Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 5.588
1.
PLoS One ; 19(5): e0300254, 2024.
Article En | MEDLINE | ID: mdl-38696450

Low back pain, knee osteoarthritis, and cancer patients suffer from chronic pain. Aberrant nerve growth into intervertebral disc, knee, and tumors, are common pathologies that lead to these chronic pain conditions. Axonal dieback induced by capsaicin (Caps) denervation has been FDA-approved to treat painful neuropathies and knee osteoarthritis but with short-term efficacy and discomfort. Herein, we propose to evaluate pyridoxine (Pyr), vincristine sulfate (Vcr) and ionomycin (Imy) as axonal dieback compounds for denervation with potential to alleviate pain. Previous literature suggests Pyr, Vcr, and Imy can cause undesired axonal degeneration, but no previous work has evaluated axonal dieback and cytotoxicity on adult rat dorsal root ganglia (DRG) explants. Thus, we performed axonal dieback screening using adult rat DRG explants in vitro with Caps as a positive control and assessed cytotoxicity. Imy inhibited axonal outgrowth and slowed axonal dieback, while Pyr and Vcr at high concentrations produced significant reduction in axon length and robust axonal dieback within three days. DRGs treated with Caps, Vcr, or Imy had increased DRG cytotoxicity compared to matched controls, but overall cytotoxicity was minimal and at least 88% lower compared to lysed DRGs. Pyr did not lead to any DRG cytotoxicity. Further, neither Pyr nor Vcr triggered intervertebral disc cell death or affected cellular metabolic activity after three days of incubation in vitro. Overall, our findings suggest Pyr and Vcr are not toxic to DRGs and intervertebral disc cells, and there is potential for repurposing these compounds for axonal dieback compounds to cause local denervation and alleviate pain.


Axons , Denervation , Ganglia, Spinal , Intervertebral Disc , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Rats , Intervertebral Disc/drug effects , Intervertebral Disc/pathology , Axons/drug effects , Capsaicin/pharmacology , Rats, Sprague-Dawley , Male , Vincristine/pharmacology
2.
Biofabrication ; 16(3)2024 May 09.
Article En | MEDLINE | ID: mdl-38565133

Spinal cord injury (SCI) can cause permanent impairment to motor or sensory functions. Pre-cultured neural stem cell (NSC) hydrogel scaffolds have emerged as a promising approach to treat SCI by promoting anti-inflammatory effects, axon regrowth, and motor function restoration. Here, in this study, we performed a coaxial extrusion process to fabricate a core-shell hydrogel microfiber with high NSC density in the core portion. Oxidized hyaluronic acid, carboxymethyl chitosan, and matrigel blend were used as a matrix for NSC growth and to facilitate the fabrication process. During thein vitrodifferentiation culture, it was found that NSC microfibers could differentiate into neurons and astrocytes with higher efficiency compared to NSC cultured in petri dishes. Furthermore, duringin vivotransplantation, NSC microfibers were coated with polylactic acid nanosheets by electrospinning for reinforcement. The coated NSC nanofibers exhibited higher anti-inflammatory effect and lesion cavity filling rate compared with the control group. Meanwhile, more neuron- and oligodendrocyte-like cells were visualized at the lesion epicenter. Finally, axon regrowth across the whole lesion site was observed, demonstrating that the microfiber could guide renascent axon regrowth. Experiment results indicate that the NSC microfiber is a promising bioactive treatment for complete SCI treatment with superior outcomes.


Axons , Cell Differentiation , Neural Stem Cells , Neurons , Spinal Cord Injuries , Tissue Scaffolds , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Axons/drug effects , Axons/physiology , Axons/metabolism , Cell Differentiation/drug effects , Neurons/cytology , Neurons/drug effects , Tissue Scaffolds/chemistry , Rats, Sprague-Dawley , Hydrogels/chemistry , Hydrogels/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Chitosan/analogs & derivatives , Cells, Cultured , Nerve Regeneration/drug effects , Nanofibers/chemistry , Rats , Female
3.
Environ Int ; 186: 108643, 2024 Apr.
Article En | MEDLINE | ID: mdl-38615544

Exposure to bisphenol S (BPS) is known to adversely affect neuronal development. As pivotal components of neuronal polarization, axons and dendrites are indispensable structures within neurons, crucial for the maintenance of nervous system function. Here, we investigated the impact of BPS exposure on axonal and dendritic development both in vivo and in vitro. Our results revealed that exposure to BPS during pregnancy and lactation led to a reduction in the complexity, density, and length of axons and dendrites in the prefrontal cortex (PFC) of offspring. Employing RNA sequencing technology to elucidate the underlying mechanisms of axonal and dendritic damage induced by BPS, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis highlighted a significant alteration in the oxidative phosphorylation (OXPHOS) pathway, essential for mitochondrial function. Subsequent experiments demonstrate BPS-induced impairment in mitochondrial function, including damaged morphology, decreased adenosine triphosphate (ATP) and superoxide dismutase (SOD) levels, and increased reactive oxygen species and malondialdehyde (MDA). These alterations coincided with the downregulated expression of OXPHOS pathway-related genes (ATP6V1B1, ATP5K, NDUFC1, NDUFC2, NDUFA3, COX6B1) and Myosin 19 (Myo19). Notably, Myo19 overexpression restored the BPS-induced mitochondrial dysfunction by alleviating the inhibition of OXPHOS pathway. Consequently, this amelioration was associated with a reduction in BPS-induced axonal and dendritic injury observed in cultured neurons of the PFC.


Axons , Dendrites , Mitochondria , Oxidative Phosphorylation , Phenols , Sulfones , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Phenols/toxicity , Dendrites/drug effects , Oxidative Phosphorylation/drug effects , Female , Sulfones/toxicity , Axons/drug effects , Pregnancy , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Mice
4.
Cell Commun Signal ; 22(1): 236, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38650003

BACKGROUND: The preservation of retinal ganglion cells (RGCs) and the facilitation of axon regeneration are crucial considerations in the management of various vision-threatening disorders. Therefore, we investigate the efficacy of interleukin-4 (IL-4), a potential therapeutic agent, in promoting neuroprotection and axon regeneration of retinal ganglion cells (RGCs) as identified through whole transcriptome sequencing in an in vitro axon growth model. METHODS: A low concentration of staurosporine (STS) was employed to induce in vitro axon growth. Whole transcriptome sequencing was utilized to identify key target factors involved in the molecular mechanism underlying axon growth. The efficacy of recombinant IL-4 protein on promoting RGC axon growth was validated through in vitro experiments. The protective effect of recombinant IL-4 protein on somas of RGCs was assessed using RBPMS-specific immunofluorescent staining in mouse models with optic nerve crush (ONC) and N-methyl-D-aspartic acid (NMDA) injury. The protective effect on RGC axons was evaluated by anterograde labeling of cholera toxin subunit B (CTB), while the promotion of RGC axon regeneration was assessed through both anterograde labeling of CTB and immunofluorescent staining for growth associated protein-43 (GAP43). RESULTS: Whole-transcriptome sequencing of staurosporine-treated 661 W cells revealed a significant upregulation in intracellular IL-4 transcription levels during the process of axon regeneration. In vitro experiments demonstrated that recombinant IL-4 protein effectively stimulated axon outgrowth. Subsequent immunostaining with RBPMS revealed a significantly higher survival rate of RGCs in the rIL-4 group compared to the vehicle group in both NMDA and ONC injury models. Axonal tracing with CTB confirmed that recombinant IL-4 protein preserved long-distance projection of RGC axons, and there was a notably higher number of surviving axons in the rIL-4 group compared to the vehicle group following NMDA-induced injury. Moreover, intravitreal delivery of recombinant IL-4 protein substantially facilitated RGC axon regeneration after ONC injury. CONCLUSION: The recombinant IL-4 protein exhibits the potential to enhance the survival rate of RGCs, protect RGC axons against NMDA-induced injury, and facilitate axon regeneration following ONC. This study provides an experimental foundation for further investigation and development of therapeutic agents aimed at protecting the optic nerve and promoting axon regeneration.


Axons , Interleukin-4 , Nerve Regeneration , Retinal Ganglion Cells , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Animals , Interleukin-4/pharmacology , Axons/drug effects , Axons/metabolism , Nerve Regeneration/drug effects , Mice , Mice, Inbred C57BL , Optic Nerve Injuries/pathology , Optic Nerve Injuries/drug therapy , N-Methylaspartate/pharmacology , Staurosporine/pharmacology , Neuroprotective Agents/pharmacology , Recombinant Proteins/pharmacology
5.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38673871

Mild traumatic brain injury (mTBI) affects millions of people in the U.S. Approximately 20-30% of those individuals develop adverse symptoms lasting at least 3 months. In a rat mTBI study, the closed-head impact model of engineered rotational acceleration (CHIMERA) produced significant axonal injury in the optic tract (OT), indicating white-matter damage. Because retinal ganglion cells project to the lateral geniculate nucleus (LGN) in the thalamus through the OT, we hypothesized that synaptic density may be reduced in the LGN of rats following CHIMERA injury. A modified SEQUIN (synaptic evaluation and quantification by imaging nanostructure) method, combined with immunofluorescent double-labeling of pre-synaptic (synapsin) and post-synaptic (PSD-95) markers, was used to quantify synaptic density in the LGN. Microglial activation at the CHIMERA injury site was determined using Iba-1 immunohistochemistry. Additionally, the effects of ketamine, a potential neuroprotective drug, were evaluated in CHIMERA-induced mTBI. A single-session repetitive (ssr-) CHIMERA (3 impacts, 1.5 joule/impact) produced mild effects on microglial activation at the injury site, which was significantly enhanced by post-injury intravenous ketamine (10 mg/kg) infusion. However, ssr-CHIMERA did not alter synaptic density in the LGN, although ketamine produced a trend of reduction in synaptic density at post-injury day 4. Further research is necessary to characterize the effects of ssr-CHIMERA and subanesthetic doses of intravenous ketamine on different brain regions and multiple time points post-injury. The current study demonstrates the utility of the ssr-CHIMERA as a rodent model of mTBI, which researchers can use to identify biological mechanisms of mTBI and to develop improved treatment strategies for individuals suffering from head trauma.


Ketamine , Microglia , Rats, Sprague-Dawley , Synapses , Animals , Ketamine/administration & dosage , Ketamine/pharmacology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats , Male , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Head Injuries, Closed/pathology , Axons/drug effects , Axons/metabolism , Axons/pathology , Disease Models, Animal , Geniculate Bodies/pathology , Geniculate Bodies/drug effects , Brain Concussion/pathology , Brain Concussion/metabolism , Disks Large Homolog 4 Protein/metabolism , Synapsins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage
6.
Transl Res ; 268: 40-50, 2024 Jun.
Article En | MEDLINE | ID: mdl-38246342

Traumatic brain injury (TBI) has a significant impact on cognitive function, affecting millions of people worldwide. Myelin loss is a prominent pathological feature of TBI, while well-functioning myelin is crucial for memory and cognition. Utilizing drug repurposing to identify effective drug candidates for TBI treatment has gained attention. Notably, recent research has highlighted the potential of clemastine, an FDA-approved allergy medication, as a promising pro-myelinating drug. Therefore, in this study, we aim to investigate whether clemastine can enhance myelination and alleviate cognitive impairment following mild TBI using a clinically relevant rat model of TBI. Mild diffuse TBI was induced using the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA). Animals were treated with either clemastine or an equivalent volume of the vehicle from day 1 to day 14 post-injury. Following treatment, memory-related behavioral tests were conducted, and myelin pathology in the cortex and hippocampus was assessed through immunofluorescence staining and ProteinSimple® capillary-based immunoassay. Our results showed that TBI leads to significant myelin loss, axonal damage, glial activation, and a decrease in mature oligodendrocytes in both the cortex and hippocampus. The TBI animals also exhibited notable deficits in memory-related tests. In contrast, animals treated with clemastine showed an increase in mature oligodendrocytes, enhanced myelination, and improved performance in the behavioral tests. These preliminary findings support the therapeutic value of clemastine in alleviating TBI-induced cognitive impairment, with substantial clinical translational potential. Our findings also underscore the potential of remyelinating therapies for TBI.


Axons , Clemastine , Cognitive Dysfunction , Disease Models, Animal , Myelin Sheath , Rats, Sprague-Dawley , Animals , Clemastine/pharmacology , Clemastine/therapeutic use , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin Sheath/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/pathology , Axons/drug effects , Axons/pathology , Male , Rats , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Diffuse/drug effects , Brain Injuries, Diffuse/pathology , Hippocampus/drug effects , Hippocampus/pathology
7.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article En | MEDLINE | ID: mdl-36835165

Emamectin benzoate (EMB) is a widely used pesticide and feed additive in agriculture and aquaculture. It easily enters the aquatic environment through various pathways, thus causing adverse effects on aquatic organisms. However, there are no systematic studies regarding the effects of EMB on the developmental neurotoxicity of aquatic organisms. Therefore, the aim of this study was to evaluate the neurotoxic effects and mechanisms of EMB at different concentrations (0.1, 0.25, 0.5, 1, 2, 4 and 8 µg/mL) using zebrafish as a model. The results showed that EMB significantly inhibited the hatching rate, spontaneous movement, body length, and swim bladder development of zebrafish embryos, as well as significantly increased the malformation rate of zebrafish larvae. In addition, EMB adversely affected the axon length of motor neurons in Tg (hb9: eGFP) zebrafish and central nervous system (CNS) neurons in Tg (HuC: eGFP) zebrafish and significantly inhibited the locomotor behavior of zebrafish larvae. Meanwhile, EMB induced oxidative damage and was accompanied by increasing reactive oxygen species in the brains of zebrafish larvae. In addition, gene expression involvement in oxidative stress-related (cat, sod and Cu/Zn-sod), GABA neural pathway-related (gat1, gabra1, gad1b, abat and glsa), neurodevelopmental-related (syn2a, gfap, elavl3, shha, gap43 and Nrd) and swim bladder development-related (foxa3, pbxla, mnx1, has2 and elovlla) genes was significantly affected by EMB exposure. In conclusion, our study shows that exposure to EMB during the early life stages of zebrafish significantly increases oxidative damage and inhibits early central neuronal development, motor neuron axon growth and swim bladder development, ultimately leading to neurobehavioral changes in juvenile zebrafish.


Ivermectin , Water Pollutants, Chemical , Zebrafish , Animals , Axons/drug effects , Axons/pathology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Larva/metabolism , Motor Neurons , Oxidative Stress , Water Pollutants, Chemical/toxicity , Zebrafish/growth & development , Zebrafish/metabolism , Ivermectin/analogs & derivatives , Ivermectin/toxicity , Neurodevelopmental Disorders/chemically induced
8.
Mol Neurodegener ; 17(1): 54, 2022 08 20.
Article En | MEDLINE | ID: mdl-35987691

BACKGROUND: Amyloid plaque deposition and axonal degeneration are early events in AD pathogenesis. Aß disrupts microtubules in presynaptic dystrophic neurites, resulting in the accumulation of impaired endolysosomal and autophagic organelles transporting ß-site amyloid precursor protein cleaving enzyme (BACE1). Consequently, dystrophic neurites generate Aß42 and significantly contribute to plaque deposition. Farnesyltransferase inhibitors (FTIs) have recently been investigated for repositioning toward the treatment of neurodegenerative disorders and block the action of farnesyltransferase (FTase) to catalyze farnesylation, a post-translational modification that regulates proteins involved in lysosome function and microtubule stability. In postmortem AD brains, FTase and its downstream signaling are upregulated. However, the impact of FTIs on amyloid pathology and dystrophic neurites is unknown. METHODS: We tested the effects of the FTIs LNK-754 and lonafarnib in the 5XFAD mouse model of amyloid pathology. RESULTS: In 2-month-old 5XFAD mice treated chronically for 3 months, LNK-754 reduced amyloid plaque burden, tau hyperphosphorylation, and attenuated the accumulation of BACE1 and LAMP1 in dystrophic neurites. In 5-month-old 5XFAD mice treated acutely for 3 weeks, LNK-754 reduced dystrophic neurite size and LysoTracker-Green accumulation in the absence of effects on Aß deposits. Acute treatment with LNK-754 improved memory and learning deficits in hAPP/PS1 amyloid mice. In contrast to LNK-754, lonafarnib treatment was less effective at reducing plaques, tau hyperphosphorylation and dystrophic neurites, which could have resulted from reduced potency against FTase compared to LNK-754. We investigated the effects of FTIs on axonal trafficking of endolysosomal organelles and found that lonafarnib and LNK-754 enhanced retrograde axonal transport in primary neurons, indicating FTIs could support the maturation of axonal late endosomes into lysosomes. Furthermore, FTI treatment increased levels of LAMP1 in mouse primary neurons and in the brains of 5XFAD mice, demonstrating that FTIs stimulated the biogenesis of endolysosomal organelles. CONCLUSIONS: We show new data to suggest that LNK-754 promoted the axonal trafficking and function of endolysosomal compartments, which we hypothesize decreased axonal dystrophy, reduced BACE1 accumulation and inhibited amyloid deposition in 5XFAD mice. Our results agree with previous work identifying FTase as a therapeutic target for treating proteinopathies and could have important therapeutic implications in treating AD.


Amyloid , Amyloidosis , Farnesyltranstransferase , Alzheimer Disease/metabolism , Amyloid/drug effects , Amyloid/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Aspartic Acid Endopeptidases/metabolism , Axons/drug effects , Axons/pathology , Disease Models, Animal , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Mice , Mice, Transgenic , Plaque, Amyloid/pathology
9.
Toxicol Appl Pharmacol ; 448: 116096, 2022 08 01.
Article En | MEDLINE | ID: mdl-35662665

Neuronal progranulin (PGRN) overexpression is an endogenous adaptive pain defense following nerve injury. It allows the survival of injured neurons to block enhanced nociceptive responses. Trimetazidine (TMZ) is widely used by cardiac patients as an anti-anginal drug, reflecting its anti-ischemic property. TMZ promotes axonal regeneration of sciatic nerves after crush injury. This study explored the interplay between PGRN and extracellular signal-regulated kinases (ERK1/2) to address mechanisms underlying neuropathic pain alleviation following paclitaxel (PTX) administration. Rats were given four injections of PTX (2 mg/kg, i.p.) every other day. Two days after the last dose, rats received TMZ (25 mg/kg) with or without the ERK inhibitor, PD98059, daily for 21 days. TMZ preserved the integrity of myelinated nerve fibers, as evidenced by an obvious reduction in axonal damage biomarkers. Accordingly, it alleviated PTX-evoked thermal, cold, and mechanical hyperalgesia/allodynia. TMZ also promoted ERK1/2 phosphorylation with a profound upsurge in PGRN content. These effects were associated with a substantial increase in Notch1 receptor gene expression and a prominent anti-inflammatory effect with a marked increase in mRNA expression of secretory leukocyte protease inhibitor. Further, TMZ decreased oxidative stress and caspase-3 activity in the sciatic nerve. Conversely, co-administration of PD98059 completely abolished these beneficial effects. Thus, the robust antinociceptive effect of TMZ is largely attributed to upregulating PGRN and Notch1 receptors via ERK1/2 activation.


MAP Kinase Signaling System , Neuralgia , Paclitaxel , Progranulins , Trimetazidine , Analgesics/pharmacology , Animals , Axons/drug effects , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , MAP Kinase Signaling System/drug effects , Neuralgia/chemically induced , Neuralgia/drug therapy , Paclitaxel/pharmacology , Progranulins/metabolism , Rats , Sciatic Nerve/drug effects , Trimetazidine/pharmacology , Up-Regulation/drug effects
10.
Nature ; 607(7919): 585-592, 2022 07.
Article En | MEDLINE | ID: mdl-35732737

The regenerative potential of mammalian peripheral nervous system neurons after injury is critically limited by their slow axonal regenerative rate1. Regenerative ability is influenced by both injury-dependent and injury-independent mechanisms2. Among the latter, environmental factors such as exercise and environmental enrichment have been shown to affect signalling pathways that promote axonal regeneration3. Several of these pathways, including modifications in gene transcription and protein synthesis, mitochondrial metabolism and the release of neurotrophins, can be activated by intermittent fasting (IF)4,5. However, whether IF influences the axonal regenerative ability remains to be investigated. Here we show that IF promotes axonal regeneration after sciatic nerve crush in mice through an unexpected mechanism that relies on the gram-positive gut microbiome and an increase in the gut bacteria-derived metabolite indole-3-propionic acid (IPA) in the serum. IPA production by Clostridium sporogenes is required for efficient axonal regeneration, and delivery of IPA after sciatic injury significantly enhances axonal regeneration, accelerating the recovery of sensory function. Mechanistically, RNA sequencing analysis from sciatic dorsal root ganglia suggested a role for neutrophil chemotaxis in the IPA-dependent regenerative phenotype, which was confirmed by inhibition of neutrophil chemotaxis. Our results demonstrate the ability of a microbiome-derived metabolite, such as IPA, to facilitate regeneration and functional recovery of sensory axons through an immune-mediated mechanism.


Indoles , Nerve Regeneration , Propionates , Wound Healing , Animals , Mice , Axons/drug effects , Axons/physiology , Chemotaxis, Leukocyte , Clostridium/metabolism , Fasting , Ganglia, Spinal/metabolism , Gastrointestinal Microbiome , Indoles/blood , Indoles/metabolism , Indoles/pharmacology , Nerve Crush , Nerve Growth Factors/metabolism , Nerve Regeneration/drug effects , Neutrophils/cytology , Neutrophils/immunology , Propionates/blood , Propionates/metabolism , Propionates/pharmacology , Recovery of Function , Sciatic Nerve/injuries , Sequence Analysis, RNA , Wound Healing/drug effects
11.
J Neuroinflammation ; 19(1): 95, 2022 Apr 16.
Article En | MEDLINE | ID: mdl-35429978

BACKGROUND: Excessively deposited fibrotic scar after spinal cord injury (SCI) inhibits axon regeneration. It has been reported that platelet-derived growth factor receptor beta (PDGFRß), as a marker of fibrotic scar-forming fibroblasts, can only be activated by platelet-derived growth factor (PDGF) B or PDGFD. However, whether the activation of the PDGFRß pathway can mediate fibrotic scar formation after SCI remains unclear. METHODS: A spinal cord compression injury mouse model was used. In situ injection of exogenous PDGFB or PDGFD in the spinal cord was used to specifically activate the PDGFRß pathway in the uninjured spinal cord, while intrathecal injection of SU16f was used to specifically block the PDGFRß pathway in the uninjured or injured spinal cord. Immunofluorescence staining was performed to explore the distributions and cell sources of PDGFB and PDGFD, and to evaluate astrocytic scar, fibrotic scar, inflammatory cells and axon regeneration after SCI. Basso Mouse Scale (BMS) and footprint analysis were performed to evaluate locomotor function recovery after SCI. RESULTS: We found that the expression of PDGFD and PDGFB increased successively after SCI, and PDGFB was mainly secreted by astrocytes, while PDGFD was mainly secreted by macrophages/microglia and fibroblasts. In addition, in situ injection of exogenous PDGFB or PDGFD can lead to fibrosis in the uninjured spinal cord, while this profibrotic effect could be specifically blocked by the PDGFRß inhibitor SU16f. We then treated the mice after SCI with SU16f and found the reduction of fibrotic scar, the interruption of scar boundary and the inhibition of lesion and inflammation, which promoted axon regeneration and locomotor function recovery after SCI. CONCLUSIONS: Our study demonstrates that activation of PDGFRß pathway can directly induce fibrotic scar formation, and specific blocking of this pathway would contribute to the treatment of SCI.


Axons , Cicatrix , Indoles , Nerve Regeneration , Pyrroles , Receptor, Platelet-Derived Growth Factor beta , Spinal Cord Injuries , Animals , Axons/drug effects , Axons/pathology , Cicatrix/drug therapy , Cicatrix/etiology , Cicatrix/metabolism , Cicatrix/pathology , Fibrosis , Indoles/pharmacology , Locomotion , Mice , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Proto-Oncogene Proteins c-sis/metabolism , Pyrroles/pharmacology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recovery of Function , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
12.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Article En | MEDLINE | ID: mdl-35298717

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Aging/metabolism , Axons/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Energy Metabolism , Insulin-Like Growth Factor I/metabolism , Sensory Receptor Cells/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Axons/drug effects , Axons/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Respiration/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neuronal Outgrowth/drug effects , Polymers/metabolism , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
13.
Life Sci ; 294: 120383, 2022 Apr 01.
Article En | MEDLINE | ID: mdl-35143827

AIMS: Small molecule compound tyrphostin A9 (A9), an inhibitor of platelet-derived growth factor (PDGF) receptor, was previously reported by our group to stimulate extracellular signal-regulated kinase 1 (ERK1) and 2 (ERK2) in neuronal cells in a PDGF receptor-irrelevant manner. The study aimed to investigate whether A9 could protect axons in experimental autoimmune encephalomyelitis through activation of ERKs. MAIN METHODS: A9 treatment on the protection on neurite outgrowth in SH-SY5Y neuroblastoma cells and primary substantia nigra neuron cultures from the neurotoxin MPP+ were analyzed. Then, clinical symptoms as well as ERK1/2 activation, axonal protection induction, and the abundance increases of the regeneration biomarker GAP-43 in the CNS in the relapsing-remitting experimental autoimmune encephalomyelitis (EAE) model were verified. KEY FINDINGS: A9 treatment could stimulate neurite outgrowth in SH-SY5Y neuroblastoma cells and protect primary substantia nigra neuron cultures from the neurotoxin MPP+. In the relapsing-remitting EAE model, oral administration of A9 successfully ameliorated clinical symptoms, activated ERK1/2, induced axonal protection, and increased the abundance of the regeneration biomarker GAP-43 in the CNS. Interestingly, gene deficiency of ERK1 or ERK2 disrupted the beneficial effects of A9 in MOG-35-55-induced EAE. SIGNIFICANCE: These results demonstrated that small molecule compounds that stimulate persistent ERK activation in vitro and in vivo may be useful in protective or restorative treatment for neurodegenerative diseases.


Axons/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Neuroblastoma/drug therapy , Tyrphostins/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Neuroblastoma/metabolism , Neuroblastoma/pathology , Rats , Rats, Sprague-Dawley
14.
Int J Mol Sci ; 23(4)2022 Feb 18.
Article En | MEDLINE | ID: mdl-35216370

The repair of severe nerve injuries requires an autograft or conduit to bridge the gap and avoid axon dispersion. Several conduits are used routinely, but their effectiveness is comparable to that of an autograft only for short gaps. Understanding nerve regeneration within short conduits could help improve their efficacy for longer gaps. Since Schwann cells are known to migrate on endothelial cells to colonize the "nerve bridge", the new tissue spontaneously forming to connect the injured nerve stumps, here we aimed to investigate whether this migratory mechanism drives Schwann cells to also proceed within the nerve conduits used to repair large nerve gaps. Injured median nerves of adult female rats were repaired with 10 mm chitosan conduits and the regenerated nerves within conduits were analyzed at different time points using confocal imaging of sequential thick sections. Our data showed that the endothelial cells formed a dense capillary network used by Schwann cells to migrate from the two nerve stumps into the conduit. We concluded that angiogenesis played a key role in the nerve conduits, not only by supporting cell survival but also by providing a pathway for the migration of newly formed Schwann cells.


Blood Vessels/physiology , Nerve Tissue/physiology , Schwann Cells/physiology , Sciatic Nerve/physiology , Animals , Axons/drug effects , Axons/physiology , Blood Vessels/drug effects , Chitosan/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Nerve Tissue/drug effects , Peripheral Nervous System Diseases/physiopathology , Rats , Rats, Wistar , Schwann Cells/drug effects , Sciatic Nerve/drug effects , Tissue Engineering/methods
15.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article En | MEDLINE | ID: mdl-35216504

Spinal cord injury (SCI) causes significant mortality and morbidity. Currently, no FDA-approved pharmacotherapy is available for treating SCI. Previously, low doses of estrogen (17ß-estradiol, E2) were shown to improve the post-injury outcome in a rat SCI model. However, the range of associated side effects makes advocating its therapeutic use difficult. Therefore, this study aimed at investigating the therapeutic efficacy of Premarin (PRM) in SCI. PRM is an FDA-approved E2 (10%) formulation, which is used for hormone replacement therapy with minimal risk of serious side effects. The effects of PRM on SCI were examined by magnetic resonance imaging, immunofluorescent staining, and western blot analysis in a rat model. SCI animals treated with vehicle alone, PRM, E2 receptor antagonist (ICI), or PRM + ICI were graded in a blinded way for locomotor function by using the Basso-Beattie-Bresnahan (BBB) locomotor scale. PRM treatment for 7 days decreased post-SCI lesion volume and attenuated neuronal cell death, inflammation, and axonal damage. PRM also altered the balance of pro- and anti-apoptotic proteins in favor of cell survival and improved angiogenesis and microvascular growth. Increased expression of estrogen receptors (ERs) ERα and ERß following PRM treatment and their inhibition by ER inhibitor indicated that the neuroprotection associated with PRM treatment might be E2-receptor mediated. The attenuation of glial activation with decreased inflammation and cell death, and increased angiogenesis by PRM led to improved functional outcome as determined by the BBB locomotor scale. These results suggest that PRM treatment has significant therapeutic implications for the improvement of post-SCI outcome.


Estrogens, Conjugated (USP)/pharmacology , Neurodegenerative Diseases/drug therapy , Spinal Cord Injuries/drug therapy , Animals , Axons/drug effects , Axons/metabolism , Disease Models, Animal , Estradiol/metabolism , Estrogens/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Locomotion/drug effects , Male , Motor Activity/drug effects , Neurodegenerative Diseases/metabolism , Neuroprotection/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
16.
Arch Toxicol ; 96(2): 431-451, 2022 02.
Article En | MEDLINE | ID: mdl-34988610

Methanol-induced optic neuropathy (Me-ION) is a serious condition that may result in long-term or irreversible visual impairment or even blindness secondary to damage and loss of function of the optic nerve and retina. Me-ION shows a tendency to occur as mass poisonings around the world with a clear predilection for poor societies in developing countries. The main mechanism underlying the molecular basis of Me-ION is the inhibition of the mitochondrial oxidative phosphorylation process through the binding of the toxic metabolite of methanol-formic acid-with the key enzyme of this process-cytochrome c oxidase. However, other mechanisms, including damage to the eye tissues by oxidative stress causing the intensification of the oxidative peroxidation process with the formation of cytotoxic compounds, as well as an increase in the synthesis of pro-inflammatory cytokines and influence on the expression of key proteins responsible for maintaining cell homeostasis, also play an important role in the pathogenesis of Me-ION. Histopathological changes in the eye tissues are mainly manifested as the degeneration of axons and glial cells of the optic nerve, often with accompanying damage of the retina that may involve all its layers. Despite the development of therapeutic approaches, persistent visual sequelae are seen in 30-40% of survivors. Thus, Me-ION continues to be an important problem for healthcare systems worldwide.


Methanol/poisoning , Optic Nerve Diseases/chemically induced , Optic Nerve/drug effects , Animals , Axons/drug effects , Axons/pathology , Formates/toxicity , Humans , Methanol/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Optic Nerve/pathology , Optic Nerve Diseases/physiopathology , Oxidative Stress/drug effects , Retina/drug effects , Retina/pathology
17.
Invest Ophthalmol Vis Sci ; 63(1): 4, 2022 01 03.
Article En | MEDLINE | ID: mdl-34982146

Purpose: Netarsudil, a Rho kinase inhibitor with norepinephrine transport inhibitory effect, lowers intraocular pressure, however, its effect on axon damage remains to be elucidated. The aim of the current study was to investigate the effect of netarsudil on TNF-induced axon loss and to examine whether it affects phosphorylated-AMP-activated kinase (p-AMPK) and autophagy in the optic nerve. Methods: Intravitreal administration of TNF or TNF with netarsudil was carried out on rats and quantification of axon number was determined. Electron microscopy determined autophagosome numbers. Localization of p-AMPK expression was examined by immunohistochemistry. The changes in p62, LC3-II, and p-AMPK levels were estimated in the optic nerve by immunoblot analysis. The effect of an AMPK activator A769662 or an AMPK inhibitor dorsomorphin on axon number was evaluated. Results: Morphometric analysis revealed apparent protection by netarsudil against TNF-induced axon degeneration. Netarsudil increased autophagosome numbers inside axons. Netarsudil treatment significantly upregulated optic nerve LC3-II levels in both the TNF-treated eyes and the control eyes. Increased p62 protein level induced by TNF was significantly ameliorated by netarsudil. The netarsudil administration alone lessened p62 levels. Netarsudil significantly upregulated the optic nerve p-AMPK levels. A769662 exhibited obvious axonal protection against TNF-induced damage. A769662 treatment upregulated LC3-II levels and the increment of p62 level induced by TNF was significantly ameliorated by A769662. Immunohistochemical analysis revealed that p-AMPK is present in axons. Netarsudil-mediated axonal protection was significantly suppressed by dorsomorphin administration. Conclusions: Netarsudil upregulated p-AMPK and autophagy. Netarsudil-mediated axonal protection may be associated with upregulated p-AMPK.


AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Axons/drug effects , Benzoates/pharmacology , Nerve Degeneration/prevention & control , Optic Nerve/drug effects , Tumor Necrosis Factor-alpha/toxicity , beta-Alanine/analogs & derivatives , rho-Associated Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Axons/enzymology , Axons/pathology , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Intravitreal Injections , Male , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/enzymology , Optic Nerve/ultrastructure , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrones/pharmacology , Rats , Rats, Wistar , Sequestosome-1 Protein/metabolism , Thiophenes/pharmacology , beta-Alanine/pharmacology
18.
J Biomater Sci Polym Ed ; 33(6): 747-768, 2022 04.
Article En | MEDLINE | ID: mdl-34865608

The repair of spinal cord injury is a great challenge in clinical. Improving the microenvironment of the injured site is the key strategy for accelerating axon regeneration and synaptic formation. Herein, a kind of silk fibroin microspheres functionalized by metformin through dopamine was developed using water-in-oil emulsification-diffusion method and surface modification technique, and the effect on cortical neuron was evaluated. The results showed that the microspheres showed a uniform size distribution with the diameter of around 60 µm and a concave structure. Moreover, the microspheres possessed good injectability and stability. In addition, the metformin could be successfully immobilized in the silk fibroin microspheres. The cell culture results displayed that the growth and morphology of cortical neurons on the microspheres with metformin concentration of 5 mg/mL and 10 mg/mL were obviously better than that on other samples. Notably, the spread area of single cortical cell on silk fibroin microspheres was increased with the ascending metformin concentration. Therefore, the results indicated that the metformin loaded silk fibroin microsphere could obviously improve the growth and spreading behavior of cortical neuron. The study may provide an important experimental basis for the development of drug loaded injectable biomaterials scaffolds for the treatment of spinal cord injury and have great potential for spinal cord regeneration.


Fibroins , Metformin , Spinal Cord Injuries , Axons/drug effects , Fibroins/administration & dosage , Fibroins/chemistry , Humans , Metformin/administration & dosage , Metformin/chemistry , Microspheres , Nerve Regeneration/drug effects , Silk/administration & dosage , Silk/blood , Spinal Cord Injuries/drug therapy , Tissue Scaffolds
19.
Cell Rep ; 37(11): 110108, 2021 12 14.
Article En | MEDLINE | ID: mdl-34910914

Activation of the pro-degenerative protein SARM1 after diverse physical and disease-relevant injuries causes programmed axon degeneration. Original studies indicate that substantially decreased SARM1 levels are required for neuroprotection. However, we demonstrate, in Sarm1 haploinsufficient mice, that lowering SARM1 levels by 50% delays programmed axon degeneration in vivo after sciatic nerve transection and partially prevents neurite outgrowth defects in mice lacking the pro-survival factor NMNAT2. In vitro, the rate of degeneration in response to traumatic, neurotoxic, and genetic triggers of SARM1 activation is also slowed. Finally, we demonstrate that Sarm1 antisense oligonucleotides decrease SARM1 levels by more than 50% in vitro, which delays or prevents programmed axon degeneration. Combining Sarm1 haploinsufficiency with antisense oligonucleotides further decreases SARM1 levels and prolongs protection after neurotoxic injury. These data demonstrate that axon protection occurs in a Sarm1 gene dose-responsive manner and that SARM1-lowering agents have therapeutic potential, making Sarm1-targeting antisense oligonucleotides a promising therapeutic strategy.


Armadillo Domain Proteins/physiology , Axons/physiology , Cytoskeletal Proteins/physiology , Haploinsufficiency , Nerve Degeneration/drug therapy , Oligonucleotides, Antisense/pharmacology , Sciatic Nerve/cytology , Wallerian Degeneration/drug therapy , Animals , Axons/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology
20.
Elife ; 102021 12 29.
Article En | MEDLINE | ID: mdl-34965204

Dopaminergic (DA) neurons exert profound influences on behavior including addiction. However, how DA axons communicate with target neurons and how those communications change with drug exposure remains poorly understood. We leverage cell type-specific labeling with large volume serial electron microscopy to detail DA connections in the nucleus accumbens (NAc) of the mouse (Mus musculus) before and after exposure to cocaine. We find that individual DA axons contain different varicosity types based on their vesicle contents. Spatially ordering along individual axons further suggests that varicosity types are non-randomly organized. DA axon varicosities rarely make specific synapses (<2%, 6/410), but instead are more likely to form spinule-like structures (15%, 61/410) with neighboring neurons. Days after a brief exposure to cocaine, DA axons were extensively branched relative to controls, formed blind-ended 'bulbs' filled with mitochondria, and were surrounded by elaborated glia. Finally, mitochondrial lengths increased by ~2.2 times relative to control only in DA axons and NAc spiny dendrites after cocaine exposure. We conclude that DA axonal transmission is unlikely to be mediated via classical synapses in the NAc and that the major locus of anatomical plasticity of DA circuits after exposure to cocaine are large-scale axonal re-arrangements with correlated changes in mitochondria.


Axons/drug effects , Cocaine/pharmacology , Connectome , Dopaminergic Neurons/drug effects , Animals , Axons/ultrastructure , Dopaminergic Neurons/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron , Mitochondria/ultrastructure , Nucleus Accumbens/drug effects
...