Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurol Sci ; 45(10): 4699-4710, 2024 Oct.
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.


Asunto(s)
Terapia Genética , Enfermedad de Parkinson , Humanos , Terapia Genética/métodos , Enfermedad de Parkinson/terapia , Enfermedad de Parkinson/genética , Animales , Ensayos Clínicos como Asunto/métodos
2.
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
3.
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
4.
Int J Psychiatry Clin Pract ; 27(2): 118-126, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35980319

RESUMEN

OBJECTIVES: We investigated the differences in serum brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) levels and clinical symptoms with first-episode depression at different ages. METHODS: Ninety patients (15-60 years old) diagnosed with first-episode depression were enrolled as the study group, and they were divided into early-onset, adult and late-onset groups. The age-matched control groups were healthy volunteers. Serum BDNF and GDNF concentrations were determined by enzyme-linked immunosorbent assay (ELISA). GraphPad Prism 9 was used for t tests, one-way ANOVAs, chi-square tests, and correlation analyses. p < 0.05 indicated significant differences. RESULTS: Serum BDNF and GDNF levels were lower in the whole study group and the three subgroups than in the healthy groups. Illness severity, anxiety and education were higher in the early-onset than late-onset patients. Serum BDNF levels were lower in the adult than late-onset patients. Serum BDNF levels were negatively correlated with patient CGI-SI scores. After the LSD test for multiple comparisons, the results were also significant. CONCLUSIONS: Low serum BDNF and GDNF levels may be involved in the pathophysiology of first-episode depression, and there were differences in serum BDNF levels at different ages, verifying that serum BDNF and GDNF could serve as potential biomarkers of depression. KEY POINTSDepression is often conceptualised as a systemic illness with different biological mechanisms, but satisfactory explanations have not been provided thus far.The aim of our study was to investigate differences in serum BDNF and GDNF levels and their relationships with clinical symptoms in patients with first-episode depression at different ages.The potential of the neurotrophic factor hypothesis to advance the diagnosis and treatment of depression will be a very exciting new strategy for future research.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Factor Neurotrófico Derivado de la Línea Celular Glial , Adolescente , Adulto , Humanos , Persona de Mediana Edad , Adulto Joven , Ansiedad , Depresión
5.
J Physiol ; 599(6): 1783-1798, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33476055

RESUMEN

KEY POINTS: Nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF) are essential for neuronal development and survival in embryo. However, after birth they play pivotal roles in the generation of hyperalgesia in many painful conditions. Both factors are believed to act on different groups of primary afferents, but interaction between them has not yet been studied. Here we show a synergism between the two factors. Intramuscular injection of a mixture of both factors at a low concentration, each of which alone had no effect, induced a significant muscular mechanical hyperalgesia in rats. We show that synergism occurs in the primary afferent neurons and find that about 25% primary afferents innervating the muscle express both TrkA (NGF receptor) and GFRα1 (GDNF receptor). We show by pharmacological means that afferent neurons with TrkA and GFRα1 express both TRPV1 and ASICs. Our data establish a basis for synergism between NGF and GDNF. In some inflammatory conditions both nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF) are upregulated and play pivotal roles in inducing hyperalgesia. However, their interaction has not been studied. We examined whether and where interaction between both neurotrophic factors occurs in SD rats. Intramuscular injection to gastrocnemius muscle (GC) of a mixture of NGF (0.1 µm) and GDNF (0.008 µm), which alone had no effect, induced a significant mechanical hyperalgesia (F(6,30)  = 13.62, P = 0.0001), demonstrating synergism between the two factors. Phosphorylated extracellular signal-regulated kinase (pERK) immunoreactivity in dorsal root ganglia (DRGs) induced by compression of GC increased after injection of the mixture (P = 0.028, compared with PBS); thus the interaction of NGF and GDNF could occur at the primary afferent level. An in situ hybridization study (n = 4) demonstrated that 23.7-29.2% of GC-innervating DRG neurons coexpressed TrkA (NGF receptor) and GFRα1 (GDNF receptor). The cell size of the coexpressing GC DRG neurons showed no skewing towards the small size range but was distributed widely from the small to the large size ranges. Therefore, some of the coexpressing neurons with thin axons are thought to contribute to this mechanical hyperalgesia. The hyperalgesia was reversed by both amiloride (F(1,13)  = 5.056, P = 0.0425, compared with PBS) and capsazepine (F(1,10)  = 8.402, P = 0.0159, compared with DMSO), suggesting that the primary afferents sensitized by the mixture express both TRPV1 and ASICs. These results showed a basis of synergism between NGF and GDNF.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Factor de Crecimiento Nervioso , Animales , Ganglios Espinales , Hiperalgesia , Neuronas Aferentes , Ratas , Ratas Sprague-Dawley
6.
Neurol Sci ; 42(7): 2865-2872, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33215334

RESUMEN

INTRODUCTION: A growing body of evidence that glial cell line-derived neurotrophic factor (GDNF) levels are probably involved in pathogenesis and disease course of Alzheimer's disease (AD) suggested that its blood levels could potentially be used as a biomarker of AD. The aim of this study was to compare serum GDNF levels in patients with AD and age-matched controls. METHODS: Serum concentrations of GDNF were compared in 25 AD patients and 25 healthy volunteers using a double-antibody sandwich enzyme-linked immunosorbent assay (ELISA). Severity of the disease in AD patients was assessed using Functional Assessment Staging (FAST). Cognitive assessment of the patients was done using the Mini-Mental State Examination (MMSE). RESULTS: Mean GDNF levels were found to be 2.45 ± 0.93 ng/ml in AD patients and 4.61 ± 3.39 ng/ml in age-matched controls. There was a statistically significant difference in GDNF serum levels in patients with AD compared to age-matched controls (p = 0.001). Moreover, GDNF serum levels were significantly correlated with disease severity (p < 0.001) and cognitive impairment (p < 0.001). CONCLUSION: This study showed that serum levels of GDNF are significantly decreased in AD patients in comparison with age-matched controls, thus suggesting a potential role of GDNF as a disease biomarker. However, a comprehensive study of changes in serum levels of multiple neurotrophic factors reflective of different neurobiological pathways in large-scale population studies is recommended.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Biomarcadores , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos
7.
Mov Disord ; 35(2): 245-255, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31840869

RESUMEN

BACKGROUND: Motor symptoms of Parkinson's disease (PD) are caused by degeneration and progressive loss of nigrostriatal dopamine neurons. Currently, no cure for this disease is available. Existing drugs alleviate PD symptoms but fail to halt neurodegeneration. Glial cell line-derived neurotrophic factor (GDNF) is able to protect and repair dopamine neurons in vitro and in animal models of PD, but the clinical use of GDNF is complicated by its pharmacokinetic properties. The present study aimed to evaluate the neuronal effects of a blood-brain-barrier penetrating small molecule GDNF receptor Rearranged in Transfection agonist, BT13, in the dopamine system. METHODS: We characterized the ability of BT13 to activate RET in immortalized cells, to support the survival of cultured dopamine neurons, to protect cultured dopamine neurons against neurotoxin-induced cell death, to activate intracellular signaling pathways both in vitro and in vivo, and to regulate dopamine release in the mouse striatum as well as BT13's distribution in the brain. RESULTS: BT13 potently activates RET and downstream signaling cascades such as Extracellular Signal Regulated Kinase and AKT in immortalized cells. It supports the survival of cultured dopamine neurons from wild-type but not from RET-knockout mice. BT13 protects cultured dopamine neurons from 6-Hydroxydopamine (6-OHDA) and 1-methyl-4-phenylpyridinium (MPP+ )-induced cell death only if they express RET. In addition, BT13 is absorbed in the brain, activates intracellular signaling cascades in dopamine neurons both in vitro and in vivo, and also stimulates the release of dopamine in the mouse striatum. CONCLUSION: The GDNF receptor RET agonist BT13 demonstrates the potential for further development of novel disease-modifying treatments against PD. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Ratones , Oxidopamina/farmacología , Enfermedad de Parkinson Secundaria/inducido químicamente , Sustancia Negra/efectos de los fármacos
8.
Int J Mol Sci ; 21(19)2020 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-32993133

RESUMEN

Rearranged during transfection (RET) is the tyrosine kinase receptor that under normal circumstances interacts with ligand at the cell surface and mediates various essential roles in a variety of cellular processes such as proliferation, differentiation, survival, migration, and metabolism. RET plays a pivotal role in the development of both peripheral and central nervous systems. RET is expressed from early stages of embryogenesis and remains expressed throughout all life stages. Mutations either activating or inhibiting RET result in several aggressive diseases, namely cancer and Hirschsprung disease. However, the physiological ligand-dependent activation of RET receptor is important for the survival and maintenance of several neuronal populations, appetite, and weight gain control, thus providing an opportunity for the development of disease-modifying therapeutics against neurodegeneration and obesity. In this review, we describe the structure of RET, its signaling, and its role in both normal conditions as well as in several disorders. We highlight the differences in the signaling and outcomes of constitutive and ligand-induced RET activation. Finally, we review the data on recently developed small molecular weight RET agonists and their potential for the treatment of various diseases.


Asunto(s)
Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Obesidad/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Humanos , Mutación , Neoplasias/genética , Neoplasias/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Obesidad/genética , Obesidad/patología , Proteínas Proto-Oncogénicas c-ret/análisis , Proteínas Proto-Oncogénicas c-ret/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
9.
Int J Mol Sci ; 21(18)2020 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-32911810

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) are able to promote the survival of multiple neuronal populations in the body and, therefore, hold considerable promise for disease-modifying treatments of diseases and conditions caused by neurodegeneration. Available data reveal the potential of GFLs for the therapy of Parkinson's disease, neuropathic pain and diseases caused by retinal degeneration but, also, amyotrophic lateral sclerosis and, possibly, Alzheimer's disease. Despite promising data collected in preclinical models, clinical translation of GFLs is yet to be conducted. The main reasons for the limited success of GFLs clinical development are the poor pharmacological characteristics of GFL proteins, such as the inability of GFLs to cross tissue barriers, poor diffusion in tissues, biphasic dose-response and activation of several receptors in the organism in different cell types, along with ethical limitations on patients' selection in clinical trials. The development of small molecules selectively targeting particular GFL receptors with improved pharmacokinetic properties can overcome many of the difficulties and limitations associated with the clinical use of GFL proteins. The current review lists several strategies to target the GFL receptor complex with drug-like molecules, discusses their advantages, provides an overview of available chemical scaffolds and peptides able to activate GFL receptors and describes the effects of these molecules in cultured cells and animal models.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Péptidos/farmacología , Animales , Células Cultivadas , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Ligandos , Neuralgia/metabolismo , Neuritas/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/efectos de los fármacos , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología
10.
Semin Cancer Biol ; 53: 212-222, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30059726

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is a potent survival factor, and a member of the transforming growth factor ß (TGF-ß) superfamily acting on different neuronal activities. GDNF was originally identified as a neurotrophic factor crucially involved in the survival of dopaminergic neurons of the nigrostriatal pathway and is currently an established therapeutic target in Parkinson's disease. However, GDNF was later reported to be highly expressed in gliomas, especially in glioblastomas, and was demonstrated as a potent proliferation factor involved in the development and migration of gliomas. Here, we review our current understanding and progress made so far by researchers in our laboratories with references to relevant articles to support our discoveries. We present past and recent discoveries on the mechanisms involved in the protection of neurons by GDNF and examine its emerging roles in gliomas, as well as reasons for the abnormal expression in Glioblastoma Multiforme (GBM). Collectively, our work establishes a paradigm by which the ability of GDNF to protect dopaminergic neurons from degradation and its corresponding effects on glioma cells points to an underlying biological vulnerability in the effects of GDNF in the normal brain which can be subverted for use by cancer cells. Hence, presenting novel opportunities for intervention in glioma therapies.


Asunto(s)
Neoplasias Encefálicas/genética , Encéfalo/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Glioma/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Movimiento Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neuronas Dopaminérgicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glioma/metabolismo , Glioma/terapia , Humanos
11.
J Cell Physiol ; 234(5): 7395-7410, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30370540

RESUMEN

We have previously shown that knockout of fibroblast growth factor-2 (FGF-2) and potential compensatory effects of other growth factors result in amelioration of disease symptoms in a transgenic mouse model of amyotrophic lateral sclerosis (ALS). ALS is a rapidly progressive neurological disorder leading to degeneration of cortical, brain stem, and spinal motor neurons followed by subsequent denervation and muscle wasting. Mutations in the superoxide dismutase 1 (SOD1) gene are responsible for approximately 20% of familial ALS cases and SOD1 mutant mice still are among the models best mimicking clinical and neuropathological characteristics of ALS. The aim of the present study was a thorough characterization of FGF-2 and other growth factors and signaling effectors in vivo in the SOD1G93A mouse model. We observed tissue-specific opposing gene regulation of FGF-2 and overall dysregulation of other growth factors, which in the gastrocnemius muscle was associated with reduced downstream extracellular-signal-regulated kinases (ERK) and protein kinase B (AKT) activation. To further investigate whether the effects of FGF-2 on motor neuron death are mediated by glial cells, astrocytes lacking FGF-2 were cocultured together with mutant SOD1 G93A motor neurons. FGF-2 had an impact on motor neuron maturation indicating that astrocytic FGF-2 affects motor neurons at a developmental stage. Moreover, neuronal gene expression patterns showed FGF-2- and SOD1 G93A -dependent changes in ciliary neurotrophic factor, glial-cell-line-derived neurotrophic factor, and ERK2, implying a potential involvement in ALS pathogenesis before the onset of clinical symptoms.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Astrocitos/enzimología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neuronas Motoras/enzimología , Músculo Esquelético/enzimología , Superóxido Dismutasa-1/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/patología , Muerte Celular , Células Cultivadas , Modelos Animales de Enfermedad , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/deficiencia , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Motoras/patología , Mutación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Superóxido Dismutasa-1/genética
12.
Cell Physiol Biochem ; 47(1): 129-139, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29763918

RESUMEN

BACKGROUND/AIMS: Transplantation of bone-marrow-derived mesenchymal stem cells (MSCs) promotes neural cell regeneration after spinal cord injury (SCI). Recently, we showed that suppression of microRNA-383 (miR-383) in MSCs increased the protein levels of glial cell line derived neurotrophic factor (GDNF), resulting in improved therapeutic effects on SCI. However, the overall effects of miR-383 suppression in MSCs on SCI therapy were not determined yet. Here, we addressed this question. METHODS: We used bioinformatics tools to predict all miR-383-targeting genes, confirmed the functional bindings in a dual luciferase reporter assay. The effects of alteration of candidate genes in MSCs on cell proliferation were analyzed by MTT assay and by Western blotting for PCNA. The effects on angiogenesis were assessed by HUVEC assay. The effects on SCI in vivo were analyzed by transplantation of the modified MSCs into nude rats that underwent SCI. RESULTS: Suppression of miR-383 in MSCs not only upregulated GDNF protein, but also increased vascular endothelial growth factor A (VEGF-A) and cyclin-dependent kinase 19 (CDK19), two other miR-383 targets. MiR-383-suppression-induced increases in CDK19 resulted in a slight but significant increase in MSC proliferation, while miR-383-suppression-induced increases in VEGF-A resulted in a slight but significant increase in MSC-mediated angiogenesis. CONCLUSIONS: Upregulation of CDK19 and VEGF-A by miR-383 suppression in MSCs further improve the therapeutic potential of MSCs in treating SCI in rats.


Asunto(s)
Regulación de la Expresión Génica , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Traumatismos de la Médula Espinal/terapia , Adulto , Animales , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Ratas , Ratas Desnudas , Traumatismos de la Médula Espinal/genética , Factor A de Crecimiento Endotelial Vascular/genética
13.
J Reprod Dev ; 64(3): 267-275, 2018 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-29657241

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) and fibroblast growth factor 2 (FGF2) are bona fide self-renewal factors for spermatogonial stem cells (SSCs). Although GDNF is indispensable for the maintenance of SSCs, the role of FGF2 in the testis remains to be elucidated. To clarify this, the expression dynamics and regulatory mechanisms of Fgf2 and Gdnf in the mouse testes were analyzed. It is well known that Sertoli cells express Gdnf, and its receptor is expressed in a subset of undifferentiated spermatogonia, including SSCs. However, we found that Fgf2 was mainly expressed in the germ cells and its receptors were expressed not only in the cultured spermatogonial cell line, but also in testicular somatic cells. Aging, hypophysectomy, retinoic acid treatment, and testicular injury induced distinct Fgf2 and Gdnf expression dynamics, suggesting a difference in the expression mechanism of Fgf2 and Gdnf in the testis. Such differences might cause a dynamic fluctuation of Gdnf/Fgf2 ratio depending on the intrinsic/extrinsic cues. Considering that FGF2-cultured spermatogonia exhibit more differentiated phenotype than those cultured with GDNF, FGF2 might play a role distinct from that of GDNF in the testis, despite the fact that both factors are self-renewal factor for SSC in vitro.


Asunto(s)
Células Madre Germinales Adultas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Espermatogonias/metabolismo , Testículo/metabolismo , Células Madre Germinales Adultas/citología , Animales , Diferenciación Celular , Proliferación Celular , Masculino , Ratones , Espermatogonias/citología , Testículo/citología
14.
Pak J Med Sci ; 34(4): 844-848, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30190739

RESUMEN

OBJECTIVES: Tic disorders are associated with neurodevelopmental origin, changes in dopaminergic neurons, and the formation of immunoreactivity, it is thought that neurotrophic factors may be crucial in the emergence of tic disorders. In this study, we targeted to explore role of neurotrophic factors in tic disorders. The aim of this study was to investigate serum Glial Cell Line-Derived Neurotrophic Factor (GDNF) and Nerve Growth Factor (NGF) levels in patients with tic disorder and healthy controls. METHODS: Thirty-four children, constituted the case group, were diagnosed with tic disorder. The control group included 34 healthy children. Development and Well-Being Assessment (DAWBA) (structured interview) and Yale Global Tic Severity Rating Scale (YGTSRS) was applied to the patients. NGF and GDNF levels were measured with ELISA kit. RESULTS: In case group, serum NGF and GDNF levels were found to be significantly higher in females than males (p = 0.042, p = 0.031). It was determined that serum NGF and GDNF levels were correlated with each other (r = 0.803, p <0.001) and there were no correlations between other parameters. There was no significant difference in NGF and GDNF in patients with tic disorder, compared to healthy controls. CONCLUSIONS: The absence of this relationship does not exclude the hypothesis that neurotrophic factors may play a role in the etiopathogenesis of tic disorders.

15.
Cell Physiol Biochem ; 41(4): 1435-1444, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28365701

RESUMEN

BACKGROUND/AIMS: Transplantation of bone-marrow-derived mesenchymal stem cells (MSCs) has been used to treat spinal cord injury (SCI) to enhance tissue repair and neural cell regeneration. Glial cell line derived neurotrophic factor (GDNF) is an identified neural growth and survival factor. Here, we examined whether modification of GDNF levels in MSCs may further increase the potential of MSCs in promoting neural cell regeneration and subsequently the therapeutic outcome. METHODS: We examined the mRNA and protein levels of GDNF in human MSCs by RT-qPCR and Western blot, respectively. Bioinformatics analyses were done to predict microRNAs (miRNAs) that target GDNF in MSCs. The functional binding of miRNAs to GDNF mRNA was examined by a dual luciferase reporter assay. MSCs were transduced with adeno-associated virus (AAV) carrying null or antisense for miR-383 (as-miR-383), which were transplanted into nude rats that underwent SCI. The intact tissue, cavity volume, and recovery of locomotor activity were assessed. RESULTS: MSCs expressed very low GDNF protein, but surprisingly high levels of GDNF mRNA. Bioinformatics analyses showed that miR-383 inhibited protein translation of GDNF, through binding to the 3'-UTR of the GDNF mRNA. MSCs transduced with AAV-as-miR-383 further increased the intact tissue percentage, decreased cavity volume, and enhanced the recovery of locomotor activity in nude rats that underwent SCI, compared to MSCs. CONCLUSIONS: Suppression of miR-383 may increase the therapeutic potential of human bone-marrow-derived MSCs in treating SCI via augmentation of GDNF protein levels.


Asunto(s)
Células de la Médula Ósea/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , MicroARNs/biosíntesis , Traumatismos de la Médula Espinal , Regiones no Traducidas 3' , Adulto , Animales , Modelos Animales de Enfermedad , Células HEK293 , Xenoinjertos , Humanos , Masculino , Ratas , Ratas Desnudas , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia
16.
Nanomedicine ; 13(7): 2209-2217, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28666950

RESUMEN

This study reports proof of concept for administering compacted DNA nanoparticles (DNPs) intracerebrally as a means to protect against neurotoxin-induced neurodegeneration of dopamine (DA) neurons. In this study we used DNPs that encoded for human glial cell line-derived neurotrophic factor (hGDNF); GDNF is a potent neurotrophic factor for DA neurons. Intracerebral injections of DNPs into the striatum and/or substantia nigra were performed 1 week before treatment with a neurotoxin. We observed that the number of surviving DA cells, the density of DA fiber terminals and recovery of motor function were greater in animals injected with GDNF-encoding DNPs than in control animals receiving DNPs encoding for an inert reporter gene. The results of these studies are one of the first to demonstrate that a non-viral, synthetic nanoparticle can be used to deliver therapeutic genes to cells in the brain as a means to protect cells against neurotoxin-induced neurodegeneration.


Asunto(s)
ADN/administración & dosificación , ADN/genética , Neuronas Dopaminérgicas/citología , Técnicas de Transferencia de Gen , Terapia Genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Neuroprotección , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Terapia Genética/métodos , Humanos , Masculino , Nanopartículas/administración & dosificación , Ratas Sprague-Dawley
17.
Mol Biol (Mosk) ; 50(4): 589-598, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-27668599

RESUMEN

Four glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) have been characterized: GDNF, neurturin (NRTN), artemin (ARTN) and persephin (PSPN). These proteins support and restore multiple neuronal populations such as dopaminergic, sensory, motor, hippocampal, basal forebrain, enteric, sympathetic and parasympathetic neurons. Therefore, GFLs attracted significant attention as a potential cure for the diseases caused by neuronal injury and degeneration. Results of multiple experiments indicate that GFLs can alleviate behavioral symptoms and restore affected neurons in animal models of several neurological disorders including, among others, Parkinson's disease (PD). During the last decade, GDNF protein and NRTN gene therapy have been tested in several clinical trials in patients with PD. Although the results of phase I clinical trials were positive, phase II clinical trials failed to reach primary end-points. Poor pharmacokinetic properties of GFLs (inability to penetrate tissues barriers, high affinity for extracellular matrix, etc.) could contribute to the absence of clear clinical benefits of these proteins for the patients. The purpose of this paper was to review therapeutic potential of GFLs and discuss possibilities to overcome difficulties associated with pharmacokinetic properties and delivery of GFLs to target neurons.

18.
J Affect Disord ; 320: 605-609, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36183819

RESUMEN

BACKGROUND: The glial cell line-derived neurotrophic factor (GDNF) has an important role in neurons and is closely associated with psychiatric disorders. The development of bipolar disorder (BD) may differ between genders. Existing studies have shown that plasma GDNF levels are altered in patients with BD. In this study, we investigate whether the GDNF levels in patients with BD differ in terms of gender. METHODS: Participants were divided into the BD group (n = 76, with 26 males and 50 females) and healthy control (HC) group (n = 89, with 35 males and 54 females). Plasma GDNF levels were detected via multifactor assay. Clinical symptoms of patients with BD were collected and assessed using the Hamilton Depression-17 Inventory, Hamilton Anxiety-17 Inventory, Young's Mania Rating Scale, and Brief Psychiatric Rating Scale. RESULTS: The GDNF levels were significantly higher in all participants in the HC group (F = 4.262, p < 0.05) compared with those in the BD group. In the HC group, the males (t = 4.814, p < 0.001) presented significantly higher levels than the females. The plasma GDNF levels in males in the BD group (t = 3.022, p < 0.05) were significantly lower than those in males in the HC group. CONCLUSION: Differences in plasma GDNF levels are associated with the gender of patients with BD.


Asunto(s)
Trastorno Bipolar , Factor Neurotrófico Derivado de la Línea Celular Glial , Femenino , Humanos , Masculino , Factor Neurotrófico Derivado de la Línea Celular Glial/sangre , Caracteres Sexuales
19.
Ann Transl Med ; 11(2): 113, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36819542

RESUMEN

Background: Asthma is a common chronic inflammatory disease of the airway, but the mechanism is still not fully understood. This study aimed to investigate the effect of glial cell line-derived neurotrophic factor (GDNF) on asthma attacks. Methods: An asthmatic rat model was established. GDNF expression in the airway and brain was observed by immunohistochemistry (IHC), and the concentration of GDNF in bronchoalveolar lavage fluid (BALF) was detected by enzyme-linked immunosorbent assay (ELISA). After injection of GDNF and its antibody into the lateral ventricle of asthmatic rats, the pulmonary function was recorded, and the levels of interferon-γ (IFN-γ) and interleukin-4 (IL-4) in BALF were tested. Results: GDNF expressions were increased significantly in the lung tissues of asthmatic rats. In the central nervous system (CNS), GDNF-positive immunoreactive substances were observed in multiple brain regions, including the medial amygdala (MeA), paraventricular nucleus (PVN), cortex, and nucleus of solitary tract (NTS). After injection of GDNF into the lateral ventricles of asthmatic rats, the symptoms of asthma and airway inflammation were significantly aggravated, which could be improved by injection of GDNF antibody into the lateral ventricles. Conclusions: GDNF expression is increased in the lung and brain in asthmatic rats. During an asthma attack, the increased GDNF expressions in the rat brain remarkably aggravate the asthmatic symptoms.

20.
J Tissue Eng Regen Med ; 16(6): 515-529, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35278347

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) has neuroprotective effects and may be a promising candidate for regenerative strategies focusing on neurodegenerative diseases. As GDNF cannot cross the blood-brain barrier to potentially regenerate damaged brain areas, continuous in situ delivery with host cells is desired. Here, a non-viral Sleeping Beauty transposon was used to achieve continuous in vitro overexpression of GDNF in immune-privileged human adipose tissue-derived mesenchymal stromal cells (GDNF-tASCs). In addition, in vivo survival, tolerance, and effectiveness of transfected cells were tested in a very mild 6-hydroxydopamine (6-OHDA)-induced dopamine depletion rat model by means of intrastriatal injection on a sample basis up to 6 months after treatment. GDNF-tASCs showed vast in vitro gene overexpression up to 13 weeks post-transfection. In vivo, GDNF was detectable 4 days following transplantation, but no longer after 1 month, although adipose tissue-derived mesenchymal stromal cells (ASCs) could be visualized histologically even after 6 months. Despite successful long-term in vitro GDNF overexpression and its in vivo detection shortly after cell transplantation, the 6-OHDA model was too mild to enable sufficient evaluation of in vivo disease improvement. Still, in vivo immunocompatibility could be further examined. ASCs initially induced a pronounced microglial accumulation at transplantation site, particularly prominent in GDNF-tASCs. However, 6-OHDA-induced pro-inflammatory immune response was attenuated by ASCs, although delayed in the GDNF-tASCs group. To further test the therapeutic potential of the generated GDNF-overexpressing cells in a disease-related context, a follow-up study using a more appropriate 6-OHDA model is needed.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial , Células Madre Mesenquimatosas , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Modelos Animales de Enfermedad , Estudios de Seguimiento , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Oxidopamina/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA