Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Mater ; 34(40): e2203643, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35980937

ABSTRACT

The learning and inference efficiencies of an artificial neural network represented by a cross-point synaptic memristor array can be achieved using a selector, with high selectivity (Ion /Ioff ) and sufficient death region, stacked vertically on a synaptic memristor. This can prevent a sneak current in the memristor array. A selector with multiple jar-shaped conductive Cu filaments in the resistive switching layer is precisely fabricated by designing the Cu ion concentration depth profile of the CuGeSe layer as a filament source, TiN diffusion barrier layer, and Ge3 Se7 switching layer. The selector performs super-linear-threshold-switching with a selectivity of > 107 , death region of -0.70-0.65 V, holding time of 300 ns, switching speed of 25 ns, and endurance cycle of > 106 . In addition, the mechanism of switching is proven by the formation of conductive Cu filaments between the CuGeSe and Ge3 Se7 layers under a positive bias on the top Pt electrode and an automatic rupture of the filaments after the holding time. Particularly, a spiking deep neural network using the designed one-selector-one-memory cross-point array improves the Modified National Institute of Standards and Technology classification accuracy by ≈3.8% by eliminating the sneak current in the cross-point array during the inference process.

2.
Glia ; 70(2): 256-272, 2022 02.
Article in English | MEDLINE | ID: mdl-34559433

ABSTRACT

Schwann cells (SCs) are known to produce extracellular vesicles (EV) that participate in cell-cell communication by transferring cargo to target cells, including mRNAs, microRNAs, and biologically active proteins. Herein, we report a novel mechanism whereby SC EVs may regulate PNS physiology, especially in injury, by controlling the activity of TNFα. SCs actively sequester tumor necrosis factor receptor-1 (TNFR1) into EVs at high density, accounting for about 2% of the total protein in SC EVs (~1000 copies TNFR1/EV). Although TNFR2 was robustly expressed by SCs in culture, TNFR2 was excluded from SC EVs. SC EV TNFR1 bound TNFα, decreasing the concentration of free TNFα available to bind to cells and thus served as a TNFα decoy. SC EV TNFR1 significantly inhibited TNFα-induced p38 MAPK phosphorylation in cultured SCs. When TNFR1 was proteolytically removed from SC EVs using tumor necrosis factor-α converting enzyme (TACE) or neutralized with antibody, the ability of TNFα to activate p38 MAPK in the presence of these EVs was restored. As further evidence of its decoy activity, SC EV TNFR1 modified TNFα activities in vitro including: (1) regulation of expression of other cytokines; (2) effects on SC morphology; and (3) effects on SC viability. SC EVs also modified the effects of TNFα on sciatic nerve morphology and neuropathic pain-related behavior in vivo. By sequestering TNFR1 in EVs, SCs may buffer against the potentially toxic effects of TNFα. SC EVs provide a novel mechanism for the spatial and temporal regulation of neuro-inflammation.


Subject(s)
Extracellular Vesicles , Receptors, Tumor Necrosis Factor, Type I , Schwann Cells , Tumor Necrosis Factor-alpha , Cells, Cultured , Extracellular Vesicles/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Schwann Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
Sensors (Basel) ; 18(8)2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30126095

ABSTRACT

Research into integrating the concept of the internet of things (IoT) into smart factories has accelerated, leading to the emergence of various smart factory solutions. Most ideas, however, focus on the automation and integration of processes in factory, rather than organic cooperation among mobile assets (e.g., the workers and manufactured products) and fixed manufacturing equipment (e.g., press molds, computer numerical controls, painting). Additionally, it is difficult to apply smart factory and IoT designs to analog factories, because such a factory would require the integration of mobile assets and smart manufacturing processes. Thus, existing analog factories remain intact and smart factories are newly constructed. To overcome this disparity and to make analog factories compatible with smart technologies and IoT, we propose the opportunistic and location-based collaboration architecture (OLCA) platform, which allows for smart devices to be attached to workers, products, and facilities to enable the collaboration of location and event information in devices. Using this system, we can monitor workers' positions and production processes in real-time to help prevent dangerous situations and better understand product movement. We evaluate the proposed OLCA platform's performance while using a simple smart factory scenario, thus confirming its suitability.

4.
PLoS Biol ; 11(1): e1001469, 2013.
Article in English | MEDLINE | ID: mdl-23349620

ABSTRACT

The cerebral cortex performs complex cognitive functions at the expense of tremendous energy consumption. Blood vessels in the brain are known to form stereotypic patterns that facilitate efficient oxygen and nutrient delivery. Yet little is known about how vessel development in the brain is normally regulated. Radial glial neural progenitors are well known for their central role in orchestrating brain neurogenesis. Here we show that, in the late embryonic cortex, radial glial neural progenitors also play a key role in brain angiogenesis, by interacting with nascent blood vessels and regulating vessel stabilization via modulation of canonical Wnt signaling. We find that ablation of radial glia results in vessel regression, concomitant with ectopic activation of Wnt signaling in endothelial cells. Direct activation of Wnt signaling also results in similar vessel regression, while attenuation of Wnt signaling substantially suppresses regression. Radial glial ablation and ectopic Wnt pathway activation leads to elevated endothelial expression of matrix metalloproteinases, while inhibition of metalloproteinase activity significantly suppresses vessel regression. These results thus reveal a previously unrecognized role of radial glial progenitors in stabilizing nascent brain vascular network and provide novel insights into the molecular cascades through which target neural tissues regulate vessel stabilization and patterning during development and throughout life.


Subject(s)
Brain/blood supply , Cerebral Cortex/blood supply , Neural Stem Cells/metabolism , Origin Recognition Complex/genetics , Radial Nerve/embryology , Animals , Cell Cycle Checkpoints , Cells, Cultured , Eye Proteins/genetics , Female , Homeodomain Proteins/genetics , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Neovascularization, Physiologic , Neurogenesis , Neuroglia/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , RNA, Messenger/biosynthesis , Radial Nerve/metabolism , Repressor Proteins/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway
5.
PLoS One ; 7(10): e48001, 2012.
Article in English | MEDLINE | ID: mdl-23110156

ABSTRACT

Astroglia are a major cell type in the brain and play a key role in many aspects of brain development and function. In the adult brain, astrocytes are known to intimately ensheath blood vessels and actively coordinate local neural activity and blood flow. During development of the neural retina, blood vessel growth follows a meshwork of astrocytic processes. Several genes have also been implicated in retinal astrocytes for regulating vessel development. This suggests a role of astrocytes in promoting angiogenesis throughout the central nervous system. To determine the roles that astrocytes may play during brain angiogenesis, we employ genetic approaches to inhibit astrogliogenesis during perinatal corticogenesis and examine its effects on brain vessel development. We find that conditional deletion from glial progenitors of orc3, a gene required for DNA replication, dramatically reduces glial progenitor cell number in the subventricular zone and astrocytes in the early postnatal cerebral cortex. This, in turn, results in severe reductions in both the density and branching frequency of cortical blood vessels. Consistent with a delayed growth but not regression of vessels, we find neither significant net decreases in vessel density between different stages after normalizing for cortical expansion nor obvious apoptosis of endothelial cells in these mutants. Furthermore, concomitant with loss of astroglial interactions, we find increased endothelial cell proliferation, enlarged vessel luminal size as well as enhanced cytoskeletal gene expression in pericytes, which suggests compensatory changes in vascular cells. Lastly, we find that blood vessel morphology in mutant cortices recovers substantially at later stages, following astrogliosis. These results thus implicate a functional requirement for astroglia in promoting blood vessel growth during brain development.


Subject(s)
Astrocytes/physiology , Blood Vessels/growth & development , Brain/blood supply , Brain/growth & development , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blood Vessels/metabolism , Brain/cytology , Cell Differentiation/genetics , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Ventricles/blood supply , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/metabolism , Neurogenesis/genetics , Origin Recognition Complex/deficiency , Origin Recognition Complex/genetics , Time Factors
6.
J Neurosci ; 32(43): 14979-93, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-23100420

ABSTRACT

The cerebellum consists of an intricate array of lobules that arises during the process of foliation. Foliation not only increases surface area, but may also facilitate organization of cerebellar neural circuitry. Defects in cerebellar foliation are associated with a number of diseases. Yet, little is known about how foliation, a process involving large-scale and simultaneous movement of several different cell types, is coordinated by cell-cell signaling at the molecular level. Here we show that Ric-8a, a guanine nucleotide exchange factor in the G-protein-coupled receptor pathway, is specifically required in Bergmann glia during cerebellar foliation. We find that ric-8a mutation in mice results in disorganized Bergmann glial scaffolding, defective granule cell migration, and disrupted Purkinje cell positioning. These abnormalities result from primary defects in Bergmann glia since mutations in granule cells do not show similar effects. They first arise during late embryogenesis, at the onset of foliation, when ric-8a mutant Bergmann glia fail to maintain adhesion to the basement membrane specifically at emerging fissures. This suggests that Ric-8a is essential for the enhanced Bergmann glia-basement membrane adhesion required for fissure formation. Indeed, we find that ric-8a-deficient cerebellar glia show decreased affinity for basement membrane components. We also find that weakening Bergmann glia-basement membrane interaction by ß1 integrin deletion results in a similar phenotype. These results thus reveal a novel role of Ric-8a in modulating Bergmann glia-basement membrane adhesion during foliation, and provide new insights into the signaling pathways that coordinate cellular movement during cerebellar morphogenesis.


Subject(s)
Basement Membrane/metabolism , Cell Adhesion/physiology , Cerebellum/cytology , Gene Expression Regulation, Developmental/physiology , Guanine Nucleotide Exchange Factors/metabolism , Neuroglia/cytology , Age Factors , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Bromodeoxyuridine/metabolism , Cell Adhesion/genetics , Cell Movement/genetics , Cell Proliferation , Cerebellum/abnormalities , Cerebellum/embryology , Cerebellum/growth & development , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Guanine Nucleotide Exchange Factors/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Histones/metabolism , Integrin beta1/metabolism , Intermediate Filament Proteins/genetics , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/metabolism , Neurons/physiology , Neuropeptides/genetics
7.
Dev Biol ; 351(1): 25-34, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21185282

ABSTRACT

The organization of neocortex, along its radial axis, into a six-layered structure is one of the most exquisite features of the brain. Because of their strategic localization in the marginal zone, and their expression of reelin, a signal that controls spatial ordering of cortical layers, Cajal-Retzius (C-R) cells play a crucial role in cortical patterning along this axis. Yet, it remains less well understood how C-R cell targeting itself is regulated. At the onset of corticogenesis when C-R cells first arrive in the cortex via tangential migration, radial glia (RG) are the main cell type present. This suggests that RG may play a role in C-R cell localization. To test this, we used genetic approaches to perturb RG scaffold during early corticogenesis. We found that disrupting RG endfoot adhesion to basal lamina consistently results in C-R cell displacement. These displacements do not appear to result from primary defects in neural progenitor cell proliferation, deficits in the meninges or basement membrane, or cell autonomous defects in C-R cells. Instead, they show close temporal and spatial correlation with RG endfoot retraction. Moreover, ablation of RG via cell cycle blockade similarly results in local displacement of C-R cells. These lines of evidence thus indicate that, during early corticogenesis, RG play a primary role in regulating spatial targeting of C-R cells. Since RG are also neural progenitors as well as neuronal migration scaffolds, these findings suggest that, during nervous system development, neuroepithelial stem cells may not only be responsible for generating a diverse array of neuronal cell types and facilitating their radial migration. They may also, through regulating the placement of guidepost cells, coordinate spatial patterning of the nervous system along its radial axis.


Subject(s)
Body Patterning , Cerebral Cortex/embryology , Neuroglia/physiology , Animals , Cell Communication , Cerebral Cortex/abnormalities , Cerebral Cortex/cytology , Integrin beta1/physiology , Mice , Origin Recognition Complex/physiology , Reelin Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...