Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 397
Filter
1.
Int J Nanomedicine ; 19: 7851-7870, 2024.
Article in English | MEDLINE | ID: mdl-39105098

ABSTRACT

Background: Inhibiting ROS overproduction is considered a very effective strategy for the treatment of peripheral nerve injuries, and Se has a remarkable antioxidant effect; however, since the difference between the effective concentration of Se and the toxic dose is not large, we synthesized a nanomaterial that can release Se slowly so that it can be used more effectively. Methods: Se@SiO2 NPs were synthesized using a mixture of Cu2-x Se nanocrystals, and the mechanism of action of Se@SiO2 NPs was initially explored by performing sequencing, immunofluorescence staining and Western blotting of cellular experiments. The mechanism of action of Se@SiO2 NPs was further determined by performing behavioral assays after animal experiments and by sampling the material for histological staining, immunofluorescence staining, and ELISA. The effects, mechanisms and biocompatibility of Se@SiO2 NPs for peripheral nerve regeneration were determined. Results: Porous Se@SiO2 was successfully synthesized, had good particle properties, and could release Se slowly. CCK-8 experiments revealed that the optimal experimental doses were 100 µM H2O2 and 200 µg/mL Se@SiO2, and RNA-seq revealed that porous Se@SiO2 was associated with cell proliferation, apoptosis, and the PI3K/AKT pathway. WB showed that porous Se@SiO2 could increase the expression of cell proliferation antigens (PCNA and S100) and antiapoptotic proteins (Bcl-2), decrease the expression of proapoptotic proteins (Bax), and increase the expression of antioxidative stress proteins (Nrf2, HO-1, and SOD2). EdU cell proliferation and ROS fluorescence assays showed that porous Se@SiO2 promoted cell proliferation and reduced ROS levels. The therapeutic effect of LY294002 (a PI3K/AKT pathway inhibitor) was decreased significantly and its effect was lost when it was added simultaneously with porous Se@SiO2. Animal experiments revealed that the regenerated nerve fiber density, myelin thickness, axon area, gastrocnemius muscle wet-to-weight ratio, myofiber area, sciatic nerve function index (SFI), CMAP, apoptotic cell ratio, and levels of antioxidative stress proteins and anti-inflammatory factors were increased following the administration of porous Se@SiO2. The levels of oxidative stress proteins and anti-inflammatory factors were significantly greater in the Se@SiO2 group than in the PNI group, and the effect of LY294002 was decreased significantly and was lost when it was added simultaneously with porous Se@SiO2. Conclusion: Se@SiO2 NPs are promising, economical and effective Se-releasing nanomaterials that can effectively reduce ROS production, inhibit apoptosis and promote cell proliferation after nerve injury via the PI3K/AKT pathway, ultimately accelerating nerve regeneration. These findings could be used to design new, promising drugs for the treatment of peripheral nerve injury.


Subject(s)
Cell Proliferation , Nerve Regeneration , Peripheral Nerve Injuries , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Selenium , Signal Transduction , Silicon Dioxide , Animals , Selenium/chemistry , Selenium/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Peripheral Nerve Injuries/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Nerve Regeneration/drug effects , Cell Proliferation/drug effects , Rats , Apoptosis/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Nanoparticles/chemistry , Male , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/chemistry , Rats, Sprague-Dawley , Oxidative Stress/drug effects , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Schwann Cells/drug effects , Schwann Cells/metabolism
2.
Biomed Microdevices ; 26(3): 34, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102047

ABSTRACT

Critical-sized peripheral nerve injuries pose a significant clinical challenge and lead to functional loss and disability. Current regeneration strategies, including autografts, synthetic nerve conduits, and biologic treatments, encounter challenges such as limited availability, donor site morbidity, suboptimal recovery, potential immune responses, and sustained stability and bioactivity. An obstacle in peripheral nerve regeneration is the immune response that can lead to inflammation and scarring that impede the regenerative process. Addressing both the immunological and regenerative needs is crucial for successful nerve recovery. Here, we introduce a novel biodegradable tacrolimus-eluting nerve guidance conduit engineered from a blend of poly (L-lactide-co-caprolactone) to facilitate peripheral nerve regeneration and report the testing of this conduit in 15-mm critical-sized gaps in the sciatic nerve of rats. The conduit's diffusion holes enable the local release of tacrolimus, a potent immunosuppressant with neuro-regenerative properties, directly into the injury site. A series of in vitro experiments were conducted to assess the ability of the conduit to maintain a controlled tacrolimus release profile that could promote neurite outgrowth. Subsequent in vivo assessments in rat models of sciatic nerve injury revealed significant enhancements in nerve regeneration, as evidenced by improved axonal growth and functional recovery compared to controls using placebo conduits. These findings indicate the synergistic effects of combining a biodegradable conduit with localized, sustained delivery of tacrolimus, suggesting a promising approach for treating peripheral nerve injuries. Further optimization of the design and long-term efficacy studies and clinical trials are needed before the potential for clinical translation in humans can be considered.


Subject(s)
Nerve Regeneration , Peripheral Nerve Injuries , Sciatic Nerve , Tacrolimus , Animals , Tacrolimus/pharmacology , Tacrolimus/administration & dosage , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/therapy , Rats , Sciatic Nerve/injuries , Sciatic Nerve/drug effects , Rats, Sprague-Dawley , Polyesters/chemistry , Disease Models, Animal , Guided Tissue Regeneration/methods
3.
Hand Clin ; 40(3): 399-408, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972684

ABSTRACT

Following nerve injury, growth factors (GFs) are transiently upregulated in injured neurons, proliferating Schwann cells, and denervated muscle and skin. They act on these same cells and tissues to promote nerve regeneration and end-organ reinnervation. Consequently, much attention has been focused on developing GF-based therapeutics. A major barrier to clinical translation of GFs is their short half-life. To provide sustained GF treatment to the affected nerve, muscle, and skin in a safe and practical manner, engineered drug delivery systems are needed. This review highlights recent advancements in GF-based therapeutics and discusses the remaining hurdles for clinical translation.


Subject(s)
Intercellular Signaling Peptides and Proteins , Nerve Regeneration , Nerve Regeneration/physiology , Nerve Regeneration/drug effects , Humans , Intercellular Signaling Peptides and Proteins/physiology , Intercellular Signaling Peptides and Proteins/therapeutic use , Peripheral Nerve Injuries/surgery , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/physiopathology , Animals , Drug Delivery Systems
4.
Int J Pharm ; 661: 124477, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39013530

ABSTRACT

Peripheral nerve injuries, predominantly affecting individuals aged 20-40, pose significant healthcare challenges, with current surgical methods often failing to achieve complete functional recovery. This study focuses on the development of 3D printed hydrogel nerve conduits using modified hyaluronic acid (HA) for potentially enhancing peripheral nerve regeneration. Hyaluronic acid was chemically altered with cysteamine HCl and methacrylic anhydride to create thiolated HA (HA-SH) and methacrylated HA (HA-MA), achieving a modification degree of approximately 20 %. This modification was crucial to maintain the receptor interaction of HA. The modified HA was rigorously tested to ensure cytocompatibility in neuronal and glial cell lines. Subsequently, various 3D printed HA formulations were evaluated, focusing on improving HA's inherent mechanical weaknesses. These formulations were assessed for cytotoxicity through direct contact and elution extract testing, confirming their safety over a 24-h period. Among the neurotrophic compounds tested, Tyrosol emerged as the most effective in promoting Schwann cell proliferation in vitro. The 3D printed HA system demonstrated proficiency in loading and releasing Tyrosol at physiological pH. The findings from this research highlight the promising role of 3D printed HA and Tyrosol in the field of nerve tissue engineering, offering a novel approach to peripheral nerve regeneration.


Subject(s)
Cell Proliferation , Hyaluronic Acid , Nerve Regeneration , Printing, Three-Dimensional , Schwann Cells , Schwann Cells/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/administration & dosage , Cell Proliferation/drug effects , Nerve Regeneration/drug effects , Animals , Cell Line , Hydrogels/chemistry , Hydrogels/administration & dosage , Humans , Rats , Tissue Engineering/methods , Peripheral Nerve Injuries/drug therapy
5.
Int J Mol Sci ; 25(13)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-39000003

ABSTRACT

Peripheral nerve injuries (PNIs) represent a significant clinical challenge, particularly in elderly populations where axonal remyelination and regeneration are impaired. Developing therapies to enhance these processes is crucial for improving PNI repair outcomes. Glutamate carboxypeptidase II (GCPII) is a neuropeptidase that plays a pivotal role in modulating glutamate signaling through its enzymatic cleavage of the abundant neuropeptide N-acetyl aspartyl glutamate (NAAG) to liberate glutamate. Within the PNS, GCPII is expressed in Schwann cells and activated macrophages, and its expression is amplified with aging. In this study, we explored the therapeutic potential of inhibiting GCPII activity following PNI. We report significant GCPII protein and activity upregulation following PNI, which was normalized by the potent and selective GCPII inhibitor 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). In vitro, 2-PMPA robustly enhanced myelination in dorsal root ganglion (DRG) explants. In vivo, using a sciatic nerve crush injury model in aged mice, 2-PMPA accelerated remyelination, as evidenced by increased myelin sheath thickness and higher numbers of remyelinated axons. These findings suggest that GCPII inhibition may be a promising therapeutic strategy to enhance remyelination and potentially improve functional recovery after PNI, which is especially relevant in elderly PNI patients where this process is compromised.


Subject(s)
Glutamate Carboxypeptidase II , Peripheral Nerve Injuries , Remyelination , Animals , Mice , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/metabolism , Remyelination/drug effects , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/metabolism , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Aging/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Mice, Inbred C57BL , Nerve Regeneration/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/drug effects , Male , Axons/drug effects , Axons/metabolism
6.
Int Immunopharmacol ; 138: 112452, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38943972

ABSTRACT

Peripheral nerve injury seriously endangers human life and health, but there is no clinical drug for the treatment of peripheral nerve injury, so it is imperative to develop drugs to promote the repair of peripheral nerve injury. Erythropoietin (EPO) not only has the traditional role of promoting erythropoiesis, but also has a tissue-protective effect. Over the past few decades, researchers have confirmed that EPO has neuroprotective effects. However, side effects caused by long-term use of EPO limited its clinical application. Therefore, EPO derivatives with low side effects have been explored. Among them, ARA290 has shown significant protective effects on the nervous system, but the biggest disadvantage of ARA290, its short half-life, limits its application. To address the short half-life issue, the researchers modified ARA290 with thioether cyclization to generate a thioether cyclized helical B peptide (CHBP). ARA290 and CHBP have promising applications as peptide drugs. The neuroprotective effects they exhibit have attracted continuous exploration of their mechanisms of action. This article will review the research on the role of EPO, ARA290 and CHBP in the nervous system around this developmental process, and provide a certain reference for the subsequent research.


Subject(s)
Erythropoietin , Neuroprotective Agents , Peripheral Nerve Injuries , Erythropoietin/therapeutic use , Humans , Peripheral Nerve Injuries/drug therapy , Animals , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Peptides/therapeutic use , Peptides/pharmacology , Oligopeptides
7.
Biomed Pharmacother ; 177: 117015, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936196

ABSTRACT

Injury of a peripheral nerve (PNI) leads to both ischemic and inflammatory alterations. Sciatic nerve injury (SNI) represents the most widely used model for PNI. Mesenchymal stem cell-based therapy (MSCs) has convenient properties on PNI by stimulating the nerve regeneration. Melatonin has cytoprotective activity. The neuroprotective characteristics of MSCs and melatonin separately or in combination remain a knowledge need. In the rats-challenged SNI, therapeutic roles of intralesional MSCs and intraperitoneal melatonin injections were evaluated by functional assessment of peripheral nerve regeneration by walking track analysis involving sciatic function index (SFI) and two electrophysiological tests, electromyography and nerve conduction velocity, as well as measurement of antioxidant markers in serum, total antioxidant capacity (TAC) and malondialdehyde, and mRNA expression of brain derived neurotrophic factor (BDNF) in nerve tissues in addition to the histopathological evaluation of nerve tissue. Both individual and combination therapy with MSCs and melatonin therapies could effectively ameliorate this SNI and promote its regeneration as evidenced by improving the SFI and two electrophysiological tests and remarkable elevation of TAC with decline in lipid peroxidation and upregulation of BDNF levels. All of these led to functional improvement of the damaged nerve tissues and good recovery of the histopathological sections of sciatic nerve tissues suggesting multifactorial synergistic approach of the concurrent usage of melatonin and MSCs in PNI. The combination regimen has the most synergistic neuro-beneficial effects in PNI that should be used as therapeutic option in patients with PNI to boost their quality of life.


Subject(s)
Antioxidants , Melatonin , Mesenchymal Stem Cell Transplantation , Nerve Regeneration , Peripheral Nerve Injuries , Sciatic Nerve , Animals , Melatonin/pharmacology , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/therapy , Mesenchymal Stem Cell Transplantation/methods , Rats , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Nerve Regeneration/drug effects , Male , Antioxidants/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Disease Models, Animal , Rats, Wistar , Neural Conduction/drug effects , Rats, Sprague-Dawley
8.
FEBS Open Bio ; 14(8): 1356-1364, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942739

ABSTRACT

Peripheral nerve injuries result in significant loss of motor and sensory function, and the slow rate of nerve regeneration can prolong recovery time. Thus, approaches that promote axonal regeneration are critical to improve the outcomes for patients with peripheral nerve injuries. In this study, we investigated the effects of Ficus carica L. (fig) and Vaccinium macrocarpon Ait. (cranberry), which are rich in phytochemicals with demonstrable and diverse medicinal properties, on nerve regeneration in a mouse model of sciatic nerve crush. Our investigation revealed that fig extract, but not cranberry extract, prevented the decline in muscle weight and nerve conduction velocity induced by nerve crush. The fig extract also mitigated motor function impairment, myelin thinning, and axon diameter reduction, indicating its potential to promote nerve regeneration. Furthermore, the fig extract enhanced macrophage infiltration into the nerve tissue, suggesting that it could ameliorate nerve injury by promoting tissue repair via increased macrophage infiltration. The study provides valuable insights into the potential of the fig extract as a novel agent promoting nerve regeneration. Further investigation into the mechanisms underlying the action of fig extracts is needed to translate these findings into clinical applications for patients with peripheral nerve injuries.


Subject(s)
Disease Models, Animal , Ficus , Nerve Regeneration , Plant Extracts , Sciatic Nerve , Animals , Nerve Regeneration/drug effects , Mice , Ficus/chemistry , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Plant Extracts/pharmacology , Peripheral Nerve Injuries/drug therapy , Nerve Crush , Male , Mice, Inbred C57BL
9.
Trials ; 25(1): 332, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773595

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is the most common non-cutaneous malignancy in men and leads to the second most common cause of cancer related mortality in men. Early detection of PCa allows for a potentially curative intervention. Most men will live over a decade from the time of their PCa diagnosis. Thus, treatments must balance curative interventions with their impact on quality of life. Radical prostatectomy (RP) is one such potentially curative intervention but often leads to erectile dysfunction (ED) and urinary incontinence (UI). Approximately 90,000 RPs are performed each year in the USA. Post-operative ED and UI is thought to occur in part from traumatic peripheral nerve injury (TPNI) to the neurovascular bundles that surround the prostate. Thus, patients undergoing RP may be a population that would benefit from clinical studies that look at TPNI. METHODS: The study is a single-institution, double-blinded placebo-controlled, randomized clinical trial in which patients immediately post-RP receive either 4-aminopyrdine (4AP) or placebo in a 1:1 fashion. The primary outcome is evaluation of the efficacy of 4AP in accelerating the early return of baseline erectile and urinary function post-radical prostatectomy. DISCUSSION: This study is critical as it could reduce the morbidity associated with RP, a commonly performed operation, and identify a patient population that may greatly benefit into further TPNI research. TRIAL REGISTRATION: ClinicalTrials.gov NCT03701581. Prospectively registered on October 10, 2018.


Subject(s)
Erectile Dysfunction , Peripheral Nerve Injuries , Prostatectomy , Prostatic Neoplasms , Urinary Incontinence , Humans , Male , Middle Aged , Double-Blind Method , Erectile Dysfunction/etiology , Erectile Dysfunction/drug therapy , Peripheral Nerve Injuries/etiology , Peripheral Nerve Injuries/drug therapy , Postoperative Complications/etiology , Prostatectomy/adverse effects , Prostatectomy/methods , Prostatic Neoplasms/surgery , Recovery of Function , Treatment Outcome , Urinary Incontinence/etiology
10.
Acta Biomater ; 182: 28-41, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38761961

ABSTRACT

The regenerative microenvironment after peripheral nerve injury is imbalanced and difficult to rebalance, which is mainly affected by inflammation, oxidative stress, and inadequate blood supply. The difficulty in remodeling the nerve regeneration microenvironment is the main reason for slow nerve regeneration. Traditional drug treatments have certain limitations, such as difficulty in penetrating the blood-nerve barrier and lack of pleiotropic effects. Therefore, there is an urgent need to build multifunctional nerve grafts that can effectively regulate the regenerative microenvironment and promote nerve regeneration. Nitric oxide (NO), a highly effective gas transmitter with diatomic radicals, is an important regulator of axonal growth and migration, synaptic plasticity, proliferation of neural precursor cells, and neuronal survival. Moreover, NO provides potential anti-inflammation, anti-oxidation, and blood vessel promotion applications. However, excess NO may cause cell death and neuroinflammatory cell damage. The prerequisite for NO treatment of peripheral nerve injury is that it is gradually released over time. In this study, we constructed an injectable NO slow-release system with two main components, including macromolecular NO donor nanoparticles (mPEG-P(MSNO-EG) nanoparticles, NO-NPs) and a carrier for the nanoparticles, mPEG-PA-PP injectable temperature-sensitive hydrogel. Due to the multiple physiological regulation of NO and better physiological barrier penetration, the conduit effectively regulates the inflammatory response and oxidative stress of damaged peripheral nerves, promotes nerve vascularization, and nerve regeneration and docking, accelerating the nerve regeneration process. STATEMENT OF SIGNIFICANCE: The slow regeneration speed of peripheral nerves is mainly due to the destruction of the regeneration microenvironment. Neural conduits with drug delivery capabilities have the potential to improve the microenvironment of nerve regeneration. However, traditional drugs are hindered by the blood nerve barrier and cannot effectively target the injured area. NO, an endogenous gas signaling molecule, can freely cross the blood nerve barrier and act on target cells. However, excessive NO can lead to cell apoptosis. In this study, a NO sustained-release system was constructed to regulate the microenvironment of nerve regeneration through various pathways and promote nerve regeneration.


Subject(s)
Delayed-Action Preparations , Nerve Regeneration , Nitric Oxide , Animals , Nitric Oxide/metabolism , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/therapy , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/metabolism , Rats, Sprague-Dawley , Rats , Peripheral Nerves/drug effects , Peripheral Nerves/pathology , Nanoparticles/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Male , Hydrogels/chemistry , Sciatic Nerve/drug effects
11.
Int J Biol Macromol ; 272(Pt 2): 132448, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821302

ABSTRACT

Peripheral nerve injury often leads to symptoms of motor and sensory impairment, and slow recovery of nerves after injury and limited treatment methods will aggravate symptoms or even lead to lifelong disability. Curcumin can promote peripheral nerve regeneration, but how to accurately deliver the appropriate concentration of curcumin in the local peripheral nerve remains to be solved. In this study, we designed a human hair keratin/chitosan (C/K) hydrogel with sodium tripolyphosphate ions crosslinked to deliver curcumin topically. Chitosan improves the mechanical properties of hydrogels and keratin improves the biocompatibility of hydrogels. C/K hydrogel showed good cytocompatibility, histocompatibility and degradability. In vitro experiments showed that hydrogels can continuously release curcumin for up to 10 days. In addition, a comprehensive analysis of behavioral, electrophysiological, histology, and target organ recovery results in animal experiments showed that locally delivered curcumin can enhance nerve regeneration in addition to hydrogels. In short, we provide a new method that combines the advantages of human hair keratin, chitosan, and curcumin for nerve damage repair.


Subject(s)
Chitosan , Curcumin , Hydrogels , Keratins , Nerve Regeneration , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/administration & dosage , Chitosan/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Nerve Regeneration/drug effects , Animals , Humans , Keratins/chemistry , Keratins/pharmacology , Rats , Peripheral Nerve Injuries/drug therapy , Mice
12.
J Biomater Sci Polym Ed ; 35(10): 1550-1570, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38630632

ABSTRACT

In recent years, mouse nerve growth factor (mNGF) has emerged as an important biological regulator to repair peripheral nerve injury, but its systemic application is restricted by low efficiency and large dosage requirement. These limitations prompted us to search for biomaterials that can be locally loaded. Oxidized sodium alginate hydrogel (OSA) exhibits good biocompatibility and physicochemical properties, and can be loaded with drugs to construct a sustained-release system that can act locally on nerve injury. Here, we constructed a sustained-release system of OSA-mouse nerve growth factor (mNGF), and investigated the loading and release of the drug through Fourier transform infrared spectroscopy and drug release curves. In vitro and in vivo experiments showed that OSA-mNGF significantly promoted the biological activities of RSC-96 cells and facilitated the recovery from sciatic nerve crush injury in rats. This observation may be attributed to the additive effect of OSA on promoting Schwann cell biological activities or its synergistic effect of cross-activating phosphoinositide 3-kinase (PI3K) through extracellular signal regulated kinase (ERK) signaling. Although the specific mechanism of OSA action needs to be explored in the future, the current results provide a valuable preliminary research basis for the clinical application of the OSA-mNGF sustained-release system for nerve repair.


Subject(s)
Alginates , Delayed-Action Preparations , Drug Liberation , Hydrogels , Nerve Growth Factor , Peripheral Nerve Injuries , Alginates/chemistry , Alginates/pharmacology , Animals , Nerve Growth Factor/chemistry , Delayed-Action Preparations/chemistry , Mice , Hydrogels/chemistry , Hydrogels/pharmacology , Rats , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/drug effects , Nerve Regeneration/drug effects , Oxidation-Reduction , Cell Line , Male , Rats, Sprague-Dawley , Drug Carriers/chemistry , Phosphatidylinositol 3-Kinases/metabolism
13.
Int J Biol Macromol ; 268(Pt 1): 131594, 2024 May.
Article in English | MEDLINE | ID: mdl-38621568

ABSTRACT

Treating severe peripheral nerve injuries is difficult. Nerve repair with conduit small gap tubulization is a treatment option but still needs to be improved. This study aimed to assess the use of microgels containing growth factors, along with chitosan-based conduits, for repairing nerves. Using the water-oil emulsion technique, microgels of methacrylic alginate (AlgMA) that contained vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF) were prepared. The effects on rat Schwann cells (RSC96) and human umbilical vein endothelial cells (HUVECs) were evaluated. Chitosan-based conduits were fabricated and used in conjunction with microgels containing two growth factors to treat complete neurotmesis in rats. The results showed that the utilization of dual growth factor microgels improved the migration and decreased the apoptosis of RSC96 cells while promoting the growth and formation of tubes in HUVECs. The utilization of dual growth factor microgels and chitosan-based conduits resulted in notable advancements in the regeneration and myelination of nerve fibers, recovery of neurons, alleviation of muscle atrophy and recovery of neuromotor function and nerve conduction. In conclusion, the use of dual growth factor AlgMA microgels in combination with chitosan-based conduits has the potential to significantly improve the effectiveness of nerve repair.


Subject(s)
Alginates , Chitosan , Human Umbilical Vein Endothelial Cells , Nerve Regeneration , Schwann Cells , Chitosan/chemistry , Chitosan/pharmacology , Alginates/chemistry , Alginates/pharmacology , Animals , Humans , Rats , Nerve Regeneration/drug effects , Schwann Cells/drug effects , Microgels/chemistry , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/therapy , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Tissue Scaffolds/chemistry , Methacrylates/chemistry , Methacrylates/pharmacology , Cell Movement/drug effects
14.
PLoS One ; 19(3): e0287390, 2024.
Article in English | MEDLINE | ID: mdl-38507417

ABSTRACT

OBJECTIVE: To determine the effective dose and therapeutic potential of maropitant using through expression of mediators of oxidative stress, inflammatory and of the unfolded protein response (UPR) (bio) markers on spinal cord using a model of neuropathic pain induced through chronic constriction injury (CCI) in rats. STUDY DESIGN: Randomized, blinded, prospective experimental study. ANIMALS: 98 male Wistar rats. METHODS: Rats were anesthetized with sevoflurane and after CCI, they were randomly assigned to the following groups that received: vehicle, 3, 6, 15, 30 e 50 mg/kg/24q of maropitant. The effect on inflammatory mediators (IL10, TNFα), oxidative stress (GPx, CAT, SOD), microglial (IBA-1) and neuronal (NeuN, TACR1) markers was evaluated though immunohistochemistry and expression levels of markers of hypoxia (HIF1α, Nrf2), antioxidant enzymes (Catalse, Sod1 and GPx1), and endoplasmic reticulum stress mediators (GRP78, CHOP and PERK) through qRT-PCR. RESULTS: Intraperitoneal injection (IP) of maropitant inhibited nociception with ID50 values of 4,1 mg/kg (5,85-19,36) in a neuropathic pain model through CCI. A dose of 30 mg/kg/24q was significantly effective in reducing mechanical allodynia 1 to 4h after treatment with nociception inhibition (145,83%). A reduction in the expression of hypoxia factors (HIF1α, Nrf2) was observed, along with an increase in antioxidant activity (CAT, SOD and GPX). Additionally, there was a reduction in inflammatory markes (IL10, TNFα), microglial (IBA-1), and neuronal markers (NeuN, TACR1). CONCLUSION AND CLINICAL RELEVANCE: These findings demonstrate that the determined dose, administered daily for seven days, had an antinociceptive effect, as well as anti-inflammatory and antioxidant activity.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Quinuclidines , Rats , Male , Animals , Antioxidants/metabolism , Rats, Wistar , Neuroinflammatory Diseases , Peripheral Nerve Injuries/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Interleukin-10/metabolism , NF-E2-Related Factor 2/metabolism , Prospective Studies , Oxidative Stress , Hyperalgesia/drug therapy , Endoplasmic Reticulum Stress , Neuralgia/drug therapy , Neuralgia/metabolism , Superoxide Dismutase/metabolism , Hypoxia/drug therapy
15.
Int J Pharm ; 655: 123978, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38458406

ABSTRACT

Peripheral nerve injury is a critical condition that can disrupt nerve functions. Despite the progress in engineering artificial nerve guidance conduits (NGCs), nerve regeneration remains challenging. Here, we developed new nanofibrous NGCs using polycaprolactone (PCL) and chitosan (CH) containing piracetam (PIR)/vitamin B12(VITB12) with an electrospinning method. The lumen of NGCs was coated by hyaluronic acid (HA) to promote regeneration in sciatic nerve injury. The NGCs were characterized via Scanning Electron Microscopy (SEM), Fourier transform infrared (FTIR), tensile, swelling, contact angle, degradation, and drug release tests. Neuronal precursor cell line (PCL12 cell) and rat mesenchymal stem cells derived from bone marrow (MSCs) were seeded on the nanofibrous conduits. After that, the biocompatibility of the NGCs was evaluated by the 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, 4',6-diamidino-2-phenylindole (DAPI) staining, and SEM images. The SEM demonstrated that PCL/CH/PIR/VITB12 NGCs had nonaligned, interconnected, smooth fibers. The mechanical properties of these NGCs were similar to rat sciatic nerve. These conduits had an appropriate swelling and degradation rate. The In Vitro studies exhibited favorable biocompatibility of the PCL/CH/PIR/VITB12 NGCs towards PC12 cells and MSCs. The in vitro studies exhibited favorable biocompatibility of the PCL/CH/PIR/VIT B12 NGCs towards MSCs and PC12 cells. To analyze functional efficacy, NGCs were implanted into a 10 mm Wistar rat sciatic nerve gap and bridged the proximal and distal stump of the defect. After three months, the results of sciatic functional index (55.3 ± 1.8), hot plate latency test (5.6 ± 0.5 s), gastrocnemius muscle wet weight-loss (38.57 ± 1.6 %) and histopathological examination using hematoxylin-eosin (H&E) /toluidine blue/ Anti-Neurofilament (NF200) staining demonstrated that the produced conduit recovered motor and sensory functions and had comparable nerve regeneration compared to the autograft that can be as the gold standard to bridge the nerve gaps.


Subject(s)
Chitosan , Nanofibers , Peripheral Nerve Injuries , Piracetam , Rats , Animals , Rats, Wistar , Hyaluronic Acid , Vitamin B 12 , Sciatic Nerve , Tissue Scaffolds , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/pathology , PC12 Cells , Nerve Regeneration
16.
J Biomed Mater Res B Appl Biomater ; 112(2): e35378, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38356051

ABSTRACT

Globally, peripheral nerve injury (PNI) is a common clinical issue. Successfully repairing severe PNIs has posed a major challenge for clinicians. GW3965 is a highly selective LXR agonist, and previous studies have demonstrated its positive protective effects in both central and peripheral nerve diseases. In this work, we examined the potential reparative effects of GW3965-loaded polylactic acid co-glycolic acid microspheres in conjunction with a chitosan nerve conduit for peripheral nerve damage. The experiment revealed that GW3965 promoted Schwann cell proliferation and neurotrophic factor release in vitro. In vivo experiments conducted on rats showed that GW3965 facilitated the restoration of motor function, promoted axon and myelin regeneration in the sciatic nerve, and enhanced the microenvironment of nerve regeneration. These results offer a novel therapeutic approach for the healing of nerve damage. Overall, this work provides valuable insights and presents a promising therapeutic strategy for addressing PNI.


Subject(s)
Benzoates , Benzylamines , Chitosan , Peripheral Nerve Injuries , Rats , Animals , Chitosan/pharmacology , Liver X Receptors/therapeutic use , Microspheres , Schwann Cells , Sciatic Nerve/injuries , Peripheral Nerve Injuries/drug therapy , Nerve Regeneration
17.
Macromol Biosci ; 24(5): e2300476, 2024 May.
Article in English | MEDLINE | ID: mdl-38245857

ABSTRACT

Peripheral nerve injuries (PNI) represent a prevalent and severe category of damage resulting from traumatic incidents. Predominantly, the deficiency in nerve regeneration can be ascribed to enduring inflammatory reactions, hence imposing substantial clinical implications for patients. Fisetin, a flavonoid derived from plants, is naturally present in an array of vegetables and fruits, including strawberries, apples, onions, and cucumbers. It exhibits immunomodulatory properties through the reduction of inflammation and oxidative stress. In the present research, a nerve defect is addressed for the first time utilizing a scaffold primed for controlled fisetin release. In this regard, fisetin-loaded chitosan hydrogels are incorporated into the lumen of polycaprolactone (PCL) nerve guide conduits (NGCs). The hydrogel maintained a steady release of an appropriate fisetin dosage. The study outcomes indicated that the fisetin/chitosan/polycaprolactone (FIS/CS/PCL) NGCs amplified Schwann cell proliferation and neural expression, curtailed oxidative stress, alleviated inflammation, and improved functions, electrophysiological properties, and morphology. This pioneering scaffold has the potential to contribute significantly to the field of neuroengineering.


Subject(s)
Chitosan , Flavonols , Hydrogels , Inflammation , Nerve Regeneration , Oxidative Stress , Polyesters , Flavonols/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Oxidative Stress/drug effects , Animals , Nerve Regeneration/drug effects , Polyesters/chemistry , Polyesters/pharmacology , Inflammation/drug therapy , Inflammation/pathology , Schwann Cells/drug effects , Schwann Cells/metabolism , Tissue Scaffolds/chemistry , Rats , Guided Tissue Regeneration/methods , Cell Proliferation/drug effects , Flavonoids/pharmacology , Flavonoids/chemistry , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/therapy
18.
Int J Mol Sci ; 25(2)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38255977

ABSTRACT

Peripheral nerve injuries lead to severe functional impairments and long recovery times, with limited effectiveness and accessibility of current treatments. This has increased interest in natural bioactive compounds, such as ursolic acid (UA). Our study evaluated the effect of an oleolyte rich in UA from white grape pomace (WGPO) on neuronal regeneration in mice with induced sciatic nerve resection, administered concurrently with the induced damage (the WGPO group) and 10 days prior (the PRE-WGPO group). The experiment was monitored at two-time points (4 and 10 days) after injury. After 10 days, the WGPO group demonstrated a reduction in muscle atrophy, evidenced by an increased number and diameter of muscle fibers and a decreased Atrogin-1 and Murf-1 expression relative to the denervated control. It was also observed that 85.7% of neuromuscular junctions (NMJs) were fully innervated, as indicated by the colocalization of α-bungarotoxin and synaptophysin, along with the significant modulation of Oct-6 and S-100. The PRE-WGPO group showed a more beneficial effect on nerve fiber reformation, with a significant increase in myelin protein zero and 95.2% fully innervated NMJs, and a pro-hypertrophic effect in resting non-denervated muscles. Our findings suggest WGPO as a potential treatment for various conditions that require the repair of nerve and muscle injuries.


Subject(s)
Peripheral Nerve Injuries , Animals , Mice , Peripheral Nerve Injuries/drug therapy , Ursolic Acid , Sciatic Nerve , Dietary Supplements , Muscle Fibers, Skeletal
19.
Adv Healthc Mater ; 13(10): e2303539, 2024 04.
Article in English | MEDLINE | ID: mdl-38233357

ABSTRACT

Peripheral nerve injury (PNI) is a common clinical problem and regenerating peripheral nerve defects remain a significant challenge. Poly(polyol sebacate) (PPS) polymers are developed as promising materials for biomedical applications due to their biodegradability, biocompatibility, elastomeric properties, and ease of production. However, the application of PPS-based biomaterials in nerve tissue engineering, especially in PNI repair, is limited. In this study, PPS-based composite nanofibers poly(l-lactic acid)-poly(polycaprolactone triol-co-sebacic acid-co-N,N-bis(2-hydroxyethyl)-2-aminoethanesulfonic acid sodium salt) (PLLA-PPSB) are aimed to construct through electrospinning and assess their in vitro biocompatibility with Schwann cells (SCs) and in vivo repair capabilities for peripheral nerve defects. For the first time, the biocompatibility and bioactivity of PPS-based nanomaterial are examined at the molecular, cellular, and animal levels for PNI repair. Electrospun PLLA-PPSB nanofibers display favorable physicochemical properties and biocompatibility, providing an effective interface for the proliferation, glial expression, and adhesion of SCs in vitro. In vivo experiments using a 10-mm rat sciatic nerve defect model show that PLLA-PPSB nanofiber nerve conduits enhance myelin formation, axonal regeneration, angiogenesis, and functional recovery. Transcriptome analysis and biological validation indicate that PLLA-PPSB nanofibers may promote SC proliferation by activating the PI3K/Akt signaling pathway. This suggests the promising potential of PLLA-PPSB nanomaterial for PNI repair.


Subject(s)
Blood Coagulation Factors , Nanofibers , Peripheral Nerve Injuries , Rats , Animals , Nanofibers/therapeutic use , Nanofibers/chemistry , Phosphatidylinositol 3-Kinases , Sciatic Nerve/physiology , Tissue Scaffolds/chemistry , Peripheral Nerve Injuries/drug therapy , Polyesters/chemistry , Nerve Regeneration
20.
J Orthop Sci ; 29(2): 653-659, 2024 Mar.
Article in English | MEDLINE | ID: mdl-36858838

ABSTRACT

BACKGROUND: Peripheral nerve injuries are common and serious conditions. The effect of Neurotropin® (NTP), a nonprotein extract derived from the inflamed skin of rabbits inoculated with vaccinia virus, on peripheral nerve regeneration has not been fully elucidated. However, it has analgesic properties via the activation of descending pain inhibitory systems. Therefore, the current study aimed to determine the effects of NTP on peripheral nerve regeneration. METHODS: We examined axonal outgrowth of dorsal root ganglion (DRG) neurons using immunocytochemistry in vitro. In addition, nerve regeneration was evaluated functionally, electrophysiologically, and histologically in a rat sciatic nerve crush injury model in vivo. Furthermore, gene expression of neurotrophic factors in the injured sciatic nerves and DRGs was evaluated. RESULTS: In the dorsal root ganglion neurons in vitro, NTP promoted axonal outgrowth at a concentration of 10 mNU/mL. Moreover, the systemic administration of NTP contributed to the recovery of motor and sensory function at 2 weeks, and of sensory function, nerve conduction velocity, terminal latency, and axon-remyelination 4 weeks after sciatic nerve injury. In the gene expression assessment, insulin-like growth factor 1 and vascular endothelial growth factor expressions were increased in the injured sciatic nerve 2 days postoperatively. CONCLUSIONS: Therefore, NTP might be effective in not only treating chronic pain but also promoting peripheral nerve regeneration after injury.


Subject(s)
Crush Injuries , Peripheral Nerve Injuries , Polysaccharides , Rats , Animals , Rabbits , Peripheral Nerve Injuries/drug therapy , Vascular Endothelial Growth Factor A , Nerve Regeneration/physiology , Sciatic Nerve/surgery , Sciatic Nerve/injuries
SELECTION OF CITATIONS
SEARCH DETAIL