Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 362
Filtrar
1.
Sci Rep ; 14(1): 17845, 2024 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090173

RESUMEN

The core of clinic treatment of Parkinson's disease (PD) is to enhance dopamine (DA) signaling within the brain. The regulation of dopamine transporter (DAT) is integral to this process. This study aims to explore the regulatory mechanism of glial cell line-derived neurotrophic factor (GDNF) on DAT, thereby gaining a profound understanding its potential value in treating PD. In this study, we investigated the effects of GDNF on both cellular and mouse models of PD, including the glycosylation and membrane transport of DAT detected by immunofluorescence and immunoblotting, DA signal measured by neurotransmitter fiber imaging technology, Golgi morphology observed by electron microscopic, as well as cognitive ability assessed by behavior tests. This study revealed that in animal trials, MPTP-induced Parkinson's Disease (PD) mice exhibited a marked decline in cognitive function. Utilizing ELISA and neurotransmitter fiber imaging techniques, we observed a decrease in dopamine levels and a significant reduction in the intensity of dopamine signal release in the Prefrontal Cortex (PFC) of PD mice induced by MPTP. Intriguingly, these alterations were reversed by Glial Cell Line-Derived Neurotrophic Factor (GDNF). In cellular experiments, following MPP + intervention, there was a decrease in Gly-DAT modification in both the cell membrane and cytoplasm, coupled with an increase in Nongly-DAT expression and aggregation of DAT within the cytoplasm. Conversely, GDNF augmented DAT glycosylation and facilitated its membrane transport in damaged dopaminergic neurons, concurrently reversing the effects of GRASP65 depletion and Golgi fragmentation, thereby reducing the accumulation of DAT in the Golgi apparatus. Furthermore, overexpression of GRASP65 enhanced DAT transport in PD cells and mice, while suppression of GRASP65 attenuated the efficacy of GDNF on DAT. Additionally, GDNF potentiated the reutilization of neurotransmitters by the PFC presynaptic membrane, boosting the effective release of dopamine following a single electrical stimulation, ultimately ameliorating the cognitive impairments in PD mice.Therefore, we propose that GDNF enhances the glycosylation and membrane trafficking of DAT by facilitating the re-aggregation of the Golgi apparatus, thereby amplifying the utilization of DA signals. This ultimately leads to the improvement of cognitive abilities in PD mouse models. Our study illuminates, from a novel angle, the beneficial role of GDNF in augmenting DA utilization and cognitive function in PD, providing fresh insights into its therapeutic potential.


Asunto(s)
Cognición , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Dopamina , Factor Neurotrófico Derivado de la Línea Celular Glial , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glicosilación , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ratones , Cognición/efectos de los fármacos , Dopamina/metabolismo , Masculino , Enfermedad de Parkinson/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Membrana Celular/metabolismo , Corteza Prefrontal/metabolismo
2.
Neurol Sci ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38795270

RESUMEN

Parkinson's disease (PD) is a chronic neurological disorder that is identified by a characteristic combination of symptoms such as bradykinesia, resting tremor, rigidity, and postural instability. It is the second most common neurodegenerative disease after Alzheimer's disease and is characterized by the progressive loss of dopamine-producing neurons in the brain. Currently, available treatments for PD are symptomatic and do not prevent the disease pathology. There is growing interest in developing disease-modifying therapy that can reduce disease progression and improve patients' quality of life. One of the promising therapeutic approaches under evaluation is gene therapy utilizing a viral vector, adeno-associated virus (AAV), to deliver transgene of interest into the central nervous system (CNS). Preclinical studies in small animals and nonhuman primates model of PD have shown promising results utilizing the gene therapy that express glial cell line-derived neurotrophic factor (GDNF), cerebral dopamine neurotrophic factor (CDNF), aromatic L-amino acid decarboxylase (AADC), and glutamic acid decarboxylase (GAD). This study provides a comprehensive review of the current state of the above-mentioned gene therapies in various phases of clinical trials for PD treatment. We have highlighted the rationale for the gene-therapy approach and the findings from the preclinical and nonhuman primates studies, evaluating the therapeutic effect, dose safety, and tolerability. The challenges associated with gene therapy for heterogeneous neurodegenerative diseases, such as PD, have also been described. In conclusion, the review identifies the ongoing promising gene therapy approaches in clinical trials and provides hope for patients with PD.

3.
Neurochem Int ; 177: 105765, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38750960

RESUMEN

BACKGROUND: Perioperative neurocognitive disorders (PND) are common complications after surgery in older patients. However, the specific mechanism of this condition remains unclear. Glial cell line-derived neurotrophic factor (GDNF) is an important neurotrophin that abundantly expressed throughout the brain. It can enhance synaptic plasticity and alleviate learning and memory impairments. Thus, the purpose of this study was to investigate the role of GDNF in PND and the mechanisms involved. METHODS: The PND animal model was established by performing left tibial fracture surgery on 18-month-old C57BL/6 mice under sevoflurane anesthesia. Recombinant adeno-associated virus (rAAV)-GDNF or empty vectors were injected bilaterally into the hippocampal CA1 region of aged mice 3 weeks before anesthesia/surgery. The open field and fear conditioning test were used to assess the behavior changes. Golgi staining and electrophysiology were utilized to evaluate the morphological and functional alterations of neuronal synaptic plasticity. Western blot analysis was carried out to measure the proteins expression levels and immunofluorescence staining was performed to probe the cellular localization of GDNF. RESULTS: Mice with surgery and anesthesia showed a significant decrease in hippocampus-dependent learning and memory, accompanied by a decline in hippocampal synaptic plasticity. Anesthesia/surgery induced a reduction of GDNF, which was colocalized with astrocytes. Overexpression of GDNF in astrocytes could ameliorate the decline in cognitive function by improving hippocampal synaptic plasticity, meanwhile astrocytic GDNF rescued the anesthesia/surgery-induced decrease in GFRα1 and NCAM. CONCLUSION: The study concludes that astrocytic GDNF may improve anesthesia/surgery-induced cognitive impairment by promoting hippocampal synaptic plasticity in aged mice via the GFRα1/NCAM pathway.


Asunto(s)
Astrocitos , Disfunción Cognitiva , Factor Neurotrófico Derivado de la Línea Celular Glial , Hipocampo , Ratones Endogámicos C57BL , Plasticidad Neuronal , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Plasticidad Neuronal/fisiología , Plasticidad Neuronal/efectos de los fármacos , Ratones , Astrocitos/metabolismo , Masculino , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Envejecimiento , Anestesia
4.
Neurotherapeutics ; 21(4): e00370, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38704311

RESUMEN

Hemorrhage-induced injury of the corticospinal tract (CST) in the internal capsule (IC) causes severe neurological dysfunction in both human patients and rodent models of intracerebral hemorrhage (ICH). A nuclear receptor Nurr1 (NR4A2) is known to exert anti-inflammatory and neuroprotective effects in several neurological disorders. Previously we showed that Nurr1 ligands prevented CST injury and alleviated neurological deficits after ICH in mice. To prove direct effect of Nurr1 on CST integrity, we examined the effect of Nurr1 overexpression in neurons of the primary motor cortex on pathological consequences of ICH in mice. ICH was induced by intrastriatal injection of collagenase type VII, where hematoma invaded into IC. Neuron-specific overexpression of Nurr1 was induced by microinjection of synapsin I promoter-driven adeno-associated virus (AAV) vector into the primary motor cortex. Nurr1 overexpression significantly alleviated motor dysfunction but showed only modest effect on sensorimotor dysfunction after ICH. Nurr1 overexpression also preserved axonal structures in IC, while having no effect on hematoma-associated inflammatory events, oxidative stress, and neuronal death in the striatum after ICH. Immunostaining revealed that Nurr1 overexpression increased the expression of Ret tyrosine kinase and phosphorylation of Akt and ERK1/2 in neurons in the motor cortex. Moreover, administration of Nurr1 ligands 1,1-bis(3'-indolyl)-1-(p-chlorophenyl)methane or amodiaquine increased phosphorylation levels of Akt and ERK1/2 as well as expression of glial cell line-derived neurotrophic factor and Ret genes in the cerebral cortex. These results suggest that the therapeutic effect of Nurr1 on striatal ICH is attributable to the preservation of CST by acting on cortical neurons.

5.
Int J Mol Sci ; 25(8)2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38674063

RESUMEN

Plasma levels of glial cell line-derived neurotrophic factor (GDNF), a pivotal regulator of differentiation and survival of dopaminergic neurons, are reportedly decreased in schizophrenia. To explore the involvement of GDNF in the pathogenesis of the disease, a case-control association analysis was performed between five non-coding single nucleotide polymorphisms (SNP) across the GDNF gene and schizophrenia. Of them, the 'G' allele of the rs11111 SNP located in the 3' untranslated region (3'-UTR) of the gene was found to associate with schizophrenia. In silico analysis revealed that the rs11111 'G' allele might create binding sites for three microRNA (miRNA) species. To explore the significance of this polymorphism, transient co-transfection assays were performed in human embryonic kidney 293T (HEK293T) cells with a luciferase reporter construct harboring either the 'A' or 'G' allele of the 3'-UTR of GDNF in combination with the hsa-miR-1185-1-3p pre-miRNA. It was demonstrated that in the presence of the rs11111 'G' (but not the 'A') allele, hsa-miR-1185-2-3p repressed luciferase activity in a dose-dependent manner. Deletion of the miRNA binding site or its substitution with the complementary sequence abrogated the modulatory effect. Our results imply that the rs11111 'G' allele occurring more frequently in patients with schizophrenia might downregulate GDNF expression in a miRNA-dependent fashion.


Asunto(s)
Regiones no Traducidas 3' , Factor Neurotrófico Derivado de la Línea Celular Glial , MicroARNs , Polimorfismo de Nucleótido Simple , Esquizofrenia , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Alelos , Sitios de Unión , Estudios de Casos y Controles , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células HEK293 , MicroARNs/genética , Esquizofrenia/genética , Esquizofrenia/metabolismo
6.
Risk Manag Healthc Policy ; 17: 935-943, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38633669

RESUMEN

Purpose: Mild cognitive impairment (MCI) and depressive disorder (DD), which are associated with unhealthy lifestyles, are prevalent worldwide. This study aimed to investigate the effects of regular aerobic exercise on cognitive function, depression, and the regulatory role of neurotrophic growth factors for providing scientific basis in preventing MCI and DD in healthy individuals. Patients and Methods: Eighty members of the fitness center and 80 community residents were recruited, who were administered by the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and the Patient Health Questionnaire (PHQ-9). Brain-derived neurotrophic factor (BDNF) and glial cell-line-derived neurotrophic factor (GDNF) in the peripheral blood were detected by enzyme-linked immunosorbent assay (ELISA). Results: The RBANS and other factor scores, except for visuospatial abilities, were higher and PHQ-9 scores were lower in the study group than in the control group. The concentrations of BDNF and GDNF in the study group were higher than those in the control group. RBANS and its factor scores positively and PHQ-9 negatively correlated with BDNF and GDNF levels. Finally, multiple regression analysis showed that BDNF, as a predictor of RBANS, could explain 59.90% of its variance and that GDNF was a predictor of PHQ-9 could explain 12.30% of the variance. Conclusion: Regular aerobic exercise can improve cognitive function and depressive symptoms by increasing the BDNF and GDNF levels.

7.
Int J Mol Sci ; 25(3)2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-38339124

RESUMEN

Peripheral nerve injury denervates muscle, resulting in muscle paralysis and atrophy. This is reversible if timely muscle reinnervation occurs. With delayed reinnervation, the muscle's reparative ability declines, and muscle-resident fibro-adipogenic progenitor cells (FAPs) proliferate and differentiate, inducing fibro-fatty muscle degradation and thereby physical disability. The mechanisms by which the peripheral nerve regulates FAPs expansion and differentiation are incompletely understood. Using the rat tibial neve transection model, we demonstrated an increased FAPs content and a changing FAPs phenotype, with an increased capacity for adipocyte and fibroblast differentiation, in gastrocnemius muscle post-denervation. The FAPs response was inhibited by immediate tibial nerve repair with muscle reinnervation via neuromuscular junctions (NMJs) and sensory organs (e.g., muscle spindles) or the sensory protection of muscle (where a pure sensory nerve is sutured to the distal tibial nerve stump) with reinnervation by muscle spindles alone. We found that both procedures reduced denervation-mediated increases in glial-cell-line-derived neurotrophic factor (GDNF) in muscle and that GDNF promoted FAPs adipogenic and fibrogenic differentiation in vitro. These results suggest that the peripheral nerve controls FAPs recruitment and differentiation via the modulation of muscle GDNF expression through NMJs and muscle spindles. GDNF can serve as a therapeutic target in the management of denervation-induced muscle injury.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Músculo Esquelético , Ratas , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Músculo Esquelético/metabolismo , Diferenciación Celular , Nervio Tibial/lesiones , Adipogénesis , Desnervación
8.
Artículo en Inglés | MEDLINE | ID: mdl-38396228

RESUMEN

Children with chronic tic disorders (CTD), including Tourette syndrome (TS), have significantly reduced serum 25-hydroxyvitamin D [25(OH)D]. While vitamin D3 supplementation (VDS) may reduce tic symptoms in these children, its mechanism is unclear. The study aim was to investigate the effects and mechanisms of vitamin D deficiency (VDD) and VDS on TS model behavior. Forty 5-week-old male Sprague-Dawley rats were randomly divided into (n = 10 each): control, TS model, TS model with VDD (TS + VDD), or TS model with VDS (TS + VDS; two intramuscular injections of 20,000 IU/200 g) groups. The VDD model was diet-induced (0 IU vitamin D/kg); the TS model was iminodipropionitrile (IDPN)-induced. All groups were tested for behavior, serum and striatal 25(OH)D and dopamine (DA), mRNA expressions of vitamin D receptor (VDR), glial cell line-derived neurotrophic factor (GDNF), protooncogene tyrosine-protein kinase receptor Ret (c-Ret), and DA D1 (DRD1) and D2 (DRD2) receptor genes in the striatum. TS + VDD had higher behavior activity scores throughout, and higher total behavior score at day 21 compared with TS model. In contrast, day 21 TS + VDS stereotyped behavior scores and total scores were lower than TS model. The serum 25(OH)D in TS + VDD was < 20 ng/mL, and lower than control. Striatal DA of TS was lower than control. Compared with TS model, striatal DA of TS + VDD was lower, while in TS + VDS it was higher than TS model. Furthermore, mRNA expression of VDR, GDNF, and c-Ret genes decreased in TS model, and GDNF expression decreased more in TS + VDD, while TS + VDS had higher GDNF and c-Ret expressions. VDD aggravates, and VDS ameliorates tic-like behavior in an IDPN-induced model. VDS may upregulate GDNF/c-Ret signaling activity through VDR, reversing the striatal DA decrease and alleviating tic-like behavior.

9.
Arch Biochem Biophys ; 753: 109893, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38309681

RESUMEN

Adipose tissue-derived stem cells (ADSCs) are a kind of stem cells with multi-directional differentiation potential, which mainly restore tissue repair function and promote cell regeneration. It can be directionally differentiated into Schwann-like cells to promote the repair of peripheral nerve injury. Glial cell line-derived neurotrophic factor (GDNF) plays an important role in the repair of nerve injury, but the underlying mechanism remains unclear, which seriously limits its further application.The study aimed to identify the molecular mechanism by which overexpression of glial cell line-derived neurotrophic factor (GDNF) facilitates the differentiation of ADSCs into Schwann cells, enhancing nerve regeneration after injury. In vitro, ADSCs overexpressing GDNF for 48 h exhibited changes in their morphology, with 80% of the cells having two or more prominences. Compared with that of ADSCs, GDNF-ADSCs exhibited increased expression of the Schwann cell marker S100, nerve damage repair-related factors.ADSC cells in normal culture and ADSC cells were overexpressing GDNF(GDNF-ADSCs) were analysed using TMT-Based Proteomic Analysis and revealed a significantly higher expression of MTA1 in GDNF-ADSCs than in control ADSCs. Hes1 expression was significantly higher in GDNF-ADSCs than in ADSCs and decreased by MTA1 silencing, along with a simultaneous decrease in the expression of S100 and nerve damage repair factors. These findings indicate that GDNF promotes the differentiation of ADSCs into Schwann cells and induces factors that accelerate peripheral nerve damage repair.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Proteómica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Regeneración Nerviosa , Tejido Adiposo , Diferenciación Celular , Células de Schwann
10.
Chin J Integr Med ; 30(3): 251-259, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38212498

RESUMEN

OBJECTIVE: To explore the mechanism of electroacupuncture (EA) in promoting recovery of the facial function with the involvement of autophagy, glial cell line-derived neurotrophic factor (GDNF), and phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway. METHODS: Seventy-two male Sprague-Dawley rats were randomly allocated into the control, sham-operated, facial nerve injury (FNI), EA, EA+3-methyladenine (3-MA), and EA+GDNF antagonist groups using a random number table, with 12 rats in each group. An FNI rat model was established with facial nerve crushing method. EA intervention was conducted at Dicang (ST 4), Jiache (ST 6), Yifeng (SJ 17), and Hegu (LI 4) acupoints for 2 weeks. The Simone's 10-Point Scale was utilized to monitor the recovery of facial function. The histopathological evaluation of facial nerves was performed using hematoxylin-eosin (HE) staining. The levels of Beclin-1, light chain 3 (LC3), and P62 were detected by immunohistochemistry (IHC), immunofluorescence, and reverse transcription-polymerase chain reaction, respectively. Additionally, IHC was also used to detect the levels of GDNF, Rai, PI3K, and mTOR. RESULTS: The facial functional scores were significantly increased in the EA group than the FNI group (P<0.05 or P<0.01). HE staining showed nerve axons and myelin sheaths, which were destroyed immediately after the injury, were recovered with EA treatment. The expressions of Beclin-1 and LC3 were significantly elevated and the expression of P62 was markedly reduced in FNI rats (P<0.01); however, EA treatment reversed these abnormal changes (P<0.01). Meanwhile, EA stimulation significantly increased the levels of GDNF, Rai, PI3K, and mTOR (P<0.01). After exogenous administration with autophagy inhibitor 3-MA or GDNF antagonist, the repair effect of EA on facial function was attenuated (P<0.05 or P<0.01). CONCLUSIONS: EA could promote the recovery of facial function and repair the facial nerve damages in a rat model of FNI. EA may exert this neuroreparative effect through mediating the release of GDNF, activating the PI3K/mTOR signaling pathway, and further regulating the autophagy of facial nerves.


Asunto(s)
Electroacupuntura , Traumatismos del Nervio Facial , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Fosfatidilinositol 3-Quinasa/metabolismo , Traumatismos del Nervio Facial/terapia , Fosfatidilinositol 3-Quinasas/metabolismo , Beclina-1 , Factor Neurotrófico Derivado de la Línea Celular Glial , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Autofagia , Mamíferos/metabolismo
11.
J Physiol Sci ; 74(1): 4, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38267849

RESUMEN

We reviewed fundamental studies on muscular pain, encompassing the characteristics of primary afferent fibers and neurons, spinal and thalamic projections, several muscular pain models, and possible neurochemical mechanisms of muscle pain. Most parts of this review were based on data obtained from animal experiments, and some researches on humans were also introduced. We focused on delayed-onset muscle soreness (DOMS) induced by lengthening contractions (LC), suitable for studying myofascial pain syndromes. The muscular mechanical withdrawal threshold (MMWT) decreased 1-3 days after LC in rats. Changing the speed and range of stretching showed that muscle injury seldom occurred, except in extreme conditions, and that DOMS occurred in parameters without muscle damage. The B2 bradykinin receptor-nerve growth factor (NGF) route and COX-2-glial cell line-derived neurotrophic factor (GDNF) route were involved in the development of DOMS. The interactions between these routes occurred at two levels. A repeated-bout effect was observed in MMWT and NGF upregulation, and this study showed that adaptation possibly occurred before B2 bradykinin receptor activation. We have also briefly discussed the prevention and treatment of DOMS.


Asunto(s)
Mialgia , Factor de Crecimiento Nervioso , Humanos , Animales , Ratas , Neuronas , Regulación hacia Arriba , Receptores de Bradiquinina
12.
Scand J Med Sci Sports ; 34(1): e14497, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37724768

RESUMEN

Delayed onset muscle soreness (DOMS) develops after performing unaccustomed eccentric exercises. Animal studies have shown that DOMS is mechanical hyperalgesia through nociceptor sensitization induced by nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF) upregulated by cyclooxygenase-2 (COX-2). However, no previous study has investigated these in relation to DOMS in humans. This study compared the first and second bouts of one-leg eccentric cycling (ECC) for changes in NGF, GDNF, and COX-2 mRNA in the vastus lateralis (VL). Seven healthy adults (18-40 years) performed two bouts of ECC (10 sets of 50 contractions) with 80% maximal voluntary concentric peak torque separated by 2 weeks (ECC1, ECC2). Muscle soreness that was assessed by a visual analog scale and maximal voluntary isometric contraction (MVC) torque of the knee extensors were measured before, immediately after (MVC only), 24 and 48 h post-exercise. Muscle biopsy was taken from the VL before the first bout from nonexercised leg (control) and 24 h after each bout from the exercised leg, and analyzed for NGF, GDNF, and COX-2 mRNA. Peak DOMS was more than two times greater and MVC torque at 48 h post-exercise was approximately 20% smaller after ECC1 than ECC2 (p < 0.05), suggesting the repeated bout effect. NGF mRNA level was higher (p < 0.05) post-ECC1 (0.79 ± 0.68 arbitrary unit) than control (0.06 ± 0.07) and post-ECC2 (0.08 ± 0.10). GDNF and COX-2 mRNA did not show significant differences between control, post-ECC1, and post-ECC2. These results suggest that an increase in NGF is associated with the development of DOMS in humans.


Asunto(s)
Músculo Esquelético , Músculo Cuádriceps , Adulto , Humanos , Músculo Cuádriceps/fisiología , Músculo Esquelético/fisiología , Mialgia , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Pierna , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Contracción Isométrica/fisiología , ARN Mensajero/metabolismo , Contracción Muscular/fisiología
13.
Neurol Sci ; 45(4): 1409-1418, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38082050

RESUMEN

Parkinson's disease is the second most common neurodegenerative condition with its prevalence projected to 8.9 million individuals globally in the year 2019. Parkinson's disease affects both motor and certain non-motor functions of an individual. Numerous research has focused on the neuroprotective effect of the glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease. Discovered in 1993, GDNF is a neurotrophic factor identified from the glial cells which was found to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. Given this property, recent studies have focused on the exogenous administration of GDNF for relieving Parkinson's disease-related symptoms both at a pre-clinical and a clinical level. This review will focus on enumerating the molecular connection between Parkinson's disease and GDNF and shed light on all the available drug delivery approaches to facilitate the selective delivery of GDNF into the brain paving the way as a potential therapeutic candidate for Parkinson's disease in the future.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Neuronas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuroglía
14.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1025617

RESUMEN

Objective:To explore the effect of Baduanjin on gait parameters and serum nerve growth factor in Parkinson disease (PD) patients with freezing of gait(FOG).Methods:From December 2021 to December 2022, thirty-eight PD patients with FOG who met the inclusion and exclusion criteria were randomly divided into observation group ( n=18) and control group ( n=20) by random number table.The patients in both two groups received 4 weeks of drug therapy combined with basic rehabilitation treatment respectively, and the patients in observation group received additional Baduanjin training.Efficacy was evaluated 1 day before intervention and after 4 weeks of intervention through unified Parkinson's disease rating scale-Ⅱ(UPDRS-Ⅱ) item 14, freezing of gait questionnaire (FOGQ), gait starting time, gait cycle, stride length, dynamic plantar peak pressure and average pressure, while the levels of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor(GDNF) in peripheral blood of patients were tested.SPSS 23.0 software was used to conduct Chi-square test, paired t-test, independent sample t-test and Mann-Whitney U test. Results:Before treatment, there were no significant differences in score of UPDRS-Ⅱ item 14, FOGQ score, gait starting time, gait cycle, stride length, dynamic planar peak pressure, average pressure, peripheral blood BDNF level and GDNF level between the two groups ( t=-0.542, 0.562, 0.490, 0.674, 0.440, 0.606, -0.835, -0.873, -0.250, all P>0.05). After treatment, compared with the control group, dynamic plantar peak pressure (control group (14.26±3.23) N/cm 2, observation group (11.40±4.13) N/cm 2, t=-2.389, P=0.022) and plantar average pressure (control group (3.34±0.72) N/cm 2, observation group (2.79±0.81) N/cm 2, t=-2.209, P=0.034) of the observation group were significantly decreased (both P<0.05). There were no significant differences in UPDRS-Ⅱ item 14, FOGQ score, gait starting time, gait cycle, stride length, BDNF and GDNF concentrations in peripheral blood between the two groups after treatment (all P>0.05). The difference between pre-treatment and post-treatment of FOGQ score (control group 1.00 (0.00, 1.00) , observation group 2.00 (0.75, 3.00), Z=-2.547, P=0.011), gait starting time (control group -1.04 (-1.86, -0.47)s, observation group -2.34 (-3.41, -1.03) s, Z=-2.280, P=0.023), gait cycle (control group 0.29 (0.08, 0.58)s, observation group 0.35 (0.16, 1.00) s, Z=-2.748, P=0.006), stride length(control group 0.19 (0.14, 0.24) m, observation group 0.26 (0.23, 0.38)m, Z=-1.360, P=0.005), the dynamic plantar peak pressure (control group -4.11 (-5.87, -2.57) N/cm 2, observation group -8.44 (-10.12, -4.81) N/cm 2, Z=-3.333, P=0.001) and average pressure (control group -0.55 (-1.00, -0.03) N/cm 2, observation group -1.11 (-1.51, -0.66) N/cm 2, Z=-2.062, P=0.009) in the observation group were better than those in the control group.After treatment, the BDNF level in peripheral blood in observation group was higher than before treatment( t=-2.315, P=0.033). Conclusion:Baduanjin can improve frozen gait score and gait parameters in PD patients with FOG, which may be related to the increase of peripheral blood BDNF.

15.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-1010332

RESUMEN

OBJECTIVE@#To explore the mechanism of electroacupuncture (EA) in promoting recovery of the facial function with the involvement of autophagy, glial cell line-derived neurotrophic factor (GDNF), and phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway.@*METHODS@#Seventy-two male Sprague-Dawley rats were randomly allocated into the control, sham-operated, facial nerve injury (FNI), EA, EA+3-methyladenine (3-MA), and EA+GDNF antagonist groups using a random number table, with 12 rats in each group. An FNI rat model was established with facial nerve crushing method. EA intervention was conducted at Dicang (ST 4), Jiache (ST 6), Yifeng (SJ 17), and Hegu (LI 4) acupoints for 2 weeks. The Simone's 10-Point Scale was utilized to monitor the recovery of facial function. The histopathological evaluation of facial nerves was performed using hematoxylin-eosin (HE) staining. The levels of Beclin-1, light chain 3 (LC3), and P62 were detected by immunohistochemistry (IHC), immunofluorescence, and reverse transcription-polymerase chain reaction, respectively. Additionally, IHC was also used to detect the levels of GDNF, Rai, PI3K, and mTOR.@*RESULTS@#The facial functional scores were significantly increased in the EA group than the FNI group (P<0.05 or P<0.01). HE staining showed nerve axons and myelin sheaths, which were destroyed immediately after the injury, were recovered with EA treatment. The expressions of Beclin-1 and LC3 were significantly elevated and the expression of P62 was markedly reduced in FNI rats (P<0.01); however, EA treatment reversed these abnormal changes (P<0.01). Meanwhile, EA stimulation significantly increased the levels of GDNF, Rai, PI3K, and mTOR (P<0.01). After exogenous administration with autophagy inhibitor 3-MA or GDNF antagonist, the repair effect of EA on facial function was attenuated (P<0.05 or P<0.01).@*CONCLUSIONS@#EA could promote the recovery of facial function and repair the facial nerve damages in a rat model of FNI. EA may exert this neuroreparative effect through mediating the release of GDNF, activating the PI3K/mTOR signaling pathway, and further regulating the autophagy of facial nerves.


Asunto(s)
Ratas , Masculino , Animales , Ratas Sprague-Dawley , Electroacupuntura , Fosfatidilinositol 3-Quinasa/metabolismo , Traumatismos del Nervio Facial/terapia , Fosfatidilinositol 3-Quinasas/metabolismo , Beclina-1 , Factor Neurotrófico Derivado de la Línea Celular Glial , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Autofagia , Mamíferos/metabolismo
16.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1007278

RESUMEN

ObjectiveTo investigate the expression of glial cell line-derived neurotrophic factor (GDNF) and androgen receptor (AR) in testicular peritubular cells (TPCs) of cryptorchidism mouse models and explore the theoretical significance of cryptorchidism-induced spermatogenesis dysfunction. MethodsA total of 30 five-week-old male ICR rats were divided randomly by using random number table method into 6 groups. Cryptorchidism was surgically induced in 3 randomly selected groups and the other 3 groups underwent sham surgery as the control groups. On days 4, 7 and 14 after surgery, we harvested the mice testes of the 3 groups and their corresponding control groups, then measured the testicular volumes, analyzed the testicular histopathology and detected the mRNA and protein expression levels of AR and GDNF in TPCs by immunofluorescence, real-time PCR and Western blot. ResultsIn normal control groups, on days 4, 7 and 14 after surgery, the testicular volumes were (125.58±19.22) mm3,(123.45±20.12) mm3, (140.09±13.62) mm3 , respectively. Clear layers of spermatogenic cells were well arranged and abundant sperm cells were found. Peritubular cells were morphologically homogeneous, with slim-spindle appearance and normal cell thickness. The mRNA expression levels of AR were 1.00±0.05, 1.06±0.07 and 1.19±0.13; GDNF mRNA 1.00±0.04, 1.09±0.05, and 1.10±0.07. The protein expression levels of AR were 1.01±0.01, 0.79±0.02 and 1.01±0.04; GDNF protein (18.68±0.43) pg/mL, (14.39±0.36) pg/mL and (16.88±0.37) pg/mL. In cryptorchidism groups, on days 4, 7 and 14 after surgery, the testicular volumes were (115.64±3.91) mm3, (69.51±14.97) mm3 and (44.86±5.56) mm3, respectively. Spermatogenic cells were disorganized, seminiferous tubules were disrupted, peritubular cells shrank, bent and fractured. The mRNA expression levels of AR were 0.76±0.06, 0.53±0.04, and 0.29±0.02; GDNF mRNA 0.72±0.05, 0.42±0.02 and 0.30±0.03. The protein expression levels of AR were 0.54±0.02, 0.98±0.04 and 0.31±0.01; GDNF protein (8.50±0.34) pg/mL, (17.44±0.32) pg/mL and (6.83±0.34) pg/mL. Statistically significant differences (P < 0.05) were found in 7-day and 14-day testicular volumes between control and cryptorchidism groups but not in the 4-day testicular volume (P > 0.05). Testicular volumes, AR and GDNF mRNA and protein expression in control groups had no statistically significant difference (P > 0.05), while those in cryptorchidism groups showed a trend of gradual decline in the amount and the differences between groups were statistically significant (P < 0.05). ConclusionsIn surgery-induced cryptorchidism mice, after the induction, the expression of AR and GDNF in TPCs showed a gradual decrease over time. AR and GDNF play a major role in mediating the TPCs damage in cryptorchidism. This study provides a theoretical basis for mechanism researches of cryptorchidism-induced spermatogenesis dysfunction.

17.
Int J Mol Sci ; 24(23)2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38069144

RESUMEN

Our previous studies showed that in patients with brain diseases, neurotrophic factors in lacrimal fluid (LF) may change more prominently than in blood serum (BS). Since glial cell line-derived neurotrophic factor (GDNF) is involved in the control of neuronal networks in an epileptic brain, we aimed to assess the GDNF levels in LF and BS as well as the BDNF and the hypothalamic-pituitary-adrenocortical and inflammation indices in BS of patients with focal epilepsy (FE) and epilepsy and comorbid depression (FE + MDD) and to compare them with those of patients with major depressive disorder (MDD) and healthy controls (HC). GDNF levels in BS were similar in patients and HC and higher in FE taking valproates. GDNF levels in LF were significantly lower in all patient groups compared to controls, and independent of drugs used. GDNF concentrations in LF and BS positively correlated in HC, but not in patient groups. BDNF level was lower in BS of patients compared with HC and higher in FE + MDD taking valproates. A reduction in the GDNF level in LF might be an important biomarker of FE. Logistic regression models demonstrated that the probability of FE can be evaluated using GDNF in LF and BDNF in BS; that of MDD using GDNF in LF and cortisol and TNF-α in BS; and that of epilepsy with MDD using GDNF in LF and TNF-α and BDNF in BS.


Asunto(s)
Trastorno Depresivo Mayor , Epilepsias Parciales , Epilepsia , Humanos , Biomarcadores , Factor Neurotrófico Derivado del Encéfalo , Depresión , Trastorno Depresivo Mayor/complicaciones , Epilepsia/complicaciones , Factor Neurotrófico Derivado de la Línea Celular Glial , Factor de Necrosis Tumoral alfa
18.
Cell Transplant ; 32: 9636897231213309, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38018498

RESUMEN

This study was designed to provide evidence of the neuroprotective of human adipose-derived mesenchymal stem cells (hADSCs) in oxygen-induced retinopathy (OIR). In vivo, hADSCs were intravitreally injected into OIR mice. Various assessments, including HE (histological evaluation), TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, electroretinogram (ERG) analysis, and retinal flat-mount examination, were performed separately at postnatal days 15 (P15) and 17 (P17) to evaluate neurological damage and functional changes. Western blot analysis of ciliary neurotrophic factor (CNTF), glial cell line-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF) was conducted at P17 to elucidate the neuroprotective mechanism. The P17 OIR group exhibited a significant increase in vascular endothelial cell nuclei and neovascularization that breached the ILM (inner limiting membrane) to the P17 control group. In addition, the retinal nonperfusion areas in the P17 OIR group and the number of apoptotic retinal cells in the P15 OIR group were significantly higher than in the corresponding hADSCs treatment group and control group. There was no significant thickness change in the inner nuclear layer (INL) but the outer nuclear layer (ONL) in the P17 OIR treatment group compared with the P17 OIR group. The cell density in the INL and ONL at P17 in the hADSCs treatment group was not significantly different from the OIR group. The amplitude of a-wave and b-wave in scotopic ERG analysis for the P17 OIR group was significantly lower than in the P17 hADSCs treatment group and the P17 control group. Furthermore, the latency of the a-wave and b-wave in the P17 OIR group was significantly longer than in the P17 hADSCs treatment group and the P17 control group. In addition, the expression levels of CNTF and BDNF in the P17 OIR group were statistically higher than those in the P17 control group, whereas the expression of GDNF was statistically lower in the P17 OIR group, compared with the P17 control group. The expression of CNTF and GDNF in the P17 hADSCs treatment group was statistically higher than in the P17 OIR group. However, the expression of BDNF in the P17 hADSCs treatment group was statistically lower than in the P17 OIR group. This study provides evidence for the neuroprotective effects of hADSCs in OIR.


Asunto(s)
Células Madre Mesenquimatosas , Fármacos Neuroprotectores , Enfermedades de la Retina , Neovascularización Retiniana , Humanos , Animales , Ratones , Oxígeno , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Ciliar , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/terapia , Células Madre Mesenquimatosas/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Neovascularización Retiniana/metabolismo
19.
Int J Mol Sci ; 24(21)2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37958856

RESUMEN

Glial-cell-line-derived neurotrophic factor (GDNF) family ligands (GFLs) contribute to the sensitization of primary afferents and are involved in the pathogenesis of inflammatory pain. The purpose of this preliminary study was to examine the expression of other GFLs (neurturin (NRTN), artemin (ARTN), persephin (PSPN)) and receptors in human IVD cells and tissues exhibiting early and advanced stages of degeneration. Human IVD cells were cultured as a monolayer after isolation from the nucleus pulposus (NP) and anulus fibrosus (AF) tissues. The mRNA expression of NRTN, ARTN, PSPN, and their receptors (GFRA2-GFRA4) was quantified using real-time PCR. Protein expression was evaluated using immunohistochemistry and Western blotting. The expression of NRTN, ARTN, PSPN, and their co-receptors (GFRA2-GFRA4) was identified in human IVD cells at both mRNA and protein levels. A trend was noted wherein the mRNA expression of ARTN, PSPN, and GFRA2 was upregulated by IL-1ß treatment in a dose-dependent manner. The percentages of immunopositive cells in the advanced degenerate stage of ARTN, PSPN, and GFRA2 were significantly higher than those in the early degenerate stage. Their expression was enhanced in advanced tissue degeneration, which suggests that GFLs (ARTN and PSPN) may be involved in the pathogenesis of discogenic pain.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Disco Intervertebral , Humanos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Disco Intervertebral/metabolismo , Factor de Crecimiento Transformador beta , ARN Mensajero/genética , Dolor , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-37754589

RESUMEN

Type 2 diabetes mellitus (T2DM) is associated with functional deterioration of the salivary gland and dental pulp, related to oxidative stress. The aim was to integrate experimental and bioinformatic findings to analyze the cellular mechanism of melatonin (MEL) action in the human parotid gland and dental pulp in diabetes. Human parotid gland tissue was obtained from 16 non-diabetic and 16 diabetic participants, as well as human dental pulp from 15 non-diabetic and 15 diabetic participants. In human non-diabetic and diabetic parotid gland cells (hPGCs) as well as in dental pulp cells (hDPCs), cultured in hyper- and normoglycemic conditions, glial cell line-derived neurotrophic factor (GDNF), MEL, inducible nitric oxide synthase (iNOS) protein expression, and superoxide dismutase (SOD) activity were measured by enzyme-linked immunosorbent assay (ELISA) and spectrophotometrically. Bioinformatic analysis was performed using ShinyGO (v.0.75) application. Diabetic participants had increased GDNF and decreased MEL in parotid (p < 0.01) and dental pulp (p < 0.05) tissues, associated with increased iNOS and SOD activity. Normoglycemic hDPCs and non-diabetic hPGCs treated with 0.1 mM MEL had increased GDNF (p < 0.05), while hyperglycemic hDPCs treated with 1 mM MEL showed a decrease in up-regulated GDNF (p < 0.05). Enrichment analyses showed interference with stress and ATF/CREB signaling. MEL induced the stress-protective mechanism in hyperglycemic hDPCs and diabetic hPGCs, suggesting MEL could be beneficial for diabetes-associated disturbances in oral tissues.


Asunto(s)
Diabetes Mellitus Tipo 2 , Melatonina , Humanos , Glándula Parótida , Factor Neurotrófico Derivado de la Línea Celular Glial , Melatonina/farmacología , Pulpa Dental , Biología Computacional , Superóxido Dismutasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA