Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.179
Filtrar
1.
Biomater Transl ; 5(2): 157-174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39351163

RESUMEN

Peripheral nerve injury poses a great threat to neurosurgery and limits the regenerative potential of sacral nerves in the neurogenic bladder. It remains unknown whether electrical stimulation can facilitate sacral nerve regeneration in addition to modulate bladder function. The objective of this study was to utilise electrical stimulation in sacra nerve crush injury with newly constructed electroconductive scaffold and explore the role of macrophages in electrical stimulation with crushed nerves. As a result, we generated a polypyrrole-coated polycaprolactone/silk fibroin scaffold through which we applied electrical stimulation. The electrical stimulation boosted nerve regeneration and polarised the macrophages towards the M2 phenotype. An in vitro test using bone marrow derived macrophages revealed that the pro-regenerative polarisation of M2 were significantly enhanced by electrical stimulation. Bioinformatics analysis showed that the expression of signal transducer and activator of transcriptions (STATs) was differentially regulated in a way that promoted M2-related genes expression. Our work indicated the feasibility of electricals stimulation used for sacral nerve regeneration and provided a firm demonstration of a pivotal role which macrophages played in electrical stimulation.

2.
Biomaterials ; 314: 122873, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39369670

RESUMEN

Spinal cord injury (SCI) triggers interconnected malignant pathological cascades culminating in structural abnormalities and composition changes of neural tissues and impairs spinal cord tissue function. Cellulose nanofibers (CNF) have considerable potential in mimicking tissue microstructure for nerve regeneration, but the effectiveness of CNF in repairing SCI remains poorly understood. In this study, we designed a Mg-Fe layered double hydroxide (LDH)-doped cellulose nanofiber (CNF) scaffold with aligned intact microchannels and homogeneously distributed pores (CNF-LDH), loaded with retinoic acid and sonic hedgehog (CNF-LDH-RS) for neuroregeneration. The aligned microchannel structure and chemical cues in the scaffold were designed further to enhance the differentiation of neural stem cells towards neurons and promote axon growth while inhibiting differentiation to astrocytes. Transplanting the scaffolds into a completely transected SCI mice model dramatically improved behavioral and electrophysiological outcomes underpinned by robust neuronal regeneration, significant axonal growth and orderly neural circuit remodeling. RNA-seq analysis revealed the pivotal roles of the RhoA/Rock/Myosin II pathway and neuroactive ligand-receptor interaction pathway in SCI repair by CNF-LDH-RS. Particularly, Myosin II emerged as a key gene for functional recovery, and its effect on negative regulation of axon growth was suppressed by the scaffolds, resulting in a distinctly oriented growth of the axons along the microchannel structure. The results indicate that CNF-LDH scaffolds rationally combined with physical and biochemical cues create promising tissue-engineered substrates to facilitate the repair of spinal cord injury.

3.
J Dent (Shiraz) ; 25(3): 215-222, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39371960

RESUMEN

Statement of the Problem: Healing of the inferior alveolar nerve injury during dental procedures is one of the biggest concerns of dentists. There are still debates on different treatment modalities. Purpose: This study aimed to compare the effect of 940nm and 810nm diode lasers on the repair of the inferior alveolar sensory nerve. Materials and Method: In this single-blinded randomized clinical trial, 39 patients with inferior alveolar nerve injury were divided into three groups: 1. 810nm laser irradiated, 2. 940nm laser irradiated, and 3. No laser irradiation (control group). All patients were treated in 12 sessions (3 days per week) and evaluated using a complete clinical neurosensory test (CNT), including brushstroke, 2-point discrimination, pinprick nociception, and thermal discrimination before and after treatment. Results: The mean dysesthesia of the patient treated with 810nm diode laser was significantly lower than the control group in all sessions (the 1st (p= 0.003), 3rd (p= 0.008), 7th (p= 0.006), and 12th sessions (p= 0.005)). The 810nm laser resulted in more satisfaction in patients than the control group in almost all sessions (1st (p< 0.001), 7th (p= 0.028), and 12th (p= 0.006)). More patient satisfaction was seen in the 1st and 3rd sessions in the 810nm laser than in the 980nm laser (p< 0.001 and p= 0.003, respectively). Conclusion: 810nm diode laser can be better than 940nm in repairing inferior alveolar sensory nerve damage.

4.
Sci Technol Adv Mater ; 25(1): 2404380, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39308888

RESUMEN

Decellularized tissues have been used as implantable materials for tissue regeneration because of their high biofunctionality. We have reported that high hydrostatic pressured (HHP) decellularized tissue developed in our laboratory exhibits good in vivo performance, but the details of the mechanism are still not known. Based on previous reports of bioactive factors called matrix bound nanovesicles (MBVs) within decellularized tissues, this study aims to investigate whether MBVs are also present in decellularized tissues prepared by HHP decellularization, which is different from the previously reported methods. In this study, we tried to extract bioactive factors from HHP decellularized brain and placenta, and evaluated their effects on nerves in vitro and in vivo, where its effects have been previously reported. The results confirmed that those factors can be extracted even if the decellularization method and tissue of origin differ, and that they have effects on a series of processes toward nerve regeneration, such as neurite outgrowth and nerve fiber repair.


In this study, we evaluated the neuroregenerative effects of matrix-bounded nanovesicles extracted from decellularized tissue using a high hydrostatic pressure method. The results indicate that bioactive factors, including matrix-bounded nanovesicles, can be extracted regardless of the decellularization method and tissue origin.

5.
Handb Clin Neurol ; 205: 243-262, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39341657

RESUMEN

Leading causes of blindness worldwide include neurodegenerative diseases of the retina, which cause irreversible loss of retinal pigment epithelium (RPE) and photoreceptors, and optic neuropathies, which result in retinal ganglion cell (RGC) death. Because photoreceptor and RGCs do not spontaneously regenerate in mammals, including humans, vision loss from these conditions is, at present, permanent. Recent advances in gene and cell-based therapies have provided new hope to patients affected by these conditions. This chapter reviews the current state and future of these approaches to treating ocular neurodegenerative disease. Gene therapies for retinal degeneration and optic neuropathies primarily focus on correcting known pathogenic mutations that cause inherited conditions to halt progression. There are multiple retinal and optic neuropathy gene therapies in clinical trials, and one retinal gene therapy is approved in the United States, Canada, Europe, and Australia. Cell-based therapies are mutation agnostic and have the potential to repopulate neurons regardless of the underlying etiology of degeneration. While photoreceptor cell replacement is nearing a human clinical trial, RPE transplantation is currently in phase I/II clinical trials. RGC replacement faces numerous logistical challenges, but preclinical research has laid the foundation for functional repair of optic neuropathies to be feasible.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Genética , Enfermedades del Nervio Óptico , Humanos , Terapia Genética/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedades del Nervio Óptico/terapia , Enfermedades de la Retina/terapia
6.
Proc Natl Acad Sci U S A ; 121(38): e2402518121, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39254997

RESUMEN

The in vivo three-dimensional genomic architecture of adult mature neurons at homeostasis and after medically relevant perturbations such as axonal injury remains elusive. Here, we address this knowledge gap by mapping the three-dimensional chromatin architecture and gene expression program at homeostasis and after sciatic nerve injury in wild-type and cohesin-deficient mouse sensory dorsal root ganglia neurons via combinatorial Hi-C, promoter-capture Hi-C, CUT&Tag for H3K27ac and RNA-seq. We find that genes involved in axonal regeneration form long-range, complex chromatin loops, and that cohesin is required for the full induction of the regenerative transcriptional program. Importantly, loss of cohesin results in disruption of chromatin architecture and severely impaired nerve regeneration. Complex enhancer-promoter loops are also enriched in the human fetal cortical plate, where the axonal growth potential is highest, and are lost in mature adult neurons. Together, these data provide an original three-dimensional chromatin map of adult sensory neurons in vivo and demonstrate a role for cohesin-dependent long-range promoter interactions in nerve regeneration.


Asunto(s)
Axones , Cromatina , Cohesinas , Regeneración Nerviosa , Regiones Promotoras Genéticas , Células Receptoras Sensoriales , Animales , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Ratones , Regiones Promotoras Genéticas/genética , Cromatina/metabolismo , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Axones/metabolismo , Axones/fisiología , Humanos , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Elementos de Facilitación Genéticos/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ganglios Espinales/metabolismo , Ganglios Espinales/citología , Nervio Ciático/metabolismo
7.
Mater Today Bio ; 28: 101250, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39318371

RESUMEN

Traumatic spinal cord injury (SCI), known for its limited intrinsic regeneration capacity, often results in considerable neurological impairment. Studies suggest that therapeutic techniques utilizing exosomes (Exo) to promote tissue regeneration and modulate immune responses are promising for SCI treatment. However, combining exosome therapy with biomaterials for SCI treatment is not very effective. This study developed an adhesive hydrogel using exosomes secreted by cortical neurons derived from human induced pluripotent stem cells (iPSCs) and decellularized extracellular matrix (dECM) from human umbilical cord mesenchymal stem cells (hUCMSCs) to enhance motor function recovery post-SCI. In vitro assessments demonstrated the excellent cytocompatibility of the dECM hydrogel. Additionally, the Exo-dECM hydrogel facilitated the polarization of early M2 macrophages, reduced neuronal apoptosis, and established a pro-regenerative microenvironment in a rodent SCI model. Subsequent analyses revealed significant activation of endogenous neural stem cells and promotion of axon regeneration and remyelination at eight weeks post-surgery. The Exo-dECM hydrogel also promoted the functional recovery and preservation of urinary tissue in SCI-afflicted rats. These findings highlighted that the Exo-dECM hydrogel is a promising therapeutic strategy for treating SCI.

8.
Cells ; 13(18)2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39329728

RESUMEN

Peripheral nerve regeneration depends on close interaction between neurons and Schwann cells (SCs). After nerve injury, SCs produce growth factors and cytokines that are crucial for axon re-growth. Previous studies revealed the supernatant of SCs exposed to nuclear magnetic resonance therapy (NMRT) treatment to increase survival and neurite formation of rat dorsal root ganglion (DRG) neurons in vitro. The aim of this study was to identify factors involved in transferring the observed NMRT-induced effects to SCs and consequently to DRG neurons. Conditioned media of NMRT-treated (CM NMRT) and untreated SCs (CM CTRL) were tested by beta-nerve growth factor (ßNGF) ELISA and multiplex cytokine panels to profile secreted factors. The expression of nociceptive transient receptor potential vanilloid 1 (TRPV1) channels was assessed and the intracellular calcium response in DRG neurons to high-potassium solution, capsaicin or adenosine triphosphate was measured mimicking noxious stimuli. NMRT induced the secretion of ßNGF and pro-regenerative-signaling factors. Blocking antibody experiments confirmed ßNGF as the main factor responsible for neurotrophic/neuritogenic effects of CM NMRT. The TRPV1 expression or sensitivity to specific stimuli was not altered, whereas the viability of cultured DRG neurons was increased. Positive effects of CM NMRT supernatant on DRG neurons are primarily mediated by increased ßNGF levels.


Asunto(s)
Ganglios Espinales , Neuritas , Células de Schwann , Ganglios Espinales/metabolismo , Ganglios Espinales/efectos de los fármacos , Animales , Células de Schwann/metabolismo , Células de Schwann/efectos de los fármacos , Neuritas/metabolismo , Neuritas/efectos de los fármacos , Ratas , Canales Catiónicos TRPV/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratas Wistar
9.
Mol Pharm ; 21(10): 4764-4785, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39235393

RESUMEN

Spinal cord injury (SCI) is a highly disabling neurological disorder. Its pathological process comprises an initial acute injury phase (primary injury) and a secondary injury phase (subsequent chronic injury). Although surgical, drug, and cell therapies have made some progress in treating SCI, there is no exact therapeutic strategy for treating SCI and promoting nerve regeneration due to the complexity of the pathological SCI process. The development of novel drug delivery systems to treat SCI is expected to significantly impact the individualized treatment of SCI due to its unique and excellent properties, such as active targeting and controlled release. In this review, we first describe the pathological progression of the SCI response, including primary and secondary injuries. Next, we provide a concise overview of newly developed nanoplatforms and their potential application in regulating and treating different pathological processes of SCI. Then, we introduce the existing potential problems and future clinical application perspectives of biomedical engineering-based therapies for SCI.


Asunto(s)
Materiales Biocompatibles , Sistemas de Liberación de Medicamentos , Traumatismos de la Médula Espinal , Traumatismos de la Médula Espinal/terapia , Humanos , Materiales Biocompatibles/química , Animales , Sistemas de Liberación de Medicamentos/métodos , Regeneración Nerviosa/efectos de los fármacos
10.
BMC Genomics ; 25(1): 896, 2024 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-39343885

RESUMEN

BACKGROUND: Decellularized extracellular matrix (dECM) is an intriguing natural biomaterial that has garnered significant attention due to its remarkable biological properties. In our study, we employed a cell-matrixed nerve graft for the repair of sciatic nerve defects in rats. The efficacy of this approach was assessed, and concurrently, the underlying molecular regulatory mechanisms were explored to elucidate how such grafts facilitate nerve regeneration. Long noncoding RNAs (lncRNAs) regulate mRNA expression via multiple mechanisms, including post-transcriptional regulation, transcription factor effects, and competitive binding with miRNAs. These interactions between lncRNAs and mRNAs facilitate precise control of gene expression, allowing organisms to adapt to varying biological environments and physiological states. By investigating the expression profiles and interaction dynamics of mRNAs and lncRNAs, we can enhance our understanding of the molecular mechanisms through which cell-matrixed nerve grafts influence neural repair. Such studies are pivotal in uncovering the intricate networks of gene regulation that underpin this process. RESULTS: Weighted gene co-expression network analysis (WGCNA) utilizes clustering algorithms, such as hierarchical clustering, to aggregate genes with similar expression profiles into modules. These modules, which potentially correspond to distinct biological functions or processes, can subsequently be analyzed for their association with external sample traits. By correlating gene modules with specific conditions, such as disease states or responses to treatments, WGCNA enables a deeper understanding of the genetic architecture underlying various phenotypic traits and their functional implications. We identified seven mRNA modules and five lncRNA modules that exhibited associations with treatment or time-related events by WGCNA. We found the blue (mRNAs) module which displayed a remarkable enrichment in "axonal guidance" and "metabolic pathways", exhibited strong co-expression with multiple lncRNA modules, including blue (related to "GnRH secretion" and "pyrimidine metabolism"), green (related to "arginine and proline metabolism"), black (related to "nitrogen metabolism"), grey60 (related to "PPAR signaling pathway"), and greenyellow (related to "steroid hormone biosynthesis"). All of the top 50 mRNAs and lncRNAs exhibiting the strongest correlation were derived from the blue module. Validation of key molecules were performed using immunohistochemistry and qRT-PCR. CONCLUSION: Revealing the principles and molecular regulatory mechanisms of the interaction between materials and biological entities, such as cells and tissues, is a direction for the development of biomimetic tissue engineering technologies and clinically effective products.


Asunto(s)
Regeneración Nerviosa , ARN Largo no Codificante , ARN Mensajero , Nervio Ciático , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Regeneración Nerviosa/genética , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Matriz Extracelular/metabolismo , Redes Reguladoras de Genes , Perfilación de la Expresión Génica , Ratas Sprague-Dawley
12.
Neuropharmacology ; 261: 110169, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39332671

RESUMEN

Peripheral nerve injury (PNI), typically caused by traumatic accidents or medical events, is currently one of the most common diseases that leads to limb disability. After PNI, tetrodotoxin-resistant voltage-gated sodium channel Nav1.8 is upregulated at the lesion site. Our earlier study suggested that ropivacaine promotes axon regrowth by regulating Nav1.8-mediated macrophage signaling. Nevertheless, the mechanism of ropivacaine in regulation of Nav1.8 expression remains incompletely understood. Kinesin family 5b (KIF5b) was reported to mediate the Nav1.8 axonal transport from dorsal root ganglia (DRGs) to lesion site. Herein, we investigated whether ropivacaine promotes axon regeneration through inhibition of KIF5b-mediated Nav1.8 transport. Reduced levels of KIF5b and Nav1.8 in DRGs coincide with their increase at the lesion site. Nav1.8 mRNA was significantly increased at the lesion site but not in DRGs. Surprisingly, ropivacaine reversed the alterations of Nav1.8 and KIF5b protein expression without affecting Nav1.8 mRNA level. Due to KIF5b overexpression in DRGs, Nav1.8 protein level was significantly decreased in DRGs and increased at the lesion site. We also found KIF5b overexpression significantly impaired behavioral functions, reduced the recovery index of compound muscle action potential (CMAP) amplitude, inhibited axonal regrowth, slowed M1 macrophage infiltration and shift to M2 phenotype, and delayed myelin debris clearance. Notably, all aforementioned results caused by KIF5b overexpression were alleviated by ropivacaine. Furthermore, we demonstrated that inhibition of Nav1.8 transport by A-803467 produced mitigating effects on the impairment of regenerative capacity induced by KIF5b overexpression similar to ropivacaine. These results suggest that ropivacaine promotes axonal regeneration at least partially by inhibiting KIF5b-mediated Nav1.8 forward transport.

13.
Front Mol Neurosci ; 17: 1411384, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39228795

RESUMEN

Retinoic acid receptor ß2 (RARß2) is an emerging therapeutic target for spinal cord injuries (SCIs) with a unique multimodal regenerative effect. We have developed a first-in-class RARß agonist drug, C286, that modulates neuron-glial pathways to induce functional recovery in a rodent model of sensory root avulsion. Here, using genome-wide and pathway enrichment analysis of avulsed rats' spinal cords, we show that C286 also influences the extracellular milieu (ECM). Protein expression studies showed that C286 upregulates tenascin-C, integrin-α9, and osteopontin in the injured cord. Similarly, C286 remodulates these ECM molecules, hampers inflammation and prevents tissue loss in a rodent model of spinal cord contusion C286. We further demonstrate C286's efficacy in human iPSC-derived neurons, with treatment resulting in a significant increase in neurite outgrowth. Additionally, we identify a putative efficacy biomarker, S100B, which plasma levels correlated with axonal regeneration in nerve-injured rats. We also found that other clinically available retinoids, that are not RARß specific agonists, did not lead to functional recovery in avulsed rats, demonstrating the requirement for RARß specific pathways in regeneration. In a Phase 1 trial, the single ascending dose (SAD) cohorts showed increases in expression of RARß2 in white blood cells correlative to increased doses and at the highest dose administered, the pharmacokinetics were similar to the rat proof of concept (POC) studies. Collectively, our data suggests that C286 signalling in neurite/axonal outgrowth is conserved between species and across nerve injuries. This warrants further clinical testing of C286 to ascertain POC in a broad spectrum of neurodegenerative conditions.

14.
Adv Mater ; : e2411628, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39268784

RESUMEN

Bioengineered nerve conduits have shown great promise for spinal cord injury (SCI) repair, while their practical values are limited by poor regenerative efficacy and lack of multi-level structural design. Here, inspired by the ingenious anatomy of natural spinal cords, a biomimetic multichannel silk nerve conduit (namely BNC@MSCs/SCs) with multicellular spatiotemporal distributions for effective SCI repair is presented. The biomimetic silk nerve conduit (BNC) with hierarchical channels and aligned pore structures is prepared via a modified directional freeze-casting strategy. Such hierarchical structures provide appropriate space for the mesenchymal stem cells (MSCs) and Schwann cells (SCs) settled in specific channels, which contributes to the generation of BNC@MSCs/SCs resembling the cellular spatiotemporal distributions of natural spinal cords. The in vitro results reveal the facilitated SC migration and MSC differentiation in such BNC@MSCs/SCs multicellular system, which further promotes the tube formation and cell migration of endothelial cells as well as M2 polarization of macrophages. Moreover, BNC@MSCs/SCs can effectively promote the tissue repair and function recovery in SCI rats by attenuating glial scar formation while promoting neuron regeneration and myelin sheath reconstruction. Thus, it is believed that the biomimetic multichannel silk nerve conduits with multicellular spatiotemporal distributions are valuable for SCI repair and other neural tissue regeneration.

15.
Adv Healthc Mater ; : e2401412, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39268836

RESUMEN

Clinically, conventional sutures for repair of short-distance nerve injuries (< 5 mm) may contribute to uncontrolled inflammation and scar formation, thus negatively impacting nerve regeneration. To repair transected peripheral nerves with short distances, a rapid-forming, robust adhesive chitosan hydrogel is prepared by synthesizing maleic and dopamine bi-functionalized fungal-sourced chitosan (DM) and subsequently photopolymerizing DM precursor solution. The hydrogel rapidly polymerized under UV light irradiation (≈2 s) and possessed a strong adhesive strength (273.33 ± 55.07 kPa), facilitating a fast bonding of nerve stump. Especially, its tailored degradation profile over 28 days supported both early gap bridging and subsequent nerve regeneration. Furthermore, deferoxamine (DFO), a pro-angiogenic drug, is loaded into the hydrogel to reach sustainable release, accelerating axonal growth synergistically. A 3 mm long sciatic nerve defects model in rats is used to investigate the efficacy of DM@DFO hydrogel for repairing peripheral nerve defects. After 60 days, the DM@DFO hydrogel significantly outperformed conventional sutures and fibrin glue, improving motor and sensory recovery by reducing inflammation, inhibiting scar formation, and accelerating vascular regeneration within 14 days post-repair. This work highlights the DM@DFO hydrogel as a promising tissue adhesive for effective short-distance peripheral nerve repair.

16.
Int J Biol Macromol ; 279(Pt 4): 135388, 2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39255892

RESUMEN

This study investigates the efficacy of a novel tissue-engineered scaffold for nerve repair and functional reconstruction following injury. Utilizing stable jet electrospinning, we fabricated aligned ultrafine fibers from dopamine and poly(L-lactic acid) (PLLA), further developing a biomimetic, oriented, and electroactive scaffold comprising poly(pyrrole) (PPy), polydopamine (PDA), and PLLA through dual in situ polymerizations. The scaffold demonstrated enhanced cell adhesion and reactive oxygen species (ROS) scavenging capabilities and promoted the differentiation of mesenchymal stem cells (MSCs) into Schwann-like cells, essential for nerve regeneration. In vivo assessments revealed significant peripheral nerve regeneration in 10 mm sciatic nerve defects in rats, with observations made 12 weeks post-transplantation. This included facilitated myelination and increased muscle density on the injured side, leading to improved motor function recovery. Our results suggest that the aligned PPy/PDA/PLLA fibrous scaffold offers a promising approach for promoting the differentiation of MSCs into Schwann-like cells conducive to nerve regeneration and represents a significant advancement in nerve repair technologies. This study provides a foundational basis for future research into tissue-engineered solutions for nerve damage, potentially impacting clinical strategies for nerve reconstruction.

17.
Heliyon ; 10(17): e37231, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39296039

RESUMEN

Objective: The purpose of this study is to investigate the role of PMMA-induced biofilm in nerve regeneration compared with silicone-induced biofilm involved in the mechanism. Methods: PMMA or silicon rods were placed next to the sciatic nerve to induce a biological membrane which was assayed by PCR, Western blot, immunohistochemistry, immunofluorescence and proteomics. A 10 mm sciatic nerve gaps were repaired with the autologous nerve wrapped in an induced biological membrane. The repair effects were observed through general observation, functional evaluation of nerve regeneration, ultrasound examination, neural electrophysiology, the wet weight ratio of bilateral pretibial muscle and histological evaluation. Cell proliferation and migration of Schwann cells co-cultured with EGF-treated fibroblasts combined with siRNA were investigated. Results: The results indicated that expression of GDNF, NGF and VEGF along with neovascularization was similar in the silicone and PMMA group and as the highest at 6 weeks after operation. Nerve injury repair mediated by toluidine blue and S100ß/NF200 expression, the sensory and motor function evaluation, ultrasound, target organ muscle wet-weight ratio, percentage of collagen fiber, electromyography and histochemical staining were not different between the two groups and better than blank group. EGF-treated fibroblasts promoted proliferation and migration may be Tnc expression dependently. Conclusion: Our study suggested that PMMA similar to silicon induced biofilm may promote autogenous nerve transplantation to repair nerve defects through EGF/Tnc/FN1 to increase Schwann cells proliferation and migration.

18.
Tissue Cell ; 91: 102556, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39293138

RESUMEN

BACKGROUND AND AIMS: Challenges in treating peripheral nerve injury include prolonged repair time and insufficient functional recovery. Stem cell therapy coupled with neural tissue engineering has been shown to induce nerve regeneration following peripheral nerve injury. Among these stem cells, adipose-derived stem cells (ADSCs) are preferred due to their accessibility, expansion, multidirectional differentiation, and production of essential nutrient factors for nerve growth. In recent years, ADSC-laden nerve guide conduit has been utilized to enhance the therapeutic effects of tissue-engineered nerve grafts. This review explores existing research that recognizes the roles played by ADSCs in inducing peripheral nerve regeneration following injury and summarizes the different methods of application of ADSC-laden nerve conduit in neural tissue engineering.

19.
Sci Rep ; 14(1): 20748, 2024 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237597

RESUMEN

The peripheral nervous system consists of ganglia, nerve trunks, plexuses, and nerve endings, that transmit afferent and efferent information. Regeneration after a peripheral nerve damage is sluggish and imperfect. Peripheral nerve injury frequently causes partial or complete loss of motor and sensory function, physical impairment, and neuropathic pain, all of which have a negative impact on patients' quality of life. Because the mechanism of peripheral nerve injury and healing is still unclear, the therapeutic efficacy is limited. As peripheral nerve injury research has processed, an increasing number of studies have revealed that biological scaffolds work in tandem with progenitor cells to repair peripheral nerve injury. Here, we fabricated collagen chitosan nerve conduit bioscaffolds together with collagen and then filled neuroepithelial stem cells (NESCs). Scanning electron microscopy showed that the NESCs grew well on the scaffold surface. Compared to the control group, the NESCs group contained more cells with bigger diameters and myelinated structures around the axons. Our findings indicated that a combination of chitosan-collagen bioscaffold and neural stem cell transplantation can facilitate the functional restoration of peripheral nerve tissue, with promising future applications and research implications.


Asunto(s)
Quitosano , Colágeno , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos , Andamios del Tejido , Quitosano/química , Regeneración Nerviosa/fisiología , Colágeno/química , Animales , Andamios del Tejido/química , Traumatismos de los Nervios Periféricos/terapia , Ratas , Células Neuroepiteliales/citología , Células-Madre Neurales/citología , Nervios Periféricos/fisiología , Nervio Ciático/fisiología
20.
Biomaterials ; 314: 122811, 2024 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-39265373

RESUMEN

Erectile dysfunction (ED) is a common male sexual disorder characterized by repeated or persistent difficulty in achieving or maintaining an erection. It can arise from various factors, with cavernous nerve injury (CNI) from radical prostatectomy being a predominant cause of iatrogenic ED, posing significant clinical concerns. The complexity of cavernous tissue damage in CNI-induced ED (CNIED) often results in poor efficacy and resistance to conventional vascular ED treatments. To address CNI-induced ED, this study developed a system of magnetic mesoporous silica nanoparticles (MSNs) loaded with peptides for targeted treatment. Core-shell Fe3O4-coated MSNs were used as drug carriers and loaded with RADA16-I/RAD-RGI peptides (PD) to create a neurotrophic microenvironment to treat peripheral nerve defects. Furthermore, the neuro-targeting peptide HLNILSTLWKYR (PT) was grafted onto MSNs. The in vivo therapeutic effect was evaluated using a rat bilateral cavernous nerve injury (BCNI) model. The results showed that the neuro-targeted Fe3O4@SiO2-PT-PD nanoparticles significantly promoted regeneration of the cavernous nerve and restored erectile function. This promising strategy offers significant clinical potential for treating CNI-induced ED. Nanomedicine technology has the potential to not only improve treatment outcomes but also reduce side effects in healthy cells, paving the way for more accurate targeted repair of cavernous nerve damage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA