Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Molecules ; 26(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071302

RESUMEN

The 3-hydroxyquinone derivative of the non-psychotrophic phytocannabinoid cannabigerol, so-called VCE-003.2, and some other derivatives have been recently investigated for neuroprotective properties in experimental models of Parkinson's disease (PD) in mice. The pharmacological effects in those models were related to the activity on the peroxisome proliferator-activated receptor-γ (PPAR-γ) and possibly other pathways. In the present study, we investigated VCE-004.8 (formulated as EHP-101 for oral administration), the 3-hydroxyquinone derivative of cannabidiol (CBD), with agonist activity at the cannabinoid receptor type-2 (CB2) receptor in addition to its activity at the PPAR-γ receptor. Studies were conducted in both in vivo (lesioned-mice) and in vitro (SH-SY5Y cells) models using the classic parkinsonian neurotoxin 6-hydroxydopamine (6-OHDA). Our data confirmed that the treatment with VCE-004.8 partially reduced the loss of tyrosine hydroxylase (TH)-positive neurons measured in the substantia nigra of 6-OHDA-lesioned mice, in parallel with an almost complete reversal of the astroglial (GFAP) and microglial (CD68) reactivity occurring in this structure. Such neuroprotective effects attenuated the motor deficiencies shown by 6-OHDA-lesioned mice in the cylinder rearing test, but not in the pole test. Next, we explored the mechanism involved in the beneficial effect of VCE-004.8 in vivo, by analyzing cell survival in cultured SH-SY5Y cells exposed to 6-OHDA. We found an important cytoprotective effect of VCE-004.8 at a concentration of 10 µM, which was completely reversed by the addition of antagonists, T0070907 and SR144528, aimed at blocking PPAR-γ and CB2 receptors, respectively. The treatment with T0070907 alone only caused a partial reversal, whereas SR144528 alone had no effect, indicating a major contribution of PPAR-γ receptors in the cytoprotective effect of VCE-004.8 at 10 µM. In summary, our data confirmed the neuroprotective potential of VCE-004.8 in 6-OHDA-lesioned mice, and in vitro studies confirmed a greater relevance for PPAR-γ receptors rather than CB2 receptors in these effects.


Asunto(s)
Cannabidiol/química , Oxidopamina/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Quinonas/química , Administración Oral , Animales , Benzamidas/farmacología , Canfanos/farmacología , Cannabinoides/química , Línea Celular Tumoral , Supervivencia Celular , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuroprotección , Oxidopamina/química , PPAR gamma/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Tirosina 3-Monooxigenasa/metabolismo
2.
Molecules ; 26(3)2021 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-33494255

RESUMEN

The damage to SH-SY5Y cells by 6-hydroxydopamine (6-OHDA) is an established cellular model of Parkinson's disease (PD). Redox nanoparticles are a promising tool for therapy, including neurodegenerative diseases. As pH of the brain tissue at sites affected by PD is lowered down to 6.5, we studied the effect of pH-responsive redox nanoparticles (poly(ethylene glycol)-b-poly[4-(2,2,6,6-tetramethylpiperidine-1-oxyl)aminomethylstyrene]), which change their structure in a pH-dependent manner and become active below pH 7 (NRNPs pH), on the viability of SH-SY5Y cells treated with 6-OHDA at pH 6.5 and 7.4. Pretreatment of the cells with NRNPs pH (15-75 µM) prior to the 6-OHDA treatment increased their survival in a concentration-dependent manner at pH 6.5, but not at pH 7.4. Among several parameters studied (ATP and GSH content, the level of reactive oxygen species, mitochondrial potential, mitochondrial mass), only the mitochondrial mass was dose-dependently protected by NRNPs pH at pH 6.5, but not at pH 7.4. These results indicate that the action of NRNPs pH on mitochondria underlies their protective effect in this cellular model of PD. These results may have potential importance for future applications of NRNPs pH in preclinical and perhaps clinical studies.


Asunto(s)
Modelos Neurológicos , Nanopartículas , Oxidopamina , Enfermedad de Parkinson , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas/química , Nanopartículas/uso terapéutico , Oxidopamina/química , Oxidopamina/farmacocinética , Oxidopamina/farmacología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/prevención & control
3.
J Parkinsons Dis ; 11(2): 515-528, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33361611

RESUMEN

BACKGROUND: Human induced pluripotent stem cells (hiPSCs) have been proposed as an alternative source for cell replacement therapy for Parkinson's disease (PD) and they provide the option of using the patient's own cells. A few studies have investigated transplantation of patient-derived dopaminergic (DA) neurons in preclinical models; however, little is known about the long-term integrity and function of grafts derived from patients with PD. OBJECTIVE: To assess the viability and function of DA neuron grafts derived from a patient hiPSC line with an α-synuclein gene triplication (AST18), using a clinical grade human embryonic stem cell (hESC) line (RC17) as a reference control. METHODS: Cells were differentiated into ventral mesencephalic (VM)-patterned DA progenitors using an established GMP protocol. The progenitors were then either terminally differentiated to mature DA neurons in vitro or transplanted into 6-hydroxydopamine (6-OHDA) lesioned rats and their survival, maturation, function, and propensity to develop α-synuclein related pathology, were assessed in vivo. RESULTS: Both cell lines generated functional neurons with DA properties in vitro. AST18-derived VM progenitor cells survived transplantation and matured into neuron-rich grafts similar to the RC17 cells. After 24 weeks, both cell lines produced DA-rich grafts that mediated full functional recovery; however, pathological changes were only observed in grafts derived from the α-synuclein triplication patient line. CONCLUSION: This data shows proof-of-principle for survival and functional recovery with familial PD patient-derived cells in the 6-OHDA model of PD. However, signs of slowly developing pathology warrants further investigation before use of autologous grafts in patients.


Asunto(s)
Células Madre Pluripotentes Inducidas , Oxidopamina/farmacología , Enfermedad de Parkinson , Sinucleinopatías , alfa-Sinucleína/química , Animales , Neuronas Dopaminérgicas/metabolismo , Humanos , Oxidopamina/química , Enfermedad de Parkinson/terapia , Ratas , alfa-Sinucleína/genética
4.
Redox Biol ; 28: 101377, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31760358

RESUMEN

Recent findings suggest that dopamine oxidation contributes to the development of Parkinson's disease (PD); however, the mechanistic details remain elusive. Here, we compare 6-hydroxydopamine (6-OHDA), a product of dopamine oxidation that commonly induces dopaminergic neurodegeneration in laboratory animals, with a synthetic alkyne-functionalized 6-OHDA variant. This synthetic molecule provides insights into the reactivity of quinone and neuromelanin formation. Employing Huisgen cycloaddition chemistry (or "click chemistry") and fluorescence imaging, we found that reactive 6-OHDA p-quinones cause widespread protein modification in isolated proteins, lysates and cells. We identified cysteine thiols as the target site and investigated the impact of proteome modification by quinones on cell viability. Mass spectrometry following cycloaddition chemistry produced a large number of 6-OHDA modified targets including proteins involved in redox regulation. Functional in vitro assays demonstrated that 6-OHDA inactivates protein disulfide isomerase (PDI), which is a central player in protein folding and redox homeostasis. Our study links dopamine oxidation to protein modification and protein folding in dopaminergic neurons and the PD model.


Asunto(s)
Neuronas Dopaminérgicas/citología , Hidroxidopaminas/efectos adversos , Enfermedad de Parkinson/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Reacción de Cicloadición , Cisteína/química , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Hidroxidopaminas/química , Masculino , Espectrometría de Masas , Ratones , Oxidopamina/efectos adversos , Oxidopamina/química , Proteómica
5.
Drug Deliv ; 25(1): 143-152, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29275639

RESUMEN

A microsphere-gel in situ forming implant (MS-Gel ISFI) dual-controlled drug delivery system was applied to a high water-soluble small-molecule compound Rasagiline mesylate (RM) for effective treatment of Parkinson's disease. This injectable complex depot system combined an in situ phase transition gel with high drug-loading and encapsulation efficiency RM-MS prepared by a modified emulsion-phase separation method and optimized by Box-Behnken design. It was evaluated for in vitro drug release, in vivo pharmacokinetics, and in vivo pharmacodynamics. We found that the RM-MS-Gel ISFI system showed no initial burst release and had a long period of in vitro drug release (60 days). An in vivo pharmacokinetic study indicated a significant reduction (p < .01) in the initial high plasma drug concentration of the RM-MS-Gel ISFI system compared to that of the single RM-MS and RM-in situ gel systems after intramuscular injection to rats. A pharmacodynamic study demonstrated a significant reduction (p < .05) in 6-hydroxydopamine-induced contralateral rotation behavior and an effective improvement (p < .05) in dopamine levels in the striatum of the lesioned side after 28 days in animals treated with the RM-MS-Gel ISFI compared with that of animals treated with saline. MS-embedded in situ phase transition gel is superior for use as a biodegradable and injectable sustained drug delivery system with a low initial burst and long period of drug release for highly hydrophilic small molecule drugs.


Asunto(s)
Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Indanos/química , Indanos/farmacología , Mesilatos/química , Mesilatos/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos/efectos de los fármacos , Geles/química , Inyecciones/métodos , Masculino , Microesferas , Oxidopamina/química , Transición de Fase , Ratas , Ratas Sprague-Dawley
6.
Neurochem Res ; 41(12): 3356-3363, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27662849

RESUMEN

Neuromelanin (NM) has long been considered as an aging pigment, perhaps an unavoidable and undesirable byproduct of dopaminergic neural transmission. However, NM is carefully packaged into double membrane-bound structures within cells of the substantia nigra and other neural tissues, suggesting a beneficial function to maintaining these stores. It is well established that NM is able to concentrate toxic xenobiotics within pigmented cells due to its unique chemical environment. In doing so, such agents may confer susceptibility to Parkinson's disease (PD) as illustrated by model PD-inducing neurotoxins such as methyl-phenyl-pyridinium ion. It is possible that high-affinity binding interactions toward NM may contribute to the adverse effects of PD-inducing toxins, as well as neuroprotective agents. Here we aim to develop a generalized assay capable of elucidating the binding constants of chemical agents to synthetic and natural neuromelanins. Toward this end, a model neuromelanin synthesized from dopamine and cysteine was prepared according to published procedure. Using a UV/Visible spectroscopic assay, we show that dopamine, 6-hydroxy dopamine, and nicotine bind to the synthetic neuromelanin, while caffeine did not. More importantly, nicotine was further found to induce a fluorescence signal in the presence of NM which was used to establish a binding constant estimated at 0.65 mM. Dopamine appears to enhance this signal, also in a saturable manner, with an estimated Kd of 0.05 mM in our isolated chemical system. In summary, the micro-scale fluorescence assay described herein will allow us to overcome many of the problems inherent in the study of chemical interaction with NM through traditional spectroscopic means. Using a single standardized signal, it should now be possible to rank a number of PD-related toxins based on NM-binding affinity and shed further light on this important problem.


Asunto(s)
Melaninas/química , Nicotina/química , Cafeína/química , Cisteína/química , Dopamina/química , Hierro/química , Melaninas/síntesis química , Oxidopamina/química , Enfermedad de Parkinson , Polimerizacion , Espectrometría de Fluorescencia
7.
J Agric Food Chem ; 64(30): 5995-6002, 2016 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-27431098

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder, which can be modeled using the neurotoxin 6-hydroxydopamine (6-OHDA) to generate oxidative stress. Here, we studied the effects of the antioxidants deferricoprogen (DFC) and dimerumic acid (DMA), produced by rice fermented with Monascus purpureus NTU 568, on 6-OHDA-induced apoptosis in SH-SY5Y cells and their potential protective mechanisms. DMA and DFC inhibited 6-OHDA-induced apoptosis and cellular reactive oxygen species (ROS) in SH-SY5Y human neuroblastoma cells. Molecular analysis demonstrated associated upregulation of the Ak mouse strain thymoma (Akt), heme oxygenase-1 (HO-1), and signal-regulated kinase (ERK) pathways along with inhibited phosphorylation of c-Jun N-terminal kinase (JNK) and p38 pathways and altered homodimeric glycoprotein, N-methyl-d-aspartate (NMDA) receptor, and immunoglobulin Fc receptor gene expression. These results suggested that the neuroprotection elicited by DMA and DFC against 6-OHDA-induced neurotoxicity was associated with the Akt, MAPK, and HO-1 pathways via regulating the gene expression of NMDA receptor, homodimeric glycoprotein, and immunoglobulin Fc receptor.


Asunto(s)
Apoptosis/efectos de los fármacos , Dicetopiperazinas/química , Hemo-Oxigenasa 1/metabolismo , Ácidos Hidroxámicos/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antioxidantes/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular , Humanos , Oryza/química , Estrés Oxidativo , Oxidopamina/química , Enfermedad de Parkinson/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
Transl Res ; 170: 40-56.e3, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26730494

RESUMEN

Although restoration of mitochondrial function in mitochondrial diseases through peptide-mediated allogeneic mitochondrial delivery (PMD) has been demonstrated in vitro, the in vivo therapeutic efficacy of PMD in Parkinson's disease (PD) has yet to be determined. In this study, we compared the functionality of mitochondrial transfer with or without Pep-1 conjugation in neurotoxin (6-hydroxydopamine, 6-OHDA)-induced PC12 cells and PD rat models. We injected mitochondria into the medial forebrain bundle (MFB) of the PD rats after subjecting the nigrostriatal pathway to a unilateral 6-OHDA lesion for 21 days, and we verified the effectiveness of the mitochondrial graft in enhancing mitochondrial function in the soma of the substantia nigra (SN) neuron through mitochondrial transport dynamics in the nigrostriatal circuit. The result demonstrated that only PMD with allogeneic and xenogeneic sources significantly sustained mitochondrial function to resist the neurotoxin-induced oxidative stress and apoptotic death in the rat PC12 cells. The remaining cells exhibited a greater capability of neurite outgrowth. Furthermore, allogeneic and xenogeneic transplantation of peptide-labeled mitochondria after 3 months improved the locomotive activity in the PD rats. This increase was accompanied by a marked decrease in dopaminergic neuron loss in the substantia nigra pars compacta (SNc) and consistent enhancement of tyrosine hydroxylase-positive immunoreaction of dopaminergic neurons in the SNc and striatum. We also observed that in the SN dopaminergic neuron in the treated PD rats, mitochondrial complex I protein and mitochondrial dynamics were restored, thus ameliorating the oxidative DNA damage. Moreover, we determined signal translocation of graft allogeneic mitochondria from the MFB to the calbindin-positive SN neuron, which demonstrated the regulatory role of mitochondrial transport in alleviating 6-OHDA-induced degeneration of dopaminergic neurons.


Asunto(s)
Cisteamina/análogos & derivados , Mitocondrias/trasplante , Oxidopamina/efectos adversos , Enfermedad de Parkinson/terapia , Péptidos/química , Animales , Calbindinas/metabolismo , Trasplante de Células , Cisteamina/química , Neuronas Dopaminérgicas/patología , Femenino , Humanos , Mitocondrias/fisiología , Estrés Oxidativo , Oxidopamina/química , Células PC12 , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Trasplante Heterólogo/métodos , Trasplante Homólogo/métodos
9.
Regen Med ; 10(5): 563-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26237701

RESUMEN

AIM: Here we investigated the neuroprotective potential of systemic CD34(+) human cord blood cells (hCBCs) in a 6-hydroxydopamine rat model of Parkinson's disease. METHODS: Purified CD34(+) hCBCs were intravenously administered to rats subjected to 6-hydroxydopamine 24 h earlier, and behavioral and immunohistological analysis performed. RESULTS: CD34(+) hCBC administration significantly prevented host nigrostriatal degeneration inducing behavioral recovery in treated rats. Although donor hCBCs did not differentiate into neural phenotypes, they stimulated the production of new neuroblasts and angiogenesis, and reduced gliosis in recipient animals. Importantly, surviving donor hCBCs were identified, and their tissue distribution pattern correlated with the observed therapeutic effects. CONCLUSION: Peripherally applied CD34(+) hCBCs can migrate into brain tissues and elicit host-based protective mechanisms to support the survival of midbrain dopamine neurons.


Asunto(s)
Antígenos CD34/metabolismo , Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Enfermedad de Parkinson/terapia , Animales , Encéfalo/patología , Diferenciación Celular , Línea Celular , Movimiento Celular , Modelos Animales de Enfermedad , Dopamina/química , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Mesencéfalo/citología , Neovascularización Patológica , Degeneración Nerviosa/patología , Neuronas/citología , Oxidopamina/química , Fenotipo , Ratas , Ratas Endogámicas F344 , Células Madre/citología , Sustancia Negra/citología , Distribución Tisular
10.
ACS Nano ; 9(5): 4850-71, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25825926

RESUMEN

Sustained and safe delivery of dopamine across the blood brain barrier (BBB) is a major hurdle for successful therapy in Parkinson's disease (PD), a neurodegenerative disorder. Therefore, in the present study we designed neurotransmitter dopamine-loaded PLGA nanoparticles (DA NPs) to deliver dopamine to the brain. These nanoparticles slowly and constantly released dopamine, showed reduced clearance of dopamine in plasma, reduced quinone adduct formation, and decreased dopamine autoxidation. DA NPs were internalized in dopaminergic SH-SY5Y cells and dopaminergic neurons in the substantia nigra and striatum, regions affected in PD. Treatment with DA NPs did not cause reduction in cell viability and morphological deterioration in SH-SY5Y, as compared to bulk dopamine-treated cells, which showed reduced viability. Herein, we report that these NPs were able to cross the BBB and capillary endothelium in the striatum and substantia nigra in a 6-hydroxydopamine (6-OHDA)-induced rat model of PD. Systemic intravenous administration of DA NPs caused significantly increased levels of dopamine and its metabolites and reduced dopamine-D2 receptor supersensitivity in the striatum of parkinsonian rats. Further, DA NPs significantly recovered neurobehavioral abnormalities in 6-OHDA-induced parkinsonian rats. Dopamine delivered through NPs did not cause additional generation of ROS, dopaminergic neuron degeneration, and ultrastructural changes in the striatum and substantia nigra as compared to 6-OHDA-lesioned rats. Interestingly, dopamine delivery through nanoformulation neither caused alterations in the heart rate and blood pressure nor showed any abrupt pathological change in the brain and other peripheral organs. These results suggest that NPs delivered dopamine into the brain, reduced dopamine autoxidation-mediated toxicity, and ultimately reversed neurochemical and neurobehavioral deficits in parkinsonian rats.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Dopamina/química , Dopamina/metabolismo , Nanopartículas/química , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/fisiopatología , Animales , Línea Celular Tumoral , Dopamina/efectos adversos , Neuronas Dopaminérgicas/metabolismo , Portadores de Fármacos/efectos adversos , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Liberación de Fármacos , Humanos , Ácido Láctico/química , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Oxidación-Reducción , Oxidopamina/química , Oxidopamina/farmacología , Oxidopamina/uso terapéutico , Enfermedad de Parkinson/metabolismo , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Wistar , Receptores Dopaminérgicos/metabolismo , Seguridad , Regulación hacia Arriba/efectos de los fármacos
11.
Proc Natl Acad Sci U S A ; 112(19): E2517-26, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25918399

RESUMEN

Parkinson's disease (PD) is characterized by severe locomotor deficits and is commonly treated with the dopamine (DA) precursor l-3,4-dihydroxyphenylalanine (L-DOPA), but its prolonged use causes dyskinesias referred to as L-DOPA-induced dyskinesias (LIDs). Recent studies in animal models of PD have suggested that dyskinesias are associated with the overactivation of G protein-mediated signaling through DA receptors. ß-Arrestins desensitize G protein signaling at DA receptors (D1R and D2R) in addition to activating their own G protein-independent signaling events, which have been shown to mediate locomotion. Therefore, targeting ß-arrestins in PD L-DOPA therapy might prove to be a desirable approach. Here we show in a bilateral DA-depletion mouse model of Parkinson's symptoms that genetic deletion of ß-arrestin2 significantly limits the beneficial locomotor effects while markedly enhancing the dyskinesia-like effects of acute or chronic L-DOPA treatment. Viral rescue or overexpression of ß-arrestin2 in knockout or control mice either reverses or protects against LIDs and its key biochemical markers. In other more conventional animal models of DA neuron loss and PD, such as 6-hydroxydopamine-treated mice or rats and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated nonhuman primates, ß-arrestin2 overexpression significantly reduced dyskinesias while maintaining the therapeutic effect of L-DOPA. Considerable efforts are being spent in the pharmaceutical industry to identify therapeutic approaches to block LIDs in patients with PD. Our results point to a potential therapeutic approach, whereby development of either a genetic or pharmacological intervention to enhance ß-arrestin2- or limit G protein-dependent D1/D2R signaling could represent a more mechanistically informed strategy.


Asunto(s)
Arrestinas/metabolismo , Discinesias/metabolismo , Levodopa/química , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/química , Animales , Arrestinas/genética , Conducta Animal , Modelos Animales de Enfermedad , Dopamina/metabolismo , Discinesia Inducida por Medicamentos/metabolismo , Eliminación de Gen , Macaca , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Oxidopamina/química , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Regulación hacia Arriba , beta-Arrestinas
12.
Exp Biol Med (Maywood) ; 240(11): 1387-95, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25769314

RESUMEN

Parkinson's disease (PD) is the first and second most prevalent motor and neurodegenerative disease, respectively. The clinical symptoms of PD result from a loss of midbrain dopaminergic (DA) neurons. However, the molecular cause of DA neuron loss remains elusive. Mounting evidence implicates enhanced inflammatory response in the development and progression of PD pathology. This review examines current research connecting PD and inflammatory response.


Asunto(s)
Inflamación/patología , Estrés Oxidativo , Enfermedad de Parkinson/inmunología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/química , Animales , Antiinflamatorios no Esteroideos/química , Citocinas/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Lipopolisacáridos/química , Neuronas/metabolismo , Oxidopamina/química , Oxígeno/química , Paraquat/química , Enfermedad de Parkinson/fisiopatología , Ubiquitina-Proteína Ligasas/metabolismo
13.
Food Chem ; 172: 640-9, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25442601

RESUMEN

ß-Carbolines are bioactive pyridoindole alkaloids occurring in foods, plants and the human body. Their activity as hydroxyl radical (OH) scavengers is reported here by using three different methods: deoxyribose degradation, hydroxylation of benzoate and hydroxylation of 2'-deoxyguanosine to give 8-hydroxy-2'-deoxyguanosine (8-OHdG) as assessed by RP-HPLC (MS). Fenton reactions (Fe(2+)/Fe(3+) plus H2O2) were used for OH generation, and the radical increased in the presence of ascorbic acid or 6-hydroxydopamine as pro-oxidants. ß-Carbolines were scavengers of OH in the three assays and in the presence of pro-oxidants. Tetrahydro-ß-carboline-3-carboxylic acids were active against the hydroxylation of 2'-deoxyguanosine. ß-Carbolines reacted with hydroxyl radicals (OH) affording hydroxy-ß-carbolines, whereas tetrahydro-ß-carbolines gave oxidative and degradation products. On the basis of IC50 and reaction rates (k), ß-carbolines (norharman and harman), and tetrahydro-ß-carbolines (tetrahydro-ß-carboline, 1-methyltetrahydro-ß-carboline and pinoline) were good OH radical scavengers and their activity was comparable to that of the indole, melatonin, which is an effective hydroxyl radical scavenger and antioxidant.


Asunto(s)
Alcaloides/farmacología , Carbolinas/farmacología , Radical Hidroxilo/farmacología , 8-Hidroxi-2'-Desoxicoguanosina , Alcaloides/química , Antioxidantes/química , Antioxidantes/farmacología , Ácido Ascórbico/química , Carbolinas/química , Cromatografía Líquida de Alta Presión , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Peróxido de Hidrógeno/química , Radical Hidroxilo/química , Melatonina/química , Melatonina/farmacología , Estrés Oxidativo/efectos de los fármacos , Oxidopamina/química
14.
Dis Model Mech ; 8(1): 57-63, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25398851

RESUMEN

Parkinson's disease (PD) is a dopaminergic-related pathology in which functioning of the basal ganglia is altered. It has been postulated that a direct receptor-receptor interaction - i.e. of dopamine D2 receptor (D2R) with adenosine A2A receptor (A2AR) (forming D2R-A2AR oligomers) - finely regulates this brain area. Accordingly, elucidating whether the pathology prompts changes to these complexes could provide valuable information for the design of new PD therapies. Here, we first resolved a long-standing question concerning whether D2R-A2AR assembly occurs in native tissue: by means of different complementary experimental approaches (i.e. immunoelectron microscopy, proximity ligation assay and TR-FRET), we unambiguously identified native D2R-A2AR oligomers in rat striatum. Subsequently, we determined that, under pathological conditions (i.e. in a rat PD model), D2R-A2AR interaction was impaired. Collectively, these results provide definitive evidence for alteration of native D2R-A2AR oligomers in experimental parkinsonism, thus conferring the rationale for appropriate oligomer-based PD treatments.


Asunto(s)
Dopamina/química , Trastornos Parkinsonianos/metabolismo , Receptores Dopaminérgicos/química , Receptores Purinérgicos P1/química , Animales , Encéfalo/patología , Membrana Celular/metabolismo , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunohistoquímica , Ligandos , Ratones , Ratones Noqueados , Microscopía Inmunoelectrónica , Oxidopamina/química , Trastornos Parkinsonianos/tratamiento farmacológico , Plásmidos/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Proc Natl Acad Sci U S A ; 111(49): 17636-41, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25413364

RESUMEN

Recurrent axon collaterals are a major means of communication between spiny projection neurons (SPNs) in the striatum and profoundly affect the function of the basal ganglia. However, little is known about the molecular and cellular mechanisms that underlie this communication. We show that intrastriatal nitric oxide (NO) signaling elevates the expression of the vesicular GABA transporter (VGAT) within recurrent collaterals of SPNs. Down-regulation of striatal NO signaling resulted in an attenuation of GABAergic signaling in SPN local collaterals, down-regulation of VGAT expression in local processes of SPNs, and impaired motor behavior. PKG1 and cAMP response element-binding protein are involved in the signal transduction that transcriptionally regulates VGAT by NO. These data suggest that transcriptional control of the vesicular GABA transporter by NO regulates GABA transmission and action selection.


Asunto(s)
Ganglios Basales/metabolismo , Guanilato Ciclasa/química , Neuronas/metabolismo , Óxido Nítrico/química , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/química , Animales , Axones/metabolismo , AMP Cíclico/metabolismo , Dopamina/metabolismo , Electrofisiología , Retroalimentación Fisiológica , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Levodopa/química , Masculino , Ratones , Plasticidad Neuronal , Oxidopamina/química , Transducción de Señal , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
16.
Biosens Bioelectron ; 59: 166-73, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24727602

RESUMEN

We propose a novel application of dielectrophoresis (DEP) to make three-dimensional (3D) methacrylated gelatin (GelMA) hydrogels with gradients of micro- and nanoparticles. DEP forces were able to manipulate micro- and nanoparticles of different sizes and materials (i.e., C2C12 myoblasts, polystyrene beads, gold microparticles, and carbon nanotubes) within GelMA hydrogels in a rapid and facile way and create 3D gradients of these particles in a microchamber. Immobilization of drugs, such as fluorescein isothiocyanate-dextran (FITC-dextran) and 6-hydroxydopamine (6-OHDA), on gold microparticles allowed us to investigate the high-throughput release of these drugs from GelMA-gold microparticle gradient systems. The latter gradient constructs were incubated with C2C12 myoblasts for 24h to examine the cell viability through the release of 6-OHDA. The drug was released from the microparticles in a gradient manner, inducing a cell viability gradient. This novel approach to create 3D chemical gradients within hydrogels is scalable to any arbitrary length scale. It is useful for making anisotropic biomimetic materials and high-throughput platforms to investigate cell-microenvironment interactions in a rapid, simple, cost-effective, and reproducible manner.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Hidrogeles/química , Animales , Técnicas Biosensibles/métodos , Supervivencia Celular/efectos de los fármacos , Dextranos/química , Electroforesis/métodos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/química , Ratones , Oxidopamina/química , Oxidopamina/farmacología , Simpaticolíticos/química , Simpaticolíticos/farmacología
17.
Neurotox Res ; 24(1): 94-101, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23385626

RESUMEN

6-Hydroxydamine has widely been used as neurotoxin in preclinical studies related on the neurodegenerative process of dopaminergic neurons in Parkinson's disease based on its ability to be neurotoxic as a consequence of free radical formation during its auto-oxidation to topaminequinone. We report that 50-µM 6-hydroxydopamine is not neurotoxic in RCSN-3 cells derived from substantia nigra incubated during 24 h contrasting with a significant sixfold increase in cell death (16 ± 2 %; P < 0.001) was observed in RCSN-3NQ7 cells expressing a siRNA against DT-diaphorase that silence the enzyme expression. To observe a significant cell death in RCSN-3 cells induced by 6-hydroxydopamine (24 ± 1 %; P < 0.01), we have to increase the concentration to 250 µm while a 45 ± 2 % cell death (P < 0.001) was observed at this concentration in RCSN-3NQ7 cells. The cell death induced by 6-hydroxydopamine in RCSN-3NQ7 cells was accompanied with a (i) significant increase in oxygen consumption (P < 0.01), (ii) depletion of reduced glutathione and (iii) a significant decrease in ATP level (P < 0.05) in comparison with RCSN-3 cells. In conclusion, our results suggest that one-electron reduction of 6-hydroxydopamine quinone seems to be the main reaction responsible for 6-hydroxydopamine neurotoxic effects in dopaminergic neurons and DT-diaphorase seems to play an important neuroprotective role by preventing one-electron reduction of topaminequinone.


Asunto(s)
Electrones , Hidroxidopaminas/química , Hidroxidopaminas/toxicidad , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , Oxidopamina/química , Oxidopamina/toxicidad , Quinonas/química , Quinonas/toxicidad , Adenosina Trifosfato/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Ratas , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-23217306

RESUMEN

Reversed-phase ion-pairing chromatography (RP-IPC) is coupled on-line with electrospray ionization-mass spectrometry (ESI-MS) through an interface comprising a four-way switch valve and an anion exchange column. Regeneration of the anion exchange column can be accomplished on-line by switching the four-way switch valve to interconnect the column to a regeneration solution. Positioning the anion exchange column between the RP-IPC and ESI-MS instruments allows the ion-pairing reagent (IPR) sodium octane sulfonate to be removed. The IPC-ESI-MS method enabled us to separate and detect four intermediates of the Fe(III)-catalyzed dopamine oxidation. In particular, 6-hydroxydopamine, which is short-lived and highly neurotoxic, was detected and quantified. Together with the separation of other intermediates, gaining insight into the mechanism and kinetics of the Fe(III)-catalyzed dopamine oxidation becomes possible.


Asunto(s)
Cromatografía de Fase Inversa/métodos , Dopamina/análogos & derivados , Dopamina/química , Hierro/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Cromatografía por Intercambio Iónico/métodos , Dopamina/análisis , Dopamina/aislamiento & purificación , Oxidación-Reducción , Oxidopamina/química , Espectrofotometría Ultravioleta
19.
Chem Res Toxicol ; 25(9): 1893-901, 2012 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-22894569

RESUMEN

Understanding the neuroprotective effects of the rosemary phenolic diterpene carnosic acid (CA) has attracted increasing attention. We explored the mechanism by which CA modulates the neurotoxic effects of 6-hydroxydopamine (6-OHDA) in SH-SY5Y cells. Cells were pretreated with CA for 12 h followed by treatment with 100 µM 6-OHDA for 12 or 24 h. Cell viability determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolim bromide (MTT) assay indicated that 0.1 to 1 µM CA dose-dependently attenuated the cell death induced by 6-OHDA, whereas the effect of 3-5 µM CA was weaker. CA at 1 µM suppressed the 6-OHDA-induced nuclear condensation, reactive oxygen species generation, and cleavage of caspase 3 and PARP. Immunoblots showed that the phosphorylation of c-Jun NH(2)-terminal kinase (JNK) and p38 by 6-OHDA was reduced in the presence of CA. Incubation of cells with CA resulted in significant increases in the total glutathione (GSH) level and the protein expression of the γ-glutamylcysteine ligase catalytic subunit and modifier subunit. L-Buthionine-sulfoximine, an inhibitor of GSH synthesis, attenuated the effect of CA on cell death and apoptosis. Treatment with CA also led to an increase in nuclear factor erythroid-2 related factor 2 (Nrf2) activation, antioxidant response element (ARE)-luciferase reporter activity, and DNA binding to the ARE. Silencing of Nrf2 expression alleviated the reversal of p38 and JNK1/2 activation by CA. These results suggest that the attenuation of 6-OHDA-induced apoptosis by CA is associated with the Nrf2-driven synthesis of GSH, which in turn down-regulates the JNK and p38 signaling pathways. The CA compound may be a promising candidate for neuroprotection in Parkinson's disease.


Asunto(s)
Abietanos/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Glutatión/metabolismo , Oxidopamina/toxicidad , Extractos Vegetales/farmacología , Abietanos/química , Antioxidantes/química , Butionina Sulfoximina/química , Butionina Sulfoximina/farmacología , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Glutamato-Cisteína Ligasa/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Oxidopamina/química , Extractos Vegetales/química , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sales de Tetrazolio/química , Sales de Tetrazolio/metabolismo , Tiazoles/química , Tiazoles/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
PLoS One ; 7(4): e35371, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22539969

RESUMEN

Parkinson's disease presents nonmotor complications such as autonomic dysfunction that do not respond to traditional anti-parkinsonian therapies. The lack of established preclinical monkey models of Parkinson's disease with cardiac dysfunction hampers development and testing of new treatments to alleviate or prevent this feature. This study aimed to assess the feasibility of developing a model of cardiac dysautonomia in nonhuman primates and preclinical evaluations tools. Five rhesus monkeys received intravenous injections of 6-hydroxydopamine (total dose: 50 mg/kg). The animals were evaluated before and after with a battery of tests, including positron emission tomography with the norepinephrine analog (11)C-meta-hydroxyephedrine. Imaging 1 week after neurotoxin treatment revealed nearly complete loss of specific radioligand uptake. Partial progressive recovery of cardiac uptake found between 1 and 10 weeks remained stable between 10 and 14 weeks. In all five animals, examination of the pattern of uptake (using Logan plot analysis to create distribution volume maps) revealed a persistent region-specific significant loss in the inferior wall of the left ventricle at 10 (P<0.001) and 14 weeks (P<0.01) relative to the anterior wall. Blood levels of dopamine, norepinephrine (P<0.05), epinephrine, and 3,4-dihydroxyphenylacetic acid (P<0.01) were notably decreased after 6-hydroxydopamine at all time points. These results demonstrate that systemic injection of 6-hydroxydopamine in nonhuman primates creates a nonuniform but reproducible pattern of cardiac denervation as well as a persistent loss of circulating catecholamines, supporting the use of this method to further develop a monkey model of cardiac dysautonomia.


Asunto(s)
Efedrina/análogos & derivados , Corazón/diagnóstico por imagen , Oxidopamina/toxicidad , Radiofármacos , Ácido 3,4-Dihidroxifenilacético/sangre , Animales , Presión Sanguínea/efectos de los fármacos , Radioisótopos de Carbono/química , Dopamina/sangre , Ecocardiografía , Efedrina/sangre , Efedrina/química , Femenino , Macaca mulatta , Masculino , Neuronas/efectos de los fármacos , Norepinefrina/sangre , Oxidopamina/química , Tomografía de Emisión de Positrones , Disautonomías Primarias/diagnóstico , Radiofármacos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...