Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 26(6): 997-1007, 2023 06.
Article in English | MEDLINE | ID: mdl-37248337

ABSTRACT

Endocannabinoids are among the most powerful modulators of synaptic transmission throughout the nervous system, and yet little is understood about the release of endocannabinoids from postsynaptic compartments. Here we report an unexpected finding that endocannabinoid release requires synucleins, key contributors to Parkinson's disease. We show that endocannabinoids are released postsynaptically by a synuclein-dependent and SNARE-dependent mechanism. Specifically, we found that synuclein deletion blocks endocannabinoid-dependent synaptic plasticity; this block is reversed by postsynaptic expression of wild-type but not of mutant α-synuclein. Whole-cell recordings and direct optical monitoring of endocannabinoid signaling suggest that the synuclein deletion specifically blocks endocannabinoid release. Given the presynaptic role of synucleins in regulating vesicle lifecycle, we hypothesize that endocannabinoids are released via a membrane interaction mechanism. Consistent with this hypothesis, postsynaptic expression of tetanus toxin light chain, which cleaves synaptobrevin SNAREs, also blocks endocannabinoid-dependent signaling. The unexpected finding that endocannabinoids are released via a synuclein-dependent mechanism is consistent with a general function of synucleins in membrane trafficking and adds a piece to the longstanding puzzle of how neurons release endocannabinoids to induce synaptic plasticity.


Subject(s)
Endocannabinoids , Signal Transduction , Endocannabinoids/metabolism , Endocannabinoids/pharmacology , Signal Transduction/physiology , Synaptic Transmission/physiology , Neurons/physiology , Cell Communication
2.
J Biomol Struct Dyn ; 40(18): 8155-8168, 2022 11.
Article in English | MEDLINE | ID: mdl-33792526

ABSTRACT

Brassica juncea (BJ) is a familiar edible crop, which has been used as a dietary ingredient and to prepare anti-inflammatory/anti-arthritic formulations in Ayurveda. But, the scientific validation or confirmation of its therapeutic properties is very limited. This study was performed to determine the efficiency of BJ leaves for the treatment of Rheumatoid arthritis using in vivo and in silico systems. Standard in vitro procedures was followed to study the total phenolic, flavonoid contents and free radical scavenging ability of the extracts of BJ. The effective extract was screened and the presence of bioactive chemicals was studied using HPLC. Further, the possible therapeutic actions of the BJ active principles against the disease targets were studied using PPI networking and docking analysis. IL2RA, IL18 and VEGFA are found to be the potential RA target and the compounds detected from BJ extract have shown great binding efficiency towards the target from molecular docking study. The resulting complexes were then subject to 100 ns molecular dynamics simulation studies with the GROMACS package to analyze the stability of docked protein-ligand complexes and to assess the fluctuation and conformational changes during protein-ligand interactions. To confirm the anti-arthritic activity of BJ, the extract was tested in CFA-induced arthritic Wistar rats. The test groups administered with BJ extract showed retrieval of altered hematological parameters and substantial recovery from inflammation and degeneration of rat hind paw.Communicated by Ramaswamy H. Sarma.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Interleukin-2 Receptor alpha Subunit/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Flavonoids/pharmacology , Free Radicals , Interleukin-18/therapeutic use , Ligands , Molecular Docking Simulation , Mustard Plant , Plant Extracts/chemistry , Plant Extracts/pharmacology , Rats , Rats, Wistar
3.
Bioorg Chem ; 103: 104227, 2020 10.
Article in English | MEDLINE | ID: mdl-32891004

ABSTRACT

Rheumatoid Arthritis is a chronic, inflammatory, and systemic autoimmune disease, it affects elders worldwide. Herbal medicines have been used for the treatment of various ailments from ancient times. Betelvine (Piper betle L.) leaves have long been used in Asian countries as a medicine to relieve pain and some metabolic diseases. The present study of methanolic extract of phytochemical analysis confirms the presence of alkaloids, tannins, terpenoids, saponins, steroids, total flavonoids and total phenols. GC-MS analysis of MeOH extract of Piper betle (PBME) revealed the presence of 40 bioactive compounds. In vitro antioxidant and anti-inflammatory assays showed greater inhibitory effect. The anti-arthritic effects of PBME at 250 and 500 mg/kg concentration showed recovery from joint damage in in vivo rat model. Among the 40 GC-MS derived bioactives, 4-Allyl-1,2-Diacetoxybenzene exhibited the higher interactions with minimized binding energy to the RA targets of MMP 1 (-6.4 kcal/mol), TGF-ß (-6.9 kcal/mol), IL-1ß (-5.9 kcal/mol). Further, the effect of PBME extract against RA molecular disease targets (IL-1ß, MMP1 and TGF- ß) were studied using Real-time PCR. These results substantiate that P. betle leaves could be a source of therapeutics for the treatment of rheumatoid arthritis.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Piper betle/chemistry , Plant Extracts/therapeutic use , Animals , Antirheumatic Agents/isolation & purification , Antirheumatic Agents/pharmacokinetics , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Female , Free Radical Scavengers/isolation & purification , Free Radical Scavengers/pharmacokinetics , Free Radical Scavengers/therapeutic use , Freund's Adjuvant , Joints/pathology , Molecular Docking Simulation , Plant Extracts/isolation & purification , Plant Extracts/pharmacokinetics , Plant Leaves/chemistry , Rats, Wistar
4.
Environ Int ; 123: 486-500, 2019 02.
Article in English | MEDLINE | ID: mdl-30622074

ABSTRACT

Mankind exposure to chemicals in the past century has increased dramatically throughout environment. There is no question that chemicals interfere with the physiology of biological system. Abundance of chemicals is documented to be detrimental to human and wildlife. The mammalian endocrine system is comprised of many interacting tissues mediate themselves through hormones that are essential for metabolism, growth and development. Humans secrete over fifty different hormones to orchestrate major physiological functions however; these vital functions can be intervened by huge number of internal and external chemical stressors that are identified as endocrine disruptors. Advanced glycation end products (AGEs), familiarly known as Maillard products, formed through non-enzymatic glycation whose production is augmented on aging as well as environmental stressors. Processed foods have become very popular today due to their taste, convenience, and inexpensiveness. Manufacture of these day-to-day foods involves extreme temperatures on processing results in the formation of AGEs could independently promote oxidative stress, aging, diabetes, cancer, degenerative diseases, more fascinatingly hormonal disruption is the subject of interest of this review. Based on some substantial observations documented till time, we discuss the emergence of dietary AGEs as potential endocrine disruptors by emphasizing their occurrence, mechanisms and participation in endocrine interruption. Both economically and in terms of human life, AGEs may represent an enormous cost for the future society. Therefore, by explicating their novel role in endocrine diseases, the review strives to make an impact on AGEs and their exposure among public as well as scientific communities.


Subject(s)
Endocrine Disruptors/adverse effects , Endocrine System/drug effects , Food/adverse effects , Glycation End Products, Advanced/adverse effects , Animals , Diet , Food Handling , Glycation End Products, Advanced/metabolism , Humans , Oxidative Stress , Receptor for Advanced Glycation End Products/metabolism , Social Responsibility
5.
Sci Signal ; 8(384): ra68, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26152695

ABSTRACT

Nitric oxide (NO) is a signaling intermediate during glutamatergic neurotransmission in the central nervous system (CNS). NO signaling is in part accomplished through cysteine S-nitrosylation, a posttranslational modification by which NO regulates protein function and signaling. In our investigation of the protein targets and functional impact of S-nitrosylation in the CNS under physiological conditions, we identified 269 S-nitrosocysteine residues in 136 proteins in the wild-type mouse brain. The number of sites was significantly reduced in the brains of mice lacking endothelial nitric oxide synthase (eNOS(-/-)) or neuronal nitric oxide synthase (nNOS(-/-)). In particular, nNOS(-/-) animals showed decreased S-nitrosylation of proteins that participate in the glutamate/glutamine cycle, a metabolic process by which synaptic glutamate is recycled or oxidized to provide energy. (15)N-glutamine-based metabolomic profiling and enzymatic activity assays indicated that brain extracts from nNOS(-/-) mice converted less glutamate to glutamine and oxidized more glutamate than those from mice of the other genotypes. GLT1 [also known as EAAT2 (excitatory amino acid transporter 2)], a glutamate transporter in astrocytes, was S-nitrosylated at Cys(373) and Cys(561) in wild-type and eNOS(-/-) mice, but not in nNOS(-/-) mice. A form of rat GLT1 that could not be S-nitrosylated at the equivalent sites had increased glutamate uptake compared to wild-type GLT1 in cells exposed to an S-nitrosylating agent. Thus, NO modulates glutamatergic neurotransmission through the selective, nNOS-dependent S-nitrosylation of proteins that govern glutamate transport and metabolism.


Subject(s)
Brain/metabolism , Cysteine/metabolism , Glutamic Acid/metabolism , Nitric Oxide/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Liquid , Cysteine/analogs & derivatives , Cysteine/genetics , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Proteome/metabolism , Proteomics/methods , Rats , S-Nitrosothiols/metabolism , Tandem Mass Spectrometry
6.
J Biol Chem ; 288(37): 26473-9, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23861393

ABSTRACT

NO is a versatile free radical that mediates numerous biological functions within every major organ system. A molecular pathway by which NO accomplishes functional diversity is the selective modification of protein cysteine residues to form S-nitrosocysteine. This post-translational modification, S-nitrosylation, impacts protein function, stability, and location. Despite considerable advances with individual proteins, the in vivo biological chemistry, the structural elements that govern the selective S-nitrosylation of cysteine residues, and the potential overlap with other redox modifications are unknown. In this minireview, we explore the functional features of S-nitrosylation at the proteome level and the structural diversity of endogenously modified residues, and we discuss the potential overlap and complementation that may exist with other cysteine modifications.


Subject(s)
Cysteine/analogs & derivatives , Gene Expression Regulation , Proteins/chemistry , S-Nitrosothiols/chemistry , Animals , Cysteine/chemistry , Humans , Mice , Myocardium/metabolism , Nitric Oxide/chemistry , Nitric Oxide Synthase/metabolism , Nitrogen/chemistry , Oxidation-Reduction , Protein Processing, Post-Translational , Proteomics/methods , Signal Transduction
7.
J Proteomics ; 92: 195-203, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-23748021

ABSTRACT

Cysteine S-nitrosylation is a post-translational modification regulating protein function and nitric oxide signaling. Herein the selectivity, reproducibility, and sensitivity of a mass spectrometry-based proteomic method for the identification of endogenous S-nitrosylated proteins are outlined. The method enriches for either S-nitrosylated proteins or peptides through covalent binding of the cysteine sulfur with phenylmercury at pH=6.0. Phenylmercury reacts selectively and efficiently with S-nitrosocysteine since no reactivity can be documented for disulfides, sulfinic or sulfonic acids, S-glutathionylated, S-alkylated or S-sulfhydrylated cysteine residues. A specificity of 97±1% for the identification of S-nitrosocysteine peptides in mouse liver tissue is achieved by the inclusion of negative controls. The method enables the detection of 36 S-nitrosocysteine peptides starting with 5pmolS-nitrosocysteine/mg of total tissue protein. Both the percentage of protein molecules modified as well as the occupancy by S-nitrosylation can be determined. Overall, selective, sensitive and reproducible enrichment of S-nitrosylated proteins and peptides is achieved by the use of phenylmercury. The inclusion of appropriate negative controls secures the precise identification of endogenous S-nitrosylated sites and proteins in biological samples. BIOLOGICAL SIGNIFICANCE: The current study describes a selective, sensitive and reproducible method for the acquisition of endogenously S-nitrosylated proteins and peptides. The acquisition of endogenous S-nitrosoproteomes provides robust data that is necessary for investigating the mechanism(s) of S-nitrosylation in vivo, the factors that govern its selectivity, the dependency of the modification on different isoforms of nitric oxide synthases (NOS), as well as the physiological functions of this protein modification. This article is part of a Special Issue entitled: Posttranslational Protein modifications in biology and Medicine.


Subject(s)
Cysteine/analogs & derivatives , Mass Spectrometry/methods , Nitric Oxide Synthase/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , S-Nitrosothiols/metabolism , Animals , Cattle , Cysteine/chemistry , Cysteine/metabolism , Mice , Nitric Oxide Synthase/chemistry , Phenylmercury Compounds/chemistry , Proteome/chemistry , Rabbits , S-Nitrosothiols/chemistry , Sensitivity and Specificity
8.
Sci Signal ; 6(256): rs1, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23281369

ABSTRACT

Cysteine S-nitrosylation is a posttranslational modification by which nitric oxide regulates protein function and signaling. Studies of individual proteins have elucidated specific functional roles for S-nitrosylation, but knowledge of the extent of endogenous S-nitrosylation, the sites that are nitrosylated, and the regulatory consequences of S-nitrosylation remains limited. We used mass spectrometry-based methodologies to identify 1011 S-nitrosocysteine residues in 647 proteins in various mouse tissues. We uncovered selective S-nitrosylation of enzymes participating in glycolysis, gluconeogenesis, tricarboxylic acid cycle, and oxidative phosphorylation, indicating that this posttranslational modification may regulate metabolism and mitochondrial bioenergetics. S-nitrosylation of the liver enzyme VLCAD [very long chain acyl-coenzyme A (CoA) dehydrogenase] at Cys(238), which was absent in mice lacking endothelial nitric oxide synthase, improved its catalytic efficiency. These data implicate protein S-nitrosylation in the regulation of ß-oxidation of fatty acids in mitochondria.


Subject(s)
Cysteine/analogs & derivatives , Energy Metabolism/physiology , Fatty Acids/metabolism , Mitochondria/metabolism , Nitric Oxide/metabolism , Protein Processing, Post-Translational/physiology , S-Nitrosothiols/metabolism , Signal Transduction/physiology , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Analysis of Variance , Animals , Cysteine/metabolism , DNA Primers/genetics , Liver/anatomy & histology , Liver/metabolism , Mass Spectrometry , Mice , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Processing, Post-Translational/genetics , Proteomics , Signal Transduction/genetics
9.
Methods ; 62(2): 165-70, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23116708

ABSTRACT

Protein S-nitrosylation is considered as one of the molecular mechanisms by which nitric oxide regulates signaling events and protein function. The present review presents an updated method which allows for the site-specific detection of S-nitrosylated proteins in vivo. The method is based on enrichment of S-nitrosylated proteins or peptides using organomercury compounds followed by LC-MS/MS detection. Technical aspects for determining the reaction and binding efficiency of the mercury resin that assists enrichment of S-nitrosylated proteins are presented and discussed. In addition, emphasis is given to the specificity of the method by providing technical details for the generation of four chemically distinct negative controls. Finally it is provided an overview of the key steps for generation and evaluation of mass spectrometry derived data.


Subject(s)
Cysteine/analogs & derivatives , Proteome/isolation & purification , S-Nitrosothiols/isolation & purification , Animals , Chromatography, Affinity , Cysteine/chemistry , Cysteine/isolation & purification , Cysteine/metabolism , Humans , Muramidase/chemistry , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Protein Binding , Protein Processing, Post-Translational , Proteome/chemistry , Proteome/metabolism , S-Nitrosothiols/chemistry , S-Nitrosothiols/metabolism , Tandem Mass Spectrometry
10.
Biochim Biophys Acta ; 1820(6): 684-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21651963

ABSTRACT

BACKGROUND: A biochemical pathway by which nitric oxide accomplishes functional diversity is the specific modification of protein cysteine residues to form S-nitrosocysteine. This post-translational modification, S-nitrosylation, impacts protein function, interactions and location. However, comprehensive studies exploring protein signaling pathways or interrelated protein clusters that are regulated by S-nitrosylation have not been performed on a global scale. SCOPE OF REVIEW: To provide insights to these important biological questions, sensitive, validated and quantitative proteomic approaches are required. This review summarizes current approaches for the global identification of S-nitrosylated proteins. MAJOR CONCLUSIONS: The application of novel methods for identifying S-nitrosylated proteins, especially when combined with mass-spectrometry based proteomics to provide site-specific identification of the modified cysteine residues, promises to deliver critical clues for the regulatory role of this dynamic posttranslational modification in cellular processes. GENERAL SIGNIFICANCE: Though several studies have established S-nitrosylation as a regulator of protein function in individual proteins, the biological chemistry and the structural elements that govern the specificity of this modification in vivo are vastly unknown. Additionally, a gap in knowledge exists concerning the potential global regulatory role(s) this modification may play in cellular physiology. By further studying S-nitrosylation at a global scale, a greater appreciation of nitric oxide and protein S-nitrosylation in cellular function can be achieved. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/metabolism , Proteins/metabolism , Cysteine/biosynthesis , Mass Spectrometry , Nitric Oxide/metabolism , Nitrosation/physiology , Protein Processing, Post-Translational , Proteins/chemistry , Proteomics , S-Nitrosothiols , Signal Transduction
11.
Mol Imaging Biol ; 11(6): 408-14, 2009.
Article in English | MEDLINE | ID: mdl-19459013

ABSTRACT

PURPOSE: Noninvasive positron emission tomography (PET) imaging of reporter gene is combined with quantitative real-time polymerase reverse transcription (RT-PCR) method to study the time course of death and proliferation of stem cells transplanted in the myocardium. METHODS: Male murine embryonic stem cells (ESCs) were stably transfected with a mutant version of herpes simplex virus type 1 thymidine kinase (HSV1-sr39tk) reporter gene; 5 x 10(6) such cells were injected into the myocardium of female athymic rats. While the transplanted cells was monitored by in vivo 9-(4-[F-18]fluoro-3-hydroxymethylbutyl)guanine ([F-18]FHBG) PET imaging of the heart, their absolute number was estimated by RT-PCR from hearts harvested at 3-5 h, 24 h, days 4, 7, and 14 after transplantation. RESULTS: (1) Forty percent of injected cells were retained in the heart while majority of injected cells were lost within a few hours after injection. Cell death was peaked at 24 h when 18% of donor cells retained in the heart were dead. (2) The substantial cell loss was reversed by significant proliferation of ESCs. This led to the recovery of cell number to 3.4 million (70% of injected dose) at day 4 and first visual observation of in vivo [F-18] signal in the heart. (3) A robust correlation (R (2) = 0.9) between percent of injected dose per gram of tissue derived from in vivo PET signal and the number of donor cells estimated by RT-PCR was revealed. CONCLUSIONS: The time course of transplanted stem cells surviving in the heart reveals a process of substantial cell loss within 24 h of injection and subsequent recovery of cell number through proliferation. Such proliferation can be noninvasively monitored by reporter gene imaging.


Subject(s)
Apoptosis , Cell Proliferation , Embryonic Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation , Myocardium/metabolism , Animals , Cell Death , Cell Survival , Cells, Cultured , Culture Media, Serum-Free , Embryonic Stem Cells/diagnostic imaging , Female , Fluorodeoxyglucose F18/metabolism , Genes, Reporter , Herpesvirus 1, Human/metabolism , In Situ Nick-End Labeling/methods , Kinetics , Male , Mice , Mice, Nude , Mutation , Phantoms, Imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Thymidine Kinase/genetics , Transfection
12.
Dev Biol ; 311(2): 408-22, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17936266

ABSTRACT

The cell adhesion molecule neurofascin (NF) has a major neuronal isoform (NF186) containing a mucin-like domain followed by a fifth fibronectin type III repeat while these domains are absent from glial NF155. Neuronal NF isoforms lacking one or both of these domains are expressed transiently in embryonic dorsal root ganglia (DRG). These two domains are co-expressed in mature NF186, which peaks in expression prior to birth and then persists almost exclusively at nodes of Ranvier on myelinated axons. In contrast, glial NF155 is only detected postnatally with the onset of myelination. All these forms of NF bound homophilically and to Schwann cells but only the mature NF186 isoform inhibits cell adhesion, and this activity may be important in formation of the node of Ranvier. Schwann cells deficient in NF155 myelinated DRG axons in a delayed manner and they showed significantly decreased clustering of both NF and Caspr in regions where paranodes normally form. The combined results suggest that NF186 is expressed prenatally on DRG neurons and it may modulate their adhesive interactions with Schwann cells, which express NF155 postnatally and require it for development of axon-glial paranodal junctions.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules/metabolism , Ganglia, Spinal , Nerve Growth Factors/metabolism , Neuroglia/metabolism , Neurons/metabolism , Protein Isoforms/metabolism , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Coculture Techniques , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Fibronectins/genetics , Fibronectins/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Humans , Myelin Sheath/physiology , Nerve Growth Factors/genetics , Neuroglia/cytology , Neurons/cytology , Protein Isoforms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Schwann Cells/cytology , Schwann Cells/metabolism
13.
Mol Cell Neurosci ; 30(1): 137-48, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16061393

ABSTRACT

Neurofascin (NF) is a neural cell adhesion molecule in the L1-family containing six Ig domains and multiple fibronectin type III (FnIII) repeats in its extracellular region. NF has many splicing variants and two of these are exemplars that have different cellular patterns of expression during development. NF186, which is expressed on neurons, contains an unusual mucin-like region and NF155, which is expressed on glia, contains a unique FnIII repeat with an RGD motif. Analysis of Fc fusion proteins representing different extracellular regions of NF indicate that NF186 inhibits cell adhesion and neurite outgrowth, and the inhibition is associated with the region containing the mucin-like domain. NF155 promotes neural cell adhesion and neurite outgrowth, and the RGD motif in its third FnIII repeat is critical for cell spreading and neurite outgrowth. The results suggest that different splicing variants of NF expressed on neurons and glia play distinct roles during neural development.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurites/physiology , Alternative Splicing , Cell Adhesion Molecules/chemistry , Cell Line , Humans , Kidney/cytology , Mutagenesis , Nerve Growth Factors/chemistry , Neuroglia/cytology , Neuroglia/physiology , Neurons/physiology , Neurons/ultrastructure , Protein Structure, Tertiary , Ranvier's Nodes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...