Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Cells ; 13(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38667284

RESUMEN

This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.


Asunto(s)
Proliferación Celular , Cognición , Proteína Doblecortina , Ketamina , Neuronas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido Y , Receptores de Neuropéptido , Animales , Masculino , Ketamina/farmacología , Ketamina/administración & dosificación , Cognición/efectos de los fármacos , Ratas , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proliferación Celular/efectos de los fármacos , Receptor trkB/agonistas , Receptor trkB/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Neurogénesis/efectos de los fármacos
2.
Expert Opin Ther Targets ; 28(4): 309-322, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38626283

RESUMEN

BACKGROUND: Major Depressive Disorder (MDD) poses a significant challenge to global health, with current treatments often limited by efficacy and onset delays. This study explores the synergistic antidepressant-like effects of an NPY1R agonist and Ketamine, targeting their neurobiological interactions within the ventral hippocampus. RESEARCH DESIGN AND METHODS: Utilizing a preclinical model, this study administered Neuropeptide Y receptor 1 (NPY1R) agonist and Ketamine, both separately and in combination, through intracerebroventricular (icv) and intranasal (i.n.) routes. The Forced Swimming Test (FST) was employed to assess antidepressant-like activity, while in situ Proximity Ligation Assay and immunohistochemistry were used to examine NPY1R/TrkB heteroreceptor complexes and BDNF expression in the ventral dentate gyrus (DG), along with neurogenesis markers. RESULTS: The combined treatment significantly reduced immobility in the FST, indicative of enhanced antidepressant-like effects, correlated with increased formation of NPY1R/TrkB complex and brain-derived neurotrophic factor (BDNF) expression in the ventral DG. These molecular alterations were associated with increased neurogenesis. CONCLUSIONS: The coadministration of an NPY1R agonist and Ketamine in a rodent model demonstrated potentiated antidepressant responses through synergistic neurobiological pathways, including TrkB signaling and hippocampal neurogenesis. This indicates a novel therapeutic strategy for MDD, warranting further clinical investigation to fully understand its implications.


Asunto(s)
Antidepresivos , Factor Neurotrófico Derivado del Encéfalo , Trastorno Depresivo Mayor , Sinergismo Farmacológico , Hipocampo , Ketamina , Neurogénesis , Receptor trkB , Receptores de Neuropéptido Y , Transducción de Señal , Animales , Neurogénesis/efectos de los fármacos , Antidepresivos/farmacología , Antidepresivos/administración & dosificación , Masculino , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Transducción de Señal/efectos de los fármacos , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Ketamina/farmacología , Ketamina/administración & dosificación , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Receptor trkB/agonistas , Receptor trkB/metabolismo , Modelos Animales de Enfermedad , Ratas , Ratones , Ratas Sprague-Dawley , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Natación
3.
Aging (Albany NY) ; 14(18): 7568-7586, 2022 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-36170028

RESUMEN

Decreased BDNF and impaired TRKB signaling contribute to neurodegeneration in Alzheimer's disease (AD). We have shown previously that coumarin derivative LM-031 enhanced CREB/BDNF/BCL2 pathway. In this study we explored if LM-031 analogs LMDS-1 to -4 may act as TRKB agonists to protect SH-SY5Y cells against Aß toxicity. By docking computation for binding with TRKB using 7,8-DHF as a control, all four LMDS compounds displayed potential of binding to domain d5 of TRKB. In addition, all four LMDS compounds exhibited anti-aggregation and neuroprotective efficacy on SH-SY5Y cells with induced Aß-GFP expression. Knock-down of TRKB significantly attenuated TRKB downstream signaling and the neurite outgrowth-promoting effects of these LMDS compounds. Among them, LMDS-1 and -2 were further examined for TRKB signaling. Treatment of ERK inhibitor U0126 or PI3K inhibitor wortmannin decreased p-CREB, BDNF and BCL2 in Aß-GFP cells, implicating the neuroprotective effects are via activating TRKB downstream ERK, PI3K-AKT and CREB signaling. LMDS-1 and -2 are blood-brain barrier permeable as shown by parallel artificial membrane permeability assay. Our results demonstrate how LMDS-1 and -2 are likely to work as TRKB agonists to exert neuroprotection in Aß cells, which may shed light on the potential application in therapeutics of AD.


Asunto(s)
Enfermedad de Alzheimer , Glicoproteínas de Membrana/agonistas , Neuroblastoma , Fármacos Neuroprotectores , Receptor trkB/agonistas , Péptidos beta-Amiloides/toxicidad , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cumarinas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Membranas Artificiales , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Wortmanina
4.
Nat Commun ; 13(1): 914, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177657

RESUMEN

Premature ovarian failure (POF) is a leading cause of women's infertility without effective treatment. Here we show that intravenous injection of Ab4B19, an agonistic antibody for the BDNF receptor TrkB, penetrates into ovarian follicles, activates TrkB signaling, and promotes ovary development. In both natural aging and cyclophosphamide-induced POF models, treatment with Ab4B19 completely reverses the reduction of pre-antral and antral follicles, and normalizes gonadal hormone. Ab4B19 also attenuates gonadotoxicity and inhibits apoptosis in cyclophosphamide-induced POF ovaries. Further, treatment with Ab4B19, but not BDNF, restores the number and quality of oocytes and enhances fertility. In human, BDNF levels are high in granulosa cells and TrkB levels increase in oocytes as they mature. Moreover, BDNF expression is down-regulated in follicles of aged women, and Ab4B19 activates TrkB signaling in human ovary tissue ex vivo. These results identify TrkB as a potential target for POF with differentiated mechanisms, and confirms superiority of TrkB activating antibody over BDNF as therapeutic agents.


Asunto(s)
Fármacos para la Fertilidad Femenina/farmacología , Glicoproteínas de Membrana/agonistas , Ovario/efectos de los fármacos , Insuficiencia Ovárica Primaria/tratamiento farmacológico , Receptor trkB/agonistas , Adulto , Envejecimiento/fisiología , Animales , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/agonistas , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular Tumoral , Ciclofosfamida/toxicidad , Modelos Animales de Enfermedad , Femenino , Fertilidad/efectos de los fármacos , Fármacos para la Fertilidad Femenina/uso terapéutico , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Persona de Mediana Edad , Técnicas de Cultivo de Órganos , Ovario/patología , Ovario/fisiopatología , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/patología , Insuficiencia Ovárica Primaria/fisiopatología , Receptor trkB/metabolismo , Adulto Joven
5.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35165147

RESUMEN

Dravet syndrome (DS) is one of the most severe childhood epilepsies, characterized by intractable seizures and comorbidities including cognitive and social dysfunction and high premature mortality. DS is mainly caused by loss-of-function mutations in the Scn1a gene encoding Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, contributing to DS phenotypes. Effective pharmacological therapy that targets defective PV interneurons is not available. The known role of brain-derived neurotrophic factor (BDNF) in the development and maintenance of interneurons, together with our previous results showing improved PV interneuronal function and antiepileptogenic effects of a TrkB receptor agonist in a posttraumatic epilepsy model, led to the hypothesis that early treatment with a TrkB receptor agonist might prevent or reduce seizure activity in DS mice. To test this hypothesis, we treated DS mice with LM22A-4 (LM), a partial agonist at the BDNF TrkB receptor, for 7 d starting at postnatal day 13 (P13), before the onset of spontaneous seizures. Results from immunohistochemistry, Western blot, whole-cell patch-clamp recording, and in vivo seizure monitoring showed that LM treatment increased the number of perisomatic PV interneuronal synapses around cortical pyramidal cells in layer V, upregulated Nav1.1 in PV neurons, increased inhibitory synaptic transmission, and decreased seizures and the mortality rate in DS mice. The results suggest that early treatment with a partial TrkB receptor agonist may be a promising therapeutic approach to enhance PV interneuron function and reduce epileptogenesis and premature death in DS.


Asunto(s)
Benzamidas/uso terapéutico , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/mortalidad , Receptor trkB/agonistas , Receptor trkB/metabolismo , Convulsiones/etiología , Convulsiones/genética , Animales , Epilepsias Mioclónicas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Neocórtex/citología , Células Piramidales/metabolismo , Receptor trkB/genética
6.
Sci Rep ; 12(1): 1378, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082359

RESUMEN

Bupivacaine, a local anaesthetic, is widely applied in the epidural or subarachnoid space to clinically manage acute and chronic pain. However, the underlying mechanisms are complex and unclear. Glycine transporter 1 (GlyT1) in the spinal cord plays a critical role in various pathologic pain conditions. Therefore, we sought to determine whether bupivacaine exerts its analgesic effect by regulating GlyT1 expression and to determine the underlying mechanisms of regulation. Primary astrocytes prepared from the spinal cord of rats were treated with bupivacaine. The protein levels of GlyT1, brain-derived neurotrophic factor (BDNF) and phosphorylated adenosine 5'-monophosphate (AMP)-activated protein kinase α (p-AMPKα) were measured by western blotting or immunofluorescence. In addition, 7,8-dihydroxyflavone (7,8-DHF, BDNF receptor agonist) and AMPK shRNA were applied to verify the relationship between the regulation of GlyT1 by bupivacaine and the p-AMPKα/BDNF signalling pathway. After treatment with bupivacaine, GlyT1 expression was diminished in a concentration-dependent manner, while the expression of BDNF and p-AMPK was increased. Moreover, 7,8-DHF decreased GlyT1 expression, and AMPK knockdown suppressed the upregulation of BDNF expression by bupivacaine. Finally, we concluded that bupivacaine reduced GlyT1 expression in spinal astrocytes by activating the p-AMPKα/BDNF signalling pathway. These results provide a new mechanism for the analgesic effect of intrathecal bupivacaine in the treatment of acute and chronic pain.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Anestésicos Locales/farmacología , Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Bupivacaína/farmacología , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Flavonas/farmacología , Técnicas de Silenciamiento del Gen/métodos , Ratas , Ratas Sprague-Dawley , Receptor trkB/agonistas , Transducción de Señal/genética , Médula Espinal/citología , Transfección
7.
Neuropharmacology ; 204: 108899, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34838815

RESUMEN

Decreased energy metabolism and mitochondrial biogenesis defects are implicated in the pathogenesis of Alzheimer's disease (AD). In present study, mitochondriomics analysis revealed significant effects of R13, a prodrug of 7,8-dihydroxyflavone, on mitochondrial protein expression profile, including the proteins related to the biological processes: fatty acid beta-oxidation, fatty acid metabolic process, mitochondrial electron transport, and mitochondrial respiratory chain. Cluster analysis demonstrated that R13 promoted mitochondrial oxidative phosphorylation (OXPHOS). The functional analysis showed that R13 increased ATP levels and enhanced OXPHOS including complex Ⅰ, Ⅱ, Ⅲ and Ⅳ. R13 treatment increased mitochondrial biogenesis by regulating the levels of p-AMPKα, p-CREB, PGC-1α, NRF1 and TFAM as a consequence of activation of TrkB receptor in the 5 × FAD mice. Finally, R13 significantly reduced the levels of tau phosphorylation and Aß plaque. Our data suggest that R13 may be used for treating AD via enhancing mitochondrial biogenesis and metabolism.


Asunto(s)
Enfermedad de Alzheimer/etiología , Metabolismo Energético , Flavanonas/farmacología , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Biogénesis de Organelos , Profármacos/farmacología , Receptor trkB/agonistas , Receptor trkB/metabolismo , Transcriptoma/genética , Adenosina Trifosfato/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Animales , Transporte de Electrón , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Flavanonas/uso terapéutico , Masculino , Ratones Transgénicos , Terapia Molecular Dirigida , Oxidación-Reducción , Fosforilación/efectos de los fármacos , Profármacos/uso terapéutico , Proteínas tau/metabolismo
8.
PLoS One ; 16(12): e0260793, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855884

RESUMEN

Retinopathy of prematurity (ROP) is one of the main causes of blindness in children worldwide. Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), play critical protective roles in the development and function of neurons and vasculature. Lack of BDNF expression results in increased endothelial cell apoptosis and reduced endothelial cell-cell contact. Premature babies who develop ROP tend to have lower serum BDNF levels. BDNF expression is also significantly lower in mouse retinas following exposure to hyperoxia compared to those reared in room air. Specifically, BDNF promotes angiogenic tube formation of endothelial cells (EC), and it is considered an EC survival factor required for stabilization of intramyocardial vessels. We hypothesized that the activation of TrkB receptor protects retinal vasculature in the mice during oxygen-induced ischemic retinopathy (OIR), a model of ROP. To test this hypothesis, we treated neonatal mice with 7,8-dihydroxyflavone (DHF) (5 mg/kg body weight), a TrkB receptor agonist. We examined its potential protective effects on retinal vessel obliteration and neovascularization, two hallmarks of ROP and OIR. We found that retinas from DHF treated postnatal day 8 (P8) and P12 mice have similar levels of vessel obliteration as retinas from age-matched control mice subjected to OIR. Similarly, DHF showed no significant effect on mitigation of retinal neovascularization during OIR in P17 mice. Collectively, our studies demonstrate that the TrkB receptor agonist DHF provides no significant protective effects during OIR.


Asunto(s)
Flavonas/farmacología , Isquemia/patología , Neovascularización Patológica/patología , Oxígeno/toxicidad , Receptor trkB/agonistas , Neovascularización Retiniana/patología , Retinopatía de la Prematuridad/patología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Isquemia/inducido químicamente , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/metabolismo , Retinopatía de la Prematuridad/inducido químicamente , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/metabolismo
9.
J Neurotrauma ; 38(20): 2896-2906, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34353120

RESUMEN

Pressure waves from explosions or other traumatic events can damage the neurons of the eye and visual centers of the brain, leading to functional loss of vision. There are currently few treatments for such injuries that can be deployed rapidly to mitigate damage. Brain-derived neurotrophic factor (BDNF) and activation of its receptor tropomycin-related kinase B (TrkB) have neuroprotective effects in a number of degeneration models. Small molecule activators of TrkB, such as N-[2-(5-hydroxy-1H-indol-3-yl)ethyl]-2-oxopiperidine-3-carboxamide (HIOC), cross the blood-brain and blood-retina barriers after systemic administration. We characterize the effects of blast-induced ocular trauma on retinal and visual function. We show that systemic administration of HIOC, a potent small molecule activator of the BDNF/TrkB receptor, preserves visual function in mice exposed to ocular blast injury. The HIOC treatment for one week preserves visual function for at least four months. The HIOC treatment effectively protected vision when the initial dose was administered up to 3 h after blast, but not if the initial treatment was delayed for 24 h. We provide evidence that the therapeutic effect of HIOC is mediated by activation of BDNF/TrkB receptors. The results indicate that HIOC may be useful for managing ocular blast injury and other forms of traumatic optic neuropathy.


Asunto(s)
Traumatismos por Explosión/complicaciones , Ceguera/tratamiento farmacológico , Ceguera/etiología , Lesiones Oculares/complicaciones , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/etiología , Receptor trkB/agonistas , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematorretinal/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuroprotección , Fármacos Neuroprotectores/farmacología , Retina/fisiopatología , Tiempo de Tratamiento , Resultado del Tratamiento
10.
Cells ; 10(8)2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34440640

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder and results in severe neurodegeneration and progressive cognitive decline. Neurotrophins are growth factors involved in the development and survival of neurons, but also in underlying mechanisms for memory formation such as hippocampal long-term potentiation. Our aim was to identify small molecules with stimulatory effects on the signaling of two neurotrophins, the nerve growth factor (NGF) and the brain derived neurotrophic factor (BDNF). To identify molecules that could potentiate neurotrophin signaling, 25,000 molecules were screened, which led to the identification of the triazinetrione derivatives ACD855 (Ponazuril) and later on ACD856, as positive allosteric modulators of tropomyosin related kinase (Trk) receptors. ACD855 or ACD856 potentiated the cellular signaling of the neurotrophin receptors with EC50 values of 1.9 and 3.2 or 0.38 and 0.30 µM, respectively, for TrkA or TrkB. ACD855 increased acetylcholine levels in the hippocampus by 40% and facilitated long term potentiation in rat brain slices. The compounds acted as cognitive enhancers in a TrkB-dependent manner in several different behavioral models. Finally, the age-induced cognitive dysfunction in 18-month-old mice could be restored to the same level as found in 2-month-old mice after a single treatment of ACD856. We have identified a novel mechanism to modulate the activity of the Trk-receptors. The identification of the positive allosteric modulators of the Trk-receptors might have implications for the treatment of Alzheimer's diseases and other diseases characterized by cognitive impairment.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Nootrópicos/farmacología , Receptores de Factor de Crecimiento Nervioso/agonistas , Factores de Edad , Animales , Encéfalo/enzimología , Encéfalo/fisiopatología , Línea Celular Tumoral , Disfunción Cognitiva/enzimología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Glicoproteínas de Membrana , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Proteínas Tirosina Quinasas , Ratas Sprague-Dawley , Receptor trkA/agonistas , Receptor trkA/metabolismo , Receptor trkB/agonistas , Receptor trkB/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas , Triazinas/farmacología
11.
Neuropharmacology ; 197: 108737, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34343610

RESUMEN

Alzheimer's disease (AD) is the most common dementia, and no disease-modifying therapeutic agents are currently available. BDNF/TrkB signaling is impaired in AD and is associated with prominent delta-secretase (δ-secretase, also known as asparaginyl endopeptidase or legumain) activation, which simultaneously cleaves both APP and Tau and promotes Aß production and neurofibrillary tangles (NFT) pathologies. Here we show that the optimized δ-secretase inhibitor (#11a) or TrkB receptor agonist (CF3CN) robustly blocks δ-secretase activity separately, and their combination synergistically blunts δ-secretase, exhibiting promising therapeutic efficacy in 3xTg AD mouse model. The optimal δ-secretase inhibitor reveals demonstrable brain exposure and oral bioavailability, suppressing APP N585 and Tau N368 cleavage by δ-secretase. Strikingly, CF3CN treatment evidently escalates BDNF levels. Both #11a and CF3CN display strong in vivo PK/PD properties and ability to suppress δ-secretase activity in the brain. Orally administrated CF3CN strongly activates TrkB that triggers active Akt to phosphorylate δ-secretase T322, preventing its proteolytic activation and mitigating AD pathologies. #11a or CF3CN significantly diminishes AD pathogenesis and improves cognitive functions with the combination exhibiting the maximal effect. Thus, our data support that these derivatives are strong pharmaceutical candidates for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Cisteína Endopeptidasas/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Glicoproteínas de Membrana/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Receptor trkB/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Cognición/efectos de los fármacos , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Glicoproteínas de Membrana/agonistas , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacocinética , Ratas , Receptor trkB/agonistas , Proteínas tau/antagonistas & inhibidores
12.
Respir Physiol Neurobiol ; 294: 103743, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34273553

RESUMEN

Neuroplasticity is a fundamental property of the respiratory control system, enabling critical adaptations in breathing to meet the challenges, but little is known whether neonates express neuroplasticity similar to adults. We tested the hypothesis that, similar to adults, tyrosine receptor kinase B (TrkB) or adenosine A2a receptor activation in neonates are independently sufficient to elicit respiratory motor facilitation, and that co-induction of TrkB and A2a receptor-dependent plasticity undermines respiratory motor facilitation. TrkB receptor activation with 7,8-dihydroxyflavone (DHF) in neonatal brainstem-spinal cord preparations induced a long-lasting increase in respiratory motor output in 55 % of preparations, whereas adenosine A2a receptor activation with CGS21680 only sporadically induced respiratory motor plasticity. CGS21680 and DHF co-application prevented DHF-dependent respiratory motor facilitation, whereas co-application of MSX-3 (adenosine A2a receptor antagonist) and DHF more rapidly induced respiratory motor plasticity. Collectively, these data suggest that mechanisms underlying respiratory neuroplasticity may be only partially operational in early neonatal life, and that adenosine A2a receptor activation undermines TrkB-induced respiratory plasticity.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Flavonas/farmacología , Plasticidad Neuronal/fisiología , Receptor de Adenosina A2A/metabolismo , Receptor trkB/agonistas , Receptor trkB/metabolismo , Fenómenos Fisiológicos Respiratorios , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Animales Recién Nacidos , Tronco Encefálico/efectos de los fármacos , Modelos Animales de Enfermedad , Plasticidad Neuronal/efectos de los fármacos , Fenetilaminas/farmacología , Ratas , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos , Médula Espinal/efectos de los fármacos
13.
Behav Pharmacol ; 32(5): 404-412, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33883449

RESUMEN

Reduced brain-derived neurotrophic factor (BDNF) signalling has been implicated in schizophrenia endophenotypes, including deficits in prepulse inhibition (PPI). Maternal immune activation (MIA) is a widely used neurodevelopmental animal model for schizophrenia but it is unclear if BDNF and its receptor, tropomyosin receptor kinase B (TrkB), are involved in PPI regulation in this model. Pregnant Long Evans rats were treated with the viral mimetic, polyinosinic-polycytidylic acid (poly I:C; 4 mg/kg i.v.), and nine male offspring from these dams were compared in adulthood to 11 male Long Evans controls. Offspring underwent PPI testing following injection with the TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF) (10 mg/kg i.p.), with or without the dopamine receptor agonist, apomorphine (APO; 1 mg/kg s.c.), or the dopamine releasing drug, methamphetamine (METH; 2 mg/kg s.c.). Acute administration of APO and METH caused the expected significant reduction of PPI. Acute administration of 7,8-DHF did not alter PPI on its own; however, it significantly reversed the effect of APO on PPI in poly I:C rats, but not in controls. A similar trend was observed in combination with METH. Western blot analysis of frontal cortex revealed significantly increased levels of BDNF protein, but not TrkB or phosphorylated TrkB/TrkB levels, in poly I:C rats. These findings suggest that, selectively in MIA offspring, 7,8-DHF has the ability to reverse PPI deficits caused by dopaminergic stimulation. This effect could be associated with increased BDNF expression in the frontal cortex. These data suggest that targeting BDNF signalling may have therapeutic potential for the treatment of certain symptoms of schizophrenia.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Flavonas/farmacología , Inhibición Prepulso , Esquizofrenia , Animales , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Lóbulo Frontal/metabolismo , Masculino , Inhibición Prepulso/efectos de los fármacos , Inhibición Prepulso/fisiología , Ratas , Ratas Long-Evans , Receptor trkB/agonistas , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Dig Dis Sci ; 66(12): 4251-4262, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33528684

RESUMEN

BACKGROUND: Whether 7,8-dihydroxyflavone (7,8-DHF), a tyrosine kinase receptor B (TrkB) agonist, modulates colonic smooth muscle motility and/or alleviates constipation has not yet been studied. AIMS: Here, we aimed to determine how 7,8-DHF influences carbachol (CCh)-stimulated contraction of colonic strips and the in vivo effect of 7,8-DHF on constipation. METHODS: Muscle strips were isolated from rat colons for recording contractile tension and performing western blotting. Constipation was induced in rats with loperamide. RESULTS: Although it specifically activated TrkB, 7,8-DHF applied alone neither activated PLCγ1 in the colonic strips nor induced colonic strip contraction. However, 7,8-DHF enhanced CCh-stimulated PLCγ1 activation and strip contraction. The PLCγ1 antagonist U73122 suppressed both CCh-stimulated and 7,8-DHF-enhanced/CCh-stimulated contraction. While clarifying the underlying mechanism, we revealed that 7,8-DHF augmented muscarinic M3 receptor expression in the colonic strips. The M3-selective antagonist tarafenacin specifically inhibited the 7,8-DHF-enhanced/CCh-stimulated contraction of the colonic strips. Since 7,8-DHF increased Akt phosphorylation, and LY294002 (an antagonist of PI3K upstream of Akt) dramatically inhibited both 7,8-DHF-augmented M3 expression and 7,8-DHF-enhanced/CCh-stimulated contractions, we assumed that 7,8-DHF/TrkB/Akt was associated with the modulation of M3 expression in the colonic strips. ANA-12, a specific TrkB antagonist, not only inhibited TrkB activation by 7,8-DHF but also suppressed 7,8-DHF-enhanced cholinergic contraction, 7,8-DHF/CCh-mediated activation of PLCγ1/Akt, and M3 overexpression in colonic strips. In vivo 7,8-DHF, also by promoting intestinal motility and M3 expression, significantly alleviated loperamide-induced functional constipation in rats. CONCLUSIONS: Our results suggest that 7,8-DHF regulates colonic motility possibly via a TrkB/Akt/M3 pathway and may be applicable for alleviating constipation.


Asunto(s)
Colon/efectos de los fármacos , Estreñimiento/tratamiento farmacológico , Defecación/efectos de los fármacos , Flavonas/farmacología , Motilidad Gastrointestinal/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Animales , Colon/metabolismo , Colon/fisiopatología , Estreñimiento/inducido químicamente , Estreñimiento/fisiopatología , Modelos Animales de Enfermedad , Técnicas In Vitro , Loperamida , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Fosfolipasa C gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/metabolismo , Receptor trkB/agonistas , Receptor trkB/metabolismo , Transducción de Señal
15.
J Neurotrauma ; 38(9): 1338-1348, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-20597685

RESUMEN

After spinal cord injury (SCI) in mammals, there is only limited repair and, consequently, only moderate recovery. One mechanism frequently discussed to be involved in this recovery is plasticity (i.e., adaptations in spared neuronal circuitries). In the current study, we tested the effect of an intrathecal application of the TrkB agonist antibody, 29D7, on plasticity after cervical SCI in adult rats. Treatment with 29D7 for 4 weeks resulted in an ∼50% increase in collateral sprouting of severed corticospinal tract fibers above the lesion compared to the control group and enhanced branching in the gray matter rostral to the injury. Growth-associated protein 43 immunoreactivity in the spinal cord rostral to the level of the injury as well as contralateral to the lesion was also increased. These indications of enhanced plasticity by 29D7 were paralleled by improved performances of the mildly affected paw, as assessed by Montoya and tray reaching tasks. The reaching behaviors of the paw ipsilateral to the side of severe injury to the cortico- and rubrospinal tract were not altered by the treatment. The present study suggests that 29D7 may be a potential candidate to promote plasticity and functional recovery, especially after moderate SCI. Future studies confirming these results, along with a potential combinatory therapy including rehabilitative training, will be needed to evaluate the clinical value of such a treatment.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Médula Cervical/lesiones , Plasticidad Neuronal/efectos de los fármacos , Receptor trkB/agonistas , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Médula Cervical/efectos de los fármacos , Médula Cervical/fisiología , Femenino , Bombas de Infusión , Inyecciones Espinales , Plasticidad Neuronal/fisiología , Ratas , Ratas Endogámicas Lew , Traumatismos de la Médula Espinal/fisiopatología
16.
Psychopharmacology (Berl) ; 237(10): 3173-3190, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32748031

RESUMEN

RATIONALE: The signaling pathways of tropomyosin-related kinase B (TrkB) receptor play a pivotal role in axonal sprouting, proliferation of dendritic arbor, synaptic plasticity, and neuronal differentiation. The levels of BDNF and TrkB receptor were reduced in patients with Alzheimer's disease (AD). OBJECTIVES: The activation of TrkB signaling pathways is a potential strategy for AD therapies. We intended to identify potential TrkB agonists to activate the neuroprotective signaling to alleviate the pathological features of AD mice. RESULTS: Both of the Aß-deteriorated hippocampal primary neurons and mouse models were generated and showed AD characteristics. We first investigated 12 potential TrkB agonists with primary hippocampal neurons of mice. Both 7,8-DHF and LMDS-1 were identified to have better effect than the other compounds on dendritic arborization of the neurons and were further applied to the Aß-injected mouse model. The short-term cognitive behavior and pathology in the mice were improved by LMDS-1. Further investigation indicated that LMDS-1 activated the TrkB through phosphorylation at Y516 rather than Y816. In addition, the ERK but not CaMKII or Akt was activated in the mouse hippocampus with LMDS-1 administration. LMDS-1 treatment also upregulated CREB and BDNF while downregulated the GSK3ß active form and tau phosphorylation. CONCLUSIONS: This study suggests that LMDS-1 upregulates the expression of BDNF and ameliorates the early-phase phenotypes of the AD-like mice through the pTrkB (Y516)-ERK-CREB pathway. In addition, LMDS-1 has better effect than 7,8-DHF in ameliorating the behavioral and pathological features of AD-like mice.


Asunto(s)
Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/toxicidad , Factores de Crecimiento Nervioso/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/toxicidad , Receptor trkB/agonistas , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/farmacología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Distribución Aleatoria , Receptor trkB/metabolismo
17.
Theranostics ; 10(15): 6854-6874, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32550908

RESUMEN

Repeated failures of "Aß-lowering" therapies call for new targets and therapeutic approaches for Alzheimer's disease (AD). We propose to treat AD by halting neuronal death and repairing synapses using a BDNF-based therapy. To overcome the poor druggability of BDNF, we have developed an agonistic antibody AS86 to mimic the function of BDNF, and evaluate its therapeutic potential for AD. Method: Biochemical, electrophysiological and behavioral techniques were used to investigate the effects of AS86 in vitro and in vivo. Results: AS86 specifically activated the BDNF receptor TrkB and its downstream signaling, without affecting its other receptor p75NTR. It promoted neurite outgrowth, enhanced spine growth and prevented Aß-induced cell death in cultured neurons, and facilitated Long-Term Potentiation (LTP) in hippocampal slices. A single-dose tail-vein injection of AS86 activated TrkB signaling in the brain, with a half-life of 6 days in the blood and brain. Bi-weekly peripheral administration of AS86 rescued the deficits in object-recognition memory in the APP/PS1 mouse model. AS86 also reversed spatial memory deficits in the 11-month, but not 14-month old AD mouse model. Conclusion: These results demonstrate the potential of AS86 in AD therapy, suggesting that neuronal and/or synaptic repair as an alternative therapeutic strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo , Plasticidad Neuronal , Receptor trkB/agonistas , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Cricetinae , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Factores Inmunológicos/farmacología , Ratones , Ratones Transgénicos , Receptor trkB/metabolismo
18.
Int Immunopharmacol ; 85: 106619, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32485352

RESUMEN

Toll-like receptor 2 (TLR2) is a primary sensor for pathogens, including those derived from gram-positive bacteria. It can also mediate the effects of endogenous inflammatory signals such as ß-amyloid peptide (Aß), thus promoting the microglial activation and subsequent neuronal dysfunction, characteristic of chronic neuroinflammatory conditions. More recently, a role for TLR2 has been proposed in the pathogenesis of disorders associated with acute inflammation, including anxiety and depression. The current study aims to characterise the acute effects of the TLR2 agonist lipoteichoic acid (LTA) on microglial activation and neuronal integrity, and to evaluate the influence of LTA exposure on sensitivity to the inflammation and neuronal dysfunction associated with Aß. Using BV2 and N2a cells as an in vitro model, we highlight that acute exposure to LTA robustly promotes inflammatory cytokine and nitric oxide (NO) production in microglia but also in neurons, similar to that reported under longer-term and chronic inflammatory conditions. Moreover, we find that exposure to LTA can enhance sensitivity to subthreshold Aß, promoting an 'M1'-like phenotype in microglia and provoking dysregulation of neuronal activity in acute hippocampal slices. Anti-inflammatory agents, including mimetics of brain-derived neurotrophic factor (BDNF), have proven effective at alleviating chronic neuroinflammatory complications. We further examined the effects of 7,8,3-trihydroxyflavone (7,8,3-THF), a small-molecule TrkB agonist, on LTA-induced microglial activation. We report that 7,8,3-THF can significantly ameliorate interleukin (IL)-6 and NO production in LTA-stimulated BV2 cells. Taken together, our findings offer support for exploration of TLR2 as a potential target for therapeutic intervention into acute neuroinflammatory conditions. Moreover we propose that exposure to gram-positive bacterial pathogens may promote sensitivity to the inflammatory changes characteristic of the aged brain.


Asunto(s)
Inflamación/metabolismo , Inflamación/fisiopatología , Lipopolisacáridos/toxicidad , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/fisiopatología , Ácidos Teicoicos/toxicidad , Receptor Toll-Like 2/agonistas , Enfermedad Aguda , Péptidos beta-Amiloides/toxicidad , Animales , Línea Celular , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Flavonas/farmacología , Inflamación/inducido químicamente , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/metabolismo , Modelos Teóricos , Enfermedades del Sistema Nervioso/inducido químicamente , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Receptor trkB/agonistas , Factor de Necrosis Tumoral alfa/metabolismo
19.
Eur Arch Psychiatry Clin Neurosci ; 270(2): 195-205, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29882089

RESUMEN

Although depressive symptoms including anhedonia (i.e., loss of pleasure) frequently accompany pain, little is known about the risk factors contributing to individual differences in pain-induced anhedonia. In this study, we examined if signaling of brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-receptor-kinase B (TrkB) contribute to individual differences in the development of neuropathic pain-induced anhedonia. Rats were randomly subjected to spared nerved ligation (SNI) or sham surgery. The SNI rats were divided into two groups based on the results of a sucrose preference test. Rats with anhedonia-like phenotype displayed lower tissue levels of BDNF in the medial prefrontal cortex (mPFC) compared with rats without anhedonia-like phenotype and sham-operated rats. In contrast, tissue levels of BDNF in the nucleus accumbens (NAc) of rats with an anhedonia-like phenotype were higher compared with those of rats without anhedonia-like phenotype and sham-operated rats. Furthermore, tissue levels of BDNF in the hippocampus, L2-5 spinal cord, muscle, and liver from both rats with or without anhedonia-like phenotype were lower compared with those of sham-operated rats. A single injection of 7,8-dihydroxyflavone (10 mg/kg; TrkB agonist), but not ANA-12 (0.5 mg/kg; TrkB antagonist), ameliorated reduced sucrose preference and reduced BDNF-TrkB signaling in the mPFC in the rats with anhedonia-like phenotype. These findings suggest that reduced BDNF-TrkB signaling in the mPFC might contribute to neuropathic pain-induced anhedonia, and that TrkB agonists could be potential therapeutic drugs for pain-induced anhedonia.


Asunto(s)
Anhedonia/fisiología , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuralgia/metabolismo , Neuralgia/fisiopatología , Corteza Prefrontal/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/fisiología , Anhedonia/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Neuralgia/etiología , Núcleo Accumbens/metabolismo , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/fisiopatología , Fenotipo , Ratas , Ratas Sprague-Dawley , Receptor trkB/agonistas , Receptor trkB/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sacarosa
20.
Proc Natl Acad Sci U S A ; 117(1): 426-431, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871166

RESUMEN

Herein we present a concept in cancer where an immune response is detrimental rather than helpful. In the cancer setting, the immune system is generally considered to be helpful in curtailing the initiation and progression of tumors. In this work we show that a patient's immune response to their tumor can, in fact, either enhance or inhibit tumor cell growth. Two closely related autoantibodies to the growth factor receptor TrkB were isolated from cancer patients' B cells. Although highly similar in sequence, one antibody was an agonist while the other was an antagonist. The agonist antibody was shown to increase breast cancer cell growth both in vitro and in vivo, whereas the antagonist antibody inhibited growth. From a mechanistic point of view, we showed that binding of the agonist antibody to the TrkB receptor was functional in that it initiated downstream signaling identical to its natural growth factor ligand, brain-derived neurotrophic factor (BDNF). Our study shows that individual autoantibodies may play a role in cancer patients.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Neoplasias de la Mama/patología , Glicoproteínas de Membrana/inmunología , Metástasis de la Neoplasia/inmunología , Receptor trkB/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/aislamiento & purificación , Autoanticuerpos/metabolismo , Autoantígenos/sangre , Autoantígenos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Factor Neurotrófico Derivado del Encéfalo/inmunología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neoplasias de la Mama/sangre , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Movimiento Celular/inmunología , Proliferación Celular , Femenino , Humanos , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/sangre , Ratones , Receptor trkB/agonistas , Receptor trkB/antagonistas & inhibidores , Receptor trkB/sangre , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA