1
|
Chandra S, Vassar R. The role of the gut microbiome in the regulation of astrocytes in Alzheimer's disease. Neurotherapeutics 2024; 21:e00425. [PMID: 39054180 PMCID: PMC11585888 DOI: 10.1016/j.neurot.2024.e00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and is the most common cause of dementia. AD is characterized pathologically by proteinaceous aggregates composed of amyloid beta (Aβ) and tau as well as progressive neurodegeneration. Concurrently with the buildup of protein aggregates, a strong neuroinflammatory response, in the form of reactive astrocytosis and microgliosis, occurs in the AD brain. It has recently been shown that the gut microbiome (GMB), composed of trillions of bacteria in the human intestine, can regulate both reactive astrocytosis and microgliosis in the context of both amyloidosis and tauopathy. Many studies have implicated microglia in these processes. However, growing evidence suggests that interactions between the GMB and astrocytes have a much larger role than previously thought. In this review, we summarize evidence regarding the gut microbiome in the control of reactive astrocytosis in AD.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA; Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Robert Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
2
|
Chung WS, Baldwin KT, Allen NJ. Astrocyte Regulation of Synapse Formation, Maturation, and Elimination. Cold Spring Harb Perspect Biol 2024; 16:a041352. [PMID: 38346858 PMCID: PMC11293538 DOI: 10.1101/cshperspect.a041352] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Astrocytes play an integral role in the development, maturation, and refinement of neuronal circuits. Astrocytes secrete proteins and lipids that instruct the formation of new synapses and induce the maturation of existing synapses. Through contact-mediated signaling, astrocytes can regulate the formation and state of synapses within their domain. Through phagocytosis, astrocytes participate in the elimination of excess synaptic connections. In this work, we will review key findings on the molecular mechanisms of astrocyte-synapse interaction with a focus on astrocyte-secreted factors, contact-mediated mechanisms, and synapse elimination. We will discuss this in the context of typical brain development and maintenance, as well as consider the consequences of dysfunction in these pathways in neurological disorders, highlighting a role for astrocytes in health and disease.
Collapse
Affiliation(s)
- Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Yuseong-gu, Daejeon 34141, Korea
| | - Katherine T Baldwin
- Department of Cell Biology and Physiology and UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| |
Collapse
|
3
|
Xunzhong Q, Miao G, Guangtao S, Huiying Z, Chenglong H, Xiaogang Z, Shunjie B, Xinyan H, Chengji W, Zuoyi H, Xiaofeng Z. Inhibition of the Rho/ROCK pathway promotes the expression of developmental and migration-related genes in astrocytes exposed to alcohol. Alcohol 2024; 115:5-12. [PMID: 37481044 DOI: 10.1016/j.alcohol.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/28/2023] [Accepted: 06/08/2023] [Indexed: 07/24/2023]
Abstract
Astrocytes are an important regulator of alcohol dependence. Furthermore, the downregulation of Rho-associated coiled coil-containing protein kinase 2 (ROCK2) attenuates alcohol-induced inflammation and oxidative stress in astrocytes. On the basis of these findings, we examined the effects of alcohol and a Rho/RACK kinases inhibitor on astrocyte function and investigated their effects on mRNA expression to further explore the protective mechanisms of a Rho/RACK kinases inhibitor in astrocytes after alcohol exposure. CTX TNA2 astrocytes were cultured with alcohol and Rho/RACK kinases inhibitor intervention before undergoing transcriptome sequencing, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and wound healing assays. Alcohol exposure modulated cell morphology and inhibited astrocyte migration, whereas Fasudil improved cell morphology and promoted astrocyte migration after alcohol exposure. Transcriptome sequencing results indicated that alcohol exposure modulates the expression of genes involved in astrocyte development. Fasudil reversed the effects of alcohol exposure on the astrocyte developmental process. Four genes related to the developmental process and migration - Ccl2, Postn, Itga8, and Serpine1 - with the highest protein-protein interaction correlations (node degree >7) were selected for verification by qRT-PCR, and the results were consistent with those of the sequencing and wound healing assays. Our results suggest that the Rho/ROCK pathway is essential for alcohol to be able to interfere with astrocyte development and migration gene expression. The Rho/ROCK pathway inhibitor Fasudil reversed the adverse effects of alcohol exposure on astrocytes and may have clinical applications.
Collapse
Affiliation(s)
- Qi Xunzhong
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Guo Miao
- Jiamusi University, Jiamusi, People's Republic of China
| | - Sun Guangtao
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Zhao Huiying
- Department of Neurology, Yichun Forestry Administration Central Hospital, Yichun, People's Republic of China
| | - Huang Chenglong
- Department of Clinical Laboratory, University-Town Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhong Xiaogang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China; College of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Bai Shunjie
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Huang Xinyan
- The Second Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Wu Chengji
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China
| | - Huang Zuoyi
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi, People's Republic of China.
| | - Zhu Xiaofeng
- Mudanjiang Medical College, Mudanjiang, People's Republic of China.
| |
Collapse
|
4
|
Liu B, Yang H, Song YS, Sorenson CM, Sheibani N. Thrombospondin-1 in vascular development, vascular function, and vascular disease. Semin Cell Dev Biol 2024; 155:32-44. [PMID: 37507331 PMCID: PMC10811293 DOI: 10.1016/j.semcdb.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
Angiogenesis is vital to developmental, regenerative and repair processes. It is normally regulated by a balanced production of pro- and anti-angiogenic factors. Alterations in this balance under pathological conditions are generally mediated through up-regulation of pro-angiogenic and/or downregulation of anti-angiogenic factors, leading to growth of new and abnormal blood vessels. The pathological manifestation of many diseases including cancer, ocular and vascular diseases are dependent on the growth of these new and abnormal blood vessels. Thrompospondin-1 (TSP1) was the first endogenous angiogenesis inhibitor identified and its anti-angiogenic and anti-inflammatory activities have been the subject of many studies. Studies examining the role TSP1 plays in pathogenesis of various ocular diseases and vascular dysfunctions are limited. Here we will discuss the recent studies focused on delineating the role TSP1 plays in ocular vascular development and homeostasis, and pathophysiology of various ocular and vascular diseases with a significant clinical relevance to human health.
Collapse
Affiliation(s)
- Bo Liu
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| | - Huan Yang
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Nader Sheibani
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
5
|
Woo AM, Sontheimer H. Interactions between astrocytes and extracellular matrix structures contribute to neuroinflammation-associated epilepsy pathology. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1198021. [PMID: 39086689 PMCID: PMC11285605 DOI: 10.3389/fmmed.2023.1198021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/31/2023] [Indexed: 08/02/2024]
Abstract
Often considered the "housekeeping" cells of the brain, astrocytes have of late been rising to the forefront of neurodegenerative disorder research. Identified as crucial components of a healthy brain, it is undeniable that when astrocytes are dysfunctional, the entire brain is thrown into disarray. We offer epilepsy as a well-studied neurological disorder in which there is clear evidence of astrocyte contribution to diseases as evidenced across several different disease models, including mouse models of hippocampal sclerosis, trauma associated epilepsy, glioma-associated epilepsy, and beta-1 integrin knockout astrogliosis. In this review we suggest that astrocyte-driven neuroinflammation, which plays a large role in the pathology of epilepsy, is at least partially modulated by interactions with perineuronal nets (PNNs), highly structured formations of the extracellular matrix (ECM). These matrix structures affect synaptic placement, but also intrinsic neuronal properties such as membrane capacitance, as well as ion buffering in their immediate milieu all of which alters neuronal excitability. We propose that the interactions between PNNs and astrocytes contribute to the disease progression of epilepsy vis a vis neuroinflammation. Further investigation and alteration of these interactions to reduce the resultant neuroinflammation may serve as a potential therapeutic target that provides an alternative to the standard anti-seizure medications from which patients are so frequently unable to benefit.
Collapse
Affiliation(s)
- AnnaLin M. Woo
- Neuroscience Graduate Program, Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| | - Harald Sontheimer
- Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
6
|
Krishna S, Choudhury A, Keough MB, Seo K, Ni L, Kakaizada S, Lee A, Aabedi A, Popova G, Lipkin B, Cao C, Nava Gonzales C, Sudharshan R, Egladyous A, Almeida N, Zhang Y, Molinaro AM, Venkatesh HS, Daniel AGS, Shamardani K, Hyer J, Chang EF, Findlay A, Phillips JJ, Nagarajan S, Raleigh DR, Brang D, Monje M, Hervey-Jumper SL. Glioblastoma remodelling of human neural circuits decreases survival. Nature 2023; 617:599-607. [PMID: 37138086 PMCID: PMC10191851 DOI: 10.1038/s41586-023-06036-1] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/31/2023] [Indexed: 05/05/2023]
Abstract
Gliomas synaptically integrate into neural circuits1,2. Previous research has demonstrated bidirectional interactions between neurons and glioma cells, with neuronal activity driving glioma growth1-4 and gliomas increasing neuronal excitability2,5-8. Here we sought to determine how glioma-induced neuronal changes influence neural circuits underlying cognition and whether these interactions influence patient survival. Using intracranial brain recordings during lexical retrieval language tasks in awake humans together with site-specific tumour tissue biopsies and cell biology experiments, we find that gliomas remodel functional neural circuitry such that task-relevant neural responses activate tumour-infiltrated cortex well beyond the cortical regions that are normally recruited in the healthy brain. Site-directed biopsies from regions within the tumour that exhibit high functional connectivity between the tumour and the rest of the brain are enriched for a glioblastoma subpopulation that exhibits a distinct synaptogenic and neuronotrophic phenotype. Tumour cells from functionally connected regions secrete the synaptogenic factor thrombospondin-1, which contributes to the differential neuron-glioma interactions observed in functionally connected tumour regions compared with tumour regions with less functional connectivity. Pharmacological inhibition of thrombospondin-1 using the FDA-approved drug gabapentin decreases glioblastoma proliferation. The degree of functional connectivity between glioblastoma and the normal brain negatively affects both patient survival and performance in language tasks. These data demonstrate that high-grade gliomas functionally remodel neural circuits in the human brain, which both promotes tumour progression and impairs cognition.
Collapse
Affiliation(s)
- Saritha Krishna
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Abrar Choudhury
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | | | - Kyounghee Seo
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Lijun Ni
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Sofia Kakaizada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Anthony Lee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Alexander Aabedi
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Galina Popova
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin Lipkin
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Caroline Cao
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Cesar Nava Gonzales
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Rasika Sudharshan
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew Egladyous
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nyle Almeida
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Yalan Zhang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | | | - Andy G S Daniel
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | | | - Jeanette Hyer
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Anne Findlay
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Srikantan Nagarajan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, USA
| | - David Brang
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Michelle Monje
- Department of Neurology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Shawn L Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Schwarz K, Schmitz F. Synapse Dysfunctions in Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24021639. [PMID: 36675155 PMCID: PMC9862173 DOI: 10.3390/ijms24021639] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the central nervous system (CNS) affecting nearly three million humans worldwide. In MS, cells of an auto-reactive immune system invade the brain and cause neuroinflammation. Neuroinflammation triggers a complex, multi-faceted harmful process not only in the white matter but also in the grey matter of the brain. In the grey matter, neuroinflammation causes synapse dysfunctions. Synapse dysfunctions in MS occur early and independent from white matter demyelination and are likely correlates of cognitive and mental symptoms in MS. Disturbed synapse/glia interactions and elevated neuroinflammatory signals play a central role. Glutamatergic excitotoxic synapse damage emerges as a major mechanism. We review synapse/glia communication under normal conditions and summarize how this communication becomes malfunctional during neuroinflammation in MS. We discuss mechanisms of how disturbed glia/synapse communication can lead to synapse dysfunctions, signaling dysbalance, and neurodegeneration in MS.
Collapse
|
8
|
Xia Y, Chen J, Ma T, Meng X, Han X, Li D. Maternal DBP exposure promotes synaptic formation in offspring by activating astrocytes via the AKT/NF-κB/IL-6/JAK2/STAT3 signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 829:154437. [PMID: 35278568 DOI: 10.1016/j.scitotenv.2022.154437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
It has been demonstrated that activated astrocytes in the hypothalamus could disrupt GnRH secretion in offspring after maternal di-n-butyl phthalate (DBP) exposure, indicating that the effect of DBP on astrocyte activation and crosstalk between astrocytes and neurons is still worthy of further investigation. In this study, pregnant mice were intragastrically administered DBP dissolved in corn oil from gestational days (GD) 12.5-21.5. Maternal DBP exposure resulted in hippocampal astrocyte activation, abnormal synaptic formation, and reduced autonomic and exploratory behavior in offspring on postnatal day (PND) 22. Further studies identified that mono-n-butyl phthalate (MBP) induced astrocyte activation and proliferation by activating the AKT/NF-κB/IL-6/JAK2/STAT3 signaling pathway. Moreover, upregulated thrombospondin 1 (TSP1) in activated astrocytes regulated synaptic-related protein expression. This study highlights the neurotoxicity of maternal DBP exposure to offspring, which provides new insights into identifying potential molecular targets for the treatment of diseases related to neurological development disorders in children.
Collapse
Affiliation(s)
- Yunhui Xia
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Junhan Chen
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou 225001, China
| | - Xiannan Meng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
9
|
Ren B, Dunaevsky A. Modeling Neurodevelopmental and Neuropsychiatric Diseases with Astrocytes Derived from Human-Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:1692. [PMID: 33567562 PMCID: PMC7915337 DOI: 10.3390/ijms22041692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Accumulating studies demonstrate the morphological and functional diversity of astrocytes, a subtype of glial cells in the central nervous system. Animal models are instrumental in advancing our understanding of the role of astrocytes in brain development and their contribution to neurological disease; however, substantial interspecies differences exist between rodent and human astrocytes, underscoring the importance of studying human astrocytes. Human pluripotent stem cell differentiation approaches allow the study of patient-specific astrocytes in the etiology of neurological disorders. In this review, we summarize the structural and functional properties of astrocytes, including the unique features of human astrocytes; demonstrate the necessity of the stem cell platform; and discuss how this platform has been applied to the research of neurodevelopmental and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Baiyan Ren
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Neurological Science, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Anna Dunaevsky
- Department of Neurological Science, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
10
|
Augusto-Oliveira M, Arrifano GP, Takeda PY, Lopes-Araújo A, Santos-Sacramento L, Anthony DC, Verkhratsky A, Crespo-Lopez ME. Astroglia-specific contributions to the regulation of synapses, cognition and behaviour. Neurosci Biobehav Rev 2020; 118:331-357. [DOI: 10.1016/j.neubiorev.2020.07.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
|
11
|
Yu X, Nagai J, Marti-Solano M, Soto JS, Coppola G, Babu MM, Khakh BS. Context-Specific Striatal Astrocyte Molecular Responses Are Phenotypically Exploitable. Neuron 2020; 108:1146-1162.e10. [PMID: 33086039 DOI: 10.1016/j.neuron.2020.09.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/13/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Astrocytes tile the central nervous system and are widely implicated in brain diseases, but the molecular mechanisms by which astrocytes contribute to brain disorders remain incompletely explored. By performing astrocyte gene expression analyses following 14 experimental perturbations of relevance to the striatum, we discovered that striatal astrocytes mount context-specific molecular responses at the level of genes, pathways, and upstream regulators. Through data mining, we also identified astrocyte pathways in Huntington's disease (HD) that were reciprocally altered with respect to the activation of striatal astrocyte G protein-coupled receptor (GPCR) signaling. Furthermore, selective striatal astrocyte stimulation of the Gi-GPCR pathway in vivo corrected several HD-associated astrocytic, synaptic, and behavioral phenotypes, with accompanying improvement of HD-associated astrocyte signaling pathways, including those related to synaptogenesis and neuroimmune functions. Overall, our data show that astrocytes are malleable, using context-specific responses that can be dissected molecularly and used for phenotypic benefit in brain disorders.
Collapse
Affiliation(s)
- Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Jun Nagai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Maria Marti-Solano
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Joselyn S Soto
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Giovanni Coppola
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
12
|
Giacobbe J, Pariante CM, Borsini A. The innate immune system and neurogenesis as modulating mechanisms of electroconvulsive therapy in pre-clinical studies. J Psychopharmacol 2020; 34:1086-1097. [PMID: 32648795 PMCID: PMC7672674 DOI: 10.1177/0269881120936538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is a powerful and fast-acting anti-depressant strategy, often used in treatment-resistant patients. In turn, patients with treatment-resistant depression often present an increased inflammatory response. The impact of ECT on several pathophysiological mechanisms of depression has been investigated, with a focus which has largely been on cellular and synaptic plasticity. Although changes in the immune system are known to influence neurogenesis, these processes have principally been explored independently from each other in the context of ECT. OBJECTIVE The aim of this review was to compare the time-dependent consequences of acute and chronic ECT on concomitant innate immune system and neurogenesis-related outcomes measured in the central nervous system in pre-clinical studies. RESULTS During the few hours following acute electroconvulsive shock (ECS), the expression of the astrocytic reactivity marker glial fibrillary acidic protein (GFAP) and inflammatory genes, such as cyclooxygenase-2 (COX2), were significantly increased together with the neurogenic brain-derived neurotrophic factor (BDNF) and cell proliferation. Similarly, chronic ECS caused an initial upregulation of the same astrocytic marker, immune genes, and neurogenic factors. Interestingly, over time, inflammation appeared to be dampened, while glial activation and neurogenesis were maintained, after either acute or chronic ECS. CONCLUSION Regardless of treatment duration ECS would seemingly trigger a rapid increase in inflammatory molecules, dampened over time, as well as a long-lasting activation of astrocytes and production of growth and neurotrophic factors, leading to cell proliferation. This suggests that both innate immune system response and neurogenesis might contribute to the efficacy of ECT.
Collapse
Affiliation(s)
| | | | - Alessandra Borsini
- Alessandra Borsini, King’s College London, Institute of Psychiatry, Psychology & Neuroscience, Division of Psychological Medicine, Stress, Psychiatry and Immunology Lab & Perinatal Psychiatry, The Maurice Wohl Clinical Neuroscience Institute, Cutcombe Road, London SE5 9RT, UK.
| |
Collapse
|
13
|
Roman C, Egert L, Di Benedetto B. Astrocytic-neuronal crosstalk gets jammed: Alternative perspectives on the onset of neuropsychiatric disorders. Eur J Neurosci 2020; 54:5717-5729. [PMID: 32644273 DOI: 10.1111/ejn.14900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/09/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
Abstract
Investigating interactions of glia cells and synapses during development and in adulthood is the focus of several research programmes which aim at understanding the neurobiology of brain physiological and pathological processes. Both glia-specific released and membrane-bound proteins play essential roles in the development, maintenance and functionality of synaptic connections. Alterations in synaptic contacts in specific brain areas are hallmarks of several brain diseases, such as major depressive disorder, autism spectrum disorder and schizophrenia. Thus, a deeper knowledge about putative astrocyte dysfunctions which might affect the synaptic compartment is warranted to improve treatment options. Here, we present the latest advances about the role of glia cells in orchestrating the arrangement of synapses and neuronal networks in physiological and pathological states. We specifically focus on the role of astrocytes in the phagocytosis of neuronal synapses as a novel mechanism which drives the refinement of neuronal circuits and might be affected in pathological conditions. Finally, we propose this astrocyte-dependent mechanism as a putative alternative target of pharmacological interventions for the treatment of brain disorders.
Collapse
Affiliation(s)
- Celia Roman
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Luisa Egert
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Barbara Di Benedetto
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| |
Collapse
|
14
|
Oyabu K, Takeda K, Kawano H, Kubota K, Watanabe T, Harata NC, Katsurabayashi S, Iwasaki K. Presynaptically silent synapses are modulated by the density of surrounding astrocytes. J Pharmacol Sci 2020; 144:76-82. [PMID: 32736867 DOI: 10.1016/j.jphs.2020.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/02/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023] Open
Abstract
Astrocytes, comprising the primary glial-cell type, are involved in the formation and maturation of synapses, and thus contribute to sustainable synaptic transmission between neurons. Given that the animals in higher phylogenetic tree have brains with a higher density of glial cells with respect to neurons, there is a possibility that the relative astrocytic density directly influences synaptic transmission. However, the notion has not been tested thoroughly. Here we addressed it, by using a primary culture preparation where single hippocampal neurons are surrounded by a variable but a countable number of cortical astrocytes in dot-patterned microislands, and recording synaptic transmission by patch-clamp electrophysiology. Neurons with a higher astrocytic density showed a higher amplitude of the evoked excitatory postsynaptic current than that of neurons with a lower astrocytic density. The size of the readily releasable pool of synaptic vesicles per neuron was significantly larger. The frequency of spontaneous synaptic transmission was higher, but the amplitude was unchanged. The number of morphologically identified glutamatergic synapses was comparable, but the percentage of functional ones was increased, indicating a lower ratio of presynaptically silent synapses. Taken together, the higher astrocytic density enhanced excitatory synaptic transmission by increasing the fraction of functional synapses through presynaptic un-silencing.
Collapse
Affiliation(s)
- Kohei Oyabu
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kotomi Takeda
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Hiroyuki Kawano
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan; A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan; A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - N Charles Harata
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan; A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
15
|
Chang X, Li J, Niu S, Xue Y, Tang M. Neurotoxicity of metal‐containing nanoparticles and implications in glial cells. J Appl Toxicol 2020; 41:65-81. [DOI: 10.1002/jat.4037] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Xiaoru Chang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Jiangyan Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| |
Collapse
|
16
|
Yamagata K. Astrocytic nutritional dysfunction associated with hypoxia-induced neuronal vulnerability in stroke-prone spontaneously hypertensive rats. Neurochem Int 2020; 138:104786. [PMID: 32579896 DOI: 10.1016/j.neuint.2020.104786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/26/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Stroke-prone spontaneously hypertensive rats (SHRSP) is a valuable animal model to investigate human strokes. SHRSP Izumo strain (Izm) neurons are highly sensitive to blood supply changes. Furthermore, SHRSP/Izm astrocytes show various abnormalities upon hypoxic stimulation compared to control Wistar Kyoto (WKY/Izm) rats. This study aimed to describe stroke-related characteristics of SHRSP/Izm-derived neurons and astrocytes. In addition, we discuss the role of astrocytes in the development of stroke in SHRSP/Izm model. In SHRSP/Izm, neuronal death is induced upon reoxygenation after hypoxia. Furthermore, it was shown that SHRSP/Izm astrocytes show significantly reduced lactate production and supply ability to nerve cells when subjected to hypoxic stimulation. In particular, decreased lactate production and monocarboxylic acid transporter (MCT) expression in SHRSP/Izm astrocytes are factors that induce neuronal cell death. Remarkable differences in glial cell line-derived neurotrophic factor (GDNF) expression and L-serine production were also observed in SHRSP/Izm-derived astrocytes compared to WKY/Izm. Reduced production of both GDNF and L-serine contributes to diminished neuronal survival. The differences between SHRSP/Izm and WKY/Izm astrocyte cellular properties may contribute to compromised neuronal nutrition and induction of neuronal death. These properties are likely to be the factors that enhance stroke in SHRSP/Izm.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Bioscience & Biotechnology, College of Bioresource Science, Nihon University (UNBS), Japan.
| |
Collapse
|
17
|
Li Z, Lu Y, Liang S, Li S, Chen B, Zhang M, Xia M, Guan D, Verkhratsky A, Li B. Fluoxetine improves behavioural deficits induced by chronic alcohol treatment by alleviating RNA editing of 5-HT 2C receptors. Neurochem Int 2020; 134:104689. [PMID: 31968217 DOI: 10.1016/j.neuint.2020.104689] [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: 09/24/2019] [Revised: 01/07/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
The alcoholism and major depressive disorder are common comorbidity, with alcohol-induced depressive symptoms being eased by selective serotonin re-uptake inhibitors (SSRIs), although the mechanisms underlying pathology and therapy are poorly understood. Chronic alcohol consumption affects the activity of serotonin 2C receptors (5-HT2CR) by regulating adenosine deaminases acting on RNA (ADARs) in neurons. Astrogliopathic changes contribute to alcohol addiction, while decreased release of ATP from astrocytes can trigger depressive-like behaviours in mice. In this study, we discovered that chronic alcohol treatment increased editing of RNA of 5-HT2CR via up-regulating the expression of ADAR2, consequently reducing the release of ATP from astrocytes induced by 5-HT2CR agonist, MK212. Moreover, SSRI antidepressant fluoxetine decreased the expression of ADAR2 through the transactivation of EGFR/PI3K/AKT/cFos signalling pathway. The increased release of astroglial ATP by MK212 which was suppressed by chronic alcohol consumption, and reduction in ADAR2 activity eliminated the RNA editing of 5-HT2CR increased by alcohol in vitro and recovered the release of ATP from astrocytes induced by MK212. Meanwhile, fluoxetine improved the behavioural and motor symptoms induced by alcohol addiction and decreased the alcohol intake. Our study suggests that the astrocytic 5-HT2CR contribute to alcohol addiction; fluoxetine thus can be used to alleviate depression, treat alcohol addiction and improve motor coordination.
Collapse
Affiliation(s)
- Zexiong Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, The Affiliated Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Shanshan Liang
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Shuai Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Beina Chen
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Manman Zhang
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, PR China
| | - Dawei Guan
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, PR China.
| | - Alexei Verkhratsky
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Baoman Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, PR China.
| |
Collapse
|
18
|
Nagai J, Rajbhandari AK, Gangwani MR, Hachisuka A, Coppola G, Masmanidis SC, Fanselow MS, Khakh BS. Hyperactivity with Disrupted Attention by Activation of an Astrocyte Synaptogenic Cue. Cell 2019; 177:1280-1292.e20. [PMID: 31031006 PMCID: PMC6526045 DOI: 10.1016/j.cell.2019.03.019] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/08/2019] [Accepted: 03/07/2019] [Indexed: 11/19/2022]
Abstract
Hyperactivity and disturbances of attention are common behavioral disorders whose underlying cellular and neural circuit causes are not understood. We report the discovery that striatal astrocytes drive such phenotypes through a hitherto unknown synaptic mechanism. We found that striatal medium spiny neurons (MSNs) triggered astrocyte signaling via γ-aminobutyric acid B (GABAB) receptors. Selective chemogenetic activation of this pathway in striatal astrocytes in vivo resulted in acute behavioral hyperactivity and disrupted attention. Such responses also resulted in upregulation of the synaptogenic cue thrombospondin-1 (TSP1) in astrocytes, increased excitatory synapses, enhanced corticostriatal synaptic transmission, and increased MSN action potential firing in vivo. All of these changes were reversed by blocking TSP1 effects. Our data identify a form of bidirectional neuron-astrocyte communication and demonstrate that acute reactivation of a single latent astrocyte synaptogenic cue alters striatal circuits controlling behavior, revealing astrocytes and the TSP1 pathway as therapeutic targets in hyperactivity, attention deficit, and related psychiatric disorders.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Abha K Rajbhandari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Mohitkumar R Gangwani
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Ayaka Hachisuka
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Giovanni Coppola
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Sotiris C Masmanidis
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Michael S Fanselow
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Baljit S Khakh
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
19
|
da Silva APB, Souza DG, Souza DO, Machado DC, Sato DK. Role of Glutamatergic Excitotoxicity in Neuromyelitis Optica Spectrum Disorders. Front Cell Neurosci 2019; 13:142. [PMID: 31031597 PMCID: PMC6473164 DOI: 10.3389/fncel.2019.00142] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/21/2019] [Indexed: 01/12/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory disorder mediated by immune-humoral responses directed against central nervous system (CNS) antigens. Most patients are positive for specific immunoglobulin G (IgG) auto-antibodies for aquaporin-4 (AQP4), a water channel present in astrocytes. Antigen-antibody binding promotes complement system cascade activation, immune system cell infiltration, IgG deposition, loss of AQP4 and excitatory amino acid transporter 2 (EAAT2) expression on the astrocytic plasma membrane, triggering necrotic destruction of spinal cord tissue and optic nerves. Astrocytes are very important cells in the CNS and, in addition to supporting other nerve cells, they also regulate cerebral homeostasis and control glutamatergic synapses by modulating neurotransmission in the cleft through the high-affinity glutamate transporters present in their cell membrane. Specific IgG binding to AQP4 in astrocytes blocks protein functions and reduces EAAT2 activity. Once compromised, EAAT2 cannot take up free glutamate from the extracellular space, triggering excitotoxicity in the cells, which is characterized by overactivation of glutamate receptors in postsynaptic neurons. Therefore, the longitudinally extensive myelitis and optic neuritis lesions observed in patients with NMOSD may be the result of primary astrocytic damage triggered by IgG binding to AQP4, which can activate the immune-system cascade and, in addition, downregulate EAAT2. All these processes may explain the destructive lesions in NMOSD secondary to neuroinflammation and glutamatergic excitotoxicity. New or repurposed existing drugs capable of controlling glutamatergic excitotoxicity may provide new therapeutic options to reduce tissue damage and permanent disability after NMOSD attacks.
Collapse
Affiliation(s)
- Ana Paula Bornes da Silva
- Molecular and Cellular Biology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Medical School, Institute of Geriatrics and Gerontology, Graduate Program in Biomedical Gerontology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Débora Guerini Souza
- Graduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Diogo Onofre Souza
- Graduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Denise Cantarelli Machado
- Molecular and Cellular Biology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Medical School, Institute of Geriatrics and Gerontology, Graduate Program in Biomedical Gerontology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Douglas Kazutoshi Sato
- Molecular and Cellular Biology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
20
|
Gong C, Xiang G, Liu K, Zhang H. [Research progress on the role of thrombospondin in synapse formation]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:124-128. [PMID: 30644272 DOI: 10.7507/1002-1892.201809006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To review the recent progress in the role of thrombospondins (TSPs) in synapse formation in the central nervous system (CNS). Methods A wide range of domestic and foreign literature on the role of TSPs in the synapse formation of the CNS was reviewed. The role of TSPs in structural features, molecules, and related diseases was reviewed. Results As an oligosaccharide protein, TSPs play important roles in angiogenesis, inflammation, osteogenesis, cell proliferation, and apoptosis. In the nervous system, they bind to voltage-dependent calcium channels, neuronectin, and other extracellular matrix proteins and cell surface receptors, and participate in and regulate multiple processes such as synapse formation, maturation, and function in the CNS. Conclusion TSPs as an oligomeric extracellular matrix protein play an important role in the formation of synapses and the repair of synapses after CNS injury.
Collapse
Affiliation(s)
- Chaoyang Gong
- Department of Orthopedics, Gansu Key Laboratory of Osteoarthropathy Research, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Gao Xiang
- Department of Orthopedics, Gansu Key Laboratory of Osteoarthropathy Research, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Kaixin Liu
- Department of Orthopedics, Gansu Key Laboratory of Osteoarthropathy Research, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Haihong Zhang
- Department of Orthopedics, Gansu Key Laboratory of Osteoarthropathy Research, Lanzhou University Second Hospital, Lanzhou Gansu, 730000,
| |
Collapse
|
21
|
Bosch ME, Kielian T. Astrocytes in juvenile neuronal ceroid lipofuscinosis (CLN3) display metabolic and calcium signaling abnormalities. J Neurochem 2018; 148:612-624. [PMID: 29964296 DOI: 10.1111/jnc.14545] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/21/2018] [Accepted: 06/22/2018] [Indexed: 12/11/2022]
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL) is a lysosomal storage disease caused by autosomal recessive mutations in ceroid lipofuscinosis 3 (CLN3). Children with JNCL experience progressive visual, cognitive, and motor deterioration with a decreased life expectancy (late teens-early 20s). Neuronal loss is thought to occur, in part, via glutamate excitotoxicity; however, little is known about astrocyte glutamate regulation in JNCL. Spontaneous Ca2+ oscillations were reduced in murine Cln3Δex7/8 astrocytes, which were also observed following glutamate or cytokine exposure. Astrocyte glutamate transport is an energy-demanding process and disruptions in metabolic pathways could influence glutamate homeostasis in Cln3Δex7/8 astrocytes. Indeed, basal mitochondrial respiration and ATP production were significantly reduced in Cln3Δex7/8 astrocytes. These changes were not attributable to reduced mitochondria, since mitochondrial DNA levels were similar between wild type and Cln3Δex7/8 astrocytes. Interestingly, despite these functional deficits in Cln3Δex7/8 astrocytes, glutamate transporter expression and glutamate uptake were not dramatically affected. Concurrent with impaired astrocyte metabolism and Ca2+ signaling, murine Cln3Δex7/8 neurons were hyper-responsive to glutamate, as reflected by heightened and prolonged Ca2+ signals. These findings identify intrinsic metabolic and Ca2+ signaling defects in Cln3Δex7/8 astrocytes that may contribute to neuronal dysfunction in CLN3 disease. This article is part of the Special Issue "Lysosomal Storage Disorders".
Collapse
Affiliation(s)
- Megan E Bosch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
22
|
Chincholkar M. Analgesic mechanisms of gabapentinoids and effects in experimental pain models: a narrative review. Br J Anaesth 2018; 120:1315-1334. [DOI: 10.1016/j.bja.2018.02.066] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 02/07/2018] [Accepted: 03/14/2018] [Indexed: 12/17/2022] Open
|
23
|
Stimulation of synapse formation between stem cell-derived neurons and native brainstem auditory neurons. Sci Rep 2017; 7:13843. [PMID: 29062015 PMCID: PMC5653851 DOI: 10.1038/s41598-017-13764-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/11/2017] [Indexed: 01/05/2023] Open
Abstract
Integration of stem cell-derived cells into native cellular environment remains a challenge in the field. This study developed novel methods to co-culture neural stem cell-derived spiral ganglion-like neurons (ScNs) and mouse auditory cochlear nucleus (CN) neurons to understand whether ScNs of the peripheral nervous system (PNS) synapse with CN neurons of the central nervous system (CNS). ScNs were obtained from neural stem cells that were derived from transgenic mouse pre-labeled with enhanced green fluorescent protein (EGFP), whereas CN neurons were from postnatal mouse primary cultures. ScNs and CN neurons were co-cultured for 4–6 days in the absence or presence of astrocyte-conditioned medium (ACM). Class III β-tubulin (TUJ1)-expressing connections were found between ScNs and CN neurons. Expression of the synaptic vesicle marker SV2 was significantly increased along connections between ScNs and CN neurons in the presence of ACM. Immunodepletion and knockout studies indicated that thrombospodin-1 played an important role in ACM-exerted synaptogenic effects. Newly-generated synapse-like structures expressed glutamatergic marker VGluT1, pre- and post-synaptic proteins. Synaptic vesicle recycling studies suggested functional synaptic vesicle retrieval. These results reveal that stem cell-derived PNS neurons are able to form functional connections with native CNS neurons, which is critical for stem cell-based neural pathway regeneration.
Collapse
|
24
|
Wang S, Hu T, Wang Z, Li N, Zhou L, Liao L, Wang M, Liao L, Wang H, Zeng L, Fan C, Zhou H, Xiong K, Huang J, Chen D. Macroglia-derived thrombospondin 2 regulates alterations of presynaptic proteins of retinal neurons following elevated hydrostatic pressure. PLoS One 2017; 12:e0185388. [PMID: 28953973 PMCID: PMC5617560 DOI: 10.1371/journal.pone.0185388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/12/2017] [Indexed: 02/03/2023] Open
Abstract
Many studies on retinal injury and repair following elevated intraocular pressure suggest that the survival ratio of retinal neurons has been improved by various measures. However, the visual function recovery is far lower than expected. The homeostasis of retinal synapses in the visual signal pathway is the key structural basis for the delivery of visual signals. Our previous studies found that complicated changes in the synaptic structure between retinal neurons occurred much earlier than obvious degeneration of retinal ganglion cells in rat retinae. The lack of consideration of these earlier retinal synaptic changes in the rescue strategy may be partly responsible for the limited visual function recovery with the types of protective methods for retinal neurons used following elevated intraocular pressure. Thus, research on the modulatory mechanisms of the synaptic changes after elevated intraocular pressure injury may give new light to visual function rescue. In this study, we found that thrombospondin 2, an important regulator of synaptogenesis in central nervous system development, was distributed in retinal macroglia cells, and its receptor α2δ-1 was in retinal neurons. Cell cultures including mixed retinal macroglia cells/neuron cultures and retinal neuron cultures were exposed to elevated hydrostatic pressure for 2 h. The expression levels of glial fibrillary acidic protein (the marker of activated macroglia cells), thrombospondin 2, α2δ-1 and presynaptic proteins were increased following elevated hydrostatic pressure in mixed cultures, but the expression levels of postsynaptic proteins were not changed. SiRNA targeting thrombospondin 2 could decrease the upregulation of presynaptic proteins induced by the elevated hydrostatic pressure. However, in retinal neuron cultures, elevated hydrostatic pressure did not affect the expression of presynaptic or postsynaptic proteins. Rather, the retinal neuron cultures with added recombinant thrombospondin 2 protein upregulated the level of presynaptic proteins. Finally, gabapentin decreased the expression of presynaptic proteins in mixed cultures by blocking the interaction of thrombospondin 2 and α2δ-1. Taken together, these results indicate that activated macroglia cells may participate in alterations of presynaptic proteins of retinal neurons following elevated hydrostatic pressure, and macroglia-derived thrombospondin 2 may modulate these changes via binding to its neuronal receptor α2δ-1.
Collapse
Affiliation(s)
- Shuchao Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Tu Hu
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Zhen Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Na Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Lihong Zhou
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Mi Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Libin Liao
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Hui Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Leping Zeng
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Chunling Fan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Hongkang Zhou
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Jufang Huang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| |
Collapse
|
25
|
Chen P, Yu N, Zhang Z, Zhang P, Yang Y, Wu N, Xu L, Zhang J, Ge J, Yu K, Zhuang J. Thrombospondin-1 might be a therapeutic target to suppress RB cells by regulating the DNA double-strand breaks repair. Oncotarget 2017; 7:6105-20. [PMID: 26756218 PMCID: PMC4868743 DOI: 10.18632/oncotarget.6835] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 12/29/2015] [Indexed: 12/04/2022] Open
Abstract
Retinoblastoma (RB) arises from the retina, and its growth usually occurs under the retina and toward the vitreous. Ideal therapy should aim to inhibit the tumor and protect neural cells, increasing the patient's life span and quality of life. Previous studies have demonstrated that Thrombospondin-1 (TSP-1) is associated with neurogenesis, neovascularization and tumorigenesis. However, at present, the bioactivity of TSP-1 in retinoblastoma has not been defined. Herein, we demonstrated that TSP-1 was silenced in RB cell lines and clinical tumor samples. HDAC inhibitor, Trichostatin A (TSA), could notably transcriptionally up-regulate TSP-1 in RB cells, WERI-Rb1 cells and Y79 cells. Moreover, we found human recombinant TSP-1 (hTSP-1) could significantly inhibit the cell viability of RB cells both in vitro and in vivo. Interestingly, hTSP-1 could significantly induce the expression of γ-H2AX, a well-characterized in situ marker of DNA double-strand breaks (DSBs) in RB cells. The DNA NHEJ pathway in WERI-Rb1 cells could be significantly inhibited by hTSP-1. A mutation in Rb1 might be involved in the hTSP-1-medicated γ-H2AX increasing in WERI-Rb1 cells. Furthermore, hTSP-1 could inhibit RB cells while promoting retinal neurocyte survival in the neuronal and retinoblastoma cell co-culture system. As such, TSP-1 may become a therapeutic target for treatment of retinoblastoma.
Collapse
Affiliation(s)
- Pei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Na Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Zhang Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Ying Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Nandan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Lijun Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, P. R. China 510060
| |
Collapse
|
26
|
Role of Matricellular Proteins in Disorders of the Central Nervous System. Neurochem Res 2016; 42:858-875. [DOI: 10.1007/s11064-016-2088-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022]
|
27
|
Filous AR, Silver J. "Targeting astrocytes in CNS injury and disease: A translational research approach". Prog Neurobiol 2016; 144:173-87. [PMID: 27026202 PMCID: PMC5035184 DOI: 10.1016/j.pneurobio.2016.03.009] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 02/03/2016] [Accepted: 03/03/2016] [Indexed: 12/31/2022]
Abstract
Astrocytes are a major constituent of the central nervous system. These glia play a major role in regulating blood-brain barrier function, the formation and maintenance of synapses, glutamate uptake, and trophic support for surrounding neurons and glia. Therefore, maintaining the proper functioning of these cells is crucial to survival. Astrocyte defects are associated with a wide variety of neuropathological insults, ranging from neurodegenerative diseases to gliomas. Additionally, injury to the CNS causes drastic changes to astrocytes, often leading to a phenomenon known as reactive astrogliosis. This process is important for protecting the surrounding healthy tissue from the spread of injury, while it also inhibits axonal regeneration and plasticity. Here, we discuss the important roles of astrocytes after injury and in disease, as well as potential therapeutic approaches to restore proper astrocyte functioning.
Collapse
Affiliation(s)
- Angela R Filous
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 216-368-4615, United States.
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 216-368-4615, United States.
| |
Collapse
|
28
|
Adermark L, Bowers MS. Disentangling the Role of Astrocytes in Alcohol Use Disorder. Alcohol Clin Exp Res 2016; 40:1802-16. [PMID: 27476876 PMCID: PMC5407469 DOI: 10.1111/acer.13168] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/02/2016] [Indexed: 01/29/2023]
Abstract
Several laboratories recently identified that astrocytes are critical regulators of addiction machinery. It is now known that astrocyte pathology is a common feature of ethanol (EtOH) exposure in both humans and animal models, as even brief EtOH exposure is sufficient to elicit long-lasting perturbations in astrocyte gene expression, activity, and proliferation. Astrocytes were also recently shown to modulate the motivational properties of EtOH and other strongly reinforcing stimuli. Given the role of astrocytes in regulating glutamate homeostasis, a crucial component of alcohol use disorder (AUD), astrocytes might be an important target for the development of next-generation alcoholism treatments. This review will outline some of the more prominent features displayed by astrocytes, how these properties are influenced by acute and long-term EtOH exposure, and future directions that may help to disentangle astrocytic from neuronal functions in the etiology of AUD.
Collapse
Affiliation(s)
- Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Box 410, SE-405 30 Gothenburg, Sweden
| | - M. Scott Bowers
- Department of Psychiatry, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, PO Box 980126, Richmond, VA 23298, USA
- Faulk Center for Molecular Therapeutics, Northwestern University; Aptinyx,, Evanston, Il 60201, USA
| |
Collapse
|
29
|
Cheng C, Lau SKM, Doering LC. Astrocyte-secreted thrombospondin-1 modulates synapse and spine defects in the fragile X mouse model. Mol Brain 2016; 9:74. [PMID: 27485117 PMCID: PMC4971702 DOI: 10.1186/s13041-016-0256-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/15/2016] [Indexed: 01/24/2023] Open
Abstract
Astrocytes are key participants in various aspects of brain development and function, many of which are executed via secreted proteins. Defects in astrocyte signaling are implicated in neurodevelopmental disorders characterized by abnormal neural circuitry such as Fragile X syndrome (FXS). In animal models of FXS, the loss in expression of the Fragile X mental retardation 1 protein (FMRP) from astrocytes is associated with delayed dendrite maturation and improper synapse formation; however, the effect of astrocyte-derived factors on the development of neurons is not known. Thrombospondin-1 (TSP-1) is an important astrocyte-secreted protein that is involved in the regulation of spine development and synaptogenesis. In this study, we found that cultured astrocytes isolated from an Fmr1 knockout (Fmr1 KO) mouse model of FXS displayed a significant decrease in TSP-1 protein expression compared to the wildtype (WT) astrocytes. Correspondingly, Fmr1 KO hippocampal neurons exhibited morphological deficits in dendritic spines and alterations in excitatory synapse formation following long-term culture. All spine and synaptic abnormalities were prevented in the presence of either astrocyte-conditioned media or a feeder layer derived from FMRP-expressing astrocytes, or following the application of exogenous TSP-1. Importantly, this work demonstrates the integral role of astrocyte-secreted signals in the establishment of neuronal communication and identifies soluble TSP-1 as a potential therapeutic target for Fragile X syndrome.
Collapse
Affiliation(s)
- Connie Cheng
- McMaster Integrative Neuroscience Discovery and Study Program (MINDS), McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada.,Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, HSC 1R15A, Hamilton, Ontario, L8N 3Z5, Canada
| | - Sally K M Lau
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, HSC 1R15A, Hamilton, Ontario, L8N 3Z5, Canada
| | - Laurie C Doering
- McMaster Integrative Neuroscience Discovery and Study Program (MINDS), McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada. .,Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, HSC 1R15A, Hamilton, Ontario, L8N 3Z5, Canada.
| |
Collapse
|
30
|
Matias I, Buosi AS, Gomes FCA. Functions of flavonoids in the central nervous system: Astrocytes as targets for natural compounds. Neurochem Int 2016; 95:85-91. [DOI: 10.1016/j.neuint.2016.01.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 01/16/2023]
|
31
|
Loss of Local Astrocyte Support Disrupts Action Potential Propagation and Glutamate Release Synchrony from Unmyelinated Hippocampal Axon Terminals In Vitro. J Neurosci 2015; 35:11105-17. [PMID: 26245971 DOI: 10.1523/jneurosci.1289-15.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Neuron-astrocyte interactions are critical for proper CNS development and function. Astrocytes secrete factors that are pivotal for synaptic development and function, neuronal metabolism, and neuronal survival. Our understanding of this relationship, however, remains incomplete due to technical hurdles that have prevented the removal of astrocytes from neuronal circuits without changing other important conditions. Here we overcame this obstacle by growing solitary rat hippocampal neurons on microcultures that were comprised of either an astrocyte bed (+astrocyte) or a collagen bed (-astrocyte) within the same culture dish. -Astrocyte autaptic evoked EPSCs, but not IPSCs, displayed an altered temporal profile, which included increased synaptic delay, increased time to peak, and severe glutamate release asynchrony, distinct from previously described quantal asynchrony. Although we observed minimal alteration of the somatically recorded action potential waveform, action potential propagation was altered. We observed a longer latency between somatic initiation and arrival at distal locations, which likely explains asynchronous EPSC peaks, and we observed broadening of the axonal spike, which likely underlies changes to evoked EPSC onset. No apparent changes in axon structure were observed, suggesting altered axonal excitability. In conclusion, we propose that local astrocyte support has an unappreciated role in maintaining glutamate release synchrony by disturbing axonal signal propagation. SIGNIFICANCE STATEMENT Certain glial cell types (oligodendrocytes, Schwann cells) facilitate the propagation of neuronal electrical signals, but a role for astrocytes has not been identified despite many other functions of astrocytes in supporting and modulating neuronal signaling. Under identical global conditions, we cultured neurons with or without local astrocyte support. Without local astrocytes, glutamate transmission was desynchronized by an alteration of the waveform and arrival time of axonal action potentials to synaptic terminals. GABA transmission was not disrupted. The disruption did not involve detectable morphological changes to axons of glutamate neurons. Our work identifies a developmental role for astrocytes in the temporal precision of excitatory signals.
Collapse
|
32
|
Fukushima K, Miura Y, Sawada K, Yamazaki K, Ito M. Establishment of a Human Neuronal Network Assessment System by Using a Human Neuron/Astrocyte Co-Culture Derived from Fetal Neural Stem/Progenitor Cells. ACTA ACUST UNITED AC 2015; 21:54-64. [PMID: 26482803 DOI: 10.1177/1087057115610055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/14/2015] [Indexed: 11/17/2022]
Abstract
Using human cell models mimicking the central nervous system (CNS) provides a better understanding of the human CNS, and it is a key strategy to improve success rates in CNS drug development. In the CNS, neurons function as networks in which astrocytes play important roles. Thus, an assessment system of neuronal network functions in a co-culture of human neurons and astrocytes has potential to accelerate CNS drug development. We previously demonstrated that human hippocampus-derived neural stem/progenitor cells (HIP-009 cells) were a novel tool to obtain human neurons and astrocytes in the same culture. In this study, we applied HIP-009 cells to a multielectrode array (MEA) system to detect neuronal signals as neuronal network functions. We observed spontaneous firings of HIP-009 neurons, and validated functional formation of neuronal networks pharmacologically. By using this assay system, we investigated effects of several reference compounds, including agonists and antagonists of glutamate and γ-aminobutyric acid receptors, and sodium, potassium, and calcium channels, on neuronal network functions using firing and burst numbers, and synchrony as readouts. These results indicate that the HIP-009/MEA assay system is applicable to the pharmacological assessment of drug candidates affecting synaptic functions for CNS drug development.
Collapse
Affiliation(s)
- Kazuyuki Fukushima
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Yuji Miura
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Kohei Sawada
- Biopharmaceutical Assessment CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Kazuto Yamazaki
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Masashi Ito
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| |
Collapse
|
33
|
Mendus D, Rankin-Gee EK, Mustapha M, Porter BE. Increased sensitivity to kindling in mice lacking TSP1. Neuroscience 2015; 305:302-8. [PMID: 26241338 PMCID: PMC6699182 DOI: 10.1016/j.neuroscience.2015.07.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/17/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022]
Abstract
The development of a hyperexcitable neuronal network is thought to be a critical event in epilepsy. Thrombospondins (TSPs) regulate synaptogenesis by binding the neuronal α2δ subunit of the voltage-gated calcium channel. TSPs regulate synapse formation during development and in the mature brain following injury. It is unclear if TSPs are involved in hyperexcitability that contributes to the development of epilepsy. Here we explore the development of epilepsy using a pentylenetetrazole (PTZ) kindling model in mice lacking TSP1 and TSP2. Unexpectedly, we found increased sensitivity to PTZ kindling in mice lacking TSP1, while mice lacking TSP2 kindled similar to wild-type. We found that the increased seizure susceptibility in the TSP1 knockout (KO) mice was not due to a compensatory increase in TSP2 mRNA as TSP1/2 KO mice were sensitive to PTZ, similar to the TSP1 KO mice. Furthermore, there were similar levels of TGF-B signal activation during kindling in the TSP1 KO mice compared to wild-type. We observed decreased expression of voltage-dependent calcium channel subunit CACNA2D1 mRNA in TSP1, TSP2, and TSP1/2 KO mice. Decreased CACNA2D2 mRNA was only detected in mice that lacked TSP1 and α2δ-1/2 protein levels in the cortex were lower in the TSP 1/2 KO mice. CACNA2D2 knockout mice have spontaneous seizures and increased PTZ seizure susceptibility. Here we report similar findings, TSP1, and TSP1/2 KO mice have low levels of CACNA2D2 mRNA expression and α2δ-1/2 receptor level in the cortex, and are more susceptible to seizures. CACNA2D2 mutations in mice and humans can cause epilepsy. Our data suggest TSP1 in particular may control CACNA2D2 levels and could be a modifier of seizure susceptibility.
Collapse
Affiliation(s)
- D Mendus
- The Department of Neurology, School of Medicine, Stanford University, Stanford, CA 94305, USA; The Department of Otolaryngology - Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - E K Rankin-Gee
- The Department of Neurology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - M Mustapha
- The Department of Otolaryngology - Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - B E Porter
- The Department of Neurology, School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
34
|
Thrombospondin-1 prevents amyloid beta-mediated synaptic pathology in Alzheimer's disease. Neurobiol Aging 2015; 36:3214-3227. [PMID: 26452999 DOI: 10.1016/j.neurobiolaging.2015.09.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 09/07/2015] [Accepted: 09/07/2015] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is characterized by impaired cognitive function and memory loss, which are often the result of synaptic pathology. Thrombospondin (TSP) is an astrocyte-secreted protein, well known for its function as a modulator of synaptogenesis and neurogenesis. Here, we investigated the effects of TSP-1 on AD pathogenesis. We found that the level of TSP-1 expression was decreased in AD brains. When we treated astrocytes with amyloid beta (Aβ), secreted TSP-1 was decreased in autophagy-dependent manner. In addition, treatment with Aβ induced synaptic pathology, such as decreased dendritic spine density and reduced synaptic activity. These effects were prevented by coincubation of TSP-1 with Aβ, which acts through the TSP-1 receptor alpha-2-delta-1 in neurons. Finally, intrasubicular injection with TSP-1 into AD model mouse brains mitigated the Aβ-mediated reduction of synaptic proteins and related signaling pathways. These results indicate that TSP-1 is a potential therapeutic target in AD pathogenesis.
Collapse
|
35
|
Rasband MN. Glial Contributions to Neural Function and Disease. Mol Cell Proteomics 2015; 15:355-61. [PMID: 26342039 DOI: 10.1074/mcp.r115.053744] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Indexed: 12/31/2022] Open
Abstract
The nervous system consists of neurons and glial cells. Neurons generate and propagate electrical and chemical signals, whereas glia function mainly to modulate neuron function and signaling. Just as there are many different kinds of neurons with different roles, there are also many types of glia that perform diverse functions. For example, glia make myelin; modulate synapse formation, function, and elimination; regulate blood flow and metabolism; and maintain ionic and water homeostasis to name only a few. Although proteomic approaches have been used extensively to understand neurons, the same cannot be said for glia. Importantly, like neurons, glial cells have unique protein compositions that reflect their diverse functions, and these compositions can change depending on activity or disease. Here, I discuss the major classes and functions of glial cells in the central and peripheral nervous systems. I describe proteomic approaches that have been used to investigate glial cell function and composition and the experimental limitations faced by investigators working with glia.
Collapse
Affiliation(s)
- Matthew N Rasband
- From the Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
36
|
Jiang P, Zhu T, Xia Z, Gao F, Gu W, Chen X, Yuan T, Yu H. Inhibition of MAPK/ERK signaling blocks hippocampal neurogenesis and impairs cognitive performance in prenatally infected neonatal rats. Eur Arch Psychiatry Clin Neurosci 2015; 265:497-509. [PMID: 25721317 DOI: 10.1007/s00406-015-0588-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 02/19/2015] [Indexed: 12/20/2022]
Abstract
Hippocampus endogenous neurogenesis has been postulated to play a favorable role in brain restoration after injury. However, the underlying molecular mechanisms have been insufficiently deciphered. Here we investigated the potential regulatory capacity of MAPK/ERK signaling on neurogenesis and the associated cognitive performance in prenatally infected neonatal rats. From our data, intrauterine infection could induce hippocampal neuronal apoptosis and promote endogenous repair by evoking neural stem cell proliferation and survival. We also found intrauterine infection could induce increased levels of p-ERK, p-CREB and BDNF, which might be responsible for the potential endogenous rescue system. Furthermore, inhibition of MAPK/ERK signaling could aggravate hippocampal neuronal apoptosis, decrease neurogenesis, and impair the offspring's cognitive performances and could also down-regulate the levels of p-ERK, p-CREB and BDNF. Our data strongly suggest that the activation of MAPK/ERK signaling may play a significant role in promoting survival of newly generated neural stem cells via an anti-apoptotic mechanism, which may be particularly important in endogenous neuroprotection associated with cognitive performance development in prenatally infected rats.
Collapse
Affiliation(s)
- Peifang Jiang
- Department of Neurology, Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Weissberg I, Wood L, Kamintsky L, Vazquez O, Milikovsky DZ, Alexander A, Oppenheim H, Ardizzone C, Becker A, Frigerio F, Vezzani A, Buckwalter MS, Huguenard JR, Friedman A, Kaufer D. Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction. Neurobiol Dis 2015; 78:115-25. [PMID: 25836421 DOI: 10.1016/j.nbd.2015.02.029] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/07/2015] [Accepted: 02/19/2015] [Indexed: 01/26/2023] Open
Abstract
Post-injury epilepsy (PIE) is a common complication following brain insults, including ischemic, and traumatic brain injuries. At present, there are no means to identify the patients at risk to develop PIE or to prevent its development. Seizures can occur months or years after the insult, do not respond to anti-seizure medications in over third of the patients, and are often associated with significant neuropsychiatric morbidities. We have previously established the critical role of blood-brain barrier dysfunction in PIE, demonstrating that exposure of brain tissue to extravasated serum albumin induces activation of inflammatory transforming growth factor beta (TGF-β) signaling in astrocytes and eventually seizures. However, the link between the acute astrocytic inflammatory responses and reorganization of neural networks that underlie recurrent spontaneous seizures remains unknown. Here we demonstrate in vitro and in vivo that activation of the astrocytic ALK5/TGF-β-pathway induces excitatory, but not inhibitory, synaptogenesis that precedes the appearance of seizures. Moreover, we show that treatment with SJN2511, a specific ALK5/TGF-β inhibitor, prevents synaptogenesis and epilepsy. Our findings point to astrocyte-mediated synaptogenesis as a key epileptogenic process and highlight the manipulation of the TGF-β-pathway as a potential strategy for the prevention of PIE.
Collapse
Affiliation(s)
- Itai Weissberg
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Lydia Wood
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Lyn Kamintsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Oscar Vazquez
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Allyson Alexander
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Hannah Oppenheim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Carolyn Ardizzone
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA
| | - Albert Becker
- Department of Neuropathology, University of Bonn Medical Center, Bonn 53105, Germany
| | - Federica Frigerio
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720-3140, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3140, USA; Canadian Institute for Advanced Research (CIFAR) Program in Child and Brain Development Toronto, ON M5G 1Z8, Canada.
| |
Collapse
|
38
|
Guillermo RB, Yang P, Vickers MH, McJarrow P, Guan J. Supplementation with complex milk lipids during brain development promotes neuroplasticity without altering myelination or vascular density. Food Nutr Res 2015; 59:25765. [PMID: 25818888 PMCID: PMC4377325 DOI: 10.3402/fnr.v59.25765] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 01/23/2015] [Accepted: 02/03/2015] [Indexed: 11/18/2022] Open
Abstract
Background Supplementation with complex milk lipids (CML) during postnatal brain development has been shown to improve spatial reference learning in rats. Objective The current study examined histo-biological changes in the brain following CML supplementation and their relationship to the observed improvements in memory. Design The study used the brain tissues from the rats (male Wistar, 80 days of age) after supplementing with either CML or vehicle during postnatal day 10–80. Immunohistochemical staining of synaptophysin, glutamate receptor-1, myelin basic protein, isolectin B-4, and glial fibrillary acidic protein was performed. The average area and the density of the staining and the numbers of astrocytes and capillaries were assessed and analysed. Results Compared with control rats, CML supplementation increased the average area of synaptophysin staining and the number of GFAP astrocytes in the CA3 sub-region of the hippocampus (p<0.01), but not in the CA4 sub-region. The supplementation also led to an increase in dopamine output in the striatum that was related to nigral dopamine expression (p<0.05), but did not alter glutamate receptors, myelination or vascular density. Conclusion CML supplementation may enhance neuroplasticity in the CA3 sub-regions of the hippocampus. The brain regions-specific increase of astrocyte may indicate a supporting role for GFAP in synaptic plasticity. CML supplementation did not associate with postnatal white matter development or vascular remodelling.
Collapse
Affiliation(s)
- Rosamond B Guillermo
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Panzao Yang
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Paul McJarrow
- Fonterra Research and Development Centre, Palmerston North, New Zealand
| | - Jian Guan
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand;
| |
Collapse
|
39
|
Chung WS, Allen NJ, Eroglu C. Astrocytes Control Synapse Formation, Function, and Elimination. Cold Spring Harb Perspect Biol 2015; 7:a020370. [PMID: 25663667 DOI: 10.1101/cshperspect.a020370] [Citation(s) in RCA: 558] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, through their close associations with synapses, can monitor and alter synaptic function, thus actively controlling synaptic transmission in the adult brain. Besides their important role at adult synapses, in the last three decades a number of critical findings have highlighted the importance of astrocytes in the establishment of synaptic connectivity in the developing brain. In this article, we will review the key findings on astrocytic control of synapse formation, function, and elimination. First, we will summarize our current structural and functional understanding of astrocytes at the synapse. Then, we will discuss the cellular and molecular mechanisms through which developing and mature astrocytes instruct the formation, maturation, and refinement of synapses. Our aim is to provide an overview of astrocytes as important players in the establishment of a functional nervous system.
Collapse
Affiliation(s)
- Won-Suk Chung
- Stanford University, School of Medicine, Department of Neurobiology, Stanford, California 94305
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, La Jolla, California 92037
| | - Cagla Eroglu
- Duke University Medical Center, Cell Biology and Neurobiology Departments, Duke Institute for Brain Sciences, Durham, North Carolina 27710
| |
Collapse
|
40
|
Jayakumar AR, Tong XY, Curtis KM, Ruiz-Cordero R, Shamaladevi N, Abuzamel M, Johnstone J, Gaidosh G, Rama Rao KV, Norenberg MD. Decreased astrocytic thrombospondin-1 secretion after chronic ammonia treatment reduces the level of synaptic proteins: in vitro and in vivo studies. J Neurochem 2014; 131:333-47. [PMID: 25040426 PMCID: PMC4364553 DOI: 10.1111/jnc.12810] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 06/16/2014] [Accepted: 06/22/2014] [Indexed: 12/23/2022]
Abstract
Chronic hepatic encephalopathy (CHE) is a major complication in patients with severe liver disease. Elevated blood and brain ammonia levels have been implicated in its pathogenesis, and astrocytes are the principal neural cells involved in this disorder. Since defective synthesis and release of astrocytic factors have been shown to impair synaptic integrity in other neurological conditions, we examined whether thrombospondin-1 (TSP-1), an astrocytic factor involved in the maintenance of synaptic integrity, is also altered in CHE. Cultured astrocytes were exposed to ammonia (NH₄Cl, 0.5-2.5 mM) for 1-10 days, and TSP-1 content was measured in cell extracts and culture media. Astrocytes exposed to ammonia exhibited a reduction in intra- and extracellular TSP-1 levels. Exposure of cultured neurons to conditioned media from ammonia-treated astrocytes showed a decrease in synaptophysin, PSD95, and synaptotagmin levels. Conditioned media from TSP-1 over-expressing astrocytes that were treated with ammonia, when added to cultured neurons, reversed the decline in synaptic proteins. Recombinant TSP-1 similarly reversed the decrease in synaptic proteins. Metformin, an agent known to increase TSP-1 synthesis in other cell types, also reversed the ammonia-induced TSP-1 reduction. Likewise, we found a significant decline in TSP-1 level in cortical astrocytes, as well as a reduction in synaptophysin content in vivo in a rat model of CHE. These findings suggest that TSP-1 may represent an important therapeutic target for CHE. Defective release of astrocytic factors may impair synaptic integrity in chronic hepatic encephalopathy. We found a reduction in the release of the astrocytic matricellular proteins thrombospondin-1 (TSP-1) in ammonia-treated astrocytes; such reduction was associated with a decrease in synaptic proteins caused by conditioned media from ammonia-treated astrocytes. Exposure of neurons to CM from ammonia-treated astrocytes, in which TSP-1 is over-expressed, reversed (by approx 75%) the reduction in synaptic proteins. NF-kB = nuclear factor kappa B; PSD95 = post-synaptic density protein 95; ONS = oxidative/nitrative stress.
Collapse
Affiliation(s)
- Arumugam R Jayakumar
- Laboratory of Neuropathology, Veterans Affairs Medical Center, Miami, Florida, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Phamduong E, Rathore MK, Crews NR, D’Angelo AS, Leinweber AL, Kappera P, Krenning TM, Rendell VR, Belcheva MM, Coscia CJ. Acute and chronic mu opioids differentially regulate thrombospondins 1 and 2 isoforms in astrocytes. ACS Chem Neurosci 2014; 5:106-14. [PMID: 24304333 DOI: 10.1021/cn400172n] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chronic opioids induce synaptic plasticity, a major neuronal adaptation. Astrocyte activation in synaptogenesis may play a critical role in opioid tolerance, withdrawal, and dependence. Thrombospondins 1 and 2 (TSP1/2) are astrocyte-secreted matricellular glycoproteins that promote neurite outgrowth as well as dendritic spine and synapse formation, all of which are inhibited by chronic μ opioids. In prior studies, we discovered that the mechanism of TSP1 regulation by μ opioids in astrocytes involves crosstalk between three different classes of receptors, μ opioid receptor, EGFR and TGFβR. Moreover, TGFβ1 stimulated TSP1 expression via EGFR and ERK/MAPK activation, indicating that EGFR is a signaling hub for opioid and TGFβ1 actions. Using various selective antagonists, and inhibitors, here we compared the mechanisms of chronic opioid regulation of TSP1/2 isoform expression in vivo and in immortalized rat cortical astrocytes. TSP1/2 release from astrocytes was also monitored. Acute and chronic μ opioids, morphine, and the prototypic μ ligand, DAMGO, modulated TSP2 protein levels. TSP2 but not TSP1 protein content was up-regulated by acute (3 h) morphine or DAMGO by an ERK/MAPK dependent mechanism. Paradoxically, TSP2 protein levels were altered neither by TGFβ1 nor by astrocytic neurotrophic factors, EGF, CNTF, and BMP4. TSP1/2 immunofluorescence was increased in astrocytes subjected to scratch-wounding, suggesting TSPs may be useful markers for the "reactive" state of these cells and potentially for different types of injury. Previously, we determined that chronic morphine attenuated both neurite outgrowth and synapse formation in cocultures of primary astrocytes and neurons under similar temporal conditions that μ opioids reduced TSP1 protein levels in astrocytes. Here we found that, after the same 8 day treatment, morphine or DAMGO diminished TSP2 protein levels in astrocytes. Therefore, μ opioids may deter synaptogenesis via both TSP1/2 isoforms, but by distinct mechanisms.
Collapse
Affiliation(s)
- Ellen Phamduong
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Maanjot K. Rathore
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Nicholas R. Crews
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Alexander S. D’Angelo
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Andrew L. Leinweber
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Pranay Kappera
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Thomas M. Krenning
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Victoria R. Rendell
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Mariana M. Belcheva
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Carmine J. Coscia
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| |
Collapse
|
42
|
Zhou L, Wang H, Luo J, Xiong K, Zeng L, Chen D, Huang J. Regulatory effects of inhibiting the activation of glial cells on retinal synaptic plasticity. Neural Regen Res 2014; 9:385-393. [PMID: 25206825 PMCID: PMC4146193 DOI: 10.4103/1673-5374.128240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2014] [Indexed: 01/09/2023] Open
Abstract
Various retinal injuries induced by ocular hypertension have been shown to induce plastic changes in retinal synapses, but the potential regulatory mechanism of synaptic plasticity after retinal injury was still unclear. A rat model of acute ocular hypertension was established by injecting saline intravitreally for an hour, and elevating the intraocular pressure to 14.63 kPa (110 mmHg). Western blot assay and immunofluorescence results showed that synaptophysin expression had a distinct spatiotemporal change that increased in the inner plexiform layer within 1 day and spread across the outer plexiform layer after 3 days. Glial fibrillary acidic protein expression in retinae was greatly increased after 3 days, and reached a peak at 7 days, which was also consistent with the peak time of synaptophysin expression in the outer plexiform layer following the increased intraocular pressure. Fluorocitrate, a glial metabolic inhibitor, was intravitreally injected to inhibit glial cell activation following high intraocular pressure. This significantly inhibited the enhanced glial fibrillary acidic protein expression induced by high intraocular pressure injury. Synaptophysin expression also decreased in the inner plexiform layer within a day and the widened distribution in the outer plexiform layer had disappeared by 3 days. The results suggested that retinal glial cell activation might play an important role in the process of retinal synaptic plasticity induced by acute high intraocular pressure through affecting the expression and distribution of synaptic functional proteins, such as synaptophysin.
Collapse
Affiliation(s)
- Lihong Zhou
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jia Luo
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Leping Zeng
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jufang Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
43
|
Electroconvulsive seizure induces thrombospondin-1 in the adult rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48:236-44. [PMID: 24121060 DOI: 10.1016/j.pnpbp.2013.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/01/2013] [Accepted: 10/02/2013] [Indexed: 01/29/2023]
Abstract
Synaptic dysfunction has recently gained attention for its involvement in mood disorders. Electroconvulsive therapy (ECT) possibly plays a role in synaptic repair. However, the underlying mechanisms remain uncertain. Thrombospondin-1 (TSP-1), a member of the TSP family, is reported to be secreted by astrocytes and to regulate synaptogenesis. We investigated the effects of electroconvulsive seizure (ECS) on the expression of TSPs in the adult rat hippocampus. Single and repeated ECS significantly increased TSP-1 mRNA expression after 2h and returned to sham levels at 24h. Conversely, the TSP-2 and -4 mRNA levels did not change. Only repeated ECS induced TSP-1 proteins. ECS also induced glial fibrillary acidic protein (GFAP) expression. The GFAP expression occurred later than the TSP-1 mRNA expression following single ECS; however, it occurred earlier and was more persistent following repeated ECS. ECS had no effect on the α2δ-1 or neuroligin-1 expressions, both of which are TSP-1 receptors. Furthermore, chronic treatment with antidepressants did not induce the expression of TSP-1 or GFAP. These findings suggest that repeated ECS, but not chronic treatment with antidepressants, induces TSP-1 expression partially via the activation of astrocytes. Therefore, TSP-1 is possibly involved in the synaptogenic effects of ECS.
Collapse
|
44
|
Lazarevic V, Pothula S, Andres-Alonso M, Fejtova A. Molecular mechanisms driving homeostatic plasticity of neurotransmitter release. Front Cell Neurosci 2013; 7:244. [PMID: 24348337 PMCID: PMC3847662 DOI: 10.3389/fncel.2013.00244] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 11/18/2013] [Indexed: 12/27/2022] Open
Abstract
Homeostatic plasticity is a process by which neurons adapt to the overall network activity to keep their firing rates in a reasonable range. At the cellular level this kind of plasticity comprises modulation of cellular excitability and tuning of synaptic strength. In this review we concentrate on presynaptic homeostatic plasticity controlling the efficacy of neurotransmitter release from presynaptic boutons. While morphological and electrophysiological approaches were successful to describe homeostatic plasticity-induced changes in the presynaptic architecture and function, cellular and molecular mechanisms underlying those modifications remained largely unknown for a long time. We summarize the latest progress made in the understanding of homeostasis-induced regulation of different steps of the synaptic vesicle cycle and the molecular machineries involved in this process. We particularly focus on the role of presynaptic scaffolding proteins, which functionally and spatially organize synaptic vesicle clusters, neurotransmitter release sites and the associated endocytic machinery. These proteins turned out to be major presynaptic substrates for remodeling during homeostatic plasticity. Finally, we discuss cellular processes and signaling pathways acting during homeostatic molecular remodeling and their potential involvement in the maladaptive plasticity occurring in multiple neuropathologic conditions such as neurodegeneration, epilepsy and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Vesna Lazarevic
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Germany
| | - Santosh Pothula
- Research Group Presynaptic Plasticity, Leibniz Institute for Neurobiology Magdeburg, Germany
| | - Maria Andres-Alonso
- Research Group Presynaptic Plasticity, Leibniz Institute for Neurobiology Magdeburg, Germany
| | - Anna Fejtova
- Research Group Presynaptic Plasticity, Leibniz Institute for Neurobiology Magdeburg, Germany ; Center for Behavioral Brain Sciences Magdeburg, Germany
| |
Collapse
|
45
|
Frias CP, Wierenga CJ. Activity-dependent adaptations in inhibitory axons. Front Cell Neurosci 2013; 7:219. [PMID: 24312009 PMCID: PMC3836028 DOI: 10.3389/fncel.2013.00219] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 10/30/2013] [Indexed: 11/13/2022] Open
Abstract
Synaptic connections in our brains change continuously and throughout our lifetime. Despite ongoing synaptic changes, a healthy balance between excitation and inhibition is maintained by various forms of homeostatic and activity-dependent adaptations, ensuring stable functioning of neuronal networks. In this review we summarize experimental evidence for activity-dependent changes occurring in inhibitory axons, in cultures as well as in vivo. Axons form many presynaptic terminals, which are dynamic structures sharing presynaptic material along the axonal shaft. We discuss how internal (e.g., vesicle sharing) and external factors (e.g., binding of cell adhesion molecules or secreted factors) may affect the formation and plasticity of inhibitory synapses.
Collapse
Affiliation(s)
| | - Corette J. Wierenga
- Division of Cell Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| |
Collapse
|
46
|
Allen NJ. Role of glia in developmental synapse formation. Curr Opin Neurobiol 2013; 23:1027-33. [PMID: 23871217 DOI: 10.1016/j.conb.2013.06.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/11/2013] [Indexed: 01/20/2023]
Abstract
Neuronal synapse formation and maturation in the developing brain is a complex multi-step process, and it is now clear that glial cells, in particular astrocytes, are key regulators of neuronal synaptogenesis. This article reviews the progress made in the past few years in identifying molecular mechanisms that glial cells use to regulate neuronal synaptogenesis. The focus is on novel glial molecules that induce synapse formation, inhibit synapse formation, or control synaptic levels of glutamate receptors. A role for glial cells in the pathology of neurodevelopmental disorders is discussed.
Collapse
Affiliation(s)
- Nicola J Allen
- Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA.
| |
Collapse
|
47
|
Huang J, Zhou L, Wang H, Luo J, Zeng L, Xiong K, Chen D. Distribution of thrombospondins and their neuronal receptor α2δ1 in the rat retina. Exp Eye Res 2013; 111:36-49. [PMID: 23541831 DOI: 10.1016/j.exer.2013.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 03/04/2013] [Accepted: 03/14/2013] [Indexed: 11/23/2022]
Abstract
The role of the extracellular matrix protein thrombospondins (TSPs) in promoting synaptogenesis is gaining more and more attention. The binding of TSP1 and TSP2 to their neuronal receptor α2δ1 stimulates excitatory synaptogenesis in the development and injury of the central nervous system; however, the specific cellular localization and expression of TSP1/2 and α2δ1 in healthy and damaged retinas is unknown. This, to a certain extent, has restricted the progress of research on the molecular mechanisms triggering synaptic plasticity after retinal injury. Here, the cellular localization and expression of TSP1/2 and their receptor α2δ1 was studied in healthy and damaged adult retina induced by elevated intraocular pressure (IOP) using double immunofluorescence labeling and confocal scanning microscopy. We showed the apparent differential distribution of TSP1 and TSP2 in the adult rat retina. TSP1 was confined to the ganglion cell layer and inner nuclear layer, in which it was preferentially expressed by ganglion cells, bipolar cells and horizontal cells but rarely expressed by glial cells. TSP2 staining was diffusely distributed in GFAP- and GS-immunopositive glial cells and processes in the inner retina. In rat retinas, α2δ1 staining was present in ganglion cells, bipolar cells, partial horizontal cells and amacrine cells and the presynaptic terminals. Müller cells and a minority of astrocytes also expressed α2δ1. On the seventh day of elevated IOP, TSP2 immunoreactivity was greatly increased, and immunopositive processes extended throughout the retinal layer and co-localized with GFAP- and GS-positive glial cells. TSP1 distribution in the retina, however, did not change distinctly. α2δ1-immunopositive processes were also increased on the seventh day after elevated IOP. Our study suggested that in the adult rat retina, TSP2, but not TSP1, secreted by glial cells may be involved in the synaptic plastic process after retinal injury through binding to its neuronal receptor α2δ1.
Collapse
Affiliation(s)
- Jufang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, 172 Tong Zipo Road, Changsha 410013, Hunan Province, PR China
| | | | | | | | | | | | | |
Collapse
|
48
|
Clarke LE, Barres BA. Emerging roles of astrocytes in neural circuit development. Nat Rev Neurosci 2013; 14:311-21. [PMID: 23595014 DOI: 10.1038/nrn3484] [Citation(s) in RCA: 704] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Astrocytes are now emerging as key participants in many aspects of brain development, function and disease. In particular, new evidence shows that astrocytes powerfully control the formation, maturation, function and elimination of synapses through various secreted and contact-mediated signals. Astrocytes are also increasingly being implicated in the pathophysiology of many psychiatric and neurological disorders that result from synaptic defects. A better understanding of how astrocytes regulate neural circuit development and function in the healthy and diseased brain might lead to the development of therapeutic agents to treat these diseases.
Collapse
Affiliation(s)
- Laura E Clarke
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | |
Collapse
|