Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Small ; 20(23): e2309793, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38148305

ABSTRACT

The nerve guidance conduits incorporated with stem cells, which can differentiate into the Schwann cells (SCs) to facilitate myelination, shows great promise for repairing the severe peripheral nerve injury. The innovation of advanced hydrogel materials encapsulating stem cells, is highly demanded for generating supportive scaffolds and adaptive microenvironment for nerve regeneration. Herein, this work demonstrates a novel strategy in regulating regenerative microenvironment for peripheral nerve repair with a biodegradable conductive hydrogel scaffold, which can offer multifunctional capabilities in immune regulation, enhancing angiogenesis, driving SCs differentiation, and promoting axon regrowth. The biodegradable conductive hydrogel is constructed by incorporation of polydopamine-modified silicon phosphorus (SiP@PDA) nanosheets into a mixture of methacryloyl gelatin and decellularized extracellular matrix (GelMA/ECM). The biomimetic electrical microenvironment performs an efficacious strategy to facilitate macrophage polarization toward a pro-healing phenotype (M2), meanwhile the conductive hydrogel supports vascularization in regenerated tissue through sustained Si element release. Furthermore, the MSCs 3D-cultured in GelMA/ECM-SiP@PDA conductive hydrogel exhibits significantly increased expression of genes associated with SC-like cell differentiation, thus facilitating the myelination and axonal regeneration. Collectively, both the in vitro and in vivo studies demonstrates that the rationally designed biodegradable multifunctional hydrogel significantly enhances nerve tissues repair.


Subject(s)
Hydrogels , Nerve Regeneration , Hydrogels/chemistry , Animals , Nerve Regeneration/drug effects , Cell Differentiation/drug effects , Gelatin/chemistry , Polymers/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Schwann Cells/cytology , Schwann Cells/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Mice , Tissue Scaffolds/chemistry , Stem Cells/cytology , Electric Conductivity , Indoles/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Methacrylates
2.
J Mater Sci Mater Med ; 35(1): 42, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073469

ABSTRACT

Studies have shown that the inhibition of phosphatase and tensin homolog deleted on chromosome 10 (PTEN)was neuroprotective against ischemia/reperfusion(I/R) injury. Bisperoxovanadium (bpV), a derivative of vanadate, is a well-established inhibitor of PTEN. However, its function islimited due to its general inadequacy in penetrating cell membranes. Mxene(Ti3C2Tx) is a novel two-dimensional lamellar nanomaterial with an excellent ability to penetrate the cell membrane. Yet, the effects of this nanomaterial on nervous system diseases have yet to be scrutinized. Here, Mxene(Ti3C2Tx) was used for the first time to carry bpV(HOpic), creating a new nanocomposite Mxene-bpV that was probed in a cerebral I/R injury model. The findings showed that this synthetic Mxene-bpV was adequately stable and can cross the cell membraneeasily. We observed that Mxene-bpV treatment significantly increased the survival rate of oxygen glucose deprivation/reperfusion(OGD/R)--insulted neurons, reduced infarct sizes and promoted the recovery of brain function after mice cerebral I/R injury. Crucially, Mxene-bpV treatment was more therapeutically efficient than bpV(HOpic) treatment alone over the same period. Mechanistically, Mxene-bpV inhibited the enzyme activity of PTEN in vitro and in vivo. It also promoted the expression of phospho-Akt (Ser473) by repressing PTEN and then activated the Akt pathway to boost cell survival. Additionally, in PTEN transgenic mice, Mxene-bpV suppressed I/R-induced inflammatory response by promoting M2 microglial polarization through PTEN inhibition. Collectively, the nanosynthetic Mxene-bpV inhibited PTEN' enzymatic activity by activating Akt pathway and promoting M2 microglial polarization, and finally exerted neuroprotection against cerebral I/R injury.


Subject(s)
Microglia , Neuroprotective Agents , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Reperfusion Injury , Signal Transduction , Vanadium Compounds , Animals , Microglia/drug effects , Microglia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Neuroprotective Agents/pharmacology , Vanadium Compounds/pharmacology , Vanadium Compounds/chemistry , PTEN Phosphohydrolase/metabolism , Male , Mice, Inbred C57BL , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell Polarity/drug effects , Neurons/drug effects , Neurons/metabolism , Nanocomposites/chemistry
3.
J Biol Chem ; 294(13): 4946-4955, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30709906

ABSTRACT

Hypoxia-inducible factor-2α (HIF2α) is a nuclear transcription factor that plays a critical role in cell survival including metabolic adaptation under hypoxia as well as normoxia, but whether HIF2α contributes to the control of whole-body metabolic balance is unclear. In this study, we found that the hypothalamic HIF2α protein level rapidly increases in young mice that are centrally stimulated with insulin. However, this insulin-induced HIF2α up-regulation is substantially attenuated in mice of advanced age. This attenuation is comparable with the effect of high-calorie feeding in young mice. Of note, unlike high-calorie feeding conditions, age-dependent HIF2α attenuation occurs without impaired activation of the hypothalamic IR/IRS-2/AKT/FOXO1 pathway in response to insulin. Molecular and physiological analyses revealed that hypothalamic HIF2α contributes to the action of central insulin in regulation of proopiomelanocortin (Pomc) gene expression and food intake. HIF2α knockout in POMC neurons led to age-dependent excess weight gain and fat increase, a phenotype that was associated with a mild degree of glucose intolerance and insulin resistance. In conclusion, hypothalamic HIF2α responds to insulin, and the up-regulation is involved in adaptive metabolic regulation as age increases, whereas impairment of HIF2α in the hypothalamus contributes to weight gain and glucose disorders in age-dependent manners.


Subject(s)
Aging/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Glucose Intolerance/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Signal Transduction , Aging/genetics , Aging/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Hypothalamus/pathology , Insulin/genetics , Mice , Mice, Transgenic , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics
4.
J Cell Mol Med ; 22(1): 25-37, 2018 01.
Article in English | MEDLINE | ID: mdl-29063730

ABSTRACT

Spinal cord injury (SCI) possesses a significant health and economic burden worldwide. Traumatic SCI is a devastating condition that evolves through two successive stages. Throughout each of these stages, disturbances in ionic homeostasis, local oedema, ischaemia, focal haemorrhage, free radicals stress and inflammatory response were observed. Although there are no fully restorative cures available for SCI patients, various molecular, cellular and rehabilitative therapies, such as limiting local inflammation, preventing secondary cell death and enhancing the plasticity of local circuits in the spinal cord, were described. Current preclinical studies have showed that fibroblast growth factors (FGFs) alone or combination therapies utilizing cell transplantation and biomaterial scaffolds are proven effective for treating SCI in animal models. More importantly, some studies further demonstrated a paucity of clinical transfer usage to promote functional recovery of numerous patients with SCI. In this review, we focus on the therapeutic capacity and pitfalls of the FGF family and its clinical application for treating SCI, including the signalling component of the FGF pathway and the role in the central nervous system, the pathophysiology of SCI and the targets for FGF treatment. We also discuss the challenges and potential for the clinical translation of FGF-based approaches into treatments for SCI.


Subject(s)
Fibroblast Growth Factors/therapeutic use , Spinal Cord Injuries/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , Humans , Molecular Targeted Therapy , Spinal Cord Injuries/physiopathology
5.
J Cell Mol Med ; 22(6): 3086-3096, 2018 06.
Article in English | MEDLINE | ID: mdl-29575654

ABSTRACT

Spermidine has therapeutic effects in many diseases including as heart diastolic function, myopathic defects and neurodegenerative disorders via autophagy activation. Autophagy has been found to mitigate cell apoptosis in intervertebral disc degeneration (IDD). Accordingly, we theorize that spermidine may have beneficial effects on IDD via autophagy stimulation. In this study, spermidine's effect on IDD was evaluated in tert-butyl hydroperoxide (TBHP)-treated nucleus pulposus cells of SD rats in vitro as well as in a puncture-induced rat IDD model. We found that autophagy was actuated by spermidine in nucleus pulposus cells. In addition, spermidine treatment weakened the apoptotic effects of TBHP in nucleus pulposus cells. Spermidine increased the expression of anabolic proteins including Collagen-II and aggrecan and decreased the expression of catabolic proteins including MMP13 and Adamts-5. Additionally, autophagy blockade using 3-MA reversed the beneficial impact of spermidine against nucleus pulposus cell apoptosis. Autophagy was thus important for spermidine's therapeutic effect on IDD. Spermidine-treated rats had an accentuated T2-weighted signal and a diminished histological degenerative grade than vehicle-treated rats, showing that spermidine inhibited intervertebral disc degeneration in vivo. Thus, spermidine protects nucleus pulposus cells against apoptosis through autophagy activation and improves disc, which may be beneficial for the treatment of IDD.


Subject(s)
Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc/drug effects , Nucleus Pulposus/drug effects , Spermidine/administration & dosage , Animals , Apoptosis/drug effects , Autophagy/drug effects , Disease Models, Animal , Extracellular Matrix/drug effects , Humans , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/chemically induced , Intervertebral Disc Degeneration/physiopathology , Nucleus Pulposus/pathology , Primary Cell Culture , Rats , tert-Butylhydroperoxide/toxicity
6.
Cell Physiol Biochem ; 47(3): 948-956, 2018.
Article in English | MEDLINE | ID: mdl-29895019

ABSTRACT

BACKGROUND/AIMS: Basic fibroblast growth factor (bFGF) and nerve growth factor (NGF) are essential for proper development, survival, growth, and maintenance of neurons in the central and peripheral nervous systems. However, because bFGF and NGF have short half-life and rapid diffusion rate, they have limited clinical efficacy. Thus, there is an urgent need to develop an effective delivery system to protect bFGF and NGF from proteolysis while maintaining their normal bioactivities. METHODS: To more efficiently deliver bFGF and NGF, we used a coacervate (synthesized with heparin and a biodegradable polycation at mass ratio of 500: 100). The maximal package loads of GFs in coacervate were determined by Western Blotting; release efficiency of bFGF and NGF was measured by ELISA. Additionally, we evaluated the effect of bFGF and NGF on the viability, survival, and proliferation of neurons by MTT assay, BrdU cell proliferation, and calcein staining. RESULTS: Our coacervate incorporated bFGF and NGF and continuously released them for at least three weeks. This enhanced the growth and proliferation of PC12 cells and SH-SY5Y cells. Moreover, co-delivery of bFGF and NGF using coacervate was more neuroprotective than free application of both factors or coacervate delivery of each GF separately. CONCLUSIONS: Dual delivery of bFGF and NGF binding coacervate was neuroprotective via stimulating the growth and proliferation of neurons.


Subject(s)
Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Nerve Growth Factor/pharmacology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Animals , Humans , Neurons/cytology , PC12 Cells , Rats
7.
J Cell Mol Med ; 21(11): 3010-3022, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28842949

ABSTRACT

In this study, we examined the neuroprotective effects and anti-inflammatory properties of Dl-3-n-butylphthalide (NBP) in Sprague-Dawley (SD) rats following traumatic spinal cord injury (SCI) as well as microglia activation and inflammatory response both in vivo and in vitro. Our results showed that NBP improved the locomotor recovery of SD rats after SCI an significantly diminished the lesion cavity area of the spinal cord, apoptotic activity in neurons, and the number of TUNEL-positive cells at 7 days post-injury. NBP inhibited activation of microglia, diminished the release of inflammatory mediators, and reduced the upregulation of microglial TLR4/NF-κB expression at 1 day post-injury. In a co-culture system with BV-2 cells and PC12 cells, NBP significantly reduced the cytotoxicity of BV-2 cells following lipopolysaccharide (LPS) stimulation. In addition, NBP reduced the activation of BV-2 cells, diminished the release of inflammatory mediators, and inhibited microglial TLR4/NF-κB expression in BV-2 cells. Our findings demonstrate that NBP may have neuroprotective and anti-inflammatory properties in the treatment of SCI by inhibiting the activation of microglia via TLR4/NF-κB signalling.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzofurans/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Cell Line , Coculture Techniques , Female , Gene Expression Regulation , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Microglia/cytology , Microglia/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/immunology , PC12 Cells , Rats , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
8.
Int J Mol Sci ; 18(2)2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28208696

ABSTRACT

Endoplasmic reticulum (ER) stress-induced apoptosis plays an important role in a range of neurological disorders, such as neurodegenerative diseases, spinal cord injury, and diabetic neuropathy. Valproate (VPA), a typical antiepileptic drug, is commonly used in the treatment of bipolar disorder and epilepsy. Recently, VPA has been reported to exert neurotrophic effects and promote neurite outgrowth, but its molecular mechanism is still unclear. In the present study, we investigated whether VPA inhibited ER stress and promoted neuroprotection and neuronal restoration in SH-SY5Y cells and in primary rat cortical neurons, respectively, upon exposure to thapsigargin (TG). In SH-SY5Y cells, cell viability was detected by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, and the expression of ER stress-related apoptotic proteins such as glucose­regulated protein (GRP78), C/EBP homologous protein (CHOP), and cleaved caspase-12/-3 were analyzed with Western blot analyses and immunofluorescence assays. To explore the pathway involved in VPA-induced cell proliferation, we also examined p-AKT, GSK3ß, p-JNK and MMP-9. Moreover, to detect the effect of VPA in primary cortical neurons, immunofluorescence staining of ß-III tubulin and Anti-NeuN was analyzed in primary cultured neurons exposed to TG. Our results demonstrated that VPA administration improved cell viability in cells exposed to TG. In addition, VPA increased the levels of GRP78 and p-AKT and decreased the levels of ATF6, XBP-1, GSK3ß, p-JNK and MMP-9. Furthermore, the levels of the ER stress-induced apoptosis response proteins CHOP, cleaved caspase-12 and cleaved caspase-3 were inhibited by VPA treatment. Meanwhile, VPA administration also increased the ratio of Bcl-2/Bax. Moreover, VPA can maintain neurite outgrowth of primary cortical neurons. Collectively, the neurotrophic effect of VPA is related to the inhibition of ER stress-induced apoptosis in SH-SY5Y cells and the maintenance of neuronal growth. Collectively, our results suggested a new approach for the therapeutic function of VPA in neurological disorders and neuroprotection.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Valproic Acid/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 9/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Thapsigargin/pharmacology , Transcription Factor CHOP/metabolism , bcl-2-Associated X Protein/metabolism
9.
J Cell Mol Med ; 20(6): 1062-75, 2016 06.
Article in English | MEDLINE | ID: mdl-26769343

ABSTRACT

After spinal cord injury (SCI), disruption of blood-spinal cord barrier (BSCB) elicits blood cell infiltration such as neutrophils and macrophages, contributing to permanent neurological disability. Previous studies show that epidermal growth factor (EGF) produces potent neuroprotective effects in SCI models. However, little is known that whether EGF contributes to the integrity of BSCB. The present study is performed to explore the mechanism of BSCB permeability changes which are induced by EGF treatment after SCI in rats. In this study, we demonstrate that EGF administration inhibits the disruption of BSCB permeability and improves the locomotor activity in SCI model rats. Inhibition of the PI3K/Akt pathways by a specific inhibitor, LY294002, suppresses EGF-induced Rac1 activation as well as tight junction (TJ) and adherens junction (AJ) expression. Furthermore, the protective effect of EGF on BSCB is related to the activation of Rac1 both in vivo and in vitro. Blockade of Rac1 activation with Rac1 siRNA downregulates EGF-induced TJ and AJ proteins expression in endothelial cells. Taken together, our results indicate that EGF treatment preserves BSCB integrity and improves functional recovery after SCI via PI3K-Akt-Rac1 signalling pathway.


Subject(s)
Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/therapeutic use , Signal Transduction , Spinal Cord Injuries/blood , Spinal Cord Injuries/drug therapy , Spinal Cord/pathology , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Animals , Chromones/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epidermal Growth Factor/administration & dosage , Female , Glucose/deficiency , Humans , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Oxygen , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects , Signal Transduction/drug effects , Spinal Cord/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL