Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 235
Filter
1.
Biotechnol J ; 19(5): e2300734, 2024 May.
Article in English | MEDLINE | ID: mdl-38719571

ABSTRACT

Self-assembly of biological elements into biomimetic cargo carriers for targeting and delivery is a promising approach. However, it still holds practical challenges. We developed a functionalization approach of DNA origami (DO) nanostructures with neuronal growth factor (NGF) for manipulating neuronal systems. NGF bioactivity and its interactions with the neuronal system were demonstrated in vitro and in vivo models. The DO elements fabricated by molecular self-assembly have manipulated the surrounding environment through static spatially and temporally controlled presentation of ligands to the cell surface receptors. Our data showed effective bioactivity in differentiating PC12 cells in vitro. Furthermore, the DNA origami NGF (DON) affected the growth directionality and spatial capabilities of dorsal root ganglion neurons in culture by introducing a chemotaxis effect along a gradient of functionalized DO structures. Finally, we showed that these elements provide enhanced axonal regeneration in a rat sciatic nerve injury model in vivo. This study is a proof of principle for the functionality of DO in neuronal manipulation and regeneration. The approach proposed here, of an engineered platform formed out of programmable nanoscale elements constructed of DO, could be extended beyond the nervous system and revolutionize the fields of regenerative medicine, tissue engineering, and cell biology.


Subject(s)
DNA , Ganglia, Spinal , Nerve Growth Factor , Nerve Regeneration , Animals , Rats , PC12 Cells , DNA/chemistry , Ganglia, Spinal/cytology , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , Nanostructures/chemistry , Neurons , Sciatic Nerve , Tissue Scaffolds/chemistry , Rats, Sprague-Dawley
2.
Biomed Mater ; 19(3)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38626779

ABSTRACT

It is well-established that multi-scale porous scaffolds can guide axonal growth and facilitate functional restoration after spinal cord injury (SCI). In this study, we developed a novel mussel shell-inspired conductive scaffold for SCI repair with ease of production, multi-scale porous structure, high flexibility, and excellent biocompatibility. By utilizing the reducing properties of polydopamine, non-conductive graphene oxide (GO) was converted into conductive reduced graphene oxide (rGO) and crosslinkedin situwithin the mussel shells.In vitroexperiments confirmed that this multi-scale porous Shell@PDA-GO could serve as structural cues for enhancing cell adhesion, differentiation, and maturation, as well as promoting the electrophysiological development of hippocampal neurons. After transplantation at the injury sites, the Shell@PDA-GO provided a pro-regenerative microenvironment, promoting endogenous neurogenesis, triggering neovascularization, and relieving glial fibrosis formation. Interestingly, the Shell@PDA-GO could induce the release of endogenous growth factors (NGF and NT-3), resulting in the complete regeneration of nerve fibers at 12 weeks. This work provides a feasible strategy for the exploration of conductive multi-scale patterned scaffold to repair SCI.


Subject(s)
Biocompatible Materials , Bivalvia , Graphite , Nerve Regeneration , Polymers , Spinal Cord Injuries , Tissue Scaffolds , Animals , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry , Porosity , Graphite/chemistry , Polymers/chemistry , Biocompatible Materials/chemistry , Indoles/chemistry , Animal Shells/chemistry , Cell Differentiation , Electric Conductivity , Neurons , Rats , Rats, Sprague-Dawley , Cell Adhesion , Neurogenesis , Tissue Engineering/methods , Nerve Growth Factor/metabolism , Nerve Growth Factor/chemistry , Hippocampus
3.
Macromol Biosci ; 24(5): e2300453, 2024 May.
Article in English | MEDLINE | ID: mdl-38224015

ABSTRACT

Spinal cord injuries are very common worldwide, leading to permanent nerve function loss with devastating effects in the affected patients. The challenges and inadequate results in the current clinical treatments are leading scientists to innovative neural regenerative research. Advances in nanoscience and neural tissue engineering have opened new avenues for spinal cord injury (SCI) treatment. In order for designed nerve guidance conduit (NGC) to be functionally useful, it must have ideal scaffold properties and topographic features that promote the linear orientation of damaged axons. In this study, it is aimed to develop channeled polycaprolactone (PCL)/Poly-D,L-lactic-co-glycolic acid (PLGA) hybrid film scaffolds, modify their surfaces by IKVAV pentapeptide/gold nanoparticles (AuNPs) or polypyrrole (PPy) and investigate the behavior of motor neurons on the designed scaffold surfaces in vitro under static/bioreactor conditions. Their potential to promote neural regeneration after implantation into the rat SCI by shaping the film scaffolds modified with neural factors into a tubular form is also examined. It is shown that channeled groups decorated with AuNPs highly promote neurite orientation under bioreactor conditions and also the developed optimal NGC (PCL/PLGA G1-IKVAV/BDNF/NGF-AuNP50) highly regenerates SCI. The results indicate that the designed scaffold can be an ideal candidate for spinal cord regeneration.


Subject(s)
Brain-Derived Neurotrophic Factor , Gold , Metal Nanoparticles , Nerve Growth Factor , Polyesters , Polylactic Acid-Polyglycolic Acid Copolymer , Spinal Cord Injuries , Tissue Scaffolds , Animals , Gold/chemistry , Gold/pharmacology , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Polyesters/chemistry , Polyesters/pharmacology , Rats , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Tissue Scaffolds/chemistry , Nerve Growth Factor/pharmacology , Nerve Growth Factor/chemistry , Nerve Regeneration/drug effects , Oligopeptides/chemistry , Oligopeptides/pharmacology , Rats, Sprague-Dawley
4.
Protein Sci ; 32(2): e4563, 2023 02.
Article in English | MEDLINE | ID: mdl-36605018

ABSTRACT

Nerve growth factor (NGF), the prototypical neurotrophic factor, is involved in the maintenance and growth of specific neuronal populations, whereas its precursor, proNGF, is involved in neuronal apoptosis. Binding of NGF or proNGF to TrkA, p75NTR , and VP10p receptors triggers complex intracellular signaling pathways that can be modulated by endogenous small-molecule ligands. Here, we show by isothermal titration calorimetry and NMR that ATP binds to the intrinsically disordered pro-peptide of proNGF with a micromolar dissociation constant. We demonstrate that Mg2+ , known to play a physiological role in neurons, modulates the ATP/proNGF interaction. An integrative structural biophysics analysis by small angle X-ray scattering and hydrogen-deuterium exchange mass spectrometry unveils that ATP binding induces a conformational rearrangement of the flexible pro-peptide domain of proNGF. This suggests that ATP may act as an allosteric modulator of the overall proNGF conformation, whose likely distinct biological activity may ultimately affect its physiological homeostasis.


Subject(s)
Nerve Growth Factor , Neurons , Nerve Growth Factor/chemistry , Nerve Growth Factor/metabolism , Protein Domains , Neurons/metabolism , Adenosine Triphosphate
5.
ACS Appl Mater Interfaces ; 14(3): 3701-3715, 2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35006667

ABSTRACT

Scarless spinal cord regeneration remains a challenge due to the complicated microenvironment at lesion sites. In this study, the nerve growth factor (NGF) was immobilized in silk protein nanofiber hydrogels with hierarchical anisotropic microstructures to fabricate bioactive systems that provide multiple physical and biological cues to address spinal cord injury (SCI). The NGF maintained bioactivity inside the hydrogels and regulated the neuronal/astroglial differentiation of neural stem cells. The aligned microstructures facilitated the migration and orientation of cells, which further stimulated angiogenesis and neuron extensions both in vitro and in vivo. In a severe rat long-span hemisection SCI model, these hydrogel matrices reduced scar formation and achieved the scarless repair of the spinal cord and effective recovery of motor functions. Histological analysis confirmed the directional regenerated neuronal tissues, with a similar morphology to that of the normal spinal cord. The in vitro and in vivo results showed promising utility for these NGF-laden silk hydrogels for spinal cord regeneration while also demonstrating the feasibility of cell-free bioactive matrices with multiple cues to regulate endogenous cell responses.


Subject(s)
Biocompatible Materials/pharmacology , Hydrogels/pharmacology , Nanofibers/chemistry , Nerve Growth Factor/chemistry , Silk/chemistry , Spinal Cord Regeneration/drug effects , Animals , Astrocytes/drug effects , Astrocytes/pathology , Biocompatible Materials/chemistry , Cell Differentiation/drug effects , Hydrogels/chemistry , Materials Testing , Neurons/drug effects , Neurons/pathology , PC12 Cells , Rats , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Tissue Scaffolds/chemistry
6.
Cells ; 10(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34943971

ABSTRACT

Experiments with cell cultures and animal models have provided solid support for the assumption that Nerve Growth Factor (NGF) plays a key role in the regulation of neuronal cell survival and death. Recently, endogenous ligands have been proposed as physiological modulators of NGF biological activity as part of this regulatory cascade. However, the structural and mechanistic determinants for NGF bioactivity remain to be elucidated. We recently unveiled, by an integrated structural biology approach, the ATP binding sites of NGF and investigated the effects on TrkA and p75NTR receptors binding. These results pinpoint ATP as a genuine endogenous modulator of NGF signaling, paving the way to the characterization of not-yet-identified chemical diverse endogenous biological active small molecules as novel modulators of NGF. The present review aims at providing an overview of the currently available 3D structures of NGF in complex with different small endogenous ligands, featuring the molecular footprints of the small molecules binding. This knowledge is essential for further understanding the functional role of small endogenous ligands in the modulation of neurotrophins signaling in physiological and pathological conditions and for better exploiting the therapeutic potentialities of NGF.


Subject(s)
Nerve Growth Factor/ultrastructure , Nerve Growth Factors/ultrastructure , Receptor, Nerve Growth Factor/genetics , Receptor, trkA/genetics , Animals , Binding Sites/genetics , Humans , Ligands , Nerve Growth Factor/chemistry , Nerve Growth Factor/genetics , Nerve Growth Factors/genetics , Neurons/metabolism , Neurons/ultrastructure , PC12 Cells , Protein Binding/genetics , Rats , Signal Transduction/genetics
7.
PLoS One ; 16(12): e0260496, 2021.
Article in English | MEDLINE | ID: mdl-34851989

ABSTRACT

The venomic profile of Asian mountain pit viper Ovophis monticola is clarified in the present study. Using mass spectrometry-based proteomics, 247 different proteins were identified in crude venom of O. monticola found in Thailand. The most abundant proteins were snake venom metalloproteases (SVMP) (36.8%), snake venom serine proteases (SVSP) (31.1%), and phospholipases A2 (PLA2) (12.1%). Less abundant proteins included L-amino acid oxidase (LAAO) (5.7%), venom nerve growth factor (3.6%), nucleic acid degrading enzymes (3.2%), C-type lectins (CTL) (1.6%), cysteine-rich secretory proteins (CRISP) (1.2%) and disintegrin (1.2%). The immunoreactivity of this viper's venom to a monovalent antivenom against green pit viper Trimeresurus albolabris, or to a polyvalent antivenom against hemotoxic venom was investigated by indirect ELISA and two-dimensional (2D) immunoblotting. Polyvalent antivenom showed substantially greater reactivity levels than monovalent antivenom. A titer for the monovalent antivenom was over 1:1.28x107 dilution while that of polyvalent antivenom was 1:5.12x107. Of a total of 89 spots comprising 173 proteins, 40 spots of predominantly SVMP, SVSP and PLA2 were specific antigens for antivenoms. The 49 unrecognized spots containing 72 proteins were characterized as non-reactive proteins, and included certain types of CTLs and CRISPs. These neglected venom constituents could limit the effectiveness of antivenom-based therapy currently available for victims of pit viper envenomation.


Subject(s)
Proteome/chemistry , Animals , Disintegrins/chemistry , L-Amino Acid Oxidase/chemistry , Lectins, C-Type/chemistry , Mass Spectrometry , Metalloproteases/chemistry , Nerve Growth Factor/chemistry , Phospholipases A2/chemistry , Proteomics , Serine Proteases/chemistry , Thailand , Trimeresurus
8.
J Mater Chem B ; 9(37): 7835-7847, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34586144

ABSTRACT

Alzheimer's disease (AD) is an incurable neurodegenerative disease. Repairing damaged nerves and promoting nerve regeneration are key ways to relieve AD symptoms. However, due to the lack of effective strategies to deliver nerve growth factor (NGF) to the brain, achieving neuron regeneration is a major challenge for curing AD. Herein, a ROS-responsive ruthenium nanoplatform (R@NGF-Se-Se-Ru) drug delivery system for AD management by promoting neuron regeneration and Aß clearance was investigated. Under near-infrared (NIR) irradiation, nanoclusters have good photothermal properties, which can effectively inhibit the aggregation of Aß and disaggregate Aß fibrils. Interestingly, the diselenide bond in the nanoclusters is broken, and the nanoclusters are degraded into small ruthenium nanoparticles in the high reactive oxygen species (ROS) environment of the diseased area. Besides, NGF can promote neuronal regeneration and repair damaged nerves. Furthermore, R@NGF-Se-Se-Ru efficiently crosses the blood-brain barrier (BBB) owing to the covalently grafted target peptides of RVG (R). In vivo studies demonstrate that R@NGF-Se-Se-Ru nanoclusters decrease Aß deposits, inhibit Aß-induced cytotoxicity, and promote neurite outgrowth. The study confirms that promoting both Aß clearance and neuron regeneration is an important therapeutic target for anti-AD drugs and provides a novel insight for AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Drug Carriers/chemistry , Nanostructures/chemistry , Nerve Growth Factor/therapeutic use , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biocompatible Materials/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Hemolysis/drug effects , Humans , Infrared Rays , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , Nerve Regeneration/drug effects , Neuronal Outgrowth/drug effects , Reactive Oxygen Species/metabolism , Ruthenium/chemistry , Selenium/chemistry
9.
Food Funct ; 12(17): 7676-7687, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34259275

ABSTRACT

The aim of the study was to evaluate the neuroprotective function of sea cucumber ovum peptide-derived NDEELNK and explore the underlying molecular mechanisms. NDEELNK exerted the neuroprotective effect by improving the acetylcholine (ACh) level and reducing the acetylcholinesterase (AChE) activity in PC12 cells. By molecular docking, we confirmed that the NDEELNK backbone and AChE interacted through hydrophobic and hydrogen bonds in contact with the amino acid residues of the cavity wall. NDEELNK increased superoxide dismutase (SOD) activity and decreased reactive oxygen species (ROS) production, thereby reducing mitochondrial dysfunction and enhancing energy metabolism. Our results demonstrated that NDEELNK supplementation alleviated scopolamine-induced PC12 cell damage by improving the cholinergic system, increasing energy metabolism and upregulating the expression of phosphorylated protein kinase A (p-PKA), brain-derived neurotrophic factor (BNDF) and nerve growth factor (NGF) signaling proteins in in vitro experiments. These results demonstrated that the sea cucumber ovum peptide-derived NDEELNK might play a protective role in PC12 cells.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Memory Disorders/metabolism , Nerve Growth Factor/metabolism , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Scopolamine/adverse effects , Sea Cucumbers/chemistry , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Animals , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/genetics , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Energy Metabolism/drug effects , Humans , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Memory Disorders/genetics , Molecular Docking Simulation , Nerve Growth Factor/chemistry , Nerve Growth Factor/genetics , Oxidative Stress/drug effects , PC12 Cells , Rats , Up-Regulation/drug effects
10.
J Biomater Appl ; 36(2): 276-288, 2021 08.
Article in English | MEDLINE | ID: mdl-34167336

ABSTRACT

Spinal cord injury (SCI) is an extremely destructive central nervous system lesion. Studies have shown that NGF can promote nerve regeneration after SCI. However, it cannot produce the desired effect due to its stability in the body and is difficulty in passing through the blood-brain barrier. In this study, we prepared nanovesicles derived from macrophage membrane encapsulating NGF (NGF-NVs) as a drug carrier for the treatment of SCI. Cell experiments showed that NGF-NVs were effectively taken up by PC12 cells and inhibited neuronal apoptosis. In vivo imaging experiments, a large quantity of NGF was delivered to the injured site with the aid of the good targeting of NVs. In animal experiments, NGF-NVs improved the survival of neurons by significantly activating the PI3K/AKT signaling pathway and had good behavioral and histological recovery effects after SCI. Therefore, NVs are a potential drug delivery vector for SCI therapy.


Subject(s)
Apoptosis , Macrophages/chemistry , Nanostructures/chemistry , Nerve Growth Factor/chemistry , Animals , Apoptosis/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biocompatible Materials/pharmacology , Cell Line , Cell Membrane/chemistry , Cell Survival/drug effects , Disease Models, Animal , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/pharmacology , Nerve Growth Factor/therapeutic use , Neurons/cytology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Spinal Cord Injuries/therapy , Tissue Distribution
11.
Angew Chem Int Ed Engl ; 60(22): 12319-12322, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33770418

ABSTRACT

We herein develop a concentration gradient generator (CGG) on a microfluidic chip for diluting different nanoparticles. Specifically designed compact disk (CD)-shaped microchannels in the CGG module could thoroughly mix the flowing solutions and generate a linear concentration gradient of nanoparticles without aggregation. We combine the CGG with a single-cell trapper array (SCA) on microfluidics to evaluate the concentration-dependent bioeffects of the nanoparticles. The precise control of the spatiotemporal generation of nanoparticle concentration on the CGG module and the single-cell-level monitoring of the cell behaviors on the SCA module by a high-content system in real time, render the CGG-SCA system a highly precise platform, which can exclude the average effect of cell population and reflect the response of individual cells to the gradient concentrations accurately. In addition, the CGG-SCA system provides an automated platform for high-throughput screening of nanomedicines with high precision and low sample consumption.


Subject(s)
High-Throughput Screening Assays/methods , Nanoparticles/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/pharmacology , Humans , Lipids/chemistry , Microfluidics , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Single-Cell Analysis
12.
World J Microbiol Biotechnol ; 36(12): 176, 2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33103226

ABSTRACT

Nerve growth factor (NGF) is an essential trophic factor for the growth and survival of neurons in the central and peripheral nervous systems. For many years, mouse NGF (mNGF) has been used to treat various neuronal and non-neuronal disorders. However, the biological activity of human NGF (hNGF) is significantly higher than that of mNGF in human cells. Using the CRISPR/Cas9 system, we constructed the transgenic mice expressing hNGF specifically in their submandibular glands. As demonstrated by fluorescence immunohistochemical staining, these mice produced hNGF successfully, with 0.8 mg produced per gram of submandibular glands. hNGF with 99% purity was successfully extracted by two-step ion-exchange chromatography and one-step size-exclusion chromatography from the submandibular glands of these transgenic mice. Further, the purified hNGF was verified by LC-MS/MS. We analyzed the NH2-terminus of hNGF using both Edman degradation and LC-MS/MS-based methods. Both results showed that the obtained hNGF lost the NH2-terminal octapeptide (SSSHPIFH). Moreover, the produced hNGF demonstrated a strong promotion in the proliferation of TF1 cells.


Subject(s)
Gene Editing/methods , Nerve Growth Factor/isolation & purification , Nerve Growth Factor/metabolism , Submandibular Gland/metabolism , Animals , Cell Line , Cell Proliferation , Chromatography, Gel , Chromatography, Ion Exchange , Humans , Mice , Mice, Transgenic , Nerve Growth Factor/chemistry , Nerve Growth Factor/genetics , Protein Domains , Protein Engineering
13.
Neurochem Int ; 140: 104812, 2020 11.
Article in English | MEDLINE | ID: mdl-32758589

ABSTRACT

The precursor of Nerve Growth Factor (proNGF) is the predominant form of NGF in the brain, where its tissue levels are increased in neurodegenerative diseases. proNGF exists in two main splicing variants, the long proNGF-A and the short proNGF-B. We demonstrated that proNGF-B is selectively increased in the hippocampus of rats affected by early diabetic encephalopathy and that native, purified proNGFs elicit different responses when used to stimulate PC12 cells. Therefore, the evaluation of the proNGF-B/proNGF-A ratio may be of important diagnostic and prognostic value in pathologies characterized by dysfunctions of NGF system. To date there is not clear pharmacological characterization of the different proNGFs variants, due to the lack of a proper recombinant proNGF-A. Using a bioinformatics approach, we predicted aminoacid sites involved in proNGF-A intracellular cleavage/conversion into proNGF-B, we cloned and expressed non-cleavable proNGF-A in HeLa cells and pursued a first characterization of their secretion modalities. Finally, we studied the biological effects of different proNGF-A mutants, stimulating PC12 cells with conditioned media from transfected HeLa cells. Based on our results, we propose the A73Y mutation as essential to obtaining an intact proNGF-A, limiting its conversion to proNGF-B. proNGF-A A73Y is probably released in an activity dependent manner and, when supplied to PC12 cells, shows a moderate differentiative capacity opposed to high neuroprotective potential. This preliminary study lays the foundation for future research aimed at uncovering the selective biological activities of proNGF-A and proNGF-B, and at developing pharmacological treatments that target the unbalance of proNGF system, induced by neurodegeneration.


Subject(s)
Computational Biology/methods , Genetic Variation/genetics , Nerve Growth Factor/genetics , Protein Precursors/genetics , Recombinant Proteins/genetics , Animals , Base Sequence , HeLa Cells , Humans , Nerve Growth Factor/chemistry , PC12 Cells , Protein Precursors/chemistry , Rats , Recombinant Proteins/chemistry
14.
J Mater Chem B ; 8(32): 7275-7287, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32638822

ABSTRACT

Easily assembled and biocompatible chitosan/hyaluronic acid nanoparticles with multiple stimuli-responsive ability are ideally suited for efficient delivery of therapeutic agents under specific endogenous triggers. We report a simple and versatile strategy to formulate oxidative stress and pH-responsive chitosan/hyaluronic acid nanocarriers with high encapsulation efficiencies of small drug molecules and nerve growth factor protein. This is achieved through invoking the dual role of a thioketal-based weak organic acid to disperse and functionalize low molecular weight chitosan in one-pot. Thioketal embedded chitosan/hyaluronic acid nanostructures respond to oxidative stress and show controlled release of quercetin, curcumin and NGF. Lowering the pH in the buffer solution led to higher quercetin release from NPs than at physiological pH, and mimicked the nanoparticle behavior in the environment of early to late endosomes. Curcumin and quercetin loaded NPs killed glioblastoma cells with high efficiency, and NGF-loaded nanoparticles retained biological activity of the protein and increased peripheral nerve outgrowth in explanted mouse dorsal root ganglia.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Drug Design , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Nerve Growth Factor/chemistry , Animals , Buffers , Ganglia, Spinal/drug effects , Ganglia, Spinal/growth & development , Mice , Nerve Growth Factor/pharmacology , Oxidative Stress/drug effects
15.
Biochimie ; 176: 31-44, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32585227

ABSTRACT

Nerve growth factor (NGF) is a minor and neglected component of snake venom. Present study describes the purification and characterization of a NGF isoform (RVV-NGFa) from Indian Russell's viper venom (RVV). RVV-NGFa showed a protonated molecular ion [MH+] at m/z 17388.725 Da. The RVV-NGFa induced neuritogenesis in PC-12 cells but did not show cytotoxicity in mammalian cells, hemolytic activity, platelet modulation, and interference in blood coagulation system which are the characteristic pharmacological properties of RVV. By ELISA and immunofluorescence microplate reader assay, the RVV-NGFa showed appreciable binding to TrkA receptor expressed in breast cancer MDA-MB-231 and MCF-7 cells; nevertheless, pre-incubation of cells with anti-TrkA (and not TrkB or TrkC) or anti-p75NTR antibody significantly decreased (p < 0.05) this binding. The RVV-NGFa demonstrated insignificant binding with non-cancerous cells (HEK-293, L6) lacking TrkA receptor. The binding of RVV-NGFa to TrkA receptor of breast cancer cells resulted in internalization of ligand (RVV-NGFa)-receptor (TrkA) complex to cell cytoplasm in a time-dependent manner. The spectrofluorometric study demonstrated an interaction between RVV-NGFa and cytosolic domain of the purified TrkA receptor. The fluorescence (FITC)-tagged RVV-NGFa depicted a strong fluorescence signal that was observed under a fluorescence microscope and determined by fluorescence microplate reader assay post binding to breast cancer cells; but no fluorescence signal was detected after incubating FITC-RVV-NGFa with non-cancerous L6 and HEK-293 cells. The clinical application of FITC/fluorescence nanoparticle tagged RVV-NGFa for the ex vivo and in vivo diagnosis of breast cancer is highly promising.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Nanoparticles , Neoplasm Proteins/biosynthesis , Nerve Growth Factor , Optical Imaging , Receptor, trkA/biosynthesis , Viper Venoms , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/enzymology , Female , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/pharmacology , HEK293 Cells , Humans , MCF-7 Cells , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , PC12 Cells , Rats , Staining and Labeling , Viper Venoms/chemistry , Viper Venoms/pharmacology
16.
PLoS One ; 15(6): e0231542, 2020.
Article in English | MEDLINE | ID: mdl-32497034

ABSTRACT

Many receptors elicit signal transduction by activating multiple intracellular pathways. This transduction can be triggered by a non-specific ligand, which simultaneously activates all the signaling pathways of the receptors. However, the binding of one biased ligand preferentially trigger one pathway over another, in a process called biased signaling. The identification the functional motions related to each of these distinct pathways has a direct impact on the development of new effective and specific drugs. We show here how to detect specific functional motions by considering the case of the NGF/TrkA-Ig2 complex. NGF-mediated TrkA receptor activation is dependent on specific structural motions that trigger the neuronal growth, development, and survival of neurons in nervous system. The R221W mutation in the ngf gene impairs nociceptive signaling. We discuss how the large-scale structural effects of this mutation lead to the suppression of collective motions necessary to induce TrkA activation of nociceptive signaling. Our results suggest that subtle changes in the NGF interaction network due to the point mutation are sufficient to inhibit the motions of TrkA receptors putatively linked to nociception. The methodological approach presented in this article, based jointly on the normal mode analysis and the experimentally observed functional alterations due to point mutations provides an essential tool to reveal the structural changes and motions linked to the disease, which in turn could be necessary for a drug design study.


Subject(s)
Models, Molecular , Nerve Growth Factor/metabolism , Point Mutation , Receptor, trkA/genetics , Receptor, trkA/metabolism , Signal Transduction , Movement , Nerve Growth Factor/chemistry , Protein Binding , Protein Conformation , Receptor, trkA/chemistry
17.
Adv Mater ; 32(25): e1908299, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32390195

ABSTRACT

Three-dimensional (3D) control over the placement of bioactive cues is fundamental to understand cell guidance and develop engineered tissues. Two-photon patterning (2PP) provides such placement at micro- to millimeter scale, but nonspecific interactions between proteins and functionalized extracellular matrices (ECMs) restrict its use. Here, a 2PP system based on nonfouling hydrophilic photocages and Sortase A (SA)-based enzymatic coupling is presented, which offers unprecedented orthogonality and signal-to-noise ratio in both inert hydrogels and complex mammalian matrices. Improved photocaged peptide synthesis and protein functionalization protocols with broad applicability are introduced. Importantly, the method enables 2PP in a single step in the presence of fragile biomolecules and cells, and is compatible with time-controlled growth factor presentation. As a corollary, the guidance of axons through 3D-patterned nerve growth factor (NGF) within brain-mimetic ECMs is demonstrated. The approach allows for the interrogation of the role of complex signaling molecules in 3D matrices, thus helping to better understand biological guidance in tissue development and regeneration.


Subject(s)
Extracellular Matrix/chemistry , Nerve Growth Factor/chemistry , Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Animals , Axons/chemistry , Axons/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Coumarins/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Extracellular Matrix/metabolism , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Microscopy, Fluorescence, Multiphoton , Nerve Growth Factor/metabolism , Photons
18.
Adv Biosyst ; 4(7): e2000047, 2020 07.
Article in English | MEDLINE | ID: mdl-32462818

ABSTRACT

Sulfated glycosaminoglycans (sGAGs) are vital molecules of the extracellular matrix (ECM) of the nervous system known to regulate proliferation, migration, and differentiation of neurons mainly through binding relevant growth factors. Alginate sulfate (AlgSulf) mimics sGAGs and binds growth factors such as basic fibroblast growth factor (FGF-2). Here, thin films of biotinylated AlgSulf (b-AlgSulfn ) are engineered with sulfation degrees (DS = 0.0 and 2.7) and the effect of polysaccharide concentration on FGF-2 and nerve growth factor (ß-NGF) binding and subsequent primary neural viability and neurite outgrowth is assessed. An increase in b-AlgSulfn concentration results in higher FGF-2 and ß-NGF binding as demonstrated by greater frequency and dissipation shifts measured with quartz crystal microbalance with dissipation monitoring (QCM-D). Primary neurons seeded on the 2D b-AlgSulfn films maintain high viability comparable to positive controls grown on poly-d-lysine. Neurons grown in 3D AlgSulf hydrogels (DS = 0.8) exhibit a significantly higher viability, neurite numbers and mean branch length compared to neurons grown in nonsulfated controls. Finally, a first step is made toward constructing 3D neuronal networks by controllably patterning neurons encapsulated in AlgSulf into an alginate carrier. The substrates and neural networks developed in the current study can be used in basic and applied neural applications.


Subject(s)
Alginates/chemistry , Fibroblast Growth Factor 2/chemistry , Nerve Growth Factor/chemistry , Nerve Net/metabolism , Neurons/metabolism , Animals , Primary Cell Culture , Rats , Rats, Sprague-Dawley
19.
J Mater Chem B ; 8(13): 2673-2688, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32147674

ABSTRACT

An effective treatment for spinal cord injury (SCI) remains a severe clinical challenge due to the intrinsically limited regenerative capacity and complex anatomical structure of the spinal cord. The combination of biomaterials, which serve as scaffolds for axonal growth, cells and neurotrophic factors, is an excellent candidate for spinal cord regeneration. Herein, a new micropatterned conductive electrospun nanofiber mesh was constructed with poly{[aniline tetramer methacrylamide]-co-[dopamine methacrylamide]-co-[poly(ethylene glycol) methyl ether methacrylate]}/PCL (PCAT) using a rotation electrospinning technology. The aim was to study the synergistic effects of electrical stimulation (ES) and a micropatterned conductive electrospun nanofiber mesh incorporated with nerve growth factor (NGF) on the differentiation of rat nerve stem cells (NSCs). The hydrophilicity of the conductive nanofiber mesh could be tailored by changing the dopamine (DA) and aniline tetramer (AT) content from 19° to 79°. A favorable electroactivity and conductivity was achieved by the AT segment of PCAT. The as-fabricated micropatterned electrospun nanofiber mesh possessed a regularly aligned valley and ridge structure, and the diameter of the nanofiber was 312 ± 58 nm, while the width of the valley and ridge was measured to be 210 ± 17 µm and 200 ± 16 µm, respectively. The growth and neurite outgrowth of differentiated NSCs were observed along the valley of the micropatterned nanofiber mesh. In addition, the NGF loaded micropatterned conductive electrospun nanofiber mesh combined with ES exhibited the highest cell viability, and effectively facilitated the differentiation of NSCs into neurons and suppressed the formation of astrocytes, thus exhibiting a great application potential for nerve tissue engineering.


Subject(s)
Nanofibers/chemistry , Nerve Regeneration , Neural Stem Cells/cytology , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Differentiation , Cells, Cultured , Electric Conductivity , Electric Stimulation , Mice , Molecular Structure , NIH 3T3 Cells , Nerve Growth Factor/chemistry , PC12 Cells , Particle Size , Polymers/chemistry , Rats , Surface Properties
20.
Biomolecules ; 10(2)2020 02 01.
Article in English | MEDLINE | ID: mdl-32024191

ABSTRACT

In the last decade, Nerve Growth Factor (NGF)-based clinical approaches have lacked specific and efficient Tyrosine Kinase A (TrkA) agonists for brain delivery. Nowadays, the characterization of novel small peptidomimetic is taking centre stage in preclinical studies, in order to overcome the main size-related limitation in brain delivery of NGF holoprotein for Central Nervous System (CNS) pathologies. Here we investigated the NGF mimetic properties of the human NGF 1-14 sequence (hNGF1-14) and its derivatives, by resorting to primary cholinergic and dorsal root ganglia (DRG) neurons. Briefly, we observed that: 1) hNGF1-14 peptides engage the NGF pathway through TrkA phosphorylation at tyrosine 490 (Y490), and activation of ShcC/PI3K and Plc-γ/MAPK signalling, promoting AKT-dependent survival and CREB-driven neuronal activity, as seen by levels of the immediate early gene c-Fos, of the cholinergic marker Choline Acetyltransferase (ChAT), and of Brain Derived Neurotrophic Factor (BDNF); 2) their NGF mimetic activity is lost upon selective TrkA inhibition by means of GW441756; 3) hNGF1-14 peptides are able to sustain DRG survival and differentiation in absence of NGF. Furthermore, the acetylated derivative Ac-hNGF1-14 demonstrated an optimal NGF mimetic activity in both neuronal paradigms and an electrophysiological profile similar to NGF in cholinergic neurons. Cumulatively, the findings here reported pinpoint the hNGF1-14 peptide, and in particular its acetylated derivative, as novel, specific and low molecular weight TrkA specific agonists in both CNS and PNS primary neurons.


Subject(s)
Cholinergic Neurons/metabolism , Ganglia, Spinal/metabolism , Nerve Growth Factor/chemistry , Receptor, trkA/agonists , Receptor, trkA/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , Animals , Biological Assay , Cell Differentiation , Cell Survival , Cells, Cultured , Humans , Peptides/chemistry , Phosphorylation , Rats , Signal Transduction , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...