Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.957
Filter
1.
Methods Mol Biol ; 2848: 135-150, 2025.
Article in English | MEDLINE | ID: mdl-39240521

ABSTRACT

Mammals do not possess the ability to spontaneously repair or regenerate damaged retinal tissue. In contrast to teleost fish which are capable of retina regeneration through the action of Müller glia, mammals undergo a process of reactive gliosis and scarring that inhibits replacement of lost neurons. Thus, it is important to discover novel methods for stimulating mammalian Müller glia to dedifferentiate and produce progenitor cells that can replace lost retinal neurons. Inducing an endogenous regenerative pathway mediated by Müller glia would provide an attractive alternative to stem cell injections or gene therapy approaches. Extracellular vesicles (EVs) are now recognized to serve as a novel form of cell-cell communication through the transfer of cargo from donor to recipient cells or by the activation of signaling cascades in recipient cells. EVs have been shown to promote proliferation and regeneration raising the possibility that delivery of EVs could be a viable treatment for visual disorders. Here, we provide protocols to isolate EVs for use in retina regeneration experiments.


Subject(s)
Extracellular Vesicles , Regeneration , Retina , Animals , Extracellular Vesicles/metabolism , Retina/metabolism , Retina/cytology , Retina/physiology , Ependymoglial Cells/metabolism , Ependymoglial Cells/cytology , Mice , Cell Communication , Cell Proliferation , Nerve Regeneration/physiology
2.
Methods Mol Biol ; 2848: 217-247, 2025.
Article in English | MEDLINE | ID: mdl-39240526

ABSTRACT

Various strategies for replacing retinal neurons lost in degenerative diseases are under investigation, including stimulating the endogenous regenerative capacity of Müller Glia (MG) as injury-inducible retinal stem cells. Inherently regenerative species, such as zebrafish, have provided key insights into mechanisms regulating MG dedifferentiation to a stem-like state and the proliferation of MG and MG-derived progenitor cells (MGPCs). Interestingly, promoting MG/MGPC proliferation is not sufficient for regeneration, yet mechanistic studies are often focused on this measure. To fully account for the regenerative process, and facilitate screens for factors regulating cell regeneration, an assay for quantifying cell replacement is required. Accordingly, we adapted an automated reporter-assisted phenotypic screening platform to quantify the pace of cellular regeneration kinetics following selective cell ablation in larval zebrafish. Here, we detail a method for using this approach to identify chemicals and genes that control the rate of retinal cell regeneration following selective retinal cell ablation.


Subject(s)
Zebrafish , Animals , Retina/cytology , Retina/metabolism , Phenotype , Cell Proliferation , Regeneration , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Kinetics , Nerve Regeneration/physiology
3.
J Nanobiotechnology ; 22(1): 590, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342236

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) often leads to a loss of motor and sensory function. Axon regeneration and outgrowth are key events for functional recovery after spinal cord injury. Endogenous growth of axons is associated with a variety of factors. Inspired by the relationship between developing nerves and blood vessels, we believe spinal cord-derived microvascular endothelial cells (SCMECs) play an important role in axon growth. RESULTS: We found SCMECs could promote axon growth when co-cultured with neurons in direct and indirect co-culture systems via downregulating the miR-323-5p expression of neurons. In rats with spinal cord injury, neuron-targeting nanoparticles were employed to regulate miR-323-5p expression in residual neurons and promote function recovery. CONCLUSIONS: Our study suggests that SCMEC can promote axon outgrowth by downregulating miR-323-5p expression within neurons, and miR-323-5p could be selected as a potential target for spinal cord injury repair.


Subject(s)
Axons , Coculture Techniques , Endothelial Cells , MicroRNAs , Rats, Sprague-Dawley , Spinal Cord Injuries , Spinal Cord , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Endothelial Cells/metabolism , Rats , Spinal Cord/metabolism , Axons/metabolism , Neurons/metabolism , Cells, Cultured , Nanoparticles/chemistry , Nerve Regeneration , Female
4.
Narra J ; 4(2): e880, 2024 08.
Article in English | MEDLINE | ID: mdl-39280316

ABSTRACT

Recent studies highlighted the role of platelet-rich plasma (PRP) in progenitor cell homing, migration, and nerve cell regeneration while also inhibiting fibrosis and apoptosis in cavernous nerve injury (CNI). The aim of this study was to investigate the effect of PRP administration on axon and collagen regeneration in CNI. A true experimental study using a post-test-only control group design was conducted. Twenty-five male Wistar rats (Rattus norvegicus), weighing 200-300 grams, were divided into five groups: two control groups (sham procedure and negative control), and three experimental groups receiving local PRP, intraperitoneal PRP, and a combination of local and intraperitoneal PRP. The cavernous nerve was injured with a hemostasis clamp for one minute before 200 µL of 200 PRP was injected locally, intraperitoneally, or both, depending on the group. After four weeks, the rats were euthanized, tissue segments (2 mm) from each cavernous nerve and mid-penis were collected and analyzed for collagen density, axon diameter, and number of myelinated axons. Our study found that collagen growth was slower in CNI group without PRP (sham procedure) compared to all PRP groups (local, intraperitoneal, and combination). The intraperitoneal PRP group had the highest collagen density at 5.62 µm; however, no significant difference was observed in collagen density among all groups (p=0.056). Similar axon diameter was found across the groups, with no statistically significant difference observed (p=0.856). In the number of myelinated axons, a significant difference was found among all groups with significantly more axons in local PRP and combined local and intraperitoneal PRP groups compared to others (p=0.026). In conclusion, PRP administration improved the number of myelinated axons in CNI, suggesting PRP role in CNI regeneration and the potential for an innovative approach to treating erectile dysfunction associated with CNI.


Subject(s)
Axons , Collagen , Erectile Dysfunction , Nerve Regeneration , Penis , Platelet-Rich Plasma , Rats, Wistar , Animals , Male , Collagen/metabolism , Rats , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Axons/physiology , Axons/pathology , Axons/drug effects , Penis/innervation , Penis/drug effects , Erectile Dysfunction/therapy , Erectile Dysfunction/drug therapy , Disease Models, Animal , Peripheral Nerve Injuries/therapy
5.
Proc Natl Acad Sci U S A ; 121(38): e2402518121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39254997

ABSTRACT

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.


Subject(s)
Axons , Chromatin , Cohesins , Nerve Regeneration , Promoter Regions, Genetic , Sensory Receptor Cells , Animals , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Mice , Promoter Regions, Genetic/genetics , Chromatin/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Axons/metabolism , Axons/physiology , Humans , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Enhancer Elements, Genetic/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Sciatic Nerve/metabolism
6.
Elife ; 132024 Sep 26.
Article in English | MEDLINE | ID: mdl-39324575

ABSTRACT

Fibro-adipogenic progenitors (FAPs) are muscle-resident mesenchymal progenitors that can contribute to muscle tissue homeostasis and regeneration, as well as postnatal maturation and lifelong maintenance of the neuromuscular system. Recently, traumatic injury to the peripheral nerve was shown to activate FAPs, suggesting that FAPs can respond to nerve injury. However, questions of how FAPs can sense the anatomically distant peripheral nerve injury and whether FAPs can directly contribute to nerve regeneration remained unanswered. Here, utilizing single-cell transcriptomics and mouse models, we discovered that a subset of FAPs expressing GDNF receptors Ret and Gfra1 can respond to peripheral nerve injury by sensing GDNF secreted by Schwann cells. Upon GDNF sensing, this subset becomes activated and expresses Bdnf. FAP-specific inactivation of Bdnf (Prrx1Cre; Bdnffl/fl) resulted in delayed nerve regeneration owing to defective remyelination, indicating that GDNF-sensing FAPs play an important role in the remyelination process during peripheral nerve regeneration. In aged mice, significantly reduced Bdnf expression in FAPs was observed upon nerve injury, suggesting the clinical relevance of FAP-derived BDNF in the age-related delays in nerve regeneration. Collectively, our study revealed the previously unidentified role of FAPs in peripheral nerve regeneration, and the molecular mechanism behind FAPs' response to peripheral nerve injury.


Subject(s)
Brain-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor , Mesenchymal Stem Cells , Nerve Regeneration , Peripheral Nerve Injuries , Animals , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Peripheral Nerve Injuries/metabolism , Mice , Mesenchymal Stem Cells/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Schwann Cells/metabolism , Male , Proto-Oncogene Proteins c-ret/metabolism , Proto-Oncogene Proteins c-ret/genetics
7.
Neuron ; 112(18): 2993-2995, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39326387

ABSTRACT

While the enteric nervous system (ENS) is highly dynamic during development, the extent to which it is capable of repair remains unclear. In this issue of Neuron, Stavely et al.1 show that enteric neurons can reinnervate damaged regions to regain functionality using a glial positioning system (GPS) as their guide.


Subject(s)
Enteric Nervous System , Nerve Regeneration , Neuroglia , Neuroglia/physiology , Enteric Nervous System/physiology , Enteric Nervous System/cytology , Animals , Nerve Regeneration/physiology , Neurites/physiology , Intestines/physiology , Humans
8.
Turk J Med Sci ; 54(4): 792-803, 2024.
Article in English | MEDLINE | ID: mdl-39295602

ABSTRACT

Background/aim: In the literature, almost all of the nerve conduits proposed for obtaining better nerve recovery were applied as graft materials. In this study, we aimed to propose a new nerve conduit model with a flap pattern and evaluate the effect of a pedicled vascularized jejunal flap on nerve regeneration after wrapping it around a sciatic nerve. Materials and methods: A total of 90 Wistar albino rats were randomly divided into nine groups with 10 rats in each. The first three groups constituted the control groups, whereas Groups 4-6 were the jejunum conduit (JC)-applied groups. A mucosa-resected JC (MRJC) was applied in Groups 7 and 8. Epineurial neurorrhaphy was performed in Groups 1, 4, and 7; repair with a nerve graft was applied in Groups 2, 5, and 8; and a 1-cm-long nerve defect was created in Groups 3, 6, and 9. After 2 months of follow-up, nerve regeneration was assessed by statistical analyses of the Sciatic Functional Index (SFI) and histopathological evaluation. Results: The MRJC groups had significantly better results in terms of SFI (p = 0.005). Statistical differences in axonal degeneration, axonal density, myelination, and disorganization were found between all control groups and MRJC groups (p = 0.022, p = 0.001, p = 0.001, and p = 0.039, respectively). Conclusion: In this study, the feasibility of wrapping around the nerve repair zones of pedicled autologous flaps designed in a tubular fashion was observed in a small rat model. The findings must be further validated with larger animals before clinical testing.


Subject(s)
Jejunum , Nerve Regeneration , Rats, Wistar , Sciatic Nerve , Surgical Flaps , Animals , Nerve Regeneration/physiology , Rats , Sciatic Nerve/surgery , Jejunum/surgery , Male
9.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(9): 1149-1156, 2024 Sep 15.
Article in Chinese | MEDLINE | ID: mdl-39300893

ABSTRACT

Objective: To describe the research progress of silk-based biomaterials in peripheral nerve repair and provide useful ideals to accelerate the regeneration of large-size peripheral nerve injury. Methods: The relative documents about silk-based biomaterials used in peripheral nerve regeneration were reviewed and the different strategies that could accelerate peripheral nerve regeneration through building bioactive microenvironment with silk fibroin were discussed. Results: Many silk fibroin tissue engineered nerve conduits have been developed to provide multiple biomimetic microstructures, and different microstructures have different mechanisms of promoting nerve repair. Biomimetic porous structures favor the nutrient exchange at wound sites and inhibit the invasion of scar tissue. The aligned structures can induce the directional growth of nerve tissue, while the multiple channels promote the axon elongation. When the fillers are introduced to the conduits, better growth, migration, and differentiation of nerve cells can be achieved. Besides biomimetic structures, different nerve growth factors and bioactive drugs can be loaded on silk carriers and released slowly at nerve wounds, providing suitable biochemical cues. Both the biomimetic structures and the loaded bioactive ingredients optimize the niches of peripheral nerves, resulting in quicker and better nerve repair. With silk biomaterials as a platform, fusing multiple ways to achieve the multidimensional regulation of nerve microenvironments is becoming a critical strategy in repairing large-size peripheral nerve injury. Conclusion: Silk-based biomaterials are useful platforms to achieve the design of biomimetic hierarchical microstructures and the co-loading of various bioactive ingredients. Silk fibroin nerve conduits provide suitable microenvironment to accelerate functional recovery of peripheral nerves. Different optimizing strategies are available for silk fibroin biomaterials to favor the nerve regeneration, which would satisfy the needs of various nerve tissue repair. Bioactive silk conduits have promising future in large-size peripheral nerve regeneration.


Subject(s)
Biocompatible Materials , Fibroins , Nerve Regeneration , Peripheral Nerves , Silk , Tissue Engineering , Tissue Scaffolds , Nerve Regeneration/drug effects , Biocompatible Materials/chemistry , Fibroins/chemistry , Tissue Scaffolds/chemistry , Peripheral Nerves/physiology , Tissue Engineering/methods , Silk/chemistry , Animals , Peripheral Nerve Injuries/therapy , Humans , Guided Tissue Regeneration/methods
10.
Theranostics ; 14(14): 5662-5681, 2024.
Article in English | MEDLINE | ID: mdl-39310103

ABSTRACT

Rationale: Spinal cord injury (SCI)-induced vascular damage causes ischemia and hypoxia at the injury site, which, in turn, leads to profound metabolic disruptions. The effects of these metabolic alterations on neural tissue remodeling and functional recovery have yet to be elucidated. The current study aimed to investigate the consequences of the SCI-induced hypoxic environment at the epicenter of the injury. Methods: This study employed metabolomics to assess changes in energy metabolism after SCI. The use of a lactate sensor identified lactate shuttle between endothelial cells (ECs) and neurons. Reanalysis of single-cell RNA sequencing data demonstrated reduced MCT1 expression in ECs after SCI. Additionally, an adeno-associated virus (AAV) overexpressing MCT1 was utilized to elucidate its role in endothelial-neuronal interactions, tissue repair, and functional recovery. Results: The findings revealed markedly decreased monocarboxylate transporter 1 (MCT1) expression that facilitates lactate delivery to neurons to support their energy metabolism in ECs post-SCI. This decreased expression of MCT1 disrupts lactate transport to neurons, resulting in a metabolic imbalance that impedes axonal regeneration. Strikingly, our results suggested that administering adeno-associated virus specifically to ECs to restore MCT1 expression enhances axonal regeneration and improves functional recovery in SCI mice. These findings indicate a novel link between lactate shuttling from endothelial cells to neurons following SCI and subsequent neural functional recovery. Conclusion: In summary, the current study highlights a novel metabolic pathway for therapeutic interventions in the treatment of SCI. Additionally, our findings indicate the potential benefits of targeting lactate transport mechanisms in recovery from SCI.


Subject(s)
Axons , Endothelial Cells , Lactic Acid , Monocarboxylic Acid Transporters , Spinal Cord Injuries , Symporters , Spinal Cord Injuries/metabolism , Animals , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Endothelial Cells/metabolism , Lactic Acid/metabolism , Mice , Axons/metabolism , Symporters/metabolism , Symporters/genetics , Recovery of Function/physiology , Dependovirus/genetics , Nerve Regeneration , Neurons/metabolism , Energy Metabolism , Mice, Inbred C57BL , Female , Disease Models, Animal , Humans
12.
Indian J Ophthalmol ; 72(10): 1404-1411, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39331430

ABSTRACT

This study aimed to comprehensively explore the intricacies of corneal neurotization (CN) and the nuanced factors that set it apart from routine clinical practice, exerting a substantial influence on its success. A symbiotic relationship is evident between corneal innervation and ocular surface health. The loss of corneal innervation results in a potentially challenging corneal condition known as neurotrophic keratopathy (NK). The majority of treatments are primarily focused on preventing epithelial breakdown rather than addressing the underlying pathogenesis. Consequently, to address the impaired corneal sensation (underlying etiology), a novel surgical approach has emerged, namely CN, which involves transferring healthy sensory nerve axons to the affected cornea. This review offers valuable insights into the existing body of supporting evidence for CN, meticulously examining clinical studies, case reports, and experimental findings. The aim is to enhance our understanding of the effectiveness and potential outcomes associated with this innovative surgical technique. The exploration of innovative therapeutic avenues holds promise for revolutionizing the management of NK, offering a potentially permanent solution to a condition once deemed incurable and severely debilitating.


Subject(s)
Cornea , Corneal Diseases , Nerve Transfer , Humans , Cornea/innervation , Cornea/surgery , Corneal Diseases/surgery , Corneal Diseases/diagnosis , Nerve Transfer/methods , Nerve Regeneration/physiology
13.
PLoS One ; 19(9): e0304773, 2024.
Article in English | MEDLINE | ID: mdl-39231134

ABSTRACT

Application of polyethylene glycol (PEG) to a peripheral nerve injury at the time of primary neurorrhaphy is thought to prevent Wallerian degeneration via direct axolemma fusion. The molecular mechanisms of nerve fusion and recovery are unclear. Our study tested the hypothesis that PEG alters gene expression in neural and muscular environments as part of its restorative properties. Lewis rats underwent unilateral sciatic nerve transection with immediate primary repair. Subjects were randomly assigned to receive either PEG treatment or standard repair at the time of neurorrhaphy. Samples of sciatic nerve distal to the injury and tibialis muscle at the site of innervation were harvested at 24 hours and 4 weeks postoperatively. Total RNA sequencing and subsequent bioinformatics analyses were used to identify significant differences in differentially expressed genes (DEGs) and their related biological pathways (p<0.05) in PEG-treated subjects compared to non-PEG controls. No significant DEGs were identified in PEG-treated sciatic nerve compared to controls after 24 hours, but 1,480 DEGs were identified in PEG-treated tibialis compared to controls. At 4 weeks, 918 DEGs were identified in PEG-treated sciatic nerve, whereas only 3 DEGs remained in PEG-treated tibialis compared to controls. DEGs in sciatic were mostly upregulated (79%) and enriched in pathways present during nervous system development and growth, whereas DEGs in muscle were mostly downregulated (77%) and related to inflammation and tissue repair. Our findings indicate that PEG application during primary neurorrhaphy leads to significant differential gene regulation in the neural and muscular environment that is associated with improved functional recovery in animals treated with PEG compared to sham non-PEG controls. A detailed understanding of key molecules underlying PEG function in recovery after peripheral nerve repair may facilitate amplification of PEG effects through systemic or focal treatments at the time of neurotmesis.


Subject(s)
Muscle, Skeletal , Peripheral Nerve Injuries , Polyethylene Glycols , Rats, Inbred Lew , Sciatic Nerve , Animals , Rats , Sciatic Nerve/injuries , Peripheral Nerve Injuries/genetics , Polyethylene Glycols/pharmacology , Muscle, Skeletal/metabolism , Muscle, Skeletal/innervation , Muscle, Skeletal/drug effects , Disease Models, Animal , Sequence Analysis, RNA , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Male , Gene Expression Regulation/drug effects , Gene Expression Profiling
14.
Ann Plast Surg ; 93(3S Suppl 2): S113-S115, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39230294

ABSTRACT

ABSTRACT: Peripheral nerve injuries (PNIs) represent a complex clinical challenge, necessitating precise diagnostic approaches for optimal management. Traditional diagnostic methods often fall short in accurately assessing nerve recovery as these methods rely on the completion of nerve reinnervation, which can prolong a patient's treatment. Diffusion tensor imaging (DTI), a noninvasive magnetic resonance imaging (MRI) technique, has emerged as a promising tool in this context. DTI offers unique advantages including the ability to quantify nerve recovery and provide in vivo visualizations of neuronal architecture. Therefore, this review aims to examine and outline DTI techniques and its utility in detecting distal nerve regeneration in both preclinical and clinical settings for peripheral nerve injury.


Subject(s)
Diffusion Tensor Imaging , Nerve Regeneration , Peripheral Nerve Injuries , Humans , Peripheral Nerve Injuries/diagnostic imaging , Diffusion Tensor Imaging/methods , Nerve Regeneration/physiology
15.
Sci Rep ; 14(1): 20748, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237597

ABSTRACT

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.


Subject(s)
Chitosan , Collagen , Nerve Regeneration , Peripheral Nerve Injuries , Tissue Scaffolds , Chitosan/chemistry , Nerve Regeneration/physiology , Collagen/chemistry , Animals , Tissue Scaffolds/chemistry , Peripheral Nerve Injuries/therapy , Rats , Neuroepithelial Cells/cytology , Neural Stem Cells/cytology , Peripheral Nerves/physiology , Sciatic Nerve/physiology
16.
Neuromolecular Med ; 26(1): 37, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266914

ABSTRACT

As the primary connection between the eye and brain, the optic nerve plays a pivotal role in visual information transmission. Injuries to the optic nerve can occur for various reasons, including trauma, glaucoma, and neurodegenerative diseases. Retinal ganglion cells (RGCs), a type of neurons that extend axons through the optic nerve, can rapidly respond to injury and initiate cell death. Additionally, following optic nerve injury microglia, which serve as markers of neuroinflammation, transition from a resting state to an activated state. The phosphorylation of collapsin response mediator protein2 (CRMP2) in the semaphorin 3A (Sema3A) signalling pathway affects several processes, including axon guidance and neuron regeneration. In this study, we used an optic nerve crush (ONC) mouse model to investigate the effects of suppressing CRMP2 phosphorylation on microglia activation. We found that CRMP2 phosphorylation inhibitor suppressed RGCs loss and promoted neuronal regeneration following ONC. In addition, CRMP2 S522A mutant (CRMP2 KI) mice exhibited decreased microglial activation in both the retina and optic nerve following ONC. These results suggest that inhibiting the phosphorylation of CRMP2 can alleviate the loss of RGCs and microglial activation after optic nerve injury, providing insight into the development of treatments for optical neuropathies and neurodegenerative diseases.


Subject(s)
Intercellular Signaling Peptides and Proteins , Microglia , Nerve Regeneration , Nerve Tissue Proteins , Optic Nerve Injuries , Optic Nerve , Retina , Retinal Ganglion Cells , Semaphorin-3A , Animals , Optic Nerve Injuries/physiopathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/drug therapy , Microglia/metabolism , Microglia/drug effects , Phosphorylation , Mice , Nerve Regeneration/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Optic Nerve/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Retina/drug effects , Retina/metabolism , Nerve Crush , Mice, Inbred C57BL , Male , Disease Models, Animal , Mice, Transgenic
17.
Neural Dev ; 19(1): 17, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267104

ABSTRACT

The complex interplay between vascular signaling and neurogenesis in the adult brain remains a subject of intense research. By exploiting the unique advantages of the zebrafish model, in particular the persistent activity of neural stem cells (NSCs) and the remarkable ability to repair brain lesions, we investigated the links between NSCs and cerebral blood vessels. In this study, we first examined the gene expression profiles of vascular endothelial growth factors aa and bb (vegfaa and vegfbb), under physiological and regenerative conditions. Employing fluorescence in situ hybridization combined with immunostaining and histology techniques, we demonstrated the widespread expression of vegfaa and vegfbb across the brain, and showed their presence in neurons, microglia/immune cells, endothelial cells and NSCs. At 1 day post-lesion (dpl), both vegfaa and vegfbb were up-regulated in neurons and microglia/peripheral immune cells (macrophages). Analysis of vegf receptors (vegfr) revealed high expression throughout the brain under homeostatic conditions, with vegfr predominantly expressed in neurons and NSCs and to a lower extent in microglia/immune cells and endothelial cells. These findings were further validated by Vegfr3 and Vegfr4 immunostainings, which showed significant expression in neurogenic radial glial cells.Following brain lesion (1 dpl), while vegfr gene expression remained stable, vegfr transcripts were detected in proliferative cells within the injured parenchyma. Collectively, our results provide a first overview of Vegf/Vegfr signaling in the brain and suggest important roles for Vegf in neurogenesis and regenerative processes.


Subject(s)
Brain , Neurogenesis , Vascular Endothelial Growth Factor A , Zebrafish Proteins , Zebrafish , Animals , Neurogenesis/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Brain/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Neural Stem Cells/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Nerve Regeneration/physiology
19.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273538

ABSTRACT

Spinal cord injury (SCI) is a catastrophic condition that disrupts neurons within the spinal cord, leading to severe motor and sensory deficits. While current treatments can alleviate pain, they do not promote neural regeneration or functional recovery. Three-dimensional (3D) bioprinting offers promising solutions for SCI repair by enabling the creation of complex neural tissue constructs. This review provides a comprehensive overview of 3D bioprinting techniques, bioinks, and stem cell applications in SCI repair. Additionally, it highlights recent advancements in 3D bioprinted scaffolds, including the integration of conductive materials, the incorporation of bioactive molecules like neurotrophic factors, drugs, and exosomes, and the design of innovative structures such as multi-channel and axial scaffolds. These innovative strategies in 3D bioprinting can offer a comprehensive approach to optimizing the spinal cord microenvironment, advancing SCI repair. This review highlights a comprehensive understanding of the current state of 3D bioprinting in SCI repair, offering insights into future directions in the field of regenerative medicine.


Subject(s)
Bioprinting , Printing, Three-Dimensional , Spinal Cord Injuries , Tissue Engineering , Tissue Scaffolds , Spinal Cord Injuries/therapy , Humans , Bioprinting/methods , Tissue Scaffolds/chemistry , Animals , Tissue Engineering/methods , Regenerative Medicine/methods , Nerve Regeneration
20.
Nutr Res ; 129: 55-67, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39217889

ABSTRACT

Peripheral nerve injuries (PNIs) are prevalent conditions mainly resulting from systemic causes, including autoimmune diseases and diabetes mellitus, or local causes, for example, chemical injury and perioperative nerve injury, which can cause a varying level of neurosensory disturbances (NSDs). Coenzyme Q10 (CoQ10) is an essential regulator of mitochondrial respiration and oxidative metabolism. Here, we review the pathophysiology of NSDs caused by PNIs, the current understanding of CoQ10's bioactivities, and its potential therapeutic roles in nerve regeneration, based on evidence from experimental and clinical studies involving CoQ10 supplementation. In summary, CoQ10 supplementation shows promise as a neuroprotective agent, potentially enhancing treatment efficacy for NSDs by reducing oxidative stress and inflammation. Future studies should focus on well-designed clinical trials with large sample sizes, using CoQ10 formulations with proven bioavailability and varying treatment duration, to further elucidate its neuroprotective effects and to optimize nerve regeneration in PNIs-induced NSDs.


Subject(s)
Neuroprotective Agents , Oxidative Stress , Peripheral Nerve Injuries , Ubiquinone , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use , Ubiquinone/pharmacology , Humans , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/complications , Animals , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Nerve Regeneration/drug effects , Dietary Supplements , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL