1
|
Koss KM, Son T, Li C, Hao Y, Cao J, Churchward MA, Zhang ZJ, Wertheim JA, Derda R, Todd KG. Toward discovering a novel family of peptides targeting neuroinflammatory states of brain microglia and astrocytes. J Neurochem 2024; 168:3386-3414. [PMID: 37171455 PMCID: PMC10640667 DOI: 10.1111/jnc.15840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
Microglia are immune-derived cells critical to the development and healthy function of the brain and spinal cord, yet are implicated in the active pathology of many neuropsychiatric disorders. A range of functional phenotypes associated with the healthy brain or disease states has been suggested from in vivo work and were modeled in vitro as surveying, reactive, and primed sub-types of primary rat microglia and mixed microglia/astrocytes. It was hypothesized that the biomolecular profile of these cells undergoes a phenotypical change as well, and these functional phenotypes were explored for potential novel peptide binders using a custom 7 amino acid-presenting M13 phage library (SX7) to identify unique peptides that bind differentially to these respective cell types. Surveying glia were untreated, reactive were induced with a lipopolysaccharide treatment, recovery was modeled with a potent anti-inflammatory treatment dexamethasone, and priming was determined by subsequently challenging the cells with interferon gamma. Microglial function was profiled by determining the secretion of cytokines and nitric oxide, and expression of inducible nitric oxide synthase. After incubation with the SX7 phage library, populations of SX7-positive microglia and/or astrocytes were collected using fluorescence-activated cell sorting, SX7 phage was amplified in Escherichia coli culture, and phage DNA was sequenced via next-generation sequencing. Binding validation was done with synthesized peptides via in-cell westerns. Fifty-eight unique peptides were discovered, and their potential functions were assessed using a basic local alignment search tool. Peptides potentially originated from proteins ranging in function from a variety of supportive glial roles, including synapse support and pruning, to inflammatory incitement including cytokine and interleukin activation, and potential regulation in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- K M Koss
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - T Son
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - C Li
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - Y Hao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - J Cao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - M A Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biology and Environmental Sciences, Concordia University of Edmonton, Alberta, Edmonton, Canada
| | - Z J Zhang
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - J A Wertheim
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - R Derda
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - K G Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biomedical Engineering, University of Alberta, Alberta, Edmonton, Canada
| |
Collapse
|
2
|
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
|
3
|
Chemistry and Function of Glycosaminoglycans in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:117-162. [DOI: 10.1007/978-3-031-12390-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
4
|
Murray TE, Richards CM, Robert-Gostlin VN, Bernath AK, Lindhout IA, Klegeris A. Potential neurotoxic activity of diverse molecules released by astrocytes. Brain Res Bull 2022; 189:80-101. [PMID: 35988785 DOI: 10.1016/j.brainresbull.2022.08.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/04/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Astrocytes are the main support cells of the central nervous system. They also participate in neuroimmune reactions. In response to pathological and immune stimuli, astrocytes transform to reactive states characterized by increased release of inflammatory mediators. Some of these molecules are neuroprotective and inflammation resolving while others, including reactive oxygen species (ROS), nitric oxide (NO), matrix metalloproteinase (MMP)- 9, L-glutamate, and tumor necrosis factor α (TNF), are well-established toxins known to cause damage to surrounding cells and tissues. We hypothesized that similar to microglia, the brain immune cells, reactive astrocytes can release a broader set of diverse molecules that are potentially neurotoxic. A literature search was conducted to identify such molecules using the following two criteria: 1) evidence of their expression and secretion by astrocytes and 2) direct neurotoxic action. This review describes 14 structurally diverse molecules as less-established astrocyte neurotoxins, including C-X-C motif chemokine ligand (CXCL)10, CXCL12/CXCL12(5-67), FS-7-associated surface antigen ligand (FasL), macrophage inflammatory protein (MIP)- 2α, TNF-related apoptosis inducing ligand (TRAIL), pro-nerve growth factor (proNGF), pro-brain-derived neurotrophic factor (proBDNF), chondroitin sulfate proteoglycans (CSPGs), cathepsin (Cat)B, group IIA secretory phospholipase A2 (sPLA2-IIA), amyloid beta peptides (Aβ), high mobility group box (HMGB)1, ceramides, and lipocalin (LCN)2. For some of these molecules, further studies are required to establish either their direct neurotoxic effects or the full spectrum of stimuli that induce their release by astrocytes. Only limited studies with human-derived astrocytes and neurons are available for most of these potential neurotoxins, which is a knowledge gap that should be addressed in the future. We also summarize available evidence of the role these molecules play in select neuropathologies where reactive astrocytes are a key feature. A comprehensive understanding of the full spectrum of neurotoxins released by reactive astrocytes is key to understanding neuroinflammatory diseases characterized by the adverse activation of these cells and may guide the development of novel treatment strategies.
Collapse
Affiliation(s)
- Taryn E Murray
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Victoria N Robert-Gostlin
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Anna K Bernath
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Ivan A Lindhout
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
5
|
Abstract
Multiple intrinsic and extrinsic factors contribute to stem and neuronal precursor cell maintenance and/or differentiation. Proteoglycans, major residents of the stem cell microenvironment, modulate key signaling cues and are of particular importance. The complexity and diversity of the glycan structure of proteoglycans make their functional characterization a challenging task. In order to test the functional role of glycosaminoglycans (GAGs) in cell self-renewal, maintenance, and differentiation, we have taken a loss-of-function approach by developing a library of both biosynthetic and degradative enzymes to specifically remodel the ECM.
Collapse
|
6
|
Domowicz MS, Chan WC, Claudio-Vázquez P, Gonzalez T, Schwartz NB. Brain transcriptome analysis of a CLN2 mouse model as a function of disease progression. J Neuroinflammation 2021; 18:262. [PMID: 34749772 PMCID: PMC8576919 DOI: 10.1186/s12974-021-02302-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neuronal ceroid lipofuscinoses, (NCLs or Batten disease) are a group of inherited, early onset, fatal neurodegenerative diseases associated with mutations in 13 genes. All forms of the disease are characterized by lysosomal accumulation of fluorescent storage material, as well as profound neurodegeneration, but the relationship of the various genes’ function to a single biological process is not obvious. In this study, we used a well-characterized mouse model of classical late infantile NCL (cLINCL) in which the tripeptidyl peptidase 1 (Tpp1) gene is disrupted by gene targeting, resulting in loss of detectable TPP1 activity and leading to progressive neurological phenotypes including ataxia, increased motor deficiency, and early death. Methods In order to identify genes and pathways that may contribute to progression of the neurodegenerative process, we analyzed forebrain/midbrain and cerebellar transcriptional differences at 1, 2, 3 and 4 months of age in control and TPP1-deficient mice by global RNA-sequencing. Results Progressive neurodegenerative inflammatory responses involving microglia, astrocytes and endothelial cells were observed, accompanied by activation of leukocyte extravasation signals and upregulation of nitric oxide production and reactive oxygen species. Several astrocytic (i.e., Gfap, C4b, Osmr, Serpina3n) and microglial (i.e., Ctss, Itgb2, Itgax, Lyz2) genes were identified as strong markers for assessing disease progression as they showed increased levels of expression in vivo over time. Furthermore, transient increased expression of choroid plexus genes was observed at 2 months in the lateral and fourth ventricle, highlighting an early role for the choroid plexus and cerebrospinal fluid in the disease pathology. Based on these gene expression changes, we concluded that neuroinflammation starts, for the most part, after 2 months in the Tpp1−/− brain and that activation of microglia and astrocytes occur more rapidly in cerebellum than in the rest of the brain; confirming increased severity of inflammation in this region. Conclusions These findings have led to a better understanding of cLINCL pathological onset and progression, which may aid in development of future therapeutic treatments for this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02302-z.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.
| | - Wen-Ching Chan
- Center for Research Informatics, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Patricia Claudio-Vázquez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Tatiana Gonzalez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.,Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
7
|
Lei T, Liao X, Chen X, Zhao T, Xu Y, Xia M, Zhang J, Xia Y, Sun X, Wei Y, Men W, Wang Y, Hu M, Zhao G, Du B, Peng S, Chen M, Wu Q, Tan S, Gao JH, Qin S, Tao S, Dong Q, He Y. Progressive Stabilization of Brain Network Dynamics during Childhood and Adolescence. Cereb Cortex 2021; 32:1024-1039. [PMID: 34378030 DOI: 10.1093/cercor/bhab263] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/14/2022] Open
Abstract
Functional brain networks require dynamic reconfiguration to support flexible cognitive function. However, the developmental principles shaping brain network dynamics remain poorly understood. Here, we report the longitudinal development of large-scale brain network dynamics during childhood and adolescence, and its connection with gene expression profiles. Using a multilayer network model, we show the temporally varying modular architecture of child brain networks, with higher network switching primarily in the association cortex and lower switching in the primary regions. This topographical profile exhibits progressive maturation, which manifests as reduced modular dynamics, particularly in the transmodal (e.g., default-mode and frontoparietal) and sensorimotor regions. These developmental refinements mediate age-related enhancements of global network segregation and are linked with the expression profiles of genes associated with the enrichment of ion transport and nucleobase-containing compound transport. These results highlight a progressive stabilization of brain dynamics, which expand our understanding of the neural mechanisms that underlie cognitive development.
Collapse
Affiliation(s)
- Tianyuan Lei
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xuhong Liao
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Xiaodan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Tengda Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Yuehua Xu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Jiaying Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Yunman Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xiaochen Sun
- Department of Linguistics, Beijing Language and Culture University, Beijing 100083, China
| | - Yongbin Wei
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Weiwei Men
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China
| | - Yanpei Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Mingming Hu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Gai Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Bin Du
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Siya Peng
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Menglu Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Shuping Tan
- Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing 100096, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,Beijing City Key Laboratory for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China.,IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.,Chinese Institute for Brain Research, Beijing 102206, China
| | - Sha Tao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.,Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
8
|
Heller GJ, Marshall MS, Issa Y, Marshall JN, Nguyen D, Rue E, Pathmasiri KC, Domowicz MS, van Breemen RB, Tai LM, Cologna SM, Crocker SJ, Givogri MI, Sands MS, Bongarzone ER. Waning efficacy in a long-term AAV-mediated gene therapy study in the murine model of Krabbe disease. Mol Ther 2021; 29:1883-1902. [PMID: 33508430 PMCID: PMC8116612 DOI: 10.1016/j.ymthe.2021.01.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Neonatal AAV9-gene therapy of the lysosomal enzyme galactosylceramidase (GALC) significantly ameliorates central and peripheral neuropathology, prolongs survival, and largely normalizes motor deficits in Twitcher mice. Despite these therapeutic milestones, new observations identified the presence of multiple small focal demyelinating areas in the brain after 6-8 months. These lesions are in stark contrast to the diffuse, global demyelination that affects the brain of naive Twitcher mice. Late-onset lesions exhibited lysosomal alterations with reduced expression of GALC and increased psychosine levels. Furthermore, we found that lesions were closely associated with the extravasation of plasma fibrinogen and activation of the fibrinogen-BMP-SMAD-GFAP gliotic response. Extravasation of fibrinogen correlated with tight junction disruptions of the vasculature within the lesioned areas. The lesions were surrounded by normal appearing white matter. Our study shows that the dysregulation of therapeutic GALC was likely driven by the exhaustion of therapeutic AAV episomal DNA within the lesions, paralleling the presence of proliferating oligodendrocyte progenitors and glia. We believe that this is the first demonstration of diminishing expression in vivo from an AAV gene therapy vector with detrimental effects in the brain of a lysosomal storage disease animal model. The development of this phenotype linking localized loss of GALC activity with relapsing neuropathology in the adult brain of neonatally AAV-gene therapy-treated Twitcher mice identifies and alerts to possible late-onset reductions of AAV efficacy, with implications to other genetic leukodystrophies.
Collapse
Affiliation(s)
- Gregory J Heller
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Michael S Marshall
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Yazan Issa
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jeffrey N Marshall
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Duc Nguyen
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emily Rue
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | | | - Miriam S Domowicz
- Department of Pediatrics, University of Chicago, Chicago, IL 60612, USA
| | | | - Leon M Tai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Maria I Givogri
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark S Sands
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
9
|
Mencio CP, Hussein RK, Yu P, Geller HM. The Role of Chondroitin Sulfate Proteoglycans in Nervous System Development. J Histochem Cytochem 2021; 69:61-80. [PMID: 32936033 PMCID: PMC7780190 DOI: 10.1369/0022155420959147] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
The orderly development of the nervous system is characterized by phases of cell proliferation and differentiation, neural migration, axonal outgrowth and synapse formation, and stabilization. Each of these processes is a result of the modulation of genetic programs by extracellular cues. In particular, chondroitin sulfate proteoglycans (CSPGs) have been found to be involved in almost every aspect of this well-orchestrated yet delicate process. The evidence of their involvement is complex, often contradictory, and lacking in mechanistic clarity; however, it remains obvious that CSPGs are key cogs in building a functional brain. This review focuses on current knowledge of the role of CSPGs in each of the major stages of neural development with emphasis on areas requiring further investigation.
Collapse
Affiliation(s)
- Caitlin P Mencio
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Rowan K Hussein
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, China
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| |
Collapse
|
10
|
Balouch B, Funnell JL, Ziemba AM, Puhl DL, Lin K, Gottipati MK, Gilbert RJ. Conventional immunomarkers stain a fraction of astrocytes in vitro: A comparison of rat cortical and spinal cord astrocytes in naïve and stimulated cultures. J Neurosci Res 2020; 99:806-826. [PMID: 33295039 DOI: 10.1002/jnr.24759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/14/2020] [Indexed: 11/05/2022]
Abstract
Astrocytes are responsible for a wide variety of essential functions throughout the central nervous system. The protein markers glial fibrillary acidic protein (GFAP), glutamate aspartate transporter (GLAST), glutamate transporter-1 (GLT-1), glutamine synthetase (GS), 10-formyltetrahydrofolate dehydrogenase (ALDH1L1), and the transcription factor SOX9 are routinely used to label astrocytes in primary rodent cultures. However, GLAST, GLT-1, GS, and SOX9 are also produced by microglia and oligodendrocytes and GFAP, GLAST, GLT-1, and GS production levels are affected by astrocyte phenotypic changes associated with reactive astrogliosis. No group has performed a comprehensive immunocytochemical evaluation to quantify the percentage of cells labeled by these markers in vitro, nor compared changes in staining between cortex- and spinal cord-derived cells in naïve and stimulated cultures. Here, we quantified the percentage of cells positively stained for these six markers in astrocyte, microglia, and oligodendrocyte cultures isolated from neonatal rat cortices and spinal cords. Additionally, we incubated the astrocytes with transforming growth factor (TGF)-β1 or TGF-β3 to determine if the labeling of these markers is altered by these stimuli. We found that only SOX9 in cortical cultures and ALDH1L1 in spinal cord cultures labeled more than 75% of the cells in naïve and stimulated astrocyte cultures and stained less than 5% of the cells in microglia and oligodendrocyte cultures. Furthermore, significantly more cortical than spinal cord astrocytes stained for GFAP, GLAST, and ALDH1L1 in naïve cultures, whereas significantly more spinal cord than cortical astrocytes stained for GLAST and GS in TGF-β1-treated cultures. These findings are important as variability in marker staining may lead to misinterpretation of the astrocyte response in cocultures, migration assays, or engineered disease models.
Collapse
Affiliation(s)
- Bailey Balouch
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jessica L Funnell
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Alexis M Ziemba
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Neuroscience Program, Smith College, Northampton, MA, USA
| | - Devan L Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Kathy Lin
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Manoj K Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
11
|
Perret R, Escuriol J, Velasco V, Mayeur L, Soubeyran I, Delfour C, Aubert S, Polivka M, Karanian M, Meurgey A, Le Guellec S, Weingertner N, Hoeller S, Coindre JM, Larousserie F, Pierron G, Tirode F, Le Loarer F. NFATc2-rearranged sarcomas: clinicopathologic, molecular, and cytogenetic study of 7 cases with evidence of AGGRECAN as a novel diagnostic marker. Mod Pathol 2020; 33:1930-1944. [PMID: 32327700 DOI: 10.1038/s41379-020-0542-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
NFATc2-rearranged sarcomas (NFATc2-Sarcomas) are infrequent round cell tumors characterized by EWSR1-NFATc2 fusions and FUS-NFATc2 fusions. Although our knowledge on these neoplasms has increased recently, novel diagnostic tools and more comprehensive series are still needed. Here, we describe the features of a series of seven molecularly confirmed NFATc2-Sarcomas (EWSR1-NFATc2, n = 4; FUS-NFATc2, n = 3) and demonstrate the utility of AGGRECAN immunohistochemistry for their identification. Patients were four males and three females, ranging in age from 19 to 66 years (median: 33). All were primary bone tumors (femur, n = 4; tibia, n = 2; ilium, n = 1), frequently infiltrating the surrounding soft tissues. Treatment often consisted of neoadjuvant chemotherapy and surgery. Follow-up was available for six patients (median 18 months, range 5-102 months), three patients died of disease and four patients are currently alive. Histologically, tumors consisted of monotonous round cells growing in lobules and sheets in variable amounts of fibrous to myxoid stroma. Other findings included spindle cells, corded and trabecular architecture, nuclear pleomorphism, cartilaginous differentiation, and osteoid-like matrix. Histological response to neoadjuvant chemotherapy was poor in all resection specimens available for review (n = 4). Tumors were diffusely positive for AGGRECAN and CD99 (7/7), and a subset expressed Pan-Keratin (AE1-AE3; 3/6), S100 (2/6), BCOR (2/6), ETV-4 (2/5), WT1 (2/6), and ERG (2/5). Desmin, NKX3-1, and SATB2 were negative (0/6). Diffuse AGGRECAN staining was also seen in 8/129 round cell sarcomas used for comparison, including mesenchymal chondrosarcoma (7/26) and CIC-sarcoma (1/26). Array-CGH showed complex karyotypes with recurrent deletions of tumor suppressor genes (CDKN2A/B, TUSC7, and DMD) in three FUS-NFATC2 cases and a simpler profile without homozygous losses in one EWSR1-NFATc2 case. Segmental chromosomal gains covering the loci of the fusion genes were detected in both variants. Overall, our study confirms and expands previous observations on NFATc2-sarcomas and supports that AGGRECAN is a useful biomarker of these tumors.
Collapse
Affiliation(s)
- Raul Perret
- Department of Biopathology, Bergonie Institute, Bordeaux, France.
| | - Julien Escuriol
- Department of Biopathology, Bergonie Institute, Bordeaux, France.,Bordeaux University, Talence, France
| | - Valérie Velasco
- Department of Biopathology, Bergonie Institute, Bordeaux, France
| | - Laetitia Mayeur
- Department of Biopathology, Bergonie Institute, Bordeaux, France
| | - Isabelle Soubeyran
- Department of Biopathology, Bergonie Institute, Bordeaux, France.,INSERM U1218, ACTION Unit, Bordeaux, France
| | - Christophe Delfour
- Department of Pathology, Montpellier University Hospital, Montpellier, France
| | - Sébastien Aubert
- Department of Pathology, Institut de Pathologie, Univ. Lille, CHU Lille, F-59000, Lille, France
| | - Marc Polivka
- Department of Pathology, APHP, Hôpital Cochin, DMU Imagina, Université de Paris, F-75014, Paris, France
| | - Marie Karanian
- Department of Pathology, Leon Berard Center, Lyon, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | | | - Sophie Le Guellec
- Department of Pathology, Claudius Regaud Institute, Toulouse-Oncopole, Toulouse, France
| | - Noelle Weingertner
- Department of Pathology, Strasbourg Regional University Hospital (Hautepierre Hospital), Strasbourg, France
| | - Sylvia Hoeller
- Department of Pathology, Hospital of the University of Basel, Basel, Switzerland
| | - Jean-Michel Coindre
- Department of Biopathology, Bergonie Institute, Bordeaux, France.,Bordeaux University, Talence, France
| | | | - Gaëlle Pierron
- Department of Tumor Biology, Curie Institute, Paris, France
| | - Franck Tirode
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - François Le Loarer
- Department of Biopathology, Bergonie Institute, Bordeaux, France. .,Bordeaux University, Talence, France. .,INSERM U1218, ACTION Unit, Bordeaux, France.
| |
Collapse
|
12
|
Hayes AJ, Melrose J. Aggrecan, the Primary Weight-Bearing Cartilage Proteoglycan, Has Context-Dependent, Cell-Directive Properties in Embryonic Development and Neurogenesis: Aggrecan Glycan Side Chain Modifications Convey Interactive Biodiversity. Biomolecules 2020; 10:E1244. [PMID: 32867198 PMCID: PMC7564073 DOI: 10.3390/biom10091244] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 02/06/2023] Open
Abstract
This review examines aggrecan's roles in developmental embryonic tissues, in tissues undergoing morphogenetic transition and in mature weight-bearing tissues. Aggrecan is a remarkably versatile and capable proteoglycan (PG) with diverse tissue context-dependent functional attributes beyond its established role as a weight-bearing PG. The aggrecan core protein provides a template which can be variably decorated with a number of glycosaminoglycan (GAG) side chains including keratan sulphate (KS), human natural killer trisaccharide (HNK-1) and chondroitin sulphate (CS). These convey unique tissue-specific functional properties in water imbibition, space-filling, matrix stabilisation or embryonic cellular regulation. Aggrecan also interacts with morphogens and growth factors directing tissue morphogenesis, remodelling and metaplasia. HNK-1 aggrecan glycoforms direct neural crest cell migration in embryonic development and is neuroprotective in perineuronal nets in the brain. The ability of the aggrecan core protein to assemble CS and KS chains at high density equips cartilage aggrecan with its well-known water-imbibing and weight-bearing properties. The importance of specific arrangements of GAG chains on aggrecan in all its forms is also a primary morphogenetic functional determinant providing aggrecan with unique tissue context dependent regulatory properties. The versatility displayed by aggrecan in biodiverse contexts is a function of its GAG side chains.
Collapse
Affiliation(s)
- Anthony J Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards 2065, NSW, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney 2052, NSW, Australia
- Sydney Medical School, Northern, The University of Sydney, Faculty of Medicine and Health at Royal North Shore Hospital, St. Leonards 2065, NSW, Australia
| |
Collapse
|
13
|
Domowicz MS, Chan WC, Claudio-Vázquez P, Henry JG, Ware CB, Andrade J, Dawson G, Schwartz NB. Global Brain Transcriptome Analysis of a Tpp1 Neuronal Ceroid Lipofuscinoses Mouse Model. ASN Neuro 2020; 11:1759091419843393. [PMID: 31003587 PMCID: PMC6475859 DOI: 10.1177/1759091419843393] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In humans, homozygous mutations in the TPP1 gene results in loss
of tripeptidyl peptidase 1 (TPP1) enzymatic activity, leading to late infantile
neuronal ceroid lipofuscinoses disease. Using a mouse model that targets the
Tpp1 gene and recapitulates the pathology and clinical
features of the human disease, we analyzed end-stage (4 months) transcriptional
changes associated with lack of TPP1 activity. Using RNA sequencing technology,
Tpp1 expression changes in the forebrain/midbrain and
cerebellum of 4-month-old homozygotes were compared with strain-related
controls. Transcriptional changes were found in 510 and 1,550 gene transcripts
in forebrain/midbrain and cerebellum, respectively, from
Tpp1-deficient brain tissues when compared with age-matched
controls. Analysis of the differentially expressed genes using the Ingenuity™
pathway software, revealed increased neuroinflammation activity in microglia and
astrocytes that could lead to neuronal dysfunction, particularly in the
cerebellum. We also observed upregulation in the production of nitric oxide and
reactive oxygen species; activation of leukocyte extravasation signals and
complement pathways; and downregulation of major transcription factors involved
in control of circadian rhythm. Several of these expression changes were
confirmed by independent quantitative polymerase chain reaction and histological
analysis by mRNA in situ hybridization, which allowed for an
in-depth anatomical analysis of the pathology and provided independent
confirmation of at least two of the major networks affected in this model. The
identification of differentially expressed genes has revealed new lines of
investigation for this complex disorder that may lead to novel therapeutic
targets.
Collapse
Affiliation(s)
- Miriam S Domowicz
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Wen-Ching Chan
- 2 Center for Research Informatics, Biological Sciences Division, The University of Chicago, IL, USA
| | | | - Judith G Henry
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Christopher B Ware
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Jorge Andrade
- 2 Center for Research Informatics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Glyn Dawson
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| | - Nancy B Schwartz
- 1 Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA.,3 Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, IL, USA
| |
Collapse
|
14
|
Deniz AAH, Abdik EA, Abdik H, Aydın S, Şahin F, Taşlı PN. Zooming in across the Skin: A Macro-to-Molecular Panorama. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1247:157-200. [DOI: 10.1007/5584_2019_442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Abstract
Proteoglycans are diverse, complex extracellular/cell surface macromolecules composed of a central core protein with covalently linked glycosaminoglycan (GAG) chains; both of these components contribute to the growing list of important bio-active functions attributed to proteoglycans. Increasingly, attention has been paid to the roles of proteoglycans in nervous tissue development due to their highly regulated spatio/temporal expression patterns, whereby they promote/inhibit neurite outgrowth, participate in specification and maturation of various precursor cell types, and regulate cell behaviors like migration, axonal pathfinding, synaptogenesis and plasticity. These functions emanate from both the environments proteoglycans create around cells by retaining ions and water or serving as scaffolds for cell shaping or motility, and from dynamic interactions that modulate signaling fields for cytokines, growth factors and morphogens, which may bind to either the protein or GAG portions. Also, genetic abnormalities impacting proteoglycan synthesis during critical steps of brain development and response to environmental insults and injuries, as well as changes in microenvironment interactions leading to tumors in the central nervous system, all suggest roles for proteoglycans in behavioral and intellectual disorders and malignancies.
Collapse
Affiliation(s)
- Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA.,Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, IL, USA
| | - Miriam S Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| |
Collapse
|
16
|
George N, Geller HM. Extracellular matrix and traumatic brain injury. J Neurosci Res 2018; 96:573-588. [PMID: 29344975 DOI: 10.1002/jnr.24151] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/21/2017] [Accepted: 08/14/2017] [Indexed: 12/27/2022]
Abstract
The brain extracellular matrix (ECM) plays a crucial role in both the developing and adult brain by providing structural support and mediating cell-cell interactions. In this review, we focus on the major constituents of the ECM and how they function in both normal and injured brain, and summarize the changes in the composition of the ECM as well as how these changes either promote or inhibit recovery of function following traumatic brain injury (TBI). Modulation of ECM composition to facilitates neuronal survival, regeneration and axonal outgrowth is a potential therapeutic target for TBI treatment.
Collapse
Affiliation(s)
- Naijil George
- Laboratory of Developmental Neurobiology, Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20892-1603, USA
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20892-1603, USA
| |
Collapse
|
17
|
Li IMH, Liu K, Neal A, Clegg PD, De Val S, Bou-Gharios G. Differential tissue specific, temporal and spatial expression patterns of the Aggrecan gene is modulated by independent enhancer elements. Sci Rep 2018; 8:950. [PMID: 29343853 PMCID: PMC5772622 DOI: 10.1038/s41598-018-19186-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023] Open
Abstract
The transcriptional mechanism through which chondrocytes control the spatial and temporal composition of the cartilage tissue has remained largely elusive. The central aim of this study was to identify whether transcriptional enhancers played a role in the organisation of the chondrocytes in cartilaginous tissue. We focused on the Aggrecan gene (Acan) as it is essential for the normal structure and function of cartilage and it is expressed developmentally in different stages of chondrocyte maturation. Using transgenic reporter studies in mice we identified four elements, two of which showed individual chondrocyte developmental stage specificity. In particular, one enhancer (-80) distinguishes itself from the others by being predominantly active in adult cartilage. Furthermore, the -62 element uniquely drove reporter activity in early chondrocytes. The remaining chondrocyte specific enhancers, +28 and -30, showed no preference to chondrocyte type. The transcription factor SOX9 interacted with all the enhancers in vitro and mutation of SOX9 binding sites in one of the enhancers (-30) resulted in a loss of its chondrocyte specificity and ectopic enhancer reporter activity. Thus, the Acan enhancers orchestrate the precise spatiotemporal expression of this gene in cartilage types at different stages of development and adulthood.
Collapse
Affiliation(s)
- Ian M H Li
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Ke Liu
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Alice Neal
- Ludwig Cancer Research Ltd, University of Oxford, Oxford, UK
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Sarah De Val
- Ludwig Cancer Research Ltd, University of Oxford, Oxford, UK
| | - George Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK.
| |
Collapse
|
18
|
Domowicz M, Wadlington NL, Henry JG, Diaz K, Munoz MJ, Schwartz NB. Glial cell responses in a murine multifactorial perinatal brain injury model. Brain Res 2017; 1681:52-63. [PMID: 29274879 DOI: 10.1016/j.brainres.2017.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/16/2017] [Accepted: 12/17/2017] [Indexed: 12/18/2022]
Abstract
The impact of traumatic brain injury during the perinatal period, which coincides with glial cell (astrocyte and oligodendrocyte) maturation was assessed to determine whether a second insult, e.g., increased inflammation due to remote bacterial exposure, exacerbates the initial injury's effects, possibly eliciting longer-term brain damage. Thus, a murine multifactorial injury model incorporating both mechanisms consisting of perinatal penetrating traumatic brain injury, with or without intraperitoneal injection of lipopolysaccharide (LPS), an analog of remote pathogen exposure has been developed. Four days after injury, gene expression changes for different cell markers were assessed using mRNA in situ hybridization (ISH) and qPCR. Astrocytic marker mRNA levels increased in the stab-alone and stab-plus-LPS treated animals indicating reactive gliosis. Activated microglial/macrophage marker levels, increased in the ipsilateral sides of stab and stab-plus LPS animals by P10, but the differences resolved by P15. Ectopic expression of glial precursor and neural stem cell markers within the cortical injury site was observed by ISH, suggesting that existing precursors and neural stem cells migrate into the injured areas to replace the cells lost in the injury process. Furthermore, single exposure to LPS concomitant with acute stab injury affected the oligodendrocyte population in both the injured and contralateral uninjured side, indicating that after compromise of the blood-brain barrier integrity, oligodendrocytes become even more susceptible to inflammatory injury. This multifactorial approach should lead to a better understanding of the pathogenic sequelae observed as a consequence of perinatal brain insult/injury, caused by combinations of trauma, intrauterine infection, hypoxia and/or ischemia in humans.
Collapse
Affiliation(s)
- Miriam Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| | - Natasha L Wadlington
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Judith G Henry
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Kasandra Diaz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Miranda J Munoz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, USA; Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
The ADAMTS hyalectanase family: biological insights from diverse species. Biochem J 2017; 473:2011-22. [PMID: 27407170 DOI: 10.1042/bcj20160148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/29/2016] [Indexed: 12/13/2022]
Abstract
The a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs (ADAMTS) family of metzincins are complex secreted proteins that have diverse functions during development. The hyalectanases (ADAMTS1, 4, 5, 8, 9, 15 and 20) are a subset of this family that have enzymatic activity against hyalectan proteoglycans, the processing of which has important implications during development. This review explores the evolution, expression and developmental functions of the ADAMTS family, focusing on the ADAMTS hyalectanases and their substrates in diverse species. This review gives an overview of how the family and their substrates evolved from non-vertebrates to mammals, the expression of the hyalectanases and substrates in different species and their functions during development, and how these functions are conserved across species.
Collapse
|
20
|
Kim SY, Porter BE, Friedman A, Kaufer D. A potential role for glia-derived extracellular matrix remodeling in postinjury epilepsy. J Neurosci Res 2016; 94:794-803. [PMID: 27265805 DOI: 10.1002/jnr.23758] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 01/04/2023]
Abstract
Head trauma and vascular injuries are known risk factors for acquired epilepsy. The sequence of events that lead from the initial injury to the development of epilepsy involves complex plastic changes and circuit rewiring. In-depth, comprehensive understanding of the epileptogenic process is critical for the identification of disease-modifying targets. Here we review the complex interactions of cellular and extracellular components that may promote epileptogenesis, with an emphasis on the role of astrocytes. Emerging evidence demonstrates that astrocytes promptly respond to brain damage and play a critical role in the development of postinjury epilepsy. Astrocytes have been shown to regulate extracellular matrix (ECM) remodeling, which can affect plasticity and stability of synapses and, in turn, contribute to the epileptogenic process. From these separate lines of evidence, we present a hypothesis suggesting a possible role for astrocyte-regulated remodeling of ECM and perineuronal nets, a specialized ECM structure around fast-spiking inhibitory interneurons, in the development and progression of posttraumatic epilepsies. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Soo Young Kim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California
| | - Brenda E Porter
- Department of Neurology, Stanford University School of Medicine, Palo Alto, California
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California.,Canadian Institute for Advanced Research Program in Child and Brain Development, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Altieri SC, Zhao T, Jalabi W, Romito-DiGiacomo RR, Maricich SM. En1 is necessary for survival of neurons in the ventral nuclei of the lateral lemniscus. Dev Neurobiol 2016; 76:1266-1274. [PMID: 26914477 DOI: 10.1002/dneu.22388] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 02/03/2016] [Accepted: 02/18/2016] [Indexed: 11/06/2022]
Abstract
The ventral nuclei of the lateral lemniscus (VNLL) are part of the central auditory system thought to participate in temporal sound processing. While the timing and location of VNLL neurogenesis have been determined, the genetic factors that regulate VNLL neuron development are unknown. Here, we use genetic fate-mapping techniques to demonstrate that all glycinergic and glycinergic/GABAergic VNLL neurons derive from a cellular lineage that expresses the homeobox transcription factor Engrailed 1 (En1). We also show that En1 deletion does not affect migration or adoption of a neuronal cell fate but does lead to VNLL neuron death during development. Furthermore, En1 deletion blocks expression of the transcription factor FoxP1 in a subset of VNLL neurons. Together, these data identify En1 as a gene important for VNLL neuron development and survival. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1266-1274, 2016.
Collapse
Affiliation(s)
- Stefanie C Altieri
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15224
| | - Tianna Zhao
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15224
| | - Walid Jalabi
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, 44106
| | | | - Stephen M Maricich
- Richard King Mellon Institute for Pediatric Research, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, 15224. .,Childrens' Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, 15224.
| |
Collapse
|
22
|
En1 directs superior olivary complex neuron positioning, survival, and expression of FoxP1. Dev Biol 2015; 408:99-108. [PMID: 26542008 DOI: 10.1016/j.ydbio.2015.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/14/2015] [Accepted: 10/02/2015] [Indexed: 01/16/2023]
Abstract
Little is known about the genetic pathways and transcription factors that control development and maturation of central auditory neurons. En1, a gene expressed by a subset of developing and mature superior olivary complex (SOC) cells, encodes a homeodomain transcription factor important for neuronal development in the midbrain, cerebellum, hindbrain and spinal cord. Using genetic fate-mapping techniques, we show that all En1-lineal cells in the SOC are neurons and that these neurons are glycinergic, cholinergic and GABAergic in neurotransmitter phenotype. En1 deletion does not interfere with specification or neural fate of these cells, but does cause aberrant positioning and subsequent death of all En1-lineal SOC neurons by early postnatal ages. En1-null cells also fail to express the transcription factor FoxP1, suggesting that FoxP1 lies downstream of En1. Our data define important roles for En1 in the development and maturation of a diverse group of brainstem auditory neurons.
Collapse
|
23
|
Translational control of myelin basic protein expression by ERK2 MAP kinase regulates timely remyelination in the adult brain. J Neurosci 2015; 35:7850-65. [PMID: 25995471 DOI: 10.1523/jneurosci.4380-14.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Successful myelin repair in the adult CNS requires the robust and timely production of myelin proteins to generate new myelin sheaths. The underlying regulatory mechanisms and complex molecular basis of myelin regeneration, however, remain poorly understood. Here, we investigate the role of ERK MAP kinase signaling in this process. Conditional deletion of Erk2 from cells of the oligodendrocyte lineage resulted in delayed remyelination following demyelinating injury to the adult mouse corpus callosum. The delayed repair occurred as a result of a specific deficit in the translation of the major myelin protein, MBP. In the absence of ERK2, activation of the ribosomal protein S6 kinase (p70S6K) and its downstream target, ribosomal protein S6 (S6RP), was impaired at a critical time when premyelinating oligodendrocytes were transitioning to mature cells capable of generating new myelin sheaths. Thus, we have described an important link between the ERK MAP kinase signaling cascade and the translational machinery specifically in remyelinating oligodendrocytes in vivo. These results suggest an important role for ERK2 in the translational control of MBP, a myelin protein that appears critical for ensuring the timely generation of new myelin sheaths following demyelinating injury in the adult CNS.
Collapse
|
24
|
Faissner A, Reinhard J. The extracellular matrix compartment of neural stem and glial progenitor cells. Glia 2015; 63:1330-49. [DOI: 10.1002/glia.22839] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/25/2015] [Accepted: 03/30/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| |
Collapse
|
25
|
Hippert C, Graca AB, Barber AC, West EL, Smith AJ, Ali RR, Pearson RA. Müller glia activation in response to inherited retinal degeneration is highly varied and disease-specific. PLoS One 2015; 10:e0120415. [PMID: 25793273 PMCID: PMC4368159 DOI: 10.1371/journal.pone.0120415] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/22/2015] [Indexed: 12/20/2022] Open
Abstract
Despite different aetiologies, most inherited retinal disorders culminate in photoreceptor loss, which induces concomitant changes in the neural retina, one of the most striking being reactive gliosis by Müller cells. It is typically assumed that photoreceptor loss leads to an upregulation of glial fibrilliary acidic protein (Gfap) and other intermediate filament proteins, together with other gliosis-related changes, including loss of integrity of the outer limiting membrane (OLM) and deposition of proteoglycans. However, this is based on a mix of both injury-induced and genetic causes of photoreceptor loss. There are very few longitudinal studies of gliosis in the retina and none comparing these changes across models over time. Here, we present a comprehensive spatiotemporal assessment of features of gliosis in the degenerating murine retina that involves Müller glia. Specifically, we assessed Gfap, vimentin and chondroitin sulphate proteoglycan (CSPG) levels and outer limiting membrane (OLM) integrity over time in four murine models of inherited photoreceptor degeneration that encompass a range of disease severities (Crb1rd8/rd8, Prph2+/Δ307, Rho-/-, Pde6brd1/rd1). These features underwent very different changes, depending upon the disease-causing mutation, and that these changes are not correlated with disease severity. Intermediate filament expression did indeed increase with disease progression in Crb1rd8/rd8 and Prph2+/Δ307, but decreased in the Prph2+/Δ307 and Pde6brd1/rd1 models. CSPG deposition usually, but not always, followed the trends in intermediate filament expression. The OLM adherens junctions underwent significant remodelling in all models, but with differences in the composition of the resulting junctions; in Rho-/- mice, the adherens junctions maintained the typical rod-Müller glia interactions, while in the Pde6brd1/rd1 model they formed predominantly between Müller cells in late stage of degeneration. Together, these results show that gliosis and its associated processes are variable and disease-dependent.
Collapse
Affiliation(s)
- Claire Hippert
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Anna B. Graca
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Amanda C. Barber
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Emma L. West
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Alexander J. Smith
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
| | - Robin R. Ali
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, City Road, London, EC1V 2PD, United Kingdom
| | - Rachael A. Pearson
- Department of Genetics, University College London Institute of Ophthalmology, 11–43 Bath Street, London, EC1V 9EL, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Agarwal R, Domowicz MS, Schwartz NB, Henry J, Medintz I, Delehanty JB, Stewart MH, Susumu K, Huston AL, Deschamps JR, Dawson PE, Palomo V, Dawson G. Delivery and tracking of quantum dot peptide bioconjugates in an intact developing avian brain. ACS Chem Neurosci 2015; 6:494-504. [PMID: 25688887 DOI: 10.1021/acschemneuro.5b00022] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Luminescent semiconductor ∼9.5 nm nanoparticles (quantum dots: QDs) have intrinsic physiochemical and optical properties which enable us to begin to understand the mechanisms of nanoparticle mediated chemical/drug delivery. Here, we demonstrate the ability of CdSe/ZnS core/shell QDs surface functionalized with a zwitterionic compact ligand to deliver a cell-penetrating lipopeptide to the developing chick embryo brain without any apparent toxicity. Functionalized QDs were conjugated to the palmitoylated peptide WGDap(Palmitoyl)VKIKKP9GGH6, previously shown to uniquely facilitate endosomal escape, and microinjected into the embryonic chick spinal cord canal at embryo day 4 (E4). We were subsequently able to follow the labeling of spinal cord extension into the ventricles, migratory neuroblasts, maturing brain cells, and complex structures such as the choroid plexus. QD intensity extended throughout the brain, and peaked between E8 and E11 when fluorescence was concentrated in the choroid plexus before declining to hatching (E21/P0). We observed no abnormalities in embryonic patterning or embryo survival, and mRNA in situ hybridization confirmed that, at key developmental stages, the expression pattern of genes associated with different brain cell types (brain lipid binding protein, Sox-2, proteolipid protein and Class III-β-Tubulin) all showed a normal labeling pattern and intensity. Our findings suggest that we can use chemically modified QDs to identify and track neural stem cells as they migrate, that the choroid plexus clears these injected QDs/nanoparticles from the brain after E15, and that they can deliver drugs and peptides to the developing brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Philip E. Dawson
- Scripps Research Institute, La
Jolla, California 92037, United States
| | - Valle Palomo
- Scripps Research Institute, La
Jolla, California 92037, United States
| | - Glyn Dawson
- Departments of Pediatrics, Biochemistry
and Molecular Biology, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
27
|
Cortes M, Cortes LK, Schwartz NB. Mapping proteoglycan functions with glycosidases. Methods Mol Biol 2015; 1229:443-55. [PMID: 25325971 DOI: 10.1007/978-1-4939-1714-3_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The intrinsic and extrinsic factors that contribute to stem and neuronal precursor cell maintenance and/or differentiation remain poorly understood. Proteoglycans, major residents of the stem cell microenvironment, modulate key signaling cues and are of particular importance. We have taken a loss-of-function approach, by developing a library of bacterial lyases and sulfatases to specifically remodel the ECM and test the functional role of glycosaminoglycans (GAGs) in cell self-renewal, maintenance, and differentiation.
Collapse
Affiliation(s)
- Mauricio Cortes
- Department of Pediatrics, University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | | | | |
Collapse
|
28
|
Lauing KL, Cortes M, Domowicz MS, Henry JG, Baria AT, Schwartz NB. Aggrecan is required for growth plate cytoarchitecture and differentiation. Dev Biol 2014; 396:224-36. [PMID: 25446537 DOI: 10.1016/j.ydbio.2014.10.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 10/10/2014] [Accepted: 10/12/2014] [Indexed: 01/28/2023]
Abstract
The proteoglycan aggrecan is a prominent component of the extracellular matrix in growth plate cartilage. A naturally occurring, recessive, perinatally lethal mutation in the aggrecan core protein gene, cmd(bc) (Acan(cmd-Bc)), that deletes the entire protein-coding sequence provided a model in which to characterize the phenotypic and morphologic effects of aggrecan deletion on skeletal development. We also generated a novel transgenic mouse, Tg(COL2A1-ACAN), that has the chick ACAN coding sequence driven by the mouse COL2A1 promoter to enable the production of cmd(bc)/cmd(bc); Tg(COL2A1-ACAN) rescue embryos. These were used to assess the impact of aggrecan on growth plate organization, chondrocyte survival and proliferation, and the expression of mRNAs encoding chondrocyte differentiation markers and growth factors. Homozygous mutant (cmd(bc)/cmd(bc)) embryos exhibited severe defects in all skeletal elements with deformed and shortened (50%) limb elements. Expression of aggrecan in rescue embryos reversed the skeletal defects to varying degrees with a 20% increase in limb element length and near-full reversal (80%) of size and diameter of the ribcage and vertebrae. Aggrecan-null growth plates were devoid of matrix and lacked chondrocyte organization and differentiation, while those of the rescue embryos exhibited matrix production concomitant with partial zonation of chondrocytes having proliferative and hypertrophic morphologies. Deformation of the trachea, likely the cause of the mutation's lethality, was reduced in the rescue embryos. Aggrecan-null embryos also had abnormal patterns of COL10A1, SOX9, IHH, PTCH1, and FGFR3 mRNA expression in the growth plate. Expression of chick aggrecan in the rescue embryos notably increased COLX expression, accompanied by the reappearance of a hypertrophic zone and IHH expression. Significantly, in transgenic rescue embryos, the cell death and decreased proliferation phenotypes exhibited by the mutants were reversed; both were restored to wild-type levels. These findings suggest that aggrecan has a major role in regulating the expression of key growth factors and signaling molecules during development of cartilaginous tissue and is essential for proper chondrocyte organization, morphology, and survival during embryonic limb development.
Collapse
Affiliation(s)
- Kristen L Lauing
- Department of Pediatrics, The University of Chicago, Chicago IL 60637, USA
| | - Mauricio Cortes
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Miriam S Domowicz
- Department of Pediatrics, The University of Chicago, Chicago IL 60637, USA
| | - Judith G Henry
- Department of Pediatrics, The University of Chicago, Chicago IL 60637, USA
| | - Alexis T Baria
- Department of Pediatrics, The University of Chicago, Chicago IL 60637, USA
| | - Nancy B Schwartz
- Department of Pediatrics, The University of Chicago, Chicago IL 60637, USA; Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
29
|
Schwartz NB, Domowicz MS. Chemistry and Function of Glycosaminoglycans in the Nervous System. ADVANCES IN NEUROBIOLOGY 2014; 9:89-115. [DOI: 10.1007/978-1-4939-1154-7_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
30
|
Eid T, Tu N, Lee TSW, Lai JCK. Regulation of astrocyte glutamine synthetase in epilepsy. Neurochem Int 2013; 63:670-81. [PMID: 23791709 DOI: 10.1016/j.neuint.2013.06.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 05/28/2013] [Accepted: 06/08/2013] [Indexed: 11/20/2022]
Abstract
Astrocytes play a crucial role in regulating and maintaining the extracellular chemical milieu of the central nervous system under physiological conditions. Moreover, proliferation of phenotypically altered astrocytes (a.k.a. reactive astrogliosis) has been associated with many neurologic and psychiatric disorders, including mesial temporal lobe epilepsy (MTLE). Glutamine synthetase (GS), which is found in astrocytes, is the only enzyme known to date that is capable of converting glutamate and ammonia to glutamine in the mammalian brain. This reaction is important, because a continuous supply of glutamine is necessary for the synthesis of glutamate and GABA in neurons. The known stoichiometry of glutamate transport across the astrocyte plasma membrane also suggests that rapid metabolism of intracellular glutamate via GS is a prerequisite for efficient glutamate clearance from the extracellular space. Several studies have indicated that the activity of GS in astrocytes is diminished in several brain disorders, including MTLE. It has been hypothesized that the loss of GS activity in MTLE leads to increased extracellular glutamate concentrations and epileptic seizures. Understanding the mechanisms by which GS is regulated may lead to novel therapeutic approaches to MTLE, which is frequently refractory to antiepileptic drugs. This review discusses several known mechanisms by which GS expression and function are influenced, from transcriptional control to enzyme modification.
Collapse
Affiliation(s)
- Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, United States.
| | | | | | | |
Collapse
|
31
|
Katsman D, Stackpole EJ, Domin DR, Farber DB. Embryonic stem cell-derived microvesicles induce gene expression changes in Müller cells of the retina. PLoS One 2012; 7:e50417. [PMID: 23226281 PMCID: PMC3511553 DOI: 10.1371/journal.pone.0050417] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/19/2012] [Indexed: 12/20/2022] Open
Abstract
Cell-derived microvesicles (MVs), recognized as important components of cell-cell communication, contain mRNAs, miRNAs, proteins and lipids and transfer their bioactive contents from parent cells to cells of other origins. We have studied the effect that MVs released from embryonic stem cells (ESMVs) have on retinal progenitor Müller cells. Cultured human Müller cells were exposed to mouse ESMVs every 48 hours for a total of 9 treatments. Morphological changes were observed by light microscopy in the treated cells, which grew as individual heterogeneous cells, compared to the uniform, spindle-like adherent cellular sheets of untreated cells. ESMVs transferred to Müller cells embryonic stem cell (ESC) mRNAs involved in the maintenance of pluripotency, including Oct4 and Sox2, and the miRNAs of the 290 cluster, important regulators of the ESC-specific cell cycle. Moreover, ESMV exposure induced up-regulation of the basal levels of endogenous human Oct4 mRNA in Müller cells. mRNA and miRNA microarrays of ESMV-treated vs. untreated Müller cells revealed the up-regulation of genes and miRNAs involved in the induction of pluripotency, cellular proliferation, early ocular genes and genes important for retinal protection and remodeling, as well as the down-regulation of inhibitory and scar-related genes and miRNAs involved in differentiation and cell cycle arrest. To further characterize the heterogeneous cell population of ESMV-treated Müller cells, their expression of retinal cell markers was compared to that in untreated control cells by immunocytochemistry. Markers for amacrine, ganglion and rod photoreceptors were present in treated but not in control Müller cells. Together, our findings indicate that ESMs induce de-differentiation and pluripotency in their target Müller cells, which may turn on an early retinogenic program of differentiation.
Collapse
Affiliation(s)
- Diana Katsman
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Emma J. Stackpole
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Daniel R. Domin
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Debora B. Farber
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
De Cegli R, Iacobacci S, Flore G, Gambardella G, Mao L, Cutillo L, Lauria M, Klose J, Illingworth E, Banfi S, di Bernardo D. Reverse engineering a mouse embryonic stem cell-specific transcriptional network reveals a new modulator of neuronal differentiation. Nucleic Acids Res 2012. [PMID: 23180766 PMCID: PMC3553984 DOI: 10.1093/nar/gks1136] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gene expression profiles can be used to infer previously unknown transcriptional regulatory interaction among thousands of genes, via systems biology ‘reverse engineering’ approaches. We ‘reverse engineered’ an embryonic stem (ES)-specific transcriptional network from 171 gene expression profiles, measured in ES cells, to identify master regulators of gene expression (‘hubs’). We discovered that E130012A19Rik (E13), highly expressed in mouse ES cells as compared with differentiated cells, was a central ‘hub’ of the network. We demonstrated that E13 is a protein-coding gene implicated in regulating the commitment towards the different neuronal subtypes and glia cells. The overexpression and knock-down of E13 in ES cell lines, undergoing differentiation into neurons and glia cells, caused a strong up-regulation of the glutamatergic neurons marker Vglut2 and a strong down-regulation of the GABAergic neurons marker GAD65 and of the radial glia marker Blbp. We confirmed E13 expression in the cerebral cortex of adult mice and during development. By immuno-based affinity purification, we characterized protein partners of E13, involved in the Polycomb complex. Our results suggest a role of E13 in regulating the division between glutamatergic projection neurons and GABAergic interneurons and glia cells possibly by epigenetic-mediated transcriptional regulation.
Collapse
Affiliation(s)
- Rossella De Cegli
- Telethon Institute of Genetics and Medicine, Via P. Castellino 111, Napoli 80131, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wiese S, Karus M, Faissner A. Astrocytes as a source for extracellular matrix molecules and cytokines. Front Pharmacol 2012; 3:120. [PMID: 22740833 PMCID: PMC3382726 DOI: 10.3389/fphar.2012.00120] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 06/06/2012] [Indexed: 12/19/2022] Open
Abstract
Research of the past 25 years has shown that astrocytes do more than participating and building up the blood-brain barrier and detoxify the active synapse by reuptake of neurotransmitters and ions. Indeed, astrocytes express neurotransmitter receptors and, as a consequence, respond to stimuli. Within the tripartite synapse, the astrocytes owe more and more importance. Besides the functional aspects the differentiation of astrocytes has gained a more intensive focus. Deeper knowledge of the differentiation processes during development of the central nervous system might help explaining and even help treating neurological diseases like Alzheimer’s disease, Amyotrophic lateral sclerosis, Parkinsons disease, and psychiatric disorders in which astrocytes have been shown to play a role. Specific differentiation of neural stem cells toward the astroglial lineage is performed as a multi-step process. Astrocytes and oligodendrocytes develop from a multipotent stem cell that prior to this has produced primarily neuronal precursor cells. This switch toward the more astroglial differentiation is regulated by a change in receptor composition on the cell surface and responsiveness to Fibroblast growth factor and Epidermal growth factor (EGF). The glial precursor cell is driven into the astroglial direction by signaling molecules like Ciliary neurotrophic factor, Bone Morphogenetic Proteins, and EGF. However, the early astrocytes influence their environment not only by releasing and responding to diverse soluble factors but also express a wide range of extracellular matrix (ECM) molecules, in particular proteoglycans of the lectican family and tenascins. Lately these ECM molecules have been shown to participate in glial development. In this regard, especially the matrix protein Tenascin C (Tnc) proved to be an important regulator of astrocyte precursor cell proliferation and migration during spinal cord development. Nevertheless, ECM molecules expressed by reactive astrocytes are also known to act mostly in an inhibitory fashion under pathophysiological conditions. Thus, we further summarize resent data concerning the role of chondroitin sulfate proteoglycans and Tnc under pathological conditions.
Collapse
Affiliation(s)
- Stefan Wiese
- Group for Molecular Cell Biology, Department for Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany
| | | | | |
Collapse
|
34
|
Abstract
Aggrecan is a large proteoglycan that plays roles in numerous tissues during vertebrate development and adult life. The 6,327-nt chick aggrecan coding sequence had been determined from overlapping clones, but a full-length cDNA, needed for use in transgenic expression studies, had not been constructed. The strategy employed to do so was to generate two overlapping cDNA subfragments that shared a unique restriction site in the overlap and then join them at that site. These subfragments were obtained and cloned into the TOPO-TA vector pCR2.1. Digestion of the two constructs with the shared-site enzyme, XbaI, produced vector/5'-cDNA and 3'-cDNA fragments with XbaI-ends; these were ligated to produce the final full-length cDNA.
Collapse
Affiliation(s)
- Mauricio Cortes
- Departments of Pediatric, The University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
35
|
Lendvai D, Morawski M, Brückner G, Négyessy L, Baksa G, Glasz T, Patonay L, Matthews RT, Arendt T, Alpár A. Perisynaptic aggrecan-based extracellular matrix coats in the human lateral geniculate body devoid of perineuronal nets. J Neurosci Res 2011; 90:376-87. [PMID: 21959900 DOI: 10.1002/jnr.22761] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/05/2011] [Accepted: 07/08/2011] [Indexed: 02/02/2023]
Abstract
The extracellular matrix surrounds different neuronal compartments in the mature nervous system. In a variety of vertebrates, most brain regions are loaded with a distinct type of extracellular matrix around the somatodendritic part of neurons, termed perineuronal nets. The present study reports that chondrotin sulfate proteoglycan-based matrix is structured differently in the human lateral geniculate body. Using various chondrotin sulfate proteoglycan-based extracellular matrix antibodies, we show that perisomatic matrix labeling is rather weak or absent, whereas dendrites are contacted by axonal coats appearing as small, oval structures. Confocal laser scanning microscopy and electron microscopy demonstrated that these typical structures are associated with synaptic loci on dendrites. Using multiple labelings, we show that different chondrotin sulfate proteoglycan components of the extracellular matrix do not associate exclusively with neuronal structures but possibly associate with glial structures as well. Finally, we confirm and extend previous findings in primates that intensity differences of various extracellular matrix markers between magno- and parvocellular layers reflect functional segregation between these layers in the human lateral geniculate body.
Collapse
Affiliation(s)
- D Lendvai
- Department of Anatomy, Histology and Embryology, Semmelweis University Medical School, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Domowicz MS, Henry JG, Wadlington N, Navarro A, Kraig RP, Schwartz NB. Astrocyte precursor response to embryonic brain injury. Brain Res 2011; 1389:35-49. [PMID: 21396923 DOI: 10.1016/j.brainres.2011.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 02/23/2011] [Accepted: 03/02/2011] [Indexed: 02/06/2023]
Abstract
Penetrating traumatic insult during pregnancy is a leading cause of human fetal demise; in particular, trauma to the brain may lead to devastating long-term cognitive sequelae. Perinatal brain injury involves glial precursors, but the neural mechanisms controlling astrocyte ontogeny after injury remain incompletely understood, partly due to a lack of appropriate markers and animal models. We analyzed astrocyte precursor response to injury at the beginning (E11) and peak (E15) of gliogenesis in an avian tectal model of penetrating embryonic brain trauma, without confounding maternal and sibling effects. At both ages, lateral ventricular dilatation, necrotic foci, periventricular cysts and intraventricular hemorrhages were observed distal to stab wounds two days after a unilateral stab injury to optic tecta. Neuronal (TUBB3) and oligodendrocyte precursor (PLP) markers were down-regulated, even far-removed from the wound site. In contrast, the mature astrocyte marker, GFAP, was up-regulated at the wound site, around necrotic areas and cysts, plus in usual areas of GFAP expression. Increased inflammatory response and apoptotic cell death were also confirmed in the injured tecta. Increased expression of NFIA, SOX9 and GLAST at the wound site and in the ventricular zone (VZ) of the injured tecta indicated an astroglial precursor response. However, cell division increased in the VZ only in early (E11) injury, but not later (E15), indicating that in late injury the astrogliogenesis occurring after acute injury is predominantly due to precursor differentiation rather than precursor proliferation. The inability to replenish the glial precursor pool during the critical period of vulnerability to injury may be an important cause of subsequent developmental abnormalities.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Department of Pediatrics, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL 60637, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Béchade C, Pascual O, Triller A, Bessis A. Nitric oxide regulates astrocyte maturation in the hippocampus: involvement of NOS2. Mol Cell Neurosci 2011; 46:762-9. [PMID: 21354308 DOI: 10.1016/j.mcn.2011.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 02/16/2011] [Accepted: 02/18/2011] [Indexed: 11/27/2022] Open
Abstract
Neurons and astrocytes are generated sequentially from radial glia. Once neurogenesis is completed, radial glia starts to differentiate into immature astrocytes. Astrocytes then maturate and change their morphology and electrophysiological properties. Neurotrophic cytokines or bone morphogenetic proteins have been identified as inducers of the developmental switch from neurogenesis to astrogenesis. However, the factors and mechanisms regulating the late differentiation of radial glia and the subsequent astrocyte maturation are poorly described. We used two independent approaches to examine the role of nitric oxide (NO) in the process of astrogenesis and maturation of astrocytes. First using a pharmacological approach, we depleted NO from developing hippocampus by intraventricular injection of a specific scavenger. Then by a genetic approach, we analyzed a nitric oxide synthase-2 (NOS2) knockout mouse. In both models, we found that differentiation of RC2-positive radial glia into late GFAP-positive radial glia was impaired. The cell-fate analysis after incorporation of BrdU demonstrated that astrogenesis was not altered upon NOS2 deficiency. Maturation of astrocytes was assessed by electrophysiological recordings at P7 and functional analysis. In wild type, 20% of astrocytes were immature as shown by their non-linear I/V relationship and high membrane resistance, whereas in NOS2-/- hippocampus, 51% of the astrocytes displayed an immature profile. The reduced branching of astrocytes upon NOS2 deficiency and their low content in connexin-43 further confirmed their immature profile. Our results highlight a novel developmental role of NO and NOS2 in the differentiation of radial glia and the maturation of astrocytes.
Collapse
Affiliation(s)
- Catherine Béchade
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Inserm U1024, Paris F-75005, France
| | | | | | | |
Collapse
|
38
|
Ali SAM, Hosaka YZ, Uehara M. Spatiotemporal distribution of chondroitin sulfate proteoglycans in the developing mouse retina and optic nerve. J Vet Med Sci 2010; 73:13-8. [PMID: 20716860 DOI: 10.1292/jvms.10-0201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of the present study was to determine the distribution of chondroitin sulfate proteoglycans in the mouse retina and optic nerve of the prenatal and postnatal mouse by immunohistochemistry. At embryonic day (E) 18, chondroitin-4-sulfate (C4S), chondroitin-6-sulfate (C6S) and biglycan were detected in the retina and optic nerve. However, aggrecan was seen in the retina but not in the optic nerve. At postnatal day (P) 7, aggrecan and biglycan were clearly observed in the optic nerve, inner nuclear layer and ganglion cell layer and diffuse in the outer retina. C4S diffusely distributed in the retina and optic nerve, but C6S was mainly confined to the photoreceptor layer and optic nerve sheath. At P42, biglycan showed diffuse distribution in the retina and optic nerve with intense staining in nerve-fiber rich layers. Aggrecan showed weak staining at the inner plexiform layer with higher density in the outer and inner nuclear layers, outer plexiform layer and ganglion cell layer. Both C4S and C6S were detected in the optic nerve and retina, but C6S showed strong immunostaining in the photoreceptor layer. The distributions of these proteoglycans with respect of time course during development of the retina and optic nerve suggest that they may have unique or overlapping roles in development and maintenance of the retina and optic nerve.
Collapse
Affiliation(s)
- Safwat Ali Mohamed Ali
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Japan
| | | | | |
Collapse
|
39
|
Extrinsic and intrinsic factors controlling axonal regeneration after spinal cord injury. Expert Rev Mol Med 2009; 11:e37. [PMID: 19968910 DOI: 10.1017/s1462399409001288] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spinal cord injury is one of the most devastating conditions that affects the central nervous system. It can lead to permanent disability and there are around two million people affected worldwide. After injury, accumulation of myelin debris and formation of an inhibitory glial scar at the site of injury leads to a physical and chemical barrier that blocks axonal growth and regeneration. The mammalian central nervous system thus has a limited intrinsic ability to repair itself after injury. To improve axonal outgrowth and promote functional recovery, it is essential to identify the various intrinsic and extrinsic factors controlling regeneration and navigation of axons within the inhibitory environment of the central nervous system. Recent advances in spinal cord research have opened new avenues for the exploration of potential targets for repairing the cord and improving functional recovery after trauma. Here, we discuss some of the important key molecules that could be harnessed for repairing spinal cord injury.
Collapse
|
40
|
Hu Z, Li T, Zhang X, Chen Y. Hepatocyte growth factor enhances the generation of high-purity oligodendrocytes from human embryonic stem cells. Differentiation 2009; 78:177-84. [DOI: 10.1016/j.diff.2009.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Revised: 04/23/2009] [Accepted: 05/20/2009] [Indexed: 01/25/2023]
|
41
|
Cortes M, Baria AT, Schwartz NB. Sulfation of chondroitin sulfate proteoglycans is necessary for proper Indian hedgehog signaling in the developing growth plate. Development 2009; 136:1697-706. [PMID: 19369399 DOI: 10.1242/dev.030742] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In contrast to the functional role of heparan sulfate proteoglycans (HSPGs), the importance of chondroitin sulfate proteoglycans (CSPGs) in modulating signaling pathways involving hedgehog proteins, wingless-related proteins and fibroblast growth factors remains unclear. To elucidate the importance of sulfated CSPGs in signaling paradigms required for endochondral bone formation, the brachymorphic (bm) mouse was used as a model for undersulfated CSPGs. The bm mouse exhibits a postnatal chondrodysplasia caused by a mutation in the phosphoadenosine phosphosulfate (PAPS) synthetase (Papss2) gene, leading to reduced levels of PAPS and undersulfated proteoglycans. Biochemical analysis of the glycosaminoglycan (GAG) content in bm cartilage via sulfate labeling and fluorophore-assisted carbohydrate electrophoresis revealed preferential undersulfation of chondroitin chains (CS) and normal sulfation of heparan sulfate chains. In situ hybridization and immunohistochemical analysis of bm limb growth plates showed diminished Indian hedgehog (Ihh) signaling and abnormal Ihh protein distribution in the extracellular matrix. Consistent with the decrease in hedgehog signaling, BrdU incorporation exhibited a significant reduction in chondrocyte proliferation. Direct measurements of Ihh binding to defined GAG chains demonstrated that Ihh interacts with CS, particularly chondroitin-4-sulfate. Furthermore, co-immunoprecipitation experiments showed that Ihh binds to the major cartilage CSPG aggrecan via its CS chains. Overall, this study demonstrates an important function for CSPGs in modulating Ihh signaling in the developing growth plate, and highlights the importance of carbohydrate sulfation in regulating growth factor signaling.
Collapse
Affiliation(s)
- Mauricio Cortes
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
42
|
Dasgupta B, Milbrandt J. AMP-activated protein kinase phosphorylates retinoblastoma protein to control mammalian brain development. Dev Cell 2009; 16:256-70. [PMID: 19217427 DOI: 10.1016/j.devcel.2009.01.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 10/29/2008] [Accepted: 01/20/2009] [Indexed: 12/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved metabolic sensor that responds to alterations in cellular energy levels to maintain energy balance. While its role in metabolic homeostasis is well documented, its role in mammalian development is less clear. Here we demonstrate that mutant mice lacking the regulatory AMPK beta1 subunit have profound brain abnormalities. The beta1(-/-) mice show atrophy of the dentate gyrus and cerebellum, and severe loss of neurons, oligodendrocytes, and myelination throughout the central nervous system. These abnormalities stem from reduced AMPK activity, with ensuing cell cycle defects in neural stem and progenitor cells (NPCs). The beta1(-/-) NPC deficits result from hypophosphorylation of the retinoblastoma protein (Rb), which is directly phosphorylated by AMPK at Ser(804). The AMPK-Rb axis is utilized by both growth factors and energy restriction to increase NPC growth. Our results reveal that AMPK integrates growth factor signaling with cell cycle control to regulate brain development.
Collapse
Affiliation(s)
- Biplab Dasgupta
- Department of Pathology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
43
|
Domowicz MS, Cortes M, Henry JG, Schwartz NB. Aggrecan modulation of growth plate morphogenesis. Dev Biol 2009; 329:242-57. [PMID: 19268444 DOI: 10.1016/j.ydbio.2009.02.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2008] [Revised: 02/02/2009] [Accepted: 02/20/2009] [Indexed: 10/21/2022]
Abstract
Chick and mouse embryos with heritable deficiencies of aggrecan exhibit severe dwarfism and premature death, demonstrating the essential involvement of aggrecan in development. The aggrecan-deficient nanomelic (nm) chick mutant E12 fully formed growth plate (GP) is devoid of matrix and exhibits markedly altered cytoarchitecture, proliferative capacity, and degree of cell death. While differentiation of chondroblasts to pre-hypertrophic chondrocytes (IHH expression) is normal up to E6, the extended periosteum expression pattern of PTCH (a downstream effector of IHH) indicates altered propagation of IHH signaling, as well as accelerated down-regulation of FGFR3 expression, decreased BrdU incorporation and higher levels of ERK phosphorylation, all indicating early effects on FGF signaling. By E7 reduced IHH expression and premature expression of COL10A1 foreshadow the acceleration of hypertrophy observed at E12. By E8, exacerbated co-expression of IHH and COL10A1 lead to delayed separation and establishment of the two GPs in each element. By E9, increased numbers of cells express P-SMAD1/5/8, indicating altered BMP signaling. These results indicate that the IHH, FGF and BMP signaling pathways are altered from the very beginning of GP formation in the absence of aggrecan, thereby inducing premature hypertrophic chondrocyte maturation, leading to the nanomelic long bone growth disorder.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Departments of Pediatrics and Biochemistry and Molecular Biology, Committee on Developmental Biology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
44
|
Virgintino D, Perissinotto D, Girolamo F, Mucignat MT, Montanini L, Errede M, Kaneiwa T, Yamada S, Sugahara K, Roncali L, Perris R. Differential distribution of aggrecan isoforms in perineuronal nets of the human cerebral cortex. J Cell Mol Med 2009; 13:3151-73. [PMID: 19220578 PMCID: PMC4516474 DOI: 10.1111/j.1582-4934.2009.00694.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aggrecan is a component of the CNS extracellular matrix (ECM) and we show here that the three primary alternative spliced transcripts of the aggrecan gene found in cartilage are also present in the adult CNS. Using a unique panel of core protein-directed antibodies against human aggrecan we further show that different aggrecan isoforms are deposited in perineuronal nets (PNNs) and neuropil ECM of Brodmann’s area 6 of the human adult cerebral cortex. According to their distribution pattern, the identified cortical aggrecan isoforms were subdivided into five clusters spanning from cluster 1, comprised isoforms that appeared widespread throughout the cortex, to cluster 5, which was an aggrecan-free subset. Comparison of brain and cartilage tissues showed a different relative abundance of aggrecan isoforms, with cartilage-specific isoforms characterizing cluster 5, and PNN-associated isoforms lacking keratan sulphate chains. In the brain, isoforms of cluster 1 were disclosed in PNNs surrounding small-medium interneurons of layers II–V, small-medium pyramidal neurons of layers III and V and large interneurons of layer VI. Aggrecan PNNs enveloped both neuron bodies and neuronal processes, encompassing pre-terminal nerve fibres, synaptic boutons and terminal processes of glial cells and aggrecan was also observed in continuous ‘coats’ associated with satellite, neuron-associated cells of a putative glial nature. Immunolabelling for calcium-binding proteins and glutamate demonstrated that aggrecan PNNs were linked to defined subsets of cortical interneurons and pyramidal cells. We suggest that in the human cerebral cortex, discrete, layer-specific PNNs are assembled through the participation of selected aggrecan isoforms that characterize defined subsets of cortical neurons.
Collapse
Affiliation(s)
- Daniela Virgintino
- Department of Human Anatomy and Histology, University of Bari School of Medicine, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yow SZ, Quek CH, Yim EKF, Lim CT, Leong KW. Collagen-based fibrous scaffold for spatial organization of encapsulated and seeded human mesenchymal stem cells. Biomaterials 2008; 30:1133-42. [PMID: 19041132 DOI: 10.1016/j.biomaterials.2008.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 11/04/2008] [Indexed: 02/08/2023]
Abstract
Living tissues consist of groups of cells organized in a controlled manner to perform a specific function. Spatial distribution of cells within a three-dimensional matrix is critical for the success of any tissue-engineering construct. Fibers endowed with cell-encapsulation capability would facilitate the achievement of this objective. Here we report the synthesis of a cell-encapsulated fibrous scaffold by interfacial polyelectrolyte complexation (IPC) of methylated collagen and a synthetic terpolymer. The collagen component was well distributed in the fiber, which had a mean ultimate tensile strength of 244.6+/-43.0 MPa. Cultured in proliferating medium, human mesenchymal stem cells (hMSCs) encapsulated in the fibers showed higher proliferation rate than those seeded on the scaffold. Gene expression analysis revealed the maintenance of multipotency for both encapsulated and seeded samples up to 7 days as evidenced by Sox 9, CBFA-1, AFP, PPARgamma2, nestin, GFAP, collagen I, osteopontin and osteonectin genes. Beyond that, seeded hMSCs started to express neuronal-specific genes such as aggrecan and MAP2. The study demonstrates the appeal of IPC for scaffold design in general and the promise of collagen-based hybrid fibers for tissue engineering in particular. It lays the foundation for building fibrous scaffold that permits 3D spatial cellular organization and multi-cellular tissue development.
Collapse
Affiliation(s)
- S Z Yow
- Graduate Program in Bioengineering, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | | | | | | | | |
Collapse
|
46
|
King LA, Schwartz NB, Domowicz MS. Glial migratory streams in the developing hindbrain: a slice culture approach. J Neurosci Methods 2008; 177:30-43. [PMID: 18948137 DOI: 10.1016/j.jneumeth.2008.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 09/05/2008] [Accepted: 09/16/2008] [Indexed: 02/05/2023]
Abstract
Compared to our knowledge of neurogenesis, relatively little is known about glial cell specification and migration during central nervous system development. We have established a novel chick hindbrain slice preparation which permits examination of gliogenesis in its native environment, providing a means to study the signaling pathways involved in glial cell specification and migration during development. Cells in the hindbrain slice preparations mature in a manner which is similar to in vivo developmental timing and patterning paradigms. To demonstrate the utility of this approach, we examined the effect of the retinoic acid signaling pathway on cells in these slices, showing that addition of exogenous trans-retinoic acid to slice cultures promotes expression of a marker of mature astrocytes, glial fibrillary acidic protein (GFAP), while the inhibition of endogenous retinoic acid synthesis reduces GFAP expression; the results suggest a role for retinoic acid in modulating glial differentiation. Using these hindbrain slice cultures, we have used two different approaches to label glial progenitors specifically at the ventricular zone and have observed for the first time the ventrally-directed migration of these cells from the ventricular zone of the hindbrain. This slice culture system is thus an innovative and robust tool for examining glial cell migration and the extracellular molecular and signaling pathways which regulate glial differentiation.
Collapse
Affiliation(s)
- Leslie A King
- Department of Pediatrics, 5841 S. Maryland Avenue, MC 5058, The University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
47
|
Extracellular matrix of the central nervous system: from neglect to challenge. Histochem Cell Biol 2008; 130:635-53. [PMID: 18696101 DOI: 10.1007/s00418-008-0485-9] [Citation(s) in RCA: 306] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2008] [Indexed: 12/13/2022]
Abstract
The basic concept, that specialized extracellular matrices rich in hyaluronan, chondroitin sulfate proteoglycans (aggrecan, versican, neurocan, brevican, phosphacan), link proteins and tenascins (Tn-R, Tn-C) can regulate cellular migration and axonal growth and thus, actively participate in the development and maturation of the nervous system, has in recent years gained rapidly expanding experimental support. The swift assembly and remodeling of these matrices have been associated with axonal guidance functions in the periphery and with the structural stabilization of myelinated fiber tracts and synaptic contacts in the maturating central nervous system. Particular interest has been focused on the putative role of chondroitin sulfate proteoglycans in suppressing central nervous system regeneration after lesions. The axon growth inhibitory properties of several of these chondroitin sulfate proteoglycans in vitro, and the partial recovery of structural plasticity in lesioned animals treated with chondroitin sulfate degrading enzymes in vivo have significantly contributed to the increased awareness of this long time neglected structure.
Collapse
|
48
|
White RE, Jakeman LB. Don't fence me in: harnessing the beneficial roles of astrocytes for spinal cord repair. Restor Neurol Neurosci 2008; 26:197-214. [PMID: 18820411 PMCID: PMC2825119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Astrocytes comprise a heterogeneous cell population that plays a complex role in repair after spinal cord injury. Reactive astrocytes are major contributors to the glial scar that is a physical and chemical barrier to axonal regeneration. Yet, consistent with a supportive role in development, astrocytes secrete neurotrophic factors and protect neurons and glia spared by the injury. In development and after injury, local cues are modulators of astrocyte phenotype and function. When multipotent cells are transplanted into the injured spinal cord, they differentiate into astrocytes and other glial cells as opposed to neurons, which is commonly viewed as a challenge to be overcome in developing stem cell technology. However, several examples show that astrocytes provide support and guidance for axonal growth and aid in improving functional recovery after spinal cord injury. Notably, transplantation of astrocytes of a developmentally immature phenotype promotes tissue sparing and axonal regeneration. Furthermore, interventions that enhance endogenous astrocyte migration or reinvasion of the injury site result in greater axonal growth. These studies demonstrate that astrocytes are dynamic, diverse cells that have the capacity to promote axon growth after injury. The ability of astrocytes to be supportive of recovery should be exploited in devising regenerative strategies.
Collapse
Affiliation(s)
- Robin E. White
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA
- The Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - Lyn B. Jakeman
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA
- The Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| |
Collapse
|