured neurons or APP/PS1 brains compared to control groups. The intrinsic neuronal hyperexcitability of APP/PS1 mice might thus be due to an increased expression of voltage-dependent sodium channels induced by Aβ1−42. These results may illuminate the mechanism of aberrant neuronal networks in AD.
Collapse
Affiliation(s)
- Xi Wang
- Department of Physiology, Dalian Medical University Dalian, China
| | - Xiao-Gang Zhang
- Department of Physiology, Dalian Medical University Dalian, China
| | - Ting-Ting Zhou
- Department of Neurology, the First Affiliated Hospital of Dalian Medical University Dalian, China
| | - Na Li
- Department of Physiology, Dalian Medical University Dalian, China
| | - Chun-Yan Jang
- Department of Physiology, Dalian Medical University Dalian, China
| | - Zhi-Cheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Kunming Medical College, Institute of Molecular and Clinical Medicine Kunming, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University Suzhou, China
| | - Shao Li
- Department of Physiology, Dalian Medical University Dalian, China
| |
Collapse
|
96
|
Rustenhoven J, Aalderink M, Scotter EL, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RLM, Curtis MA, Park TIH, Dragunow M. TGF-beta1 regulates human brain pericyte inflammatory processes involved in neurovasculature function. J Neuroinflammation 2016; 13:37. [PMID: 26867675 PMCID: PMC4751726 DOI: 10.1186/s12974-016-0503-0] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/03/2016] [Indexed: 12/17/2022] Open
Abstract
Background Transforming growth factor beta 1 (TGFβ1) is strongly induced following brain injury and polarises microglia to an anti-inflammatory phenotype. Augmentation of TGFβ1 responses may therefore be beneficial in preventing inflammation in neurological disorders including stroke and neurodegenerative diseases. However, several other cell types display immunogenic potential and identifying the effect of TGFβ1 on these cells is required to more fully understand its effects on brain inflammation. Pericytes are multifunctional cells which ensheath the brain vasculature and have garnered recent attention with respect to their immunomodulatory potential. Here, we sought to investigate the inflammatory phenotype adopted by TGFβ1-stimulated human brain pericytes. Methods Microarray analysis was performed to examine transcriptome-wide changes in TGFβ1-stimulated pericytes, and results were validated by qRT-PCR and cytometric bead arrays. Flow cytometry, immunocytochemistry and LDH/Alamar Blue® viability assays were utilised to examine phagocytic capacity of human brain pericytes, transcription factor modulation and pericyte health. Results TGFβ1 treatment of primary human brain pericytes induced the expression of several inflammatory-related genes (NOX4, COX2, IL6 and MMP2) and attenuated others (IL8, CX3CL1, MCP1 and VCAM1). A synergistic induction of IL-6 was seen with IL-1β/TGFβ1 treatment whilst TGFβ1 attenuated the IL-1β-induced expression of CX3CL1, MCP-1 and sVCAM-1. TGFβ1 was found to signal through SMAD2/3 transcription factors but did not modify nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) translocation. Furthermore, TGFβ1 attenuated the phagocytic ability of pericytes, possibly through downregulation of the scavenger receptors CD36, CD47 and CD68. Whilst TGFβ did decrease pericyte number, this was due to a reduction in proliferation, not apoptotic death or compromised cell viability. Conclusions TGFβ1 attenuated pericyte expression of key chemokines and adhesion molecules involved in CNS leukocyte trafficking and the modulation of microglial function, as well as reduced the phagocytic ability of pericytes. However, TGFβ1 also enhanced the expression of classical pro-inflammatory cytokines and enzymes which can disrupt BBB functioning, suggesting that pericytes adopt a phenotype which is neither solely pro- nor anti-inflammatory. Whilst the effects of pericyte modulation by TGFβ1 in vivo are difficult to infer, the reduction in pericyte proliferation together with the elevated IL-6, MMP-2 and NOX4 and reduced phagocytosis suggests a detrimental action of TGFβ1 on neurovasculature. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0503-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | - Miranda Aalderink
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | | | - Peter S Bergin
- Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - E Scott Graham
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | - Richard L M Faull
- Department of Anatomy, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | - Maurice A Curtis
- Department of Anatomy, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, 1023, New Zealand.,Department of Anatomy, The University of Auckland, Auckland, 1023, New Zealand.,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, 1023, New Zealand. .,Centre for Brain Research, The University of Auckland, Auckland, 1023, New Zealand.
| |
Collapse
|
97
|
Couturier J, Stancu IC, Schakman O, Pierrot N, Huaux F, Kienlen-Campard P, Dewachter I, Octave JN. Activation of phagocytic activity in astrocytes by reduced expression of the inflammasome component ASC and its implication in a mouse model of Alzheimer disease. J Neuroinflammation 2016; 13:20. [PMID: 26818951 PMCID: PMC4729126 DOI: 10.1186/s12974-016-0477-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/06/2016] [Indexed: 12/19/2022] Open
Abstract
Background The proinflammatory cytokine interleukin-1β (IL-1β) is overexpressed in Alzheimer disease (AD) as a key regulator of neuroinflammation. Amyloid-β (Aβ) peptide triggers activation of inflammasomes, protein complexes responsible for IL-1β maturation in microglial cells. Downregulation of NALP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome has been shown to decrease amyloid load and rescue cognitive deficits in a mouse model of AD. Whereas activation of inflammasome in microglial cells has been described in AD, no data are currently available concerning activation of inflammasome in astrocytes, although they are involved in inflammatory response and phagocytosis. Here, by targeting the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD domain), we investigated the influence of activation of the inflammasome on the phagocytic activity of astrocytes. Methods We used an ASC knockout mouse model, as ASC is a central protein in the inflammasome, acting as an adaptor and stabilizer of the complex and thus critical for its activation. Lipopolysaccharide (LPS)-primed primary cultures of astrocytes from newborn mice were utilized to evaluate Aβ-induced inflammasome activation by measuring IL-1β release by ECLIA (electro-chemiluminescence immunoassay). Phagocytosis efficiency was measured by incorporation of bioparticles, and the release of the chemokine CCL3 (C-C motif ligand 3) was measured by ECLIA. ASC mice were crossbred with 5xFAD (familial Alzheimer disease) mice and tested for spatial reference memory using the Morris water maze (MWM) at 7–8 months of age. Amyloid load and CCL3 were quantified by thioflavine S staining and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. Results Cultured astrocytes primed with LPS and treated with Aβ showed an ASC-dependent production of IL-1β resulting from inflammasome activation mediated by Aβ phagocytosis and cathepsin B enzymatic activity. ASC+/− astrocytes displayed a higher phagocytic activity as compared to ASC+/+ and ASC −/− cells, resulting from a higher release of the chemokine CCL3. A significant decrease in amyloid load was measured in the brain of 7–8-month-old 5xFAD mice carrying the ASC +/− genotype, correlated with an increase in CCL3 gene expression. In addition, the ASC +/− genotype rescued spatial reference memory deficits observed in 5xFAD mice. Conclusions Our results demonstrate that Aβ is able to activate astrocytic inflammasome. Downregulation of inflammasome activity increases phagocytosis in astrocytes due to the release of CCL3. This could explain why downregulation of inflammasome activity decreases amyloid load and rescues memory deficits in a mouse model of AD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0477-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julien Couturier
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Ilie-Cosmin Stancu
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Olivier Schakman
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Nathalie Pierrot
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - François Huaux
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Université catholique de Louvain, Brussels, Belgium.
| | - Pascal Kienlen-Campard
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Ilse Dewachter
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Jean-Noël Octave
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| |
Collapse
|
98
|
Bryson KJ, Lynch MA. Linking T cells to Alzheimer's disease: from neurodegeneration to neurorepair. Curr Opin Pharmacol 2015; 26:67-73. [PMID: 26516722 DOI: 10.1016/j.coph.2015.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 12/14/2022]
Abstract
The overly-simplistic view that inflammatory and anti-inflammatory influences in the brain were respectively detrimental and advantageous in Alzheimer's disease (AD) is being challenged by advances in methodologies, and a debate relating to immune surveillance mechanisms in the brain. In contrast with previous findings, increasing interleukin (IL)-4 and IL-10 in brain by a recently-developed adenoviral delivery method, had detrimental effects in an animal model of AD, and the ability to isolate the choroid plexus has opened the debate on the role of this specialized tissue in immune surveillance. Delivery of polarized T cells to animal models of AD by different routes has yielded contrasting results; analysis of these diverse responses is vital to understand the role of T cells in the brain in AD, first reported over 25 years ago.
Collapse
Affiliation(s)
- Karen J Bryson
- Trinity College Institute for Neuroscience, Trinity College, Dublin 2, Ireland.
| | - Marina A Lynch
- Trinity College Institute for Neuroscience, Trinity College, Dublin 2, Ireland
| |
Collapse
|
99
|
Jones RS, Minogue AM, Fitzpatrick O, Lynch MA. Inhibition of JAK2 attenuates the increase in inflammatory markers in microglia from APP/PS1 mice. Neurobiol Aging 2015; 36:2716-24. [PMID: 26227742 DOI: 10.1016/j.neurobiolaging.2015.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/27/2015] [Accepted: 04/29/2015] [Indexed: 11/30/2022]
Abstract
There is a wealth of evidence indicating that macrophages adopt distinct phenotypes when exposed to specific stimuli and, in the past few years, accumulating data suggest that microglia behave somewhat similarly. Therefore, microglia can adopt the so-called M1 or M2 phenotypes in response to interferon-γ (IFNγ) and interleukin-4, respectively. Although it has yet to be unequivocally proven in the context of microglia, acutely activated M1 cells are probably protective, although a persistent M1 state is likely to be damaging, whereas M2 cells may be reparative and restorative. In this case, particularly because the current evidence suggests the development of a predominantly M1 state with age and in neurodegenerative diseases, it is important to identify mechanisms by which polarization of microglia can be modulated. The present findings indicate that exposure of cultured microglia to IFNγ increased expressions of the archetypal markers of the M1 phenotype, tumour necrosis factor-α, and inducible nitric oxide synthase, and preexposure of cells to amyloid-β (Aβ) sensitized microglia to subsequent stimulation with IFNγ. Importantly, this synergy was also evident in microglia prepared from the brains of transgenic mice that overexpress amyloid precursor protein (APP) and presenilin 1 (PS1, APP/PS1 mice) and are exposed to a combination of increasing concentrations of endogenous Aβ from 4 or 5 months of age and an age-related increase in IFNγ. Significantly, the JAK2 inhibitor, TG101209, attenuated the IFNγ-induced changes in cultured microglia and in isolated microglia prepared from APP/PS1 mice. These findings suggest that targeting JAK2 may be a potential strategy for reducing neuroinflammation in Alzheimer's disease.
Collapse
Affiliation(s)
- Raasay S Jones
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Aedín M Minogue
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Orla Fitzpatrick
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Marina A Lynch
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland.
| |
Collapse
|
100
|
Lööv C, Mitchell CH, Simonsson M, Erlandsson A. Slow degradation in phagocytic astrocytes can be enhanced by lysosomal acidification. Glia 2015; 63:1997-2009. [PMID: 26095880 DOI: 10.1002/glia.22873] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 05/28/2015] [Indexed: 11/07/2022]
Abstract
Inefficient lysosomal degradation is central in the development of various brain disorders, but the underlying mechanisms and the involvement of different cell types remains elusive. We have previously shown that astrocytes effectively engulf dead cells, but then store, rather than degrade the ingested material. In the present study we identify reasons for the slow digestion and ways to accelerate degradation in primary astrocytes. Our results show that actin-rings surround the phagosomes for long periods of time, which physically inhibit the phago-lysosome fusion. Furthermore, astrocytes express high levels of Rab27a, a protein known to reduce the acidity of lysosomes by Nox2 recruitment, in order to preserve antigens for presentation. We found that Nox2 colocalizes with the ingested material, indicating that it may influence antigen processing also in astrocytes, as they express MHC class II. By inducing long-time acidification of astrocytic lysosomes using acidic nanoparticles, we could increase the digestion of astrocyte-ingested, dead cells. The degradation was, however, normalized over time, indicating that inhibitory pathways are up-regulated in response to the enhanced acidification. GLIA 2015;63:1997-2009.
Collapse
Affiliation(s)
- Camilla Lööv
- Department of Neuroscience, Uppsala University, Uppsala University Hospital Ent 85, 2nd Fl., Uppsala, Sweden
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin Simonsson
- SciLifeLab and Department of Computer Science, Electrical & Space Engineering, Luleå, University of Technology, Luleå, Sweden
| | - Anna Erlandsson
- Department of Neuroscience, Uppsala University, Uppsala University Hospital Ent 85, 2nd Fl., Uppsala, Sweden
- Department of Public Health and Caring Sciences, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| |
Collapse
|