51
|
Pre-Treatment with Laminarin Protects Hippocampal CA1 Pyramidal Neurons and Attenuates Reactive Gliosis Following Transient Forebrain Ischemia in Gerbils. Mar Drugs 2020; 18:md18010052. [PMID: 31940961 PMCID: PMC7024340 DOI: 10.3390/md18010052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
Transient brain ischemia triggers selective neuronal death/loss, especially in vulnerable regions of the brain including the hippocampus. Laminarin, a polysaccharide originating from brown seaweed, has various pharmaceutical properties including an antioxidant function. To the best of our knowledge, few studies have been conducted on the protective effects of laminarin against ischemic injury induced by ischemic insults. In this study, we histopathologically investigated the neuroprotective effects of laminarin in the Cornu Ammonis 1 (CA1) field of the hippocampus, which is very vulnerable to ischemia-reperfusion injury, following transient forebrain ischemia (TFI) for five minutes in gerbils. The neuroprotective effect was examined by cresyl violet staining, Fluoro-Jade B histofluorescence staining and immunohistochemistry for neuronal-specific nuclear protein. Additionally, to study gliosis (glial changes), we performed immunohistochemistry for glial fibrillary acidic protein to examine astrocytes, and ionized calcium-binding adaptor molecule 1 to examine microglia. Furthermore, we examined alterations in pro-inflammatory M1 microglia by using double immunofluorescence. Pretreatment with 10 mg/kg laminarin failed to protect neurons in the hippocampal CA1 field and did not attenuate reactive gliosis in the field following TFI. In contrast, pretreatment with 50 or 100 mg/kg laminarin protected neurons, attenuated reactive gliosis and reduced pro-inflammatory M1 microglia in the CA1 field following TFI. Based on these results, we firmly propose that 50 mg/kg laminarin can be strategically applied to develop a preventative against injuries following cerebral ischemic insults.
Collapse
|
52
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2020. [PMCID: PMC6954730 DOI: 10.1002/sctm.19-0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
53
|
Overview of Current Drug Delivery Methods Across the Blood-Brain Barrier for the Treatment of Primary Brain Tumors. CNS Drugs 2020; 34:1121-1131. [PMID: 32965590 PMCID: PMC7658069 DOI: 10.1007/s40263-020-00766-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2020] [Indexed: 01/05/2023]
Abstract
Existing drug delivery methods have not led to a significant increase in survival for patients with malignant primary brain tumors. While the combination of conventional therapies consisting of surgery, radiotherapy, and chemotherapy has improved survival for some types of brain tumors (e.g., WNT medulloblastoma), other types of brain tumors (e.g., glioblastoma and diffuse midline glioma) still have a poor prognosis. The reason for the differences in response can be largely attributed to the blood-brain barrier (BBB), a specialized structure at the microvasculature level that regulates the transport of molecules across the blood vessels into the brain parenchyma. This structure hampers the delivery of most chemotherapeutic agents for the treatment of primary brain tumors. Several drug delivery methods such as nanoparticles, convection enhanced delivery, focused ultrasound, intranasal delivery, and intra-arterial delivery have been developed to overcome the BBB in primary brain tumors. However, prognosis of most primary brain tumors still remains poor. The heterogeneity of the BBB in primary brain tumors and the distinct vasculature of tumors make it difficult to design a drug delivery method that targets the entire tumor. Drug delivery methods that combine strategies such as focused ultrasound and nanoparticles might be a more successful approach. However, more research is needed to optimize and develop new drug delivery techniques to improve survival of patients with primary brain tumors.
Collapse
|
54
|
Miccoli B, Braeken D, Li YCE. Brain-on-a-chip Devices for Drug Screening and Disease Modeling Applications. Curr Pharm Des 2019; 24:5419-5436. [PMID: 30806304 DOI: 10.2174/1381612825666190220161254] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
Neurodegenerative disorders are related to the progressive functional loss of the brain, often connected to emotional and physical disability and, ultimately, to death. These disorders, strongly connected to the aging process, are becoming increasingly more relevant due to the increase of life expectancy. Current pharmaceutical treatments poorly tackle these diseases, mainly acting only on their symptomology. One of the main reasons of this is the current drug development process, which is not only expensive and time-consuming but, also, still strongly relies on animal models at the preclinical stage. Organ-on-a-chip platforms have the potential to strongly impact and improve the drug screening process by recreating in vitro the functionality of human organs. Patient-derived neurons from different regions of the brain can be directly grown and differentiated on a brain-on-a-chip device where the disease development, progression and pharmacological treatments can be studied and monitored in real time. The model reliability is strongly improved by using human-derived cells, more relevant than animal models for pharmacological screening and disease monitoring. The selected cells will be then capable of proliferating and organizing themselves in the in vivo environment thanks to the device architecture, materials selection and bio-chemical functionalization. In this review, we start by presenting the fundamental strategies adopted for brain-on-a-chip devices fabrication including e.g., photolithography, micromachining and 3D printing technology. Then, we discuss the state-of-theart of brain-on-a-chip platforms including their role in the study of the functional architecture of the brain e.g., blood-brain barrier, or of the most diffuse neurodegenerative diseases like Alzheimer's and Parkinson's. At last, the current limitations and future perspectives of this approach for the development of new drugs and neurodegenerative diseases modeling will be discussed.
Collapse
Affiliation(s)
- Beatrice Miccoli
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium.,Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Dries Braeken
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium
| | - Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung City, Taiwan
| |
Collapse
|
55
|
Paulo JA, Gygi SP. mTMT: An Alternative, Nonisobaric, Tandem Mass Tag Allowing for Precursor-Based Quantification. Anal Chem 2019; 91:12167-12172. [PMID: 31490667 DOI: 10.1021/acs.analchem.9b03162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Stable isotope labeling of peptides is the basis for numerous mass-spectrometry-based quantification strategies. Isobaric tagging and metabolic labeling, namely, tandem mass tagging (TMT) and SILAC, are among the most widely used techniques for relative protein quantification. Here we report an alternative, precursor-based quantification method using nonisobaric TMT variants: TMTzero (TMT0) and superheavy TMT (shTMT). We term this strategy mass difference tandem mass tagging (mTMT). These TMT variants differ by 11 mass units; however, peptides labeled with these reagents coelute, analogous to SILAC-labeled peptide pairs. As a proof-of-concept, we profiled the proteomes of two cell lines that are frequently used in neuroscience studies, SH-SY5Y and SVGp12, using mTMT and standard SILAC-labeling approaches. We show similar quantified proteins and peptides for each method, with highly correlated fold-changes between workflows. We conclude that mTMT is a suitable alternative for precursor-based protein quantification.
Collapse
Affiliation(s)
- Joao A Paulo
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Steven P Gygi
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
56
|
Chen PY, Tsai YW, Cheng YJ, Giangrande A, Chien CT. Glial response to hypoxia in mutants of NPAS1/3 homolog Trachealess through Wg signaling to modulate synaptic bouton organization. PLoS Genet 2019; 15:e1007980. [PMID: 31381576 PMCID: PMC6695205 DOI: 10.1371/journal.pgen.1007980] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/15/2019] [Accepted: 07/15/2019] [Indexed: 11/18/2022] Open
Abstract
Synaptic structure and activity are sensitive to environmental alterations. Modulation of synaptic morphology and function is often induced by signals from glia. However, the process by which glia mediate synaptic responses to environmental perturbations such as hypoxia remains unknown. Here, we report that, in the mutant for Trachealess (Trh), the Drosophila homolog for NPAS1 and NPAS3, smaller synaptic boutons form clusters named satellite boutons appear at larval neuromuscular junctions (NMJs), which is induced by the reduction of internal oxygen levels due to defective tracheal branches. Thus, the satellite bouton phenotype in the trh mutant is suppressed by hyperoxia, and recapitulated in wild-type larvae raised under hypoxia. We further show that hypoxia-inducible factor (HIF)-1α/Similar (Sima) is critical in mediating hypoxia-induced satellite bouton formation. Sima upregulates the level of the Wnt/Wingless (Wg) signal in glia, leading to reorganized microtubule structures within presynaptic sites. Finally, hypoxia-induced satellite boutons maintain normal synaptic transmission at the NMJs, which is crucial for coordinated larval locomotion.
Collapse
Affiliation(s)
- Pei-Yi Chen
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Wei Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ying-Ju Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Cheng-Ting Chien
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
57
|
Park H, Choi SH, Kong MJ, Kang TC. Dysfunction of 67-kDa Laminin Receptor Disrupts BBB Integrity via Impaired Dystrophin/AQP4 Complex and p38 MAPK/VEGF Activation Following Status Epilepticus. Front Cell Neurosci 2019; 13:236. [PMID: 31178701 PMCID: PMC6542995 DOI: 10.3389/fncel.2019.00236] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Status epilepticus (SE, a prolonged seizure activity) impairs brain-blood barrier (BBB) integrity, which results in secondary complications following SE. The non-integrin 67-kDa laminin receptor (67-kDa LR) plays a role in cell adherence to laminin (a major glycoprotein component in basement membrane), and participates laminin-mediated signaling pathways including p38 mitogen-activated protein kinase (p38 MAPK). Thus, we investigated the role of 67-kDa LR in SE-induced vasogenic edema formation in the rat piriform cortex (PC). SE diminished 67-kDa LR expression, but increased laminin expression, in endothelial cells accompanied by the reduced SMI-71 (a rat BBB barrier marker) expression. Astroglial 67-kDa LR expression was also reduced in the PC due to massive astroglial loss. 67-kDa LR neutralization led to serum extravasation in the PC concomitant with the reduced SMI-71 expression. 67-kDa LR neutralization also decreased expressions of dystrophin and aquaporin-4 (AQP4). In addition, it increased p38 MAPK phosphorylation and expressions of vascular endothelial growth factor (VEGF), laminin and endothelial nitric oxide synthase (eNOS), which were abrogated by SB202190, a p38 MAPK inhibitor. Therefore, our findings indicate that 67-kDa LR dysfunction may disrupt dystrophin-AQP4 complex, which would evoke vasogenic edema formation and subsequent laminin over-expression via activating p38 MAPK/VEGF axis.
Collapse
Affiliation(s)
- Hana Park
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Seo-Hyeon Choi
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Min-Jeong Kong
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
58
|
Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol 2019; 320:112957. [PMID: 31108085 DOI: 10.1016/j.expneurol.2019.112957] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of both acute and long-term morbidity in the pediatric population, leading to a substantial, long-term socioeconomic burden. Despite the increase in the amount of pre-clinical and clinical research, treatment options for TBI rely heavily on supportive care with very limited targeted interventions that improve the acute and chronic sequelae of TBI. Other than injury prevention, not much can be done to limit the primary injury, which consists of tissue damage and cellular destruction. Secondary injury is the result of the ongoing complex inflammatory pathways that further exacerbate tissue damage, resulting in the devastating chronic outcomes of TBI. On the other hand, some level of inflammation is essential for neuronal regeneration and tissue repair. In this review article we discuss the various stages of the neuroimmune response in the immature, pediatric brain in the context of normal maturation and development of the immune system. The developing brain has unique features that distinguish it from the adult brain, and the immune system plays an integral role in CNS development. Those features could potentially make the developing brain more susceptible to worse outcomes, both acutely and in the long-term. The neuroinflammatory reaction which is triggered by TBI can be described as a highly intricate interaction between the cells of the innate and the adaptive immune systems. The innate immune system is triggered by non-specific danger signals that are released from damaged cells and tissues, which in turn leads to neutrophil infiltration, activation of microglia and astrocytes, complement release, as well as histamine release by mast cells. The adaptive immune response is subsequently activated leading to the more chronic effects of neuroinflammation. We will also discuss current attempts at modulating the TBI-induced neuroinflammatory response. A better understanding of the role of the immune system in normal brain development and how immune function changes with age is crucial for designing therapies to appropriately target the immune responses following TBI in order to enhance repair and plasticity.
Collapse
Affiliation(s)
- Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Young Chun
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America.
| |
Collapse
|
59
|
Lee SR, Hyung S, Bang S, Lee Y, Ko J, Lee S, Kim HJ, Jeon NL. Modeling neural circuit, blood–brain barrier, and myelination on a microfluidic 96 well plate. Biofabrication 2019; 11:035013. [DOI: 10.1088/1758-5090/ab1402] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
60
|
Ho TY, Wu WH, Hung SJ, Liu T, Lee YM, Liu YH. Expressional Profiling of Carpet Glia in the Developing Drosophila Eye Reveals Its Molecular Signature of Morphology Regulators. Front Neurosci 2019; 13:244. [PMID: 30983950 PMCID: PMC6449730 DOI: 10.3389/fnins.2019.00244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/01/2019] [Indexed: 12/11/2022] Open
Abstract
Homeostasis in the nervous system requires intricate regulation and is largely accomplished by the blood-brain barrier (BBB). The major gate keeper of the vertebrate BBB is vascular endothelial cells, which form tight junctions (TJs). To gain insight into the development of the BBB, we studied the carpet glia, a subperineurial glial cell type with vertebrate TJ-equivalent septate junctions, in the developing Drosophila eye. The large and flat, sheet-like carpet glia, which extends along the developing eye following neuronal differentiation, serves as an easily accessible experimental system to understand the cell types that exhibit barrier function. We profiled transcribed genes in the carpet glia using targeted DNA adenine methyl-transferase identification, followed by next-generation sequencing (targeted DamID-seq) and found that the majority of genes expressed in the carpet glia function in cellular activities were related to its dynamic morphological changes in the developing eye. To unravel the morphology regulators, we silenced genes selected from the carpet glia transcriptome using RNA interference. The Rho1 gene encoding a GTPase was previously reported as a key regulator of the actin cytoskeleton. The expression of the pathetic (path) gene, encoding a solute carrier transporter in the developing eye, is specific to the carpet glia. The reduced expression of Rho1 severely disrupted the formation of intact carpet glia, and the silencing path impaired the connection between the two carpet glial cells, indicating the pan-cellular and local effects of Rho1 and Path on carpet glial cell morphology, respectively. Our study molecularly characterized a particular subperineurial cell type providing a resource for a further understanding of the cell types comprising the BBB.
Collapse
Affiliation(s)
- Tsung-Ying Ho
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Hang Wu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Jou Hung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Ming Lee
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ya-Hsin Liu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
61
|
Yeh PA, Liu YH, Chu WC, Liu JY, Sun YH. Glial expression of disease-associated poly-glutamine proteins impairs the blood-brain barrier in Drosophila. Hum Mol Genet 2019; 27:2546-2562. [PMID: 29726932 DOI: 10.1093/hmg/ddy160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/25/2018] [Indexed: 12/25/2022] Open
Abstract
Expansion of poly-glutamine (polyQ) stretches in several proteins has been linked to neurodegenerative diseases. The effects of polyQ-expanded proteins on neurons have been extensively studied, but their effects on glia remain unclear. We found that expression of distinct polyQ proteins exclusively in all glia or specifically in the blood-brain barrier (BBB) and blood-retina barrier (BRB) glia caused cell-autonomous impairment of BBB/BRB integrity, suggesting that BBB/BRB glia are most vulnerable to polyQ-expanded proteins. Furthermore, we also found that BBB/BRB leakage in Drosophila is reflected in reversed waveform polarity on the basis of electroretinography (ERG), making ERG a sensitive method to detect BBB/BRB leakage. The polyQ-expanded protein Atxn3-84Q forms aggregates, induces BBB/BRB leakage, restricts Drosophila lifespan and reduces the level of Repo (a pan-glial transcriptional factor required for glial differentiation). Expression of Repo in BBB/BRB glia can rescue BBB/BRB leakage, suggesting that the reduced expression of Repo is important for the effect of polyQ on BBB/BRB impairment. Coexpression of the chaperon HSP40 and HSP70 effectively rescues the effects of Atxn3-84Q, indicating that polyQ protein aggregation in glia is deleterious. Intriguingly, coexpression of wild-type Atxn3-27Q can also rescue BBB/BRB impairment, suggesting that normal polyQ protein may have a protective function.
Collapse
Affiliation(s)
- Po-An Yeh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.,Department of Bioscience Technology, Chung Yuan Christian University, Chung Li, Taiwan
| | - Ya-Hsin Liu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Chen Chu
- Laboratory for Morphogenetic Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Japan
| | - Jia-Yu Liu
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y Henry Sun
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
62
|
An introduction to innate immunity in the central nervous system. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
63
|
Papaneophytou CP, Georgiou E, Karaiskos C, Sargiannidou I, Markoullis K, Freidin MM, Abrams CK, Kleopa KA. Regulatory role of oligodendrocyte gap junctions in inflammatory demyelination. Glia 2018; 66:2589-2603. [PMID: 30325069 PMCID: PMC6519212 DOI: 10.1002/glia.23513] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/27/2022]
Abstract
Gap junctions (GJs) coupling oligodendrocytes to astrocytes and to other oligodendrocytes are formed mainly by connexin47 (Cx47) and a smaller portion by connexin32 (Cx32). Mutations in both connexins cause inherited demyelinating disorders, but their expression is also disrupted in multiple sclerosis (MS). To clarify whether the loss of either Cx47 or Cx32 could modify the outcome of inflammation and myelin loss, we induced experimental autoimmune encephalomyelitis (EAE) in fully backcrossed Cx32 knockout (KO) and Cx47KO mice and compared their outcome with wild type (WT, C57BI/6 N) mice. Cx47KO EAE mice developed the most severe phenotype assessed by clinical scores and behavioral testing, followed by Cx32KO and WT mice. Cx47KO more than Cx32KO EAE mice developed more microglial activation, myelin, and axonal loss than did WT mice. Oligodendrocyte apoptosis and precursor proliferation was also higher in Cx47KO than in Cx32KO or WT EAE mice. Similarly, blood-spinal cord barrier (BSCB) disruption and inflammatory infiltrates of macrophages, T- and B-cells were more severe in Cx47KO than either Cx32KO or WT EAE groups. Finally, expression profiling revealed that several proinflammatory cytokines were higher at the peak of inflammation in the Cx47KO mice and persisted at later stages of EAE in contrast to reduction of their levels in WT EAE mice. Thus, loss of oligodendrocyte GJs aggravates BSCB disruption and inflammatory myelin loss, likely due to dysregulation of proinflammatory cytokines. This mechanism may play an important role in MS brain with reduced connexin expression, as well as in patients with inherited mutations in oligodendrocyte connexins and secondary inflammation.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Astrocytes/metabolism
- Astrocytes/pathology
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/physiopathology
- Calcium-Binding Proteins/metabolism
- Cell Proliferation/genetics
- Connexins/genetics
- Connexins/metabolism
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Freund's Adjuvant/toxicity
- Gap Junctions/metabolism
- Gap Junctions/pathology
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Gene Expression Regulation/physiology
- Hand Strength/physiology
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/metabolism
- Motor Activity/drug effects
- Motor Activity/genetics
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Oligodendroglia/metabolism
- Oligodendroglia/pathology
- Peptide Fragments/toxicity
- Gap Junction beta-1 Protein
Collapse
Affiliation(s)
- Christos P. Papaneophytou
- Neuroscience LaboratoryThe Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
- Department of Life and Health Sciences, School of Sciences and EngineeringUniversity of NicosiaNicosiaCyprus
| | - Elena Georgiou
- Neuroscience LaboratoryThe Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
| | - Christos Karaiskos
- Neuroscience LaboratoryThe Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
| | - Irene Sargiannidou
- Neuroscience LaboratoryThe Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
| | - Kyriaki Markoullis
- Neuroscience LaboratoryThe Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
| | - Mona M. Freidin
- Department of Neurology and RehabilitationUniversity of Illinois ChicagoChicagoIllinois
| | - Charles K. Abrams
- Department of Neurology and RehabilitationUniversity of Illinois ChicagoChicagoIllinois
| | - Kleopas A. Kleopa
- Neuroscience LaboratoryThe Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
- Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular MedicineNicosiaCyprus
| |
Collapse
|
64
|
Banerjee S, Riordan M. Coordinated Regulation of Axonal Microtubule Organization and Transport by Drosophila Neurexin and BMP Pathway. Sci Rep 2018; 8:17337. [PMID: 30478335 PMCID: PMC6255869 DOI: 10.1038/s41598-018-35618-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/07/2018] [Indexed: 01/23/2023] Open
Abstract
Neurexins are well known trans-synaptic cell adhesion molecules that are required for proper synaptic development and function across species. Beyond synapse organization and function, little is known about other roles Neurexins might have in the nervous system. Here we report novel phenotypic consequences of mutations in Drosophila neurexin (dnrx), which alters axonal microtubule organization and transport. We show that dnrx mutants display phenotypic similarities with the BMP receptor wishful thinking (wit) and one of the downstream effectors, futsch, which is a known regulator of microtubule organization and stability. dnrx has genetic interactions with wit and futsch. Loss of Dnrx also results in reduced levels of other downstream effectors of BMP signaling, phosphorylated-Mad and Trio. Interestingly, postsynaptic overexpression of the BMP ligand, Glass bottom boat, in dnrx mutants partially rescues the axonal transport defects but not the synapse undergrowth at the neuromuscular junctions. These data suggest that Dnrx and BMP signaling are involved in many diverse functions and that regulation of axonal MT organization and transport might be distinct from regulation of synaptic growth in dnrx mutants. Together, our work uncovers a novel function of Drosophila Neurexin and may provide insights into functions of Neurexins in vertebrates.
Collapse
Affiliation(s)
- Swati Banerjee
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| | - Maeveen Riordan
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.,University of Colorado School of Medicine, 12631 E. 17th Avenue B177, Aurora, CO, 80045, USA
| |
Collapse
|
65
|
Yildirim K, Petri J, Kottmeier R, Klämbt C. Drosophila glia: Few cell types and many conserved functions. Glia 2018; 67:5-26. [DOI: 10.1002/glia.23459] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Kerem Yildirim
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Johanna Petri
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Rita Kottmeier
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Christian Klämbt
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| |
Collapse
|
66
|
Delsing L, Dönnes P, Sánchez J, Clausen M, Voulgaris D, Falk A, Herland A, Brolén G, Zetterberg H, Hicks R, Synnergren J. Barrier Properties and Transcriptome Expression in Human iPSC-Derived Models of the Blood-Brain Barrier. Stem Cells 2018; 36:1816-1827. [PMID: 30171748 DOI: 10.1002/stem.2908] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/09/2018] [Accepted: 08/18/2018] [Indexed: 11/09/2022]
Abstract
Cell-based models of the blood-brain barrier (BBB) are important for increasing the knowledge of BBB formation, degradation and brain exposure of drug substances. Human models are preferred over animal models because of interspecies differences in BBB structure and function. However, access to human primary BBB tissue is limited and has shown degeneration of BBB functions in vitro. Human induced pluripotent stem cells (iPSCs) can be used to generate relevant cell types to model the BBB with human tissue. We generated a human iPSC-derived model of the BBB that includes endothelial cells in coculture with pericytes, astrocytes and neurons. Evaluation of barrier properties showed that the endothelial cells in our coculture model have high transendothelial electrical resistance, functional efflux and ability to discriminate between CNS permeable and non-permeable substances. Whole genome expression profiling revealed transcriptional changes that occur in coculture, including upregulation of tight junction proteins, such as claudins and neurotransmitter transporters. Pathway analysis implicated changes in the WNT, TNF, and PI3K-Akt pathways upon coculture. Our data suggest that coculture of iPSC-derived endothelial cells promotes barrier formation on a functional and transcriptional level. The information about gene expression changes in coculture can be used to further improve iPSC-derived BBB models through selective pathway manipulation. Stem Cells 2018;36:1816-12.
Collapse
Affiliation(s)
- Louise Delsing
- Department of Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden.,Discovery Sciences, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | | | - José Sánchez
- Biostatistics, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Maryam Clausen
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Dimitrios Voulgaris
- Department of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Herland
- Department of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gabriella Brolén
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Ryan Hicks
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
| |
Collapse
|
67
|
Campisi M, Shin Y, Osaki T, Hajal C, Chiono V, Kamm RD. 3D self-organized microvascular model of the human blood-brain barrier with endothelial cells, pericytes and astrocytes. Biomaterials 2018; 180:117-129. [PMID: 30032046 PMCID: PMC6201194 DOI: 10.1016/j.biomaterials.2018.07.014] [Citation(s) in RCA: 441] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) regulates molecular trafficking, protects against pathogens, and prevents efficient drug delivery to the brain. Models to date failed to reproduce the human anatomical complexity of brain barriers, contributing to misleading results in clinical trials. To overcome these limitations, a novel 3-dimensional BBB microvascular network model was developed via vasculogenesis to accurately replicate the in vivo neurovascular organization. This microfluidic system includes human induced pluripotent stem cell-derived endothelial cells, brain pericytes, and astrocytes as self-assembled vascular networks in fibrin gel. Gene expression of membrane transporters, tight junction and extracellular matrix proteins, was consistent with computational analysis of geometrical structures and quantitative immunocytochemistry, indicating BBB maturation and microenvironment remodelling. Confocal microscopy validated microvessel-pericyte/astrocyte dynamic contact-interactions. The BBB model exhibited perfusable and selective microvasculature, with permeability lower than conventional in vitro models, and similar to in vivo measurements in rat brain. This robust and physiologically relevant BBB microvascular model offers an innovative and valuable platform for drug discovery to predict neuro-therapeutic transport efficacy in pre-clinical applications as well as recapitulate patient-specific and pathological neurovascular functions in neurodegenerative disease.
Collapse
Affiliation(s)
- Marco Campisi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, MA, 02139, USA
| | - Yoojin Shin
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, MA, 02139, USA
| | - Tatsuya Osaki
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, MA, 02139, USA
| | - Cynthia Hajal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, MA, 02139, USA
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, MA, 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, MA, 02139, USA; Singapore-MIT Alliance for Research&Technology (SMART), BioSytems and Micromechanics (BioSyM), Singapore, Singapore.
| |
Collapse
|
68
|
Yuan T, York JR, McCauley DW. Gliogenesis in lampreys shares gene regulatory interactions with oligodendrocyte development in jawed vertebrates. Dev Biol 2018; 441:176-190. [DOI: 10.1016/j.ydbio.2018.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 01/09/2023]
|
69
|
Chen X, Patra A, Sadowska GB, Stonestreet BS. Ischemic-Reperfusion Injury Increases Matrix Metalloproteinases and Tissue Metalloproteinase Inhibitors in Fetal Sheep Brain. Dev Neurosci 2018; 40:234-245. [PMID: 30048980 DOI: 10.1159/000489700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/29/2018] [Indexed: 12/31/2022] Open
Abstract
Hypoxic-ischemic brain injury is a leading cause of neurodevelopmental morbidities in preterm and full-term infants. Blood-brain barrier dysfunction represents an important component of perinatal hypoxic-ischemic brain injury. The extracellular matrix (ECM) is a vital component of the blood-brain barrier. Matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) are important ECM components. They contribute to brain development, blood-brain barrier maintenance, and to regenerative and repair processes after hypoxic-ischemic brain injury. We hypothesized that ischemia at different durations of reperfusion affects the ECM protein composition of MMPs and TIMPs in the cerebral cortex of fetal sheep. Cerebral cortical samples were snap-frozen from sham control fetuses at 127 days of gestation and from fetuses after exposure to 30-min carotid occlusion and 4-, 24-, and 48-h of reperfusion. Protein expression of MMP-2, -8, -9, and -13 and TIMP-1, -2, -3, and -4 was measured by Western immunoblotting along with the gelatinolytic activity of MMP-2 and MMP-9 by zymography. The expression of MMP-8 was increased (Kruskal-Wallis, p = 0.04) in fetuses 48 h after ischemia. In contrast, changes were not observed in the protein expression of MMP-2, -9, or -13. The gelatinolytic activity of pro-MMP-2 was increased (ANOVA, p = 0.02, Tukey HSD, p = 0.05) 24 h after ischemia. TIMP-1 and -3 expression levels were also higher (TIMP-1, ANOVA, p = 0.003, Tukey HSD, p = 0.01; TIMP-3, ANOVA, p = 0.006, Tukey HSD, p = 0.01) 24 h after ischemia compared with both the sham controls and with fetuses exposed to 4 h of reperfusion. The changes in the expression of TIMP-1, -2, and -3 correlated with the changes in the MMP-8 and -13 protein expression. We speculate that regulation of MMP-8, MMP-13, and TIMPs contributes to ECM remodeling after is chemic-reperfusion injury in the fetal brain.
Collapse
|
70
|
Maronpot RR, Nyska A, Troth SP, Gabrielson K, Sysa-Shah P, Kalchenko V, Kuznetsov Y, Harmelin A, Schiffenbauer YS, Bonnel D, Stauber J, Ramot Y. Regulatory Forum Opinion Piece*: Imaging Applications in Toxicologic Pathology-Recommendations for Use in Regulated Nonclinical Toxicity Studies. Toxicol Pathol 2018. [PMID: 28641506 DOI: 10.1177/0192623317710014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Available imaging systems for use in preclinical toxicology studies increasingly show utility as important tools in the toxicologic pathologist's armamentarium, permit longitudinal evaluation of functional and morphological changes in tissues, and provide important information such as organ and lesion volume not obtained by conventional toxicology study parameters. Representative examples of practical imaging applications in toxicology research and preclinical studies are presented for ultrasound, positron emission tomography/single-photon emission computed tomography, optical, magnetic resonance imaging, and matrix-assisted laser desorption ionization-imaging mass spectrometry imaging. Some of the challenges for making imaging systems good laboratory practice-compliant for regulatory submission are presented. Use of imaging data on a case-by-case basis as part of safety evaluation in regulatory submissions is encouraged.
Collapse
Affiliation(s)
| | - Abraham Nyska
- 2 Toxicologic Pathology, Sackler School of Medicine, Tel Aviv University, Timrat, Israel
| | - Sean P Troth
- 3 Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Kathleen Gabrielson
- 4 Department of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Polina Sysa-Shah
- 4 Department of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vyacheslav Kalchenko
- 5 Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Yuri Kuznetsov
- 5 Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Harmelin
- 5 Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | - Yuval Ramot
- 8 Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
71
|
Liebner S, Dijkhuizen RM, Reiss Y, Plate KH, Agalliu D, Constantin G. Functional morphology of the blood-brain barrier in health and disease. Acta Neuropathol 2018; 135:311-336. [PMID: 29411111 PMCID: PMC6781630 DOI: 10.1007/s00401-018-1815-1] [Citation(s) in RCA: 558] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/24/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
The adult quiescent blood-brain barrier (BBB), a structure organised by endothelial cells through interactions with pericytes, astrocytes, neurons and microglia in the neurovascular unit, is highly regulated but fragile at the same time. In the past decade, there has been considerable progress in understanding not only the molecular pathways involved in BBB development, but also BBB breakdown in neurological diseases. Specifically, the Wnt/β-catenin, retinoic acid and sonic hedgehog pathways moved into the focus of BBB research. Moreover, angiopoietin/Tie2 signalling that is linked to angiogenic processes has gained attention in the BBB field. Blood vessels play an essential role in initiation and progression of many diseases, including inflammation outside the central nervous system (CNS). Therefore, the potential influence of CNS blood vessels in neurological diseases associated with BBB alterations or neuroinflammation has become a major focus of current research to understand their contribution to pathogenesis. Moreover, the BBB remains a major obstacle to pharmaceutical intervention in the CNS. The complications may either be expressed by inadequate therapeutic delivery like in brain tumours, or by poor delivery of the drug across the BBB and ineffective bioavailability. In this review, we initially describe the cellular and molecular components that contribute to the steady state of the healthy BBB. We then discuss BBB alterations in ischaemic stroke, primary and metastatic brain tumour, chronic inflammation and Alzheimer's disease. Throughout the review, we highlight common mechanisms of BBB abnormalities among these diseases, in particular the contribution of neuroinflammation to BBB dysfunction and disease progression, and emphasise unique aspects of BBB alteration in certain diseases such as brain tumours. Moreover, this review highlights novel strategies to monitor BBB function by non-invasive imaging techniques focussing on ischaemic stroke, as well as novel ways to modulate BBB permeability and function to promote treatment of brain tumours, inflammation and Alzheimer's disease. In conclusion, a deep understanding of signals that maintain the healthy BBB and promote fluctuations in BBB permeability in disease states will be key to elucidate disease mechanisms and to identify potential targets for diagnostics and therapeutic modulation of the BBB.
Collapse
Affiliation(s)
- Stefan Liebner
- Institute of Neurology, Goethe University Clinic, Frankfurt am Main, Germany.
- Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner site Frankfurt, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research (DZHK), Partner site Frankfurt/Mainz, Frankfurt am Main, Germany.
| | - Rick M Dijkhuizen
- Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Yvonne Reiss
- Institute of Neurology, Goethe University Clinic, Frankfurt am Main, Germany
- Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner site Frankfurt, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl H Plate
- Institute of Neurology, Goethe University Clinic, Frankfurt am Main, Germany
- Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner site Frankfurt, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dritan Agalliu
- Departments of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Departments of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
- Departments of Pharmacology, Columbia University Medical Center, New York, NY, 10032, USA
- Departments of Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, 10032, USA
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| |
Collapse
|
72
|
Kanai MI, Kim MJ, Akiyama T, Takemura M, Wharton K, O'Connor MB, Nakato H. Regulation of neuroblast proliferation by surface glia in the Drosophila larval brain. Sci Rep 2018; 8:3730. [PMID: 29487331 PMCID: PMC5829083 DOI: 10.1038/s41598-018-22028-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 02/15/2018] [Indexed: 01/19/2023] Open
Abstract
Despite the importance of precisely regulating stem cell division, the molecular basis for this control is still elusive. Here, we show that surface glia in the developing Drosophila brain play essential roles in regulating the proliferation of neural stem cells, neuroblasts (NBs). We found that two classes of extracellular factors, Dally-like (Dlp), a heparan sulfate proteoglycan, and Glass bottom boat (Gbb), a BMP homologue, are required for proper NB proliferation. Interestingly, Dlp expressed in perineural glia (PG), the most outer layer of the surface glia, is responsible for NB proliferation. Consistent with this finding, functional ablation of PG using a dominant-negative form of dynamin showed that PG has an instructive role in regulating NB proliferation. Gbb acts not only as an autocrine proliferation factor in NBs but also as a paracrine survival signal in the PG. We propose that bidirectional communication between NBs and glia through TGF-β signaling influences mutual development of these two cell types. We also discuss the possibility that PG and NBs communicate via direct membrane contact or transcytotic transport of membrane components. Thus, our study shows that the surface glia acts not only as a simple structural insulator but also a dynamic regulator of brain development.
Collapse
Affiliation(s)
- Makoto I Kanai
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Takuya Akiyama
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Masahiko Takemura
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Kristi Wharton
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hiroshi Nakato
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
73
|
Castro V, Skowronska M, Lombardi J, He J, Seth N, Velichkovska M, Toborek M. Occludin regulates glucose uptake and ATP production in pericytes by influencing AMP-activated protein kinase activity. J Cereb Blood Flow Metab 2018; 38:317-332. [PMID: 28718701 PMCID: PMC5951017 DOI: 10.1177/0271678x17720816] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Energetic regulation at the blood-brain barrier is critical for maintaining its integrity, transport capabilities, and brain demands for glucose. However, the underlying mechanisms that regulate these processes are still poorly explored. We recently characterized the protein occludin as a NADH oxidase and demonstrated its influence on the expression and activation of the histone deacetylase SIRT-1. Because SIRT-1 works in concert with AMP-activated protein kinase (AMPK) (AMPK), we investigated the impact of occludin on this metabolic switch. Here we show that in blood-brain barrier pericytes, occludin promotes AMPK expression and activation, influencing the expression of glucose transporters GLUT-1 and GLUT-4, glucose uptake, and ATP content. Furthermore, occludin expression, AMP-dependent protein kinase activity, and glucose uptake were altered under inflammatory (TNFα) and infectious (HIV) conditions. We also show that pericytes share glucose and mitochondria with astrocytes, and that occludin levels modify the ability of pericytes to share those energetic resources. In addition, we demonstrate that murine mitochondria can be transferred from live brain microvessels to energetically impaired human astrocytes, promoting their survival. Our findings demonstrate that occludin plays an important role in blood-brain barrier pericyte metabolism by influencing AMPK protein kinase activity, glucose uptake, ATP production, and by regulating the ability of pericytes to interact metabolically with astrocytes.
Collapse
Affiliation(s)
- Victor Castro
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Marta Skowronska
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jorge Lombardi
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jane He
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Neil Seth
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Martina Velichkovska
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
74
|
Cabrera-Maqueda JM, Fuentes Rumí L, López GV, Guerrero AEB, Molina EG, Pérez JD, García-Vázquez E. [Antibiotic diffusion to central nervous system]. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2018; 31:1-12. [PMID: 29390599 PMCID: PMC6159365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Central nervous system (CNS) infections caused by pathogens with a reduced sensitivity to drugs are a therapeutic challenge. Transport of fluid and solutes is tightly controlled within CNS, where vasculature exhibits a blood-brain barrier (BBB).The entry of drugs, including antibiotics, into the cerebro-spinal fluid (CSF) is governed by molecular size, lipophilicity, plasma protein binding and their affinity to transport systems at the BBB. The ratio of the AUCCSF (Area under the curve in CSF)/AUCS (Area under the curve in serum) is the most accurate parameter to characterize drug penetration into the CSF. Linezolid, some fluoroquinolones and metronidazole get high CSF concentrations and are useful for treating susceptible pathogens. Some highly active antibiotic compounds with low BBB permeability can be directly administered into the ventricles together with concomitant intravenous therapy. The ideal antibiotic to treat CNS infections should be that with a small moderately lipophilic molecule, low plasma protein binding and low affinity to efflux pumps at BBB. Knowledge of the pharmacokinetics and pharmacodynamics of antibiotics at the BBB will assist to optimize antibiotic treatment in CNS infections. This article reviews the physicochemical properties of the main groups of antibiotics to assess which compounds are most promising for the treatment of CNS infections and how to use them in the daily clinical practice.
Collapse
Affiliation(s)
| | - Luna Fuentes Rumí
- Servicio de Neurología. Hospital Clínico Universitario Virgen de la Arrixaca de Murcia
| | - Gabriel Valero López
- Servicio de Neurología. Hospital Clínico Universitario Virgen de la Arrixaca de Murcia
| | | | | | - José Díaz Pérez
- Servicio de Neurología. Hospital Clínico Universitario Virgen de la Arrixaca de Murcia
| | - Elisa García-Vázquez
- Servicio de Medicina Interna-Infecciosas. Facultad de Medicina. Universidad de Murcia. IMIB-Murcia
| |
Collapse
|
75
|
Role of the peripheral innate immune system in the development of Alzheimer's disease. Exp Gerontol 2017; 107:59-66. [PMID: 29275160 DOI: 10.1016/j.exger.2017.12.019] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/27/2017] [Accepted: 12/20/2017] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease is one of the most devastating neurodegenerative diseases. The exact cause of the disease is still not known although many scientists believe in the beta amyloid hypothesis which states that the accumulation of the amyloid peptide beta (Aβ) in brain is the initial cause which consequently leads to pathological neuroinflammation. However, it was recently shown that Aβ may have an important role in defending the brain against infections. Thus, the balance between positive and negative impact of Aβ may determine disease progression. Microglia in the brain are innate immune cells, and brain-initiated inflammatory responses reflected in the periphery suggests that Alzheimer's disease is to some extent also a systemic inflammatory disease. Greater permeability of the blood brain barrier facilitates the transport of peripheral immune cells to the brain and vice versa so that a vicious circle originating on the periphery may contribute to the development of overt clinical AD. Persistent inflammatory challenges by pathogens in the periphery, increasing with age, may also contribute to the central propagation of the pathological changes seen clinically. Therefore, the activation status of peripheral innate immune cells may represent an early biomarker of the upcoming impact on the brain. The modulation of these cells may thus become a useful mechanism for modifying disease progression.
Collapse
|
76
|
Bang S, Lee SR, Ko J, Son K, Tahk D, Ahn J, Im C, Jeon NL. A Low Permeability Microfluidic Blood-Brain Barrier Platform with Direct Contact between Perfusable Vascular Network and Astrocytes. Sci Rep 2017; 7:8083. [PMID: 28808270 PMCID: PMC5556097 DOI: 10.1038/s41598-017-07416-0] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/26/2017] [Indexed: 12/14/2022] Open
Abstract
A novel three dimensional blood brain barrier (BBB) platform was developed by independently supplying different types of media to separate cell types within a single device. One channel (vascular channel, VC) is connected to the inner lumen of the vascular network while the other supplies media to the neural cells (neural channel, NC). Compared to co-cultures supplied with only one type of medium (or 1:1 mixture), best barrier properties and viability were obtained with culturing HUVECs with endothelial growth medium (EGM) and neural cells with neurobasal medium supplemented with fetal bovine serum (NBMFBS) independently. The measured vascular network permeability were comparable to reported in vivo values (20 kDa FITC-dextran, 0.45 ± 0.11 × 10−6 cm/s; 70 kDa FITC-dextran, 0.36 ± 0.05 × 10−6 cm/s) and a higher degree of neurovascular interfacing (astrocytic contact with the vascular network, GFAP-CD31 stain overlap) and presence of synapses (stained with synaptophysin). The BBB platform can dependably imitate the perivascular network morphology and synaptic structures characteristic of the NVU. This microfluidic BBB model can find applications in screening pharmaceuticals that target the brain for in neurodegenerative diseases.
Collapse
Affiliation(s)
- Seokyoung Bang
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Seung-Ryeol Lee
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Jihoon Ko
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Kyungmin Son
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Dongha Tahk
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Jungho Ahn
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Changkyun Im
- BK21 Plus Transformative Training Program for Creative Mechanical Engineers, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea. .,Institute of Advanced Machinery and Design, Seoul National University, Seoul, South Korea.
| |
Collapse
|
77
|
4,4′-Diisothiocyanatostilbene-2,2′-disulfonic acid attenuates spontaneous recurrent seizures and vasogenic edema following lithium-pilocarpine induced status epilepticus. Neurosci Lett 2017; 653:51-57. [DOI: 10.1016/j.neulet.2017.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/30/2017] [Accepted: 05/08/2017] [Indexed: 11/18/2022]
|
78
|
|
79
|
Tian X, Brookes O, Battaglia G. Pericytes from Mesenchymal Stem Cells as a model for the blood-brain barrier. Sci Rep 2017; 7:39676. [PMID: 28098158 PMCID: PMC5241806 DOI: 10.1038/srep39676] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/25/2016] [Indexed: 12/15/2022] Open
Abstract
Blood brain-barrier (BBB) in vitro models have been widely reported in studies of the BBB phenotype. However, established co-culture systems involve brain endothelial cells, astrocytes, neurons and pericytes, and therefore are often consuming and technically challenging. Here we use mesenchymal system cells (MSC) as a potential substitute for pericytes in a BBB model. Both MSC and pericyte markers in 2D culture environment were evaluated on different extracellular matrix compositions. Further experiments indicated that MSC contributed in a similar manner to pericytes in a co-cultured 3D model on increasing trans-endothelial electric resistance (TEER) and decreasing permeability against macromolecules.
Collapse
Affiliation(s)
- Xiaohe Tian
- School of Life Science, Anhui University, Hefei, China.,Department of Chemistry, University College London, London, UK
| | - Oliver Brookes
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | | |
Collapse
|
80
|
Marcoli M, Agnati LF, Benedetti F, Genedani S, Guidolin D, Ferraro L, Maura G, Fuxe K. On the role of the extracellular space on the holistic behavior of the brain. Rev Neurosci 2016; 26:489-506. [PMID: 26103627 DOI: 10.1515/revneuro-2015-0007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/11/2015] [Indexed: 12/18/2022]
Abstract
Multiple players are involved in the brain integrative action besides the classical neuronal and astrocyte networks. In the past, the concept of complex cellular networks has been introduced to indicate that all the cell types in the brain can play roles in its integrative action. Intercellular communication in the complex cellular networks depends not only on well-delimited communication channels (wiring transmission) but also on diffusion of signals in physically poorly delimited extracellular space pathways (volume transmission). Thus, the extracellular space and the extracellular matrix are the main players in the intercellular communication modes in the brain. Hence, the extracellular matrix is an 'intelligent glue' that fills the brain and, together with the extracellular space, contributes to the building-up of the complex cellular networks. In addition, the extracellular matrix is part of what has been defined as the global molecular network enmeshing the entire central nervous system, and plays important roles in synaptic contact homeostasis and plasticity. From these premises, a concept is introduced that the global molecular network, by enmeshing the central nervous system, contributes to the brain holistic behavior. Furthermore, it is suggested that plastic 'brain compartments' can be detected in the central nervous system based on the astrocyte three-dimensional tiling of the brain volume and on the existence of local differences in cell types and extracellular space fluid and extracellular matrix composition. The relevance of the present view for neuropsychiatry is discussed. A glossary box with terms and definitions is provided.
Collapse
|
81
|
Induction of Perivascular Neural Stem Cells and Possible Contribution to Neurogenesis Following Transient Brain Ischemia/Reperfusion Injury. Transl Stroke Res 2016; 8:131-143. [PMID: 27352866 DOI: 10.1007/s12975-016-0479-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/12/2016] [Accepted: 06/17/2016] [Indexed: 12/16/2022]
Abstract
Recent therapeutic advances have increased the likelihood of recanalizing the obstructed brain arteries in patients with stroke. Therefore, it is important to understand the fate of neural cells under transient ischemia/reperfusion injury. Accumulating evidence shows that neurogenesis occurs in perivascular regions following brain injury, although the precise mechanism and origin of these newborn neurons under transient ischemia/reperfusion injury remain unclear. Using a mouse model of transient brain ischemia/reperfusion injury, we found that neural stem cells (NSCs) develop within injured areas. This induction of NSCs following ischemia/reperfusion injury was observed even in response to nonlethal ischemia, although massive numbers of NSCs were induced by lethal ischemia. Immunohistochemical and immunoelectron microscopic studies indicated that platelet-derived growth factor receptor beta-positive (PDGFRβ+) pericytes within injured areas following nonlethal ischemia began to express the NSC marker nestin as early as 3 days after transient ischemia/reperfusion. Some PDGFRβ+ pericytes expressed the immature neuronal marker doublecortin at day 7. These findings indicate that brain pericytes are a potential source of the perivascular NSCs that generate neuronal cells under lethal and nonlethal ischemic conditions following transient ischemia/reperfusion. Thus, brain pericytes might be a target for neurogenesis mediation in patients with nonlethal and lethal ischemia following transient ischemia/reperfusion injury.
Collapse
|
82
|
Potential Therapies by Stem Cell-Derived Exosomes in CNS Diseases: Focusing on the Neurogenic Niche. Stem Cells Int 2016; 2016:5736059. [PMID: 27195011 PMCID: PMC4853949 DOI: 10.1155/2016/5736059] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/27/2016] [Indexed: 12/31/2022] Open
Abstract
Neurodegenerative disorders are one of the leading causes of death and disability and one of the biggest burdens on health care systems. Novel approaches using various types of stem cells have been proposed to treat common neurodegenerative disorders such as Alzheimer's Disease, Parkinson's Disease, or stroke. Moreover, as the secretome of these cells appears to be of greater benefit compared to the cells themselves, the extracellular components responsible for its therapeutic benefit have been explored. Stem cells, as well as most cells, release extracellular vesicles such as exosomes, which are nanovesicles able to target specific cell types and thus to modify their function by delivering proteins, lipids, and nucleic acids. Exosomes have recently been tested in vivo and in vitro as therapeutic conveyors for the treatment of diseases. As such, they could be engineered to target specific populations of cells within the CNS. Considering the fact that many degenerative brain diseases have an impact on adult neurogenesis, we discuss how the modulation of the adult neurogenic niches may be a therapeutic target of stem cell-derived exosomes. These novel approaches should be examined in cellular and animal models to provide better, more effective, and specific therapeutic tools in the future.
Collapse
|
83
|
Wootla B, Watzlawik JO, Stavropoulos N, Wittenberg NJ, Dasari H, Abdelrahim MA, Henley JR, Oh SH, Warrington AE, Rodriguez M. Recent Advances in Monoclonal Antibody Therapies for Multiple Sclerosis. Expert Opin Biol Ther 2016; 16:827-839. [PMID: 26914737 DOI: 10.1517/14712598.2016.1158809] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating disease of the CNS and results in neurological disability. Existing immunomodulatory and immunosuppressive approaches lower the number of relapses but do not cure or reverse existing deficits nor improve long-term disability in MS patients. AREAS COVERED Monogenic antibodies were described as treatment options for MS, however the immunogenicity of mouse antibodies hampered the efficacy of potential therapeutics in humans. Availability of improved antibody production technologies resulted in a paradigm shift in MS treatment strategies. In this review, an overview of immunotherapies for MS that use conventional monoclonal antibodies reactive to immune system and their properties and mechanisms of action will be discussed, including recent advances in MS therapeutics and highlight natural autoantibodies (NAbs) that directly target CNS cells. EXPERT OPINION Recent challenges for MS therapy are the identification of relevant molecular and cellular targets, time frame of treatment, and antibody toxicity profiles to identify safe treatment options for MS patients. The application of monoclonal antibody therapies with better biological efficacy associated with minimum side effects possesses huge clinical potential. Advances in monoclonal antibody technologies that directly target cells of nervous system may promote the CNS regeneration field from bench to bedside.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Jens O Watzlawik
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224, USA
| | - Nikolaos Stavropoulos
- Department of General Medicine, Charles University in Prague, Faculty of Medicine in Hradec Kralove, Simkova 870, Hradec Kralove 1, 500 38, Czech Republic
| | - Nathan J Wittenberg
- Department of Electrical and Computer Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA.,Department of Biomedical Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA
| | - Harika Dasari
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Murtada A Abdelrahim
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - John R Henley
- Department of Neurologic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA.,Department of Biomedical Engineering, University of Minnesota, 200 Union Street SE, 4-174 Keller Hall Minneapolis, MN 55455, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
84
|
Kopitar-Jerala N. Innate Immune Response in Brain, NF-Kappa B Signaling and Cystatins. Front Mol Neurosci 2015; 8:73. [PMID: 26696821 PMCID: PMC4673337 DOI: 10.3389/fnmol.2015.00073] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/16/2015] [Indexed: 12/29/2022] Open
Abstract
Recently several reports have demonstrated that innate immune response and inflammation have an important role in major neurodegenerative diseases. The activation of the NF-κB family of transcription factors is a key step in the regulation of pro inflammatory cytokine expression. Microglia and other cell types in the brain can be activated in response to endogenous danger molecules as well as aggregated proteins and brain injury. During the past couple of years several studies reported the role of cystatins in neuroinflammation and neurodegeneration. In the present review, I will summarize and analyze recent findings regarding the role of cystatins in inflammation and NF-κB activation. Type I cystatin stefin B (cystatin B) is an endogenous cysteine cathepsin inhibitor localized in the cytosol, mitochondria and nucleus. Mutations in the gene of stefin B are associated with the neurodegenerative disease known as Unverricht-Lundborg disease and microglial activation plays an important role in the pathogenesis of the disease. Stefin B deficient mice have increased caspase-11 expression and secreted higher amounts of pro-inflammatory cytokines. The increased caspase-11 gene expression, was a consequence of increased NF-κB activation.
Collapse
Affiliation(s)
- Nataša Kopitar-Jerala
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute Ljubljana, Slovenia
| |
Collapse
|
85
|
Wong CT, Wais J, Crawford DA. Prenatal exposure to common environmental factors affects brain lipids and increases risk of developing autism spectrum disorders. Eur J Neurosci 2015. [DOI: 10.1111/ejn.13028] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Christine T. Wong
- School of Kinesiology and Health Science; York University; Toronto ON Canada M3J 1P3
- Neuroscience Graduate Diploma Program; York University; Toronto ON Canada M3J 1P3
| | - Joshua Wais
- School of Kinesiology and Health Science; York University; Toronto ON Canada M3J 1P3
| | - Dorota A. Crawford
- School of Kinesiology and Health Science; York University; Toronto ON Canada M3J 1P3
- Neuroscience Graduate Diploma Program; York University; Toronto ON Canada M3J 1P3
- Department of Biology; York University; Toronto ON Canada M3J 1P3
| |
Collapse
|
86
|
Kim JY, Ko AR, Hyun HW, Kang TC. ETB receptor-mediated MMP-9 activation induces vasogenic edema via ZO-1 protein degradation following status epilepticus. Neuroscience 2015; 304:355-67. [PMID: 26232046 DOI: 10.1016/j.neuroscience.2015.07.065] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/08/2015] [Accepted: 07/24/2015] [Indexed: 01/08/2023]
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells with specialized tight junctions (TJs) lining the blood vessels and astroglial endfeet surrounding the blood vessels. Although BBB disruption during brain insults leads to vasogenic edema as one of the primary steps in the epileptogenic process, little is known about the molecular and physiological events concerning vasogenic edema formation. In the present study, status epilepticus (SE) changed the expressions and subcellular localizations of TJ proteins (claudin-5, occludin and zonula occludens-1 (ZO-1)) in endothelial cells of the rat piriform cortex. Among TJ proteins, the alteration in ZO-1 expression was relevant to endothelin B (ETB) receptor-mediated endothelial nitric oxide synthase (eNOS) activation, which increased matrix metalloproteinase-9 (MMP-9) activity. Indeed, BQ788 (an ETB receptor antagonist) effectively attenuated SE-induced vasogenic edema by inhibiting eNOS-mediated MMP-9 activation and ZO-1 protein degradation in endothelial cells, although astroglial endfeet were detached from endothelial cells. Therefore, we suggest that SE-induced ETB receptor/eNOS-mediated MMP-9 activation may lead to impairments of endothelial cell function via TJ protein degradation, which are involved in vasogenic edema formation independent of perivascular astroglial functions.
Collapse
Affiliation(s)
- J Y Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | - A-R Ko
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | - H-W Hyun
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | - T-C Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea.
| |
Collapse
|
87
|
Ko AR, Kim JY, Hyun HW, Kim JE. Endothelial NOS activation induces the blood-brain barrier disruption via ER stress following status epilepticus. Brain Res 2015; 1622:163-73. [PMID: 26115585 DOI: 10.1016/j.brainres.2015.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/03/2015] [Accepted: 06/06/2015] [Indexed: 11/19/2022]
Abstract
The blood-brain barrier (BBB) maintains the unique brain microenvironment, which is separated from the systemic circulating system. Since the endoplasmic reticulum (ER) is an important cell organelle that is responsible for protein synthesis, the correct folding and sorting of proteins contributing to cell survivals, ER stress is a potential cause of cell damage in various diseases. Therefore, it would be worthy to explore the the relationship between the ER stress and BBB disruption during vasogenic edema formation induced by epileptogenic insults. In the present study, we investigated the roles of ER stress in vasogenic edema and its related events in rat epilepsy models provoked by pilocarpine-induced status epilepticus (SE). SE-induced eNOS activation induces BBB breakdown via up-regulation of GRP78 expression and dysfunction of SMI-71 (an endothelial BBB marker) in the piriform cortex (PC). In addition, caveolin-1 peptide (an eNOS inhibitor) effectively attenuated GRP78 expression and down-regulation of SMI-71. Taken together, our findings suggest that eNOS-mediated ER stress may participate in SE-induced vasogenic edema formation. Therefore, the modulation of ER stress may be a considerable strategy for therapy in impairments of endothelial cell function.
Collapse
Affiliation(s)
- Ah-Reum Ko
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea
| | - Ji Yang Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea
| | - Hye-Won Hyun
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea.
| |
Collapse
|
88
|
Nakagomi T, Nakano-Doi A, Kawamura M, Matsuyama T. Do Vascular Pericytes Contribute to Neurovasculogenesis in the Central Nervous System as Multipotent Vascular Stem Cells? Stem Cells Dev 2015; 24:1730-9. [PMID: 25900222 DOI: 10.1089/scd.2015.0039] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence suggests that multipotent stem cells are harbored within a vascular niche inside various organs. Although a precise phenotype of resident vascular stem cells (VSCs) that can function as multipotent stem cells remains unclear, accumulating evidence shows that multipotent VSCs are likely vascular pericytes (PCs) that localize within blood vessels. These PCs are multipotent, possessing the ability to differentiate into various cell types, including vascular lineage cells. In addition, brain PCs are unique: They are derived from neural crest and can differentiate into neural lineage cells. Because PCs in the central nervous system (CNS) can contribute to both neurogenesis and vasculogenesis, they may mediate the reparative process of neurovascular units that are constructed by neural and vascular cells. Here, we describe the activity of PCs when viewed as multipotent VSCs, primarily regarding their neurogenic and vasculogenic potential in the CNS. We also discuss similarities between PCs and other candidates for multipotent VSCs, including perivascular mesenchymal stem cells, neural crest-derived stem cells, adventitial progenitor cells, and adipose-derived stem cells.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| | - Akiko Nakano-Doi
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| | - Miki Kawamura
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan .,2 Department of Neurology, Osaka University Graduate School of Medicine , Osaka, Japan
| | - Tomohiro Matsuyama
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| |
Collapse
|
89
|
Nakagomi T, Kubo S, Nakano-Doi A, Sakuma R, Lu S, Narita A, Kawahara M, Taguchi A, Matsuyama T. Brain vascular pericytes following ischemia have multipotential stem cell activity to differentiate into neural and vascular lineage cells. Stem Cells 2015; 33:1962-74. [PMID: 25694098 DOI: 10.1002/stem.1977] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 01/27/2015] [Indexed: 01/01/2023]
Abstract
Brain vascular pericytes (PCs) are a key component of the blood-brain barrier (BBB)/neurovascular unit, along with neural and endothelial cells. Besides their crucial role in maintaining the BBB, increasing evidence shows that PCs have multipotential stem cell activity. However, their multipotency has not been considered in the pathological brain, such as after an ischemic stroke. Here, we examined whether brain vascular PCs following ischemia (iPCs) have multipotential stem cell activity and differentiate into neural and vascular lineage cells to reconstruct the BBB/neurovascular unit. Using PCs extracted from ischemic regions (iPCs) from mouse brains and human brain PCs cultured under oxygen/glucose deprivation, we show that PCs developed stemness presumably through reprogramming. The iPCs revealed a complex phenotype of angioblasts, in addition to their original mesenchymal properties, and multidifferentiated into cells from both a neural and vascular lineage. These data indicate that under ischemic/hypoxic conditions, PCs can acquire multipotential stem cell activity and can differentiate into major components of the BBB/neurovascular unit. Thus, these findings support the novel concept that iPCs can contribute to both neurogenesis and vasculogenesis at the site of brain injuries.
Collapse
Affiliation(s)
| | - Shuji Kubo
- Department of Genetics, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan
| | - Rika Sakuma
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan
| | - Shan Lu
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan.,Department of Neurology of Hangzhou First People's Hospital, Hangzhou, People's Republic of China
| | - Aya Narita
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan
| | - Maiko Kawahara
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan.,Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | | |
Collapse
|
90
|
Matzat T, Sieglitz F, Kottmeier R, Babatz F, Engelen D, Klämbt C. Axonal wrapping in the Drosophila PNS is controlled by glia-derived neuregulin homolog Vein. Development 2015; 142:1336-45. [PMID: 25758464 DOI: 10.1242/dev.116616] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Efficient neuronal conductance requires that axons are insulated by glial cells. For this, glial membranes need to wrap around axons. Invertebrates show a relatively simple extension of glial membranes around the axons, resembling Remak fibers formed by Schwann cells in the mammalian peripheral nervous system. To unravel the molecular pathways underlying differentiation of glial cells that provide axonal wrapping, we are using the genetically amenable Drosophila model. At the end of larval life, the wrapping glia differentiates into very large cells, spanning more than 1 mm of axonal length. The extension around axonal membranes is not influenced by the caliber of the axon or its modality. Using cell type-specific gene knockdown we show that the extension of glial membranes around the axons is regulated by an autocrine activation of the EGF receptor through the neuregulin homolog Vein. This resembles the molecular mechanism employed during cell-autonomous reactivation of glial differentiation after injury in mammals. We further demonstrate that Vein, produced by the wrapping glia, also regulates the formation of septate junctions in the abutting subperineurial glia. Moreover, the wrapping glia indirectly controls the proliferation of the perineurial glia. Thus, the wrapping glia appears center stage to orchestrate the development of the different glial cell layers in a peripheral nerve.
Collapse
Affiliation(s)
- Till Matzat
- Institut für Neurobiologie, Universität Münster, Badestr. 9, Münster D-48149, Germany
| | - Florian Sieglitz
- Institut für Neurobiologie, Universität Münster, Badestr. 9, Münster D-48149, Germany
| | - Rita Kottmeier
- Institut für Neurobiologie, Universität Münster, Badestr. 9, Münster D-48149, Germany
| | - Felix Babatz
- Institut für Neurobiologie, Universität Münster, Badestr. 9, Münster D-48149, Germany
| | - Daniel Engelen
- Institut für Neurobiologie, Universität Münster, Badestr. 9, Münster D-48149, Germany
| | - Christian Klämbt
- Institut für Neurobiologie, Universität Münster, Badestr. 9, Münster D-48149, Germany
| |
Collapse
|
91
|
Barbosa DJ, Capela JP, de Lourdes Bastos M, Carvalho F. In vitro models for neurotoxicology research. Toxicol Res (Camb) 2015; 4:801-842. [DOI: 10.1039/c4tx00043a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
The nervous system has a highly complex organization, including many cell types with multiple functions, with an intricate anatomy and unique structural and functional characteristics; the study of its (dys)functionality following exposure to xenobiotics, neurotoxicology, constitutes an important issue in neurosciences.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - João Paulo Capela
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
92
|
Schapansky J, Nardozzi JD, LaVoie MJ. The complex relationships between microglia, alpha-synuclein, and LRRK2 in Parkinson's disease. Neuroscience 2014; 302:74-88. [PMID: 25284317 DOI: 10.1016/j.neuroscience.2014.09.049] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 12/19/2022]
Abstract
The proteins alpha-synuclein (αSyn) and leucine rich repeat kinase 2 (LRRK2) are both key players in the pathogenesis of the neurodegenerative disorder Parkinson's disease (PD), but establishing a functional link between the two proteins has proven elusive. Research studies for these two proteins have traditionally and justifiably focused in neuronal cells, but recent studies indicate that each protein could play a greater pathological role elsewhere. αSyn is expressed at high levels within neurons, but they also secrete the protein into the extracellular milieu, where it can have broad ranging effects in the nervous system and relevance to disease etiology. Similarly, low neuronal LRRK2 expression and activity suggests that LRRK2-related functions could be more relevant in cells with higher expression, such as brain-resident microglia. Microglia are monocytic immune cells that protect neurons from noxious stimuli, including pathological αSyn species, and microglial activation is believed to contribute to neuroinflammation and neuronal death in PD. Interestingly, both αSyn and LRRK2 can be linked to microglial function. Secreted αSyn can directly activate microglia, and can be taken up by microglia for clearance, while LRRK2 has been implicated in the intrinsic regulation of microglial activation and of lysosomal degradation processes. Based on these observations, the present review will focus on how PD-associated mutations in LRRK2 could potentially alter microglial biology with respect to neuronally secreted αSyn, resulting in cell dysfunction and neurodegeneration.
Collapse
Affiliation(s)
- J Schapansky
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States
| | - J D Nardozzi
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States
| | - M J LaVoie
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States.
| |
Collapse
|
93
|
Kim YJ, Kim JY, Ko AR, Kang TC. Over-expression of laminin correlates to recovery of vasogenic edema following status epilepticus. Neuroscience 2014; 275:146-61. [DOI: 10.1016/j.neuroscience.2014.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/03/2014] [Accepted: 06/05/2014] [Indexed: 11/29/2022]
|
94
|
Andersson O, Badisco L, Hansen AH, Hansen SH, Hellman K, Nielsen PA, Olsen LR, Verdonck R, Abbott NJ, Vanden Broeck J, Andersson G. Characterization of a novel brain barrier ex vivo insect-based P-glycoprotein screening model. Pharmacol Res Perspect 2014; 2:e00050. [PMID: 25505597 PMCID: PMC4186439 DOI: 10.1002/prp2.50] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 01/16/2023] Open
Abstract
In earlier studies insects were proposed as suitable models for vertebrate blood–brain barrier (BBB) permeability prediction and useful in early drug discovery. Here we provide transcriptome and functional data demonstrating the presence of a P-glycoprotein (Pgp) efflux transporter in the brain barrier of the desert locust (Schistocerca gregaria). In an in vivo study on the locust, we found an increased uptake of the two well-known Pgp substrates, rhodamine 123 and loperamide after co-administration with the Pgp inhibitors cyclosporine A or verapamil. Furthermore, ex vivo studies on isolated locust brains demonstrated differences in permeation of high and low permeability compounds. The vertebrate Pgp inhibitor verapamil did not affect the uptake of passively diffusing compounds but significantly increased the brain uptake of Pgp substrates in the ex vivo model. In addition, studies at 2°C and 30°C showed differences in brain uptake between Pgp-effluxed and passively diffusing compounds. The transcriptome data show a high degree of sequence identity of the locust Pgp transporter protein sequences to the human Pgp sequence (37%), as well as the presence of conserved domains. As in vertebrates, the locust brain–barrier function is morphologically confined to one specific cell layer and by using a whole-brain ex vivo drug exposure technique our locust model may retain the major cues that maintain and modulate the physiological function of the brain barrier. We show that the locust model has the potential to act as a robust and convenient model for assessing BBB permeability in early drug discovery.
Collapse
Affiliation(s)
- Olga Andersson
- EntomoPharm, R&D Medicon Village, S-223 81, Lund, Sweden
| | - Liesbeth Badisco
- Department of Animal Physiology and Neurobiology, Katholieke Universiteit Leuven Leuven, Belgium
| | | | - Steen Honoré Hansen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen DK-2100, Copenhagen, Denmark
| | - Karin Hellman
- EntomoPharm, R&D Medicon Village, S-223 81, Lund, Sweden
| | | | - Line Rørbæk Olsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen DK-2100, Copenhagen, Denmark
| | - Rik Verdonck
- Department of Animal Physiology and Neurobiology, Katholieke Universiteit Leuven Leuven, Belgium
| | - N Joan Abbott
- BBB Group, Institute of Pharmaceutical Science, King's College London Franklin Wilkins Building, London, SE1 9NH, United Kingdom
| | - Jozef Vanden Broeck
- Department of Animal Physiology and Neurobiology, Katholieke Universiteit Leuven Leuven, Belgium
| | | |
Collapse
|
95
|
Glial wingless/Wnt regulates glutamate receptor clustering and synaptic physiology at the Drosophila neuromuscular junction. J Neurosci 2014; 34:2910-20. [PMID: 24553932 DOI: 10.1523/jneurosci.3714-13.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glial cells are emerging as important regulators of synapse formation, maturation, and plasticity through the release of secreted signaling molecules. Here we use chromatin immunoprecipitation along with Drosophila genomic tiling arrays to define potential targets of the glial transcription factor Reversed polarity (Repo). Unexpectedly, we identified wingless (wg), a secreted morphogen that regulates synaptic growth at the Drosophila larval neuromuscular junction (NMJ), as a potential Repo target gene. We demonstrate that Repo regulates wg expression in vivo and that local glial cells secrete Wg at the NMJ to regulate glutamate receptor clustering and synaptic function. This work identifies Wg as a novel in vivo glial-secreted factor that specifically modulates assembly of the postsynaptic signaling machinery at the Drosophila NMJ.
Collapse
|
96
|
Muoio V, Persson PB, Sendeski MM. The neurovascular unit - concept review. Acta Physiol (Oxf) 2014; 210:790-8. [PMID: 24629161 DOI: 10.1111/apha.12250] [Citation(s) in RCA: 364] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/01/2013] [Accepted: 01/27/2014] [Indexed: 01/01/2023]
Abstract
The cerebral hyperaemia is one of the fundamental mechanisms for the central nervous system homeostasis. Due also to this mechanism, oxygen and nutrients are maintained in satisfactory levels, through vasodilation and vasoconstriction. The brain hyperaemia, or coupling, is accomplished by a group of cells, closely related to each other; called neurovascular unit (NVU). The neurovascular unit is composed by neurones, astrocytes, endothelial cells of blood-brain barrier (BBB), myocytes, pericytes and extracellular matrix components. These cells, through their intimate anatomical and chemical relationship, detect the needs of neuronal supply and trigger necessary responses (vasodilation or vasoconstriction) for such demands. Here, we review the concepts of NVU, the coupling mechanisms and research strategies.
Collapse
Affiliation(s)
- V. Muoio
- Institut für Vegetative Physiologie; Charite- Universisitätmedizin Berlin; Berlin Germany
| | - P. B. Persson
- Institut für Vegetative Physiologie; Charite- Universisitätmedizin Berlin; Berlin Germany
| | - M. M. Sendeski
- Institut für Vegetative Physiologie; Charite- Universisitätmedizin Berlin; Berlin Germany
| |
Collapse
|
97
|
Chang YC, Olson J, Beasley FC, Tung C, Zhang J, Crocker PR, Varki A, Nizet V. Group B Streptococcus engages an inhibitory Siglec through sialic acid mimicry to blunt innate immune and inflammatory responses in vivo. PLoS Pathog 2014; 10:e1003846. [PMID: 24391502 PMCID: PMC3879367 DOI: 10.1371/journal.ppat.1003846] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 11/06/2013] [Indexed: 12/15/2022] Open
Abstract
Group B Streptococcus (GBS) is a common agent of bacterial sepsis and meningitis in newborns. The GBS surface capsule contains sialic acids (Sia) that engage Sia-binding immunoglobulin-like lectins (Siglecs) on leukocytes. Here we use mice lacking Siglec-E, an inhibitory Siglec of myelomonocytic cells, to study the significance of GBS Siglec engagement during in vivo infection. We found GBS bound to Siglec-E in a Sia-specific fashion to blunt NF-κB and MAPK activation. As a consequence, Siglec-E-deficient macrophages had enhanced pro-inflammatory cytokine secretion, phagocytosis and bactericidal activity against the pathogen. Following pulmonary or low-dose intravenous GBS challenge, Siglec-E KO mice produced more pro-inflammatory cytokines and exhibited reduced GBS invasion of the central nervous system. In contrast, upon high dose lethal challenges, cytokine storm in Siglec-E KO mice was associated with accelerated mortality. We conclude that GBS Sia mimicry influences host innate immune and inflammatory responses in vivo through engagement of an inhibitory Siglec, with the ultimate outcome of the host response varying depending upon the site, stage and magnitude of infection. The bacterium Group B Streptococcus (GBS) causes serious infections such as meningitis in human newborn babies. The surface of GBS is coated with a capsule made of sugar molecules. Prominent among these is sialic acid (Sia), a human-like sugar that interacts with protein receptors called Siglecs on the surface of our white blood cells. In a test tube, GBS Sia binding to human Siglecs can suppress white blood cell activation, reducing their bacterial killing abilities; however, the significance of this during actual infection was unknown. To answer this question, we studied mice for which a key white blood cell Siglec has been genetically deleted. When infected with GBS, white blood cells from the mutant mice are not shut off by the pathogen's Sia-containing sugar capsule. The white blood cells from the Siglec-deficient mice are better at killing GBS and are able to clear infection more quickly than a normal mouse. However, if the mice are given an overwhelming dose of GBS bacteria, exaggerated white blood activation can trigger shock and more rapid death. These studies show how “molecular mimicry” of sugar molecules in the host can influence a bacterial pathogen's interaction with the immune system and the outcome of infection.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/immunology
- Cytokines/genetics
- Cytokines/immunology
- Humans
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/pathology
- Macrophages/immunology
- Macrophages/pathology
- Mice
- Mice, Knockout
- Molecular Mimicry/immunology
- N-Acetylneuraminic Acid/genetics
- N-Acetylneuraminic Acid/immunology
- Pneumonia, Bacterial/genetics
- Pneumonia, Bacterial/immunology
- Pneumonia, Bacterial/pathology
- Streptococcal Infections/genetics
- Streptococcal Infections/immunology
- Streptococcal Infections/pathology
- Streptococcus agalactiae/immunology
Collapse
Affiliation(s)
- Yung-Chi Chang
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, United States of America
- Department of Pediatrics, University of California, San Diego, La Jolla, California, United States of America
| | - Joshua Olson
- Department of Pediatrics, University of California, San Diego, La Jolla, California, United States of America
| | - Federico C. Beasley
- Department of Pediatrics, University of California, San Diego, La Jolla, California, United States of America
| | - Christine Tung
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Jiquan Zhang
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Paul R. Crocker
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ajit Varki
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, United States of America
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (AV); (VN)
| | - Victor Nizet
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, United States of America
- Department of Pediatrics, University of California, San Diego, La Jolla, California, United States of America
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
- Rady Children's Hospital, San Diego, California, United States of America
- * E-mail: (AV); (VN)
| |
Collapse
|
98
|
Disease Influence on BBB Transport in Neurodegenerative Disorders. DRUG DELIVERY TO THE BRAIN 2014. [DOI: 10.1007/978-1-4614-9105-7_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
99
|
Abstract
The neurovascular unit (NVU) is a dynamic structure assembled by endothelial cells (EC), a basement membrane (BM), perivascular astrocytes (PA), pericytes, and surrounding neurons. The NVU regulates the passage of substances and cellular elements from the intravascular space into the brain parenchyma. This function, also known as blood-brain barrier (BBB), is regulated by the integrity of tight junctions proteins between EC, and the interaction between PA and the basal lamina. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) are abundantly expressed in the NVU. Here we will review data indicating that the interaction between TWEAK and Fn14 in the endothelial cell-BM-astrocyte interface regulates the function of the BBB following an ischemic/hypoxic injury, and that pharmacological inhibition of TWEAK-Fn14 is a promising target for the treatment of patients with neurological diseases that have a direct impact on the structure and function of the NVU.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine , Atlanta, GA , USA ; Department of Neurology, Veterans Affairs Medical Center, Emory University School of Medicine , Atlanta, GA , USA
| |
Collapse
|
100
|
Synapse location during growth depends on glia location. Cell 2013; 154:337-50. [PMID: 23870123 DOI: 10.1016/j.cell.2013.06.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 04/09/2013] [Accepted: 06/19/2013] [Indexed: 11/22/2022]
Abstract
Synaptic contacts are largely established during embryogenesis and are then maintained during growth. To identify molecules involved in this process, we conducted a forward genetic screen in C. elegans and identified cima-1. In cima-1 mutants, synaptic contacts are correctly established during embryogenesis, but ectopic synapses emerge during postdevelopmental growth. cima-1 encodes a solute carrier in the SLC17 family of transporters that includes sialin, a protein that when mutated in humans results in neurological disorders. cima-1 does not function in neurons but rather functions in the nearby epidermal cells to correctly position glia during postlarval growth. Our findings indicate that CIMA-1 antagonizes the FGF receptor (FGFR), and does so most likely by inhibiting FGFR's role in epidermal-glia adhesion rather than signaling. Our data suggest that epidermal-glia crosstalk, in this case mediated by a transporter and the FGF receptor, is vital to preserve embryonically derived circuit architecture during postdevelopmental growth.
Collapse
|