Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.231
Filter
1.
Biomed Pharmacother ; 174: 116460, 2024 May.
Article in English | MEDLINE | ID: mdl-38520864

ABSTRACT

Ischemic stroke is a common intravascular disease and one of the leading causes of death and disability. The salidroside derivative SHPL-49, which we previously synthesized, significantly attenuates cerebral ischemic injury in a rat model of permanent middle cerebral artery occlusion. To explore the neuroprotective mechanism of SHPL-49, the effects of SHPL-49 on the expression levels of neurotrophic factors in neurons and microglia and the polarization of microglia were investigated in the present study. SHPL-49 activated the brain-derived neurotrophic factor (BDNF) pathway, decreased the number of degenerated neurons, and accelerated neurogenesis in rats with cerebral ischemia. In addition, SHPL-49 promoted the polarization of microglia toward the M2 phenotype to alleviate neuroinflammation. In BV2 cells, SHPL-49 upregulated CD206 mRNA and protein levels and inhibited CD86 mRNA and protein levels. SHPL-49 also increased neurotrophic factor secretion in BV2 cells, which indirectly promoted the survival of primary neurons after oxygen-glucose deprivation (OGD). Proteomics analysis revealed that SHPL-49 promoted growth-associated protein 43 (Gap43) expression. SHPL-49 enhanced synaptic plasticity and increased Gap43 protein levels via activation of the BDNF pathway in the OGD primary neuron model. These results indicate that SHPL-49 prevents cerebral ischemic injury by activating neurotrophic factor pathways and altering microglial polarization. Thus, SHPL-49 is a potential neuroprotective agent.


Subject(s)
Brain Ischemia , Brain-Derived Neurotrophic Factor , GAP-43 Protein , Glucosides , Microglia , Neurons , Neuroprotective Agents , Phenols , Rats, Sprague-Dawley , Receptor, trkB , Signal Transduction , Animals , Brain-Derived Neurotrophic Factor/metabolism , Neuroprotective Agents/pharmacology , Glucosides/pharmacology , Phenols/pharmacology , Male , Rats , GAP-43 Protein/metabolism , Microglia/drug effects , Microglia/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Signal Transduction/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Receptor, trkB/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Cell Line , Disease Models, Animal , Neurogenesis/drug effects , Mice
2.
CNS Neurosci Ther ; 30(4): e14535, 2024 04.
Article in English | MEDLINE | ID: mdl-38168094

ABSTRACT

INTRODUCTION: Self-repair of spinal cord injury (SCI) has been found in humans and experimental animals with partial recovery of neurological functions. However, the regulatory mechanisms underlying the spontaneous locomotion recovery after SCI are elusive. AIMS: This study was aimed at evaluating the pathological changes in injured spinal cord and exploring the possible mechanism related to the spontaneous recovery. RESULTS: Immunofluorescence staining was performed to detect GAP43 expression in lesion site after spinal cord transection (SCT) in rats. Then RNA sequencing and gene ontology (GO) analysis were employed to predict lncRNA that correlates with GAP43. LncRNA smart-silencing was applied to verify the function of lncRNA vof16 in vitro, and knockout rats were used to evaluate its role in neurobehavioral functions after SCT. MicroRNA sequencing, target scan, and RNA22 prediction were performed to further explore the underlying regulatory mechanisms, and miR-185-5p stands out. A miR-185-5p site-regulated relationship with GAP43 and vof16 was determined by luciferase activity analysis. GAP43-silencing, miR-185-5p-mimic/inhibitor, and miR-185-5p knockout rats were also applied to elucidate their effects on spinal cord neurite growth and neurobehavioral function after SCT. We found that a time-dependent increase of GAP43 corresponded with the limited neurological recovery in rats with SCT. CRNA chip and GO analysis revealed lncRNA vof16 was the most functional in targeting GAP43 in SCT rats. Additionally, silencing vof16 suppressed neurite growth and attenuated the motor dysfunction in SCT rats. Luciferase reporter assay showed that miR-185-5p competitively bound the same regulatory region of vof16 and GAP43. CONCLUSIONS: Our data indicated miR-185-5p could be a detrimental factor in SCT, and vof16 may function as a ceRNA by competitively binding miR-185-5p to modulate GAP43 in the process of self-recovery after SCT. Our study revealed a novel vof16-miR-185-5p-GAP43 regulatory network in neurological self-repair after SCT and may underlie the potential treatment target for SCI.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Spinal Cord Injuries , Animals , Rats , Luciferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/pathology , GAP-43 Protein/genetics , GAP-43 Protein/metabolism
3.
Nat Commun ; 15(1): 202, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38172114

ABSTRACT

In Alzheimer's disease, amyloid-beta (Aß) triggers the trans-synaptic spread of tau pathology, and aberrant synaptic activity has been shown to promote tau spreading. Aß induces aberrant synaptic activity, manifesting in increases in the presynaptic growth-associated protein 43 (GAP-43), which is closely involved in synaptic activity and plasticity. We therefore tested whether Aß-related GAP-43 increases, as a marker of synaptic changes, drive tau spreading in 93 patients across the aging and Alzheimer's spectrum with available CSF GAP-43, amyloid-PET and longitudinal tau-PET assessments. We found that (1) higher GAP-43 was associated with faster Aß-related tau accumulation, specifically in brain regions connected closest to subject-specific tau epicenters and (2) that higher GAP-43 strengthened the association between Aß and connectivity-associated tau spread. This suggests that GAP-43-related synaptic changes are linked to faster Aß-related tau spread across connected regions and that synapses could be key targets for preventing tau spreading in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/metabolism , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Positron-Emission Tomography , Cognitive Dysfunction/metabolism , Biomarkers/metabolism
4.
Neurobiol Aging ; 132: 209-219, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37852045

ABSTRACT

Apolipoprotein E-ε4 (APOE-ε4) carriers had elevated cerebrospinal fluid (CSF) presynaptic protein growth-associated protein-43 (GAP-43), but the underlying mechanism is not fully understood. We investigated how the APOE-ε4 genotype affects the baseline and longitudinal changes in CSF GAP-43 and their associations with ß-amyloid positron emission tomography (Aß PET), CSF phosphorylated tau 181 (p-Tau181), neurodegeneration, and cognitive decline. Compared to APOE-ε4 non-carriers, APOE-ε4 carriers had higher baseline levels and faster rates of increases in Aß PET, CSF p-Tau181, and CSF GAP-43. Both higher baseline levels and faster rates of increase in CSF GAP-43 were associated with greater baseline Aß PET and CSF p-Tau181, which fully mediated the APOE-ε4 effect on CSF GAP-43 elevations. Independent of Aß PET and CSF p-Tau181, APOE-ε4 carriage was associated with exacerbated GAP-43-related longitudinal hippocampal atrophy and cognitive decline, especially in Aß+ participants (GAP-43 × time × APOE-ε4). These findings suggest that the APOE-ε4 effect on GAP-43-related presynaptic dysfunction is mediated by primary Alzheimer's pathologies and independently correlates to hippocampal atrophy and cognitive decline in the future.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Cognitive Dysfunction , GAP-43 Protein , Humans , Alzheimer Disease/genetics , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoprotein E4/genetics , Atrophy , Biomarkers/cerebrospinal fluid , Cognitive Dysfunction/pathology , GAP-43 Protein/cerebrospinal fluid , GAP-43 Protein/metabolism , tau Proteins/cerebrospinal fluid
5.
Neuroscience ; 529: 62-72, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37591334

ABSTRACT

Cerebral ischemia is one of the major problems threatening global health. Many of the cerebral ischemia survivors would suffer from the physical and cognitive disabilities for their whole lifetime. Cell based-therapies have been introduced as a therapeutic approach for alleviating ischemia-enforced limitations. Photothrombotic stroke model was applied on the left medial prefrontal cortex (mPFC) of adult male BALB/c mice. Then, pericytes isolated from brain microvessels of adult male BALB/c mice, microglia isolated from brain cortices of the neonatal male BALB/c mice, and M2 phenotype shifted microglia by IL-4 treatment were used for transplantation into the injured area after 24 h of ischemia induction. The behavioural outcomes evaluated by social interaction and Barnes tests and the levels of growth associated protein (GAP)-43 and inflammatory cytokine interleukin (IL)-1 protein were assessed by western blotting 7 days after cell transplantation. Animals in both of the microglia + pericytes and microglia M2 + pericytes transplanted groups showed better performance in social memory as well as enhanced spatial learning and memory compared to ischemic controls. Also, improved escape latency was only observed in microglia M2 + pericytes (p < 0.01) group compared to ischemic controls. GAP-43 showed significant protein expression in microglia + pericytes and microglia M2 + pericytes groups compared to the control group. Conversely, IL-1 levels diminished in all of the pericytes microglia + pericytes, and microglia M2 + pericytes groups compared to the ischemic controls. Current study highlights efficiency of M2 microglia and pericytes combinatory transplantation therapeutic role on relieving ischemic stroke outcomes.


Subject(s)
Brain Ischemia , Microglia , Mice , Animals , Male , Microglia/metabolism , Pericytes/metabolism , Brain Ischemia/metabolism , Prefrontal Cortex/metabolism , Cognition , GAP-43 Protein/metabolism , Ischemia/metabolism
6.
Environ Sci Pollut Res Int ; 30(38): 88685-88703, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37442924

ABSTRACT

This study investigated the possible beneficial role of the bee venom (BV, Apis mellifera L.) against zinc oxide nanoparticles (ZNPs)-induced neurobehavioral and neurotoxic impacts in rats. Fifty male Sprague Dawley rats were alienated into five groups. Three groups were intraperitoneally injected distilled water (C 28D group), ZNPs (100 mg/kg b.wt) (ZNPs group), or ZNPs (100 mg/kg.wt) and BV (1 mg/ kg.bwt) (ZNPs + BV group) for 28 days. One group was intraperitoneally injected with 1 mL of distilled water for 56 days (C 56D group). The last group was intraperitoneally injected with ZNPs for 28 days, then BV for another 28 days at the same earlier doses and duration (ZNPs/BV group). Depression, anxiety, locomotor activity, spatial learning, and memory were evaluated using the forced swimming test, elevated plus maze, open field test, and Morris water maze test, respectively. The brain contents of dopamine, serotonin, total antioxidant capacity (TAC), malondialdehyde (MDA), and Zn were estimated. The histopathological changes and immunoexpressions of neurofilament and GAP-43 protein in the brain tissues were followed. The results displayed that BV significantly decreased the ZNPs-induced depression, anxiety, memory impairment, and spatial learning disorders. Moreover, the ZNPs-induced increment in serotonin and dopamine levels and Zn content was significantly suppressed by BV. Besides, BV significantly restored the depleted TAC but minimized the augmented MDA brain content associated with ZNPs exposure. Likewise, the neurodegenerative changes induced by ZNPs were significantly abolished by BV. Also, the increased neurofilament and GAP-43 immunoexpression due to ZNPs exposure were alleviated with BV. Of note, BV achieved better results in the ZNPs + BV group than in the ZNPs/BV group. Conclusively, these results demonstrated that BV could be employed as a biologically effective therapy to mitigate the neurotoxic and neurobehavioral effects of ZNPs, particularly when used during ZNPs exposure.


Subject(s)
Bee Venoms , Nanoparticles , Neurotoxicity Syndromes , Zinc Oxide , Rats , Animals , Male , Bees , Rats, Sprague-Dawley , GAP-43 Protein/metabolism , GAP-43 Protein/pharmacology , Zinc Oxide/metabolism , Bee Venoms/pharmacology , Bee Venoms/toxicity , Dopamine/metabolism , Dopamine/pharmacology , Serotonin/metabolism , Intermediate Filaments/metabolism , Antioxidants/metabolism , Neurotoxicity Syndromes/metabolism , Brain
7.
Zhongguo Zhong Yao Za Zhi ; 48(7): 1739-1750, 2023 Apr.
Article in Chinese | MEDLINE | ID: mdl-37282948

ABSTRACT

This study investigated the effect of Lianmei Qiwu Decoction(LMQWD) on the improvement of cardiac autonomic nerve remodeling in the diabetic rat model induced by the high-fat diet and explored the underlying mechanism of LMQWD through the AMP-activated protein kinase(AMPK)/tropomyosin receptor kinase A(TrkA)/transient receptor potential melastatin 7(TRPM7) signaling pathway. The diabetic rats were randomly divided into a model group, an LMQWD group, an AMPK agonist group, an unloaded TRPM7 adenovirus group(TRPM7-N), an overexpressed TRPM7 adenovirus group(TRPM7), an LMQWD + unloaded TRPM7 adenovirus group(LMQWD+TRPM7-N), an LMQWD + overexpressed TRPM7 adenovirus group(LMQWD+TRPM7), and a TRPM7 channel inhibitor group(TRPM7 inhibitor). After four weeks of treatment, programmed electrical stimulation(PES) was employed to detect the arrhythmia susceptibility of rats. The myocardial cell structure and myocardial tissue fibrosis of myocardial and ganglion samples in diabetic rats were observed by hematoxylin-eosin(HE) staining and Masson staining. The immunohistochemistry, immunofluorescence, real-time quantitative polymerase chain reaction(RT-PCR), and Western blot were adopted to detect the distribution and expression of TRPM7, tyrosine hydroxylase(TH), choline acetyltransferase(ChAT), growth associated protein-43(GAP-43), nerve growth factor(NGF), p-AMPK/AMPK, and other genes and related neural markers. The results showed that LMQWD could significantly reduce the arrhythmia susceptibility and the degree of fibrosis in myocardial tissues, decrease the levels of TH, ChAT, and GAP-43 in the myocardium and ganglion, increase NGF, inhibit the expression of TRPM7, and up-regulate p-AMPK/AMPK and p-TrkA/TrkA levels. This study indicated that LMQWD could attenuate cardiac autonomic nerve remodeling in the diabetic state, and its mechanism was associated with the activation of AMPK, further phosphorylation of TrkA, and inhibition of TRPM7 expression.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , TRPM Cation Channels , Rats , Animals , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Diabetes Mellitus, Experimental/drug therapy , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , GAP-43 Protein/metabolism , Signal Transduction , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/genetics , Fibrosis
8.
Neurochem Res ; 48(9): 2826-2834, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37148458

ABSTRACT

Although the beneficial effects of curcumin, extracted from rhizomes of the ginger family genus Curcuma, on the repair and regeneration of nerves have been evaluated in vitro, there are few studies concerning its effects on axon myelination. Here, we used pheochromocytoma cells as an in vitro model of peripheral nerves. Pheochromocytoma cells were cultured alone or cocultured with Schwann cells and treated with increasing concentrations of curcumin. Cell growth was observed, and the expression levels of growth-associated protein 43 (GAP-43), microtubule-associated protein 2 (MAP-2), myelin basic protein (MBP), myelin protein zero (MPZ), Krox-20, and octamer binding factor 6 (Oct-6) were quantified. We found a significant increase in expression of all six proteins following curcumin treatment, with a corresponding increase in the levels of MBP, MPZ, Krox-20, and Oct-6 mRNA. Upregulation was greater with increasing curcumin concentration, showing a concentration-dependent effect. The results suggested that curcumin can promote the growth of axons by upregulating the expression of GAP-43 and MAP-2, stimulate synthesis and secretion of myelin-related proteins, and facilitate formation of the myelin sheath in axons by upregulating the expression of Krox-20 and Oct-6. Therefore, curcumin could be widely applied in future strategies for the treatment of nerve injuries.


Subject(s)
Adrenal Gland Neoplasms , Curcumin , Pheochromocytoma , Humans , Myelin Sheath/metabolism , Curcumin/pharmacology , GAP-43 Protein/metabolism , Pheochromocytoma/metabolism , Schwann Cells/metabolism , Myelin Proteins/metabolism , Axons/metabolism , Myelin P0 Protein/metabolism , Adrenal Gland Neoplasms/metabolism
9.
Nat Cancer ; 4(5): 648-664, 2023 05.
Article in English | MEDLINE | ID: mdl-37169842

ABSTRACT

The transfer of intact mitochondria between heterogeneous cell types has been confirmed in various settings, including cancer. However, the functional implications of mitochondria transfer on tumor biology are poorly understood. Here we show that mitochondria transfer is a prevalent phenomenon in glioblastoma (GBM), the most frequent and malignant primary brain tumor. We identified horizontal mitochondria transfer from astrocytes as a mechanism that enhances tumorigenesis in GBM. This transfer is dependent on network-forming intercellular connections between GBM cells and astrocytes, which are facilitated by growth-associated protein 43 (GAP43), a protein involved in neuron axon regeneration and astrocyte reactivity. The acquisition of astrocyte mitochondria drives an increase in mitochondrial respiration and upregulation of metabolic pathways linked to proliferation and tumorigenicity. Functionally, uptake of astrocyte mitochondria promotes cell cycle progression to proliferative G2/M phases and enhances self-renewal and tumorigenicity of GBM. Collectively, our findings reveal a host-tumor interaction that drives proliferation and self-renewal of cancer cells, providing opportunities for therapeutic development.


Subject(s)
Glioblastoma , Humans , Astrocytes/metabolism , Astrocytes/pathology , GAP-43 Protein/metabolism , GAP-43 Protein/therapeutic use , Axons/metabolism , Axons/pathology , Cell Line, Tumor , Nerve Regeneration , Mitochondria/metabolism , Mitochondria/pathology
10.
CNS Neurosci Ther ; 29(11): 3378-3390, 2023 11.
Article in English | MEDLINE | ID: mdl-37208955

ABSTRACT

AIMS: Few treatments are available in the subacute phase of traumatic brain injury (TBI) except rehabilitation training. We previously reported that transient CO2 inhalation applied within minutes after reperfusion has neuroprotective effects against cerebral ischemia/reperfusion injury. In this study, it was hypothesized that delayed CO2 postconditioning (DCPC) starting at the subacute phase may promote neurological recovery of TBI. METHODS: Using a cryogenic TBI (cTBI) model, mice received DCPC daily by inhaling 5%/10%/20% CO2 for various time-courses (one/two/three cycles of 10-min inhalation/10-min break) at Days 3-7, 3-14 or 7-18 after cTBI. Beam walking and gait tests were used to assess the effect of DCPC. Lesion size, expression of GAP-43 and synaptophysin, amoeboid microglia number and glia scar area were detected. Transcriptome and recombinant interferon regulatory factor 7 (Irf7) adeno-associated virus were applied to investigate the molecular mechanisms. RESULTS: DCPC significantly promoted recovery of motor function in a concentration and time-course dependent manner with a wide therapeutic time window of at least 7 days after cTBI. The beneficial effects of DCPC were blocked by intracerebroventricular injection of NaHCO3 . DCPC also increased puncta density of GAP-43 and synaptophysin, and reduced amoeboid microglia number and glial scar formation in the cortex surrounding the lesion. Transcriptome analysis showed many inflammation-related genes and pathways were altered by DCPC, and Irf7 was a hub gene, while overexpression of IRF7 blocked the motor function improvement of DCPC. CONCLUSIONS: We first showed that DCPC promoted functional recovery and brain tissue repair, which opens a new therapeutic time window of postconditioning for TBI. Inhibition of IRF7 is a key molecular mechanism for the beneficial effects of DCPC, and IRF7 may be a potential therapeutic target for rehabilitation after TBI.


Subject(s)
Brain Injuries, Traumatic , Carbon Dioxide , Interferon Regulatory Factor-7 , Animals , Mice , Brain Injuries, Traumatic/metabolism , Carbon Dioxide/metabolism , Carbon Dioxide/therapeutic use , Disease Models, Animal , GAP-43 Protein/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/therapeutic use , Synaptophysin/metabolism , Synaptophysin/therapeutic use
11.
Biochem Biophys Res Commun ; 656: 104-114, 2023 05 14.
Article in English | MEDLINE | ID: mdl-36963347

ABSTRACT

Podocyte injury is a crucial factor in the pathogenesis of diabetic kidney disease (DKD), and finding potential therapeutic interventions that can mitigate podocyte injury holds significant clinical relevance. This study was to elucidate the role of growth associated protein-43(Gap43) in podocyte injury of high glucose (HG). We confirmed the expression of Gap43 in human glomerulus and found that Gap43 expression was downregulated in podocytes of patients with DKD and HG-treated podocytes in vitro. Gap43 knockdown in podocytes promoted podocyte apoptosis, increased migration ability and decreased nephrin expression, while overexpression of Gap43 markedly suppressed HG-induced injury. Moreover, the increased expression and activity of calcineurin (CaN) were also abrogated by overexpression Gap43 in HG. Pretreatment with a typical CaN inhibitor FK506 in Gap43 knockdown podocytes restored the injury. Mechanistically, co-immunoprecipitation experiments suggested that Gap43 could bind to calmodulin (CaM). Pull-down assay further demonstrated that Gap43 and CaM directly interacts with each other via amino acids 30-52 of Gap43 and amino acids 133-197 of CaM. In addition, we also identified Pax5 as potential transcription inhibitor factor mediating Gap43 expression. In conclusion, the study indicated that the Gap43/CaM-CaN pathway may be exploited as a promising therapeutic target for protecting against podocyte injury in high glucose.


Subject(s)
Diabetic Nephropathies , GAP-43 Protein , Podocytes , Humans , Apoptosis , Calcineurin/metabolism , Calmodulin/metabolism , Diabetic Nephropathies/metabolism , GAP-43 Protein/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Podocytes/metabolism
14.
Brain Behav ; 13(4): e2949, 2023 04.
Article in English | MEDLINE | ID: mdl-36942730

ABSTRACT

OBJECTIVE: Prenatal stress (PS) is a problematic situation resulting in psychological implications such as social anxiety. Ubiquitous extremely low-frequency electromagnetic fields (ELF-EMF) have been confirmed as a potential physiological stressor; however, useful neuroregenerative effect of these types of electromagnetic fields has also frequently been reported. The aim of the present study was to survey the interaction of PS and ELF-EMF on anxiety-like behavior. METHOD: A total of 24 female rats 40 days of age were distributed into four groups of 6 rats each: control, stress (their mothers were exposed to stress), EMF (their mothers underwent to ELF-EMF), and EMF/stress (their mothers concurrently underwent to stress and ELF-EMF). The rats were assayed using elevated plus-maze and open field tests. RESULTS: Expressions of the hippocampus GAP-43, BDNF, and caspase-3 (cas-3) were detected by immunohistochemistry in Cornu Ammonis 1 (CA1) and dentate gyrus (DG) of the hippocampus and prefrontal cortex (PFC). Anxiety-like behavior increased in all treatment groups. Rats in the EMF/stress group presented more serious anxiety-like behavior. In all treatment groups, upregulated expression of cas-3 was seen in PFC, DG, and CA1 and downregulated expression of BDNF and GAP-43 was seen in PFC and DG and the CA1. Histomorphological study showed vast neurodegenerative changes in the hippocampus and PFC. CONCLUSION: The results showed ,female rats that underwent PS or/and EMF exhibited critical anxiety-like behavior and this process may be attributed to neurodegeneration in PFC and DG of the hippocampus and possibly decreased synaptic plasticity so-called areas.


Subject(s)
Brain-Derived Neurotrophic Factor , Electromagnetic Fields , Female , Rats , Animals , Electromagnetic Fields/adverse effects , Brain-Derived Neurotrophic Factor/metabolism , GAP-43 Protein/metabolism , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Anxiety/etiology
15.
Mol Pain ; 19: 17448069231158287, 2023.
Article in English | MEDLINE | ID: mdl-36733259

ABSTRACT

Important neurotrophic factors that are potentially involved in degenerative intervertebral disc (IVD) disease of the spine's lumbosacral (L/S) region include glial cell-derived neurotrophic factor (GDNF) and growth associated protein 43 (GAP-43). The aim of this study was to determine and compare the concentrations of GAP-43 and GDNF in degenerated and healthy IVDs and to quantify and compare the GAP-43-positive and GDNF-positive nerve fibers. The study group consisted of 113 Caucasian patients with symptomatic lumbosacral discopathy (confirmed by a specialist surgeon), an indication for surgical treatment. The control group included 81 people who underwent postmortem examination. GAP-43 and GDNF concentrations were significantly higher in IVD samples from the study group compared with the control group, and the highest concentrations were observed in the degenerated IVDs that were graded 4 on the Pfirrmann scale. In the case of GAP-43, it was found that as the degree of IVD degeneration increased, the number of GAP-43-positive nerve fibers decreased. In the case of GDNF, the greatest number of fibers per mm2 of surface area was found in the IVD samples graded 3 on the Pfirrmann scale, and the number was found to be lower in samples graded 4 and 5. Hence, GAP-43 and GDNF are promising targets for analgesic treatment of degenerative IVD disease of the lumbosacral region of the spine.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Humans , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , GAP-43 Protein/metabolism , Lumbosacral Region
16.
Ecotoxicol Environ Saf ; 252: 114576, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36736231

ABSTRACT

Nonylphenol (NP) is a typical environmental endocrine disruptor with estrogenic effects. It serves as an emulsifier and as the main ingredient of detergents and has become an increasingly common pollutant in both fresh and salt water, vegetables, and fruits. This study aimed to clarify whether NP exposure could lead to cognitive dysfunction and synaptic plasticity impairment, and also explore the mechanism of microRNA (miR)- 219a-5p regulation of N-methyl-D-aspartate receptor (NMDAR) in NP-induced synaptic plasticity impairment in vivo and in vitro. In vivo, 30 male Sprague-Dawley rats were randomly divided into 2 groups: blank control group (pure corn oil) and NP-exposed group [NP 80 mg/(kg·d)], with 15 rats in each group. In vitro, the extracted hippocampal neurons were divided into 6 groups: blank control group, mimics NC group, miR-219 mimics group, NP group (70 µmol/L NP), NP + mimics NC group, and NP + miR-219 mimics group. In vivo, the content of NP in hippocampal tissues after 90 days of NP exposure was significantly higher in the NP-stained group than in the blank control group. NP exposure could lead to a decrease in the ability to learn and memory, ability to remember, and space spatial memory ability in rats. The dendrites in the NP-stained group were disordered, with few dendritic spines and significantly decreased dendritic spine density. The postsynaptic densities were loosely arranged, the thickness and length of the postsynaptic densities shortened, and the length and width of the synaptic gap increased. Glutamine (Glu) and γ-aminobutyric acid (GABA) contents in hippocampal tissues decreased in the NP-stained group. The expression of miR-219a-5p mRNA decreased in the NP-stained group after 3 months of NP exposure. The expression of NMDAR1, NMDAR2A, NMDAR2B, nerve growth-associated protein (GAP-43), and Ca/calmodulin-dependent kinase II (CaMKII) mRNA/proteins decreased in the NP-stained group. In vitro, NMDAR protein expression decreased, while GAP-43 and CaMKII protein expression increased in the miR-219 mimics group compared with the control group. The expression levels of NMDAR and GAP-43 and CaMKII proteins were higher in the NP + miR-219 mimics group compared with the NP group. The levels of neurotransmitters Glu and GABA decreased in the NP and NP + mimics NC groups compared with the blank group. Shortened synaptic active band length, decreased thickness of postsynaptic densities, and shortened length of postsynaptic densities were observed in the NP, NP + mimics NC, and NP + miR-219 mimics groups compared with the blank control group. In vivo, NP exposure reduced learning memory capacity and neurotransmitter content in rats and caused a decrease in dendritic spine density and synaptic number density and a decrease in miR-219a-5p expression. In vitro, high expression of miR-219a-5p inhibited the expression of NMDAR, thus reducing the effect of NP on synaptic plasticity impairment in hippocampal neurons. Our study provided a scientific basis for the prevention of cognitive impairment owing to NP exposure and the development of targeted drug treatment strategies.


Subject(s)
MicroRNAs , Receptors, N-Methyl-D-Aspartate , Rats , Male , Animals , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Rats, Sprague-Dawley , MicroRNAs/genetics , MicroRNAs/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , GAP-43 Protein/metabolism , GAP-43 Protein/pharmacology , Neuronal Plasticity/physiology , RNA, Messenger
17.
Tissue Cell ; 81: 102014, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36621294

ABSTRACT

AIMS: Oxidative stress also plays an important role in the pathogenesis of diabetic neuropathy (DN). Both resveratrol (RES) and exercise (EX) have potent anti-oxidative benefits. Low levels of nerve growth factor (NGF) and SIRT1 (a member of sirtuin family) have been reported in patients with DN. The current study has been designed to investigate the role of serum NGF and SIRT1 on DN-induced hyperalgesia and motor incoordination and to evaluate the possible protective role of RES and/or EX. MAIN METHODS: A total of 40 male adult albino rats divided into five groups; control, DN, DN + RES, DN + EX, and DN + RES and EX. DN was confirmed by sensorimotor disturbance and diminished nerve conduction velocity (NCV). NGF and SIRT1 levels were measured by western blot. Calcitonin gene-related peptide (CGRP) was measured by PCR. Myofibrillar degeneration and inflammation scores were revealed via H&E microscopic analysis of the gastrocnemius muscle. Immunohistochemical evaluation of caspase3 and TNF-α was performed in the lumber segment of spinal cord and gastrocnemius muscle sections. Ultrastructural evaluation of sciatic nerve axonal degeneration has also been assessed. KEY FINDINGS: DN group showed decreased SIRT1 level, decreased NGF level and correlated with CGRP level and Na+/K+ ATPase. Treatment with RES and/or EX resulted in improvement of sensorimotor disturbance. DN characterized by reduced SOD level, whereas RES and/or EX could limit oxidative damage by up-regulation Bcl2, Akt and GAP-43 and down-regulation of caspase3 and TNF-α. In conclusion, increased level of SIRT1and NGF by incorporation of RES (natural supplementation) and EX (life style modification) could improve the neuroinflammatory state in DN.


Subject(s)
Diabetic Neuropathies , Exercise , Muscular Diseases , Resveratrol , Male , Calcitonin Gene-Related Peptide , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/therapy , Muscular Diseases/drug therapy , Muscular Diseases/therapy , Nerve Growth Factor/metabolism , Resveratrol/pharmacology , Sirtuin 1/metabolism , Tumor Necrosis Factor-alpha , Rats , Diabetes Complications/drug therapy , Diabetes Complications/metabolism , Diabetes Complications/therapy , GAP-43 Protein/metabolism , Animals
18.
Neurochem Res ; 48(2): 519-536, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36309937

ABSTRACT

Recent reports have suggested that abnormal miR-29c expression in hippocampus have been implicated in the pathophysiology of some neurodegenerative and neuropsychiatric diseases. However, the underlying effect of miR-29c in regulating hippocampal neuronal function is not clear. In this study, HT22 cells were infected with lentivirus containing miR-29c or miR-29c sponge. Cell counting kit-8 (CCK8) and lactate dehydrogenase (LDH) assay kit were applied to evaluate cell viability and toxicity before and after TNF-α administration. Reactive oxygen species (ROS) generation and mitochondrial membrane potential (MMP) were measured with fluorescent probes. Hoechst 33258 staining and TUNEL assay were used to evaluate cell apoptosis. The expression of key mRNA/proteins (TNFR1, Bcl-2, Bax, TRADD, FADD, caspase-3, -8 and -9) in the apoptosis pathway was detected by PCR or WB. In addition, the protein expression of microtubule-associated protein-2 (MAP-2), nerve growth-associated protein 43 (GAP-43) and synapsin-1 (SYN-1) was detected by WB. As a result, we found that miR-29c overexpression could improve cell viability, attenuate LDH release, reduce ROS production and inhibit MMP depolarization in TNF-α-treated HT22 cells. Furthermore, miR-29c overexpression was found to decrease apoptotic rate, along with decreased expression of Bax, cleaved caspase-3, cleaved caspase-9, and increased expression of Bcl-2 in TNF-α-treated HT22 cells. However, miR-29c sponge exhibited an opposite effects. In addition, in TNF-α-treated HT22 cells, miR-29c overexpression could decrease the expressions of TNFR1, TRADD, FADD and cleaved caspase-8. However, in HT22 cells transfected with miR-29c sponge, TNF-α-induced the expressions of TNFR1, TRADD, FADD and cleaved caspase-8 was significantly exacerbated. At last, TNF-α-induced the decreased expression of MAP-2, GAP-43 and SYN-1 was reversed by miR-29c but exacerbated by miR-29c sponge. Overall, our study demonstrated that miR-29c protects against TNF-α-induced HT22 cells injury through alleviating ROS production and reduce neuronal apoptosis. Therefore, miR-29c might be a potential therapeutic agent for TNF-α accumulation and toxicity-related brain diseases.


Subject(s)
MicroRNAs , Tumor Necrosis Factor-alpha , Mice , Animals , Reactive Oxygen Species/metabolism , Caspase 3/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Caspase 8/metabolism , Caspase 8/pharmacology , Receptors, Tumor Necrosis Factor, Type I , bcl-2-Associated X Protein/metabolism , GAP-43 Protein/metabolism , Cell Line , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism , MicroRNAs/metabolism , Hippocampus/metabolism
19.
Morphologie ; 107(356): 67-79, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35715368

ABSTRACT

Adverse experiences during pregnancy have a negative impact on the neuronal structure and behavior of offspring, but the effects of a father's life events on the outcome of progeny are scarce. The present study is intended to investigate whether paternal stress affects the offspring brain structure, especially those regions concerned with learning and formation of memory, namely the hippocampus (HC) and prefrontal cortex (PFC), and also the expression of certain genes linked to learning and memory in the offspring. Induced stress to male rats by five stressors, one per day followed by allowing them to mate with the normal, unstressed female. Synaptophysin immunoreactivity was assessed in the tissue sections of the HC and PFC as well as expression of genes concerned with learning and memory was evaluated by RT-PCR in the progeny of stress-received males. The progeny of stressed rats had reduced antisynaptophysin immunoreactivity in the HC and PFC. The synaptic density in HC was less in the A-S (Offspring of male rats who received stress during adulthood) and PA-S (offspring of male rats who received stress during both adolescence and adulthood) than in P-S (offspring of male rats who received stress during adolescence) and C-C (offspring of control) groups. Similar results were observed even in the PFC. The results of post hoc tests proved that the HC and PFC of the progeny of stress-exposed rats exhibited considerably less synaptic density than control (P<0.05), and the levels of expression of GAP-43, GRIN1, M1, and SYP genes in HC and PFC were down-regulated. This study concludes that paternal adverse experiences can affect the offspring's synaptic plasticity and also the genes, which can regulate learning and formation of memory.


Subject(s)
Hippocampus , Prefrontal Cortex , Pregnancy , Rats , Animals , Male , Female , Humans , GAP-43 Protein/metabolism , GAP-43 Protein/pharmacology , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Learning , Fathers , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Synaptophysin/pharmacology
20.
Auris Nasus Larynx ; 50(3): 358-364, 2023 Jun.
Article in English | MEDLINE | ID: mdl-35999123

ABSTRACT

OBJECTIVE: Olfactory neuroblastoma (ONB) is often difficult to pathologically distinguish from other small round cell tumors (SRCTs) arising in the nasal cavities. Although there are several diagnostic markers used for differential diagnosis of ONB, these molecules are also expressed in various neuronal derived tumors. Here, we examined the expression of NeuroD, GAP43, and olfactory marker protein (OMP) in ONB and non-ONB SRCT to determine their utility in the differential diagnosis of ONB. METHODS: Twenty-six patients diagnosed with and treated for ONB at Kobe University Hospital between 1997 and 2017 with formalin-fixed, paraffin-embedded biopsy or surgical resection specimens were included. The expressions of NeuroD, GAP43, and OMP were immunohistochemically examined in these 26 ONB specimens and specimens from 13 SRCTs arising in the nasal cavities for reference. RESULTS: Among the 26 ONB samples, focal, patchy, and marked staining for NeuroD was observed in 4, 3, and 9 samples, respectively. Focal, patchy, and marked GAP43 staining was observed in 5, 3, and 11 samples, respectively. Consequently, marked positive staining for either NeuroD or GAP43 was observed in 54% (14/26) of ONBs. Among the 13 SRCTs, marked staining for NeuroD was observed in two small cell carcinomas, one undifferentiated carcinoma, and one neuroendocrine carcinoma, whereas marked positive staining for GAP43 was observed only in one undifferentiated carcinoma. No specimen in this study exhibited OMP staining. CONCLUSIONS: Our results suggest possible roles of GAP43 immunostaining in the differential diagnosis of ONB.


Subject(s)
Carcinoma, Neuroendocrine , Esthesioneuroblastoma, Olfactory , Nose Neoplasms , Humans , Carcinoma, Neuroendocrine/pathology , Esthesioneuroblastoma, Olfactory/pathology , Nasal Cavity/pathology , Nose Neoplasms/pathology , Olfactory Marker Protein , GAP-43 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...