Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Neurotoxicology ; 97: 120-132, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37302585

RESUMEN

Copper (Cu) and Zinc (Zn) are required in small concentrations for metabolic functions, but are also toxic. There is a great concern about soil pollution by heavy metals, which may exposure the population to these toxicants, either by inhalation of dust or exposure to toxicants through ingestion of food derived from contaminated soils. In addition, the toxicity of metals in combination is questionable, as soil quality guidelines only assess them separately. It is well known that metal accumulation is often found in the pathologically affected regions of many neurodegenerative diseases, including Huntington's disease (HD). HD is caused by an autosomal dominantly inherited CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. This results in the formation of a mutant huntingtin (mHTT) protein with an abnormally long polyglutamine (polyQ) repeat. The pathology of HD results in loss of neuronal cells, motor changes, and dementia. Rutin is a flavonoid found in various food sources, and previous studies indicate it has protective effects in HD models and acts as a metal chelator. However, further studies are needed to unravel its effects on metal dyshomeostasis and to discern the underlying mechanisms. In the present study, we investigated the toxic effects of long-term exposure to copper, zinc, and their mixture, and the relationship with the progression of neurotoxicity and neurodegeneration in a C. elegans-based HD model. Furthermore, we investigated the effects of rutin post metal exposure. Overall, we demonstrate that chronic exposure to the metals and their mixture altered body parameters, locomotion, and developmental delay, in addition to increasing polyQ protein aggregates in muscles and neurons causing neurodegeneration. We also propose that rutin has protective effects acting through mechanisms involving antioxidant and chelating properties. Altogether, our data provides new indications about the higher toxicity of metals in combination, the chelating potential of rutin in the C. elegans model of HD and possible strategies for future treatments of neurodegenerative diseases caused by the aggregation of proteins related to metals.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Animales , Humanos , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/prevención & control , Enfermedad de Huntington/genética , Caenorhabditis elegans , Cobre/toxicidad , Zinc , Rutina/farmacología , Modelos Animales de Enfermedad
3.
Eur J Pharmacol ; 927: 175046, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35623405

RESUMEN

Oxidative stress induced neurotoxicity is increasingly perceived as an important neuropathologic mechanism underlying the motor and behavioral phenotypes associated with Huntington's disease (HD). Repeated exposure to 3-nitropropionic acid (3-NP) induces neurotoxic changes which closely simulate the neuropathological and behavioral characteristics of HD. This study aimed at evaluating the prophylactic effects of the dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) inhibitor "harmine" against 3-NP-indued neurotoxicity and HD-like symptoms. The potential prophylactic effect of harmine (10 mg/kg/day; intraperitoneal) was investigated on 3-NP-induced motor and cognitive HD-like deficits, nuclear factor erythroid 2 like 2 (NRF2), AMP kinase (AMPK) and p21 protein levels and the gene expression of haem oxygenase-1 (Ho-1), NAD(P)H: quinone oxidoreductase-1 (Nqo-1) and p62 in addition to redox imbalance and histological neurotoxic changes in the striatum, prefrontal cortex, and hippocampus of male Wistar rats. Harmine successfully increased the protein levels of NRF2, AMPK and p21 and the gene expression of Ho-1, Nqo-1 and p62, restored redox homeostasis, and reduced CASPASE-3 level. This was reflected in attenuation of 3-NP-induced neurodegenerative changes and improvement of rats' motor and cognitive performance. This study draws attention to the protective role of harmine against 3-NP-induced motor and cognitive dysfunction that could be mediated via enhancing NRF2-mediated signaling with subsequent amelioration of oxidative stress injury via NRF2 activators, p21 and AMPK, in the striatum, prefrontal cortex, and hippocampus which could offer a promising therapeutic tool to slow the progression of HD.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Harmina , Enfermedad de Huntington , Fármacos Neuroprotectores , Síndromes de Neurotoxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Adenilato Quinasa/metabolismo , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Harmina/farmacología , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/prevención & control , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/efectos adversos , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/prevención & control , Nitrocompuestos/antagonistas & inhibidores , Nitrocompuestos/farmacología , Estrés Oxidativo , Propionatos/antagonistas & inhibidores , Propionatos/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
4.
ACS Chem Neurosci ; 13(6): 721-732, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35226456

RESUMEN

Oxidative stress and mitochondrial dysfunction are among the mechanisms expected to explain the pathogenesis of Huntington's disease. Erythropoietin (EPO) and the Bacillus Calmette-Guérin (BCG) vaccine have neuroprotective effects against neurodegenerative diseases; however, the full mechanisms of their action are currently unclear. Here, for the first time, we investigated the neuroprotective effect of BCG vaccination in Huntington-like disease induced by 3-nitropropionic acid (3-NP) and its combination with EPO. Male Wistar rats were randomized into five groups: saline-treated control; 3-NP group (20 mg/kg/day, i.p.) for 7 days; EPO-treated group (5000 IU/kg/day, i.p.) for 14 days after 3-NP administration; live BCG vaccine prophylactic group (5000 cfu/g, i.p.) 10 days prior to 3-NP administration; and live BCG vaccine (5000 cfu/g, i.p.) 10 days before 3-NP administration, followed by EPO treatment (5000 IU/kg/day, i.p.) for 14 days. In a histopathological examination, striatum neurodegeneration was evidenced in the 3-NP injected rats. Administration of 3-NP elevated the levels of p-PI3K, p-Akt, p-mTOR, p-P70S6K, BAX, malondialdehyde, nitric oxide, and cytochrome oxidase while reduced the levels of BCL-2, superoxide dismutase, reduced glutathione, and the autophagy marker microtubule-associated protein light chain 3 in the striatum. EPO and BCG ameliorated the biochemical, histopathological, and behavioral derangements induced by 3-NP, with prominent neuroprotection observed in rats administered the BCG prophylactic combined with EPO treatment. These results highlight the role played by EPO and BCG in the management of 3-NP-induced Huntington-like disease by inhibiting the PI3K/Akt/mTOR/P70S6K pathway and enhancing the autophagy.


Asunto(s)
Eritropoyetina , Enfermedad de Huntington , Fármacos Neuroprotectores , Animales , Autofagia , Vacuna BCG , Eritropoyetina/uso terapéutico , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/prevención & control , Masculino , Fármacos Neuroprotectores/uso terapéutico , Nitrocompuestos/efectos adversos , Fosfatidilinositol 3-Quinasas , Propionatos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Proteínas Quinasas S6 Ribosómicas 70-kDa/uso terapéutico , Serina-Treonina Quinasas TOR , Vacunación
5.
Nutr Neurosci ; 25(9): 1898-1908, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33856270

RESUMEN

BACKGROUND: Naringenin is a powerful antioxidant and anti-inflammatory flavonoid which has been widely used as a therapeutic agent in various toxic models. However, few studies have clearly discussed the neuromodulatory effects of naringenin against different neurodegenerative disorders. AIM: We investigated the neuroprotective efficacy of naringenin against 3-nitropropionic acid (3-NP)-induced neurobehavioral, biochemical and histopathological alterations in rats. METHODS: Albino Wistar rats were randomly divided into three experimental groups. Group 1, the vehicle administered group, received saline. Group 2 received 3-NP (20 mg/kg body weight, i.p.) for 4 consecutive days. Group 3 received naringenin (50 mg/kg body weight, p.o.) twice daily for a period of 4 days, 30 min before and 6 h after the 3-NP administration. On the 5th day, neurobehavioral experiments were performed to access the behavioral outcomes and the striatum tissue was used for analysis of the monoamine oxidase (MAO) activity and serotonin (5-HT) levels. In addition, astrocytes activation was observed by glial fibrillary acidic protein (GFAP) immunostaining. RESULTS: Our results showed that naringenin co-treatment provides neuroprotection against 3-NP-induced neurological disorders. Naringenin also increased the MAO activity and 5-HT levels in the striatum. Moreover, co-treatment with naringenin reduced the expression of GFAP protein in the striatal part and significantly attenuated the neuronal cell death. The findings of the present study suggest that naringenin provides neuroprotection and mitigates neurobehavioral alterations in experimental rats. CONCLUSION: The results show that co-treatment with naringenin ameliorates 3-NP-induced HD-like symptoms in rats.


Asunto(s)
Flavanonas , Enfermedad de Huntington , Fármacos Neuroprotectores , Animales , Antioxidantes/uso terapéutico , Peso Corporal , Cuerpo Estriado , Modelos Animales de Enfermedad , Flavanonas/uso terapéutico , Proteína Ácida Fibrilar de la Glía/metabolismo , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/prevención & control , Monoaminooxidasa/metabolismo , Monoaminooxidasa/farmacología , Monoaminooxidasa/uso terapéutico , Actividad Motora , Neuroprotección , Fármacos Neuroprotectores/farmacología , Nitrocompuestos/toxicidad , Propionatos/toxicidad , Ratas , Ratas Wistar , Serotonina/metabolismo
6.
Naunyn Schmiedebergs Arch Pharmacol ; 394(9): 1917-1928, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34061228

RESUMEN

Mitochondrial abnormalities and redox imbalance are major pathogenic factors in progression of Huntington's disease (HD), manifested clinically by affective, motor, cognitive, and psychiatric incompetence. Antioxidants behold much promise in mitigation of several pathological facets in HD. Ellagic acid (EA) is a naturally derived polyphenol acknowledged for potent neuroprotective abilities that enabled its significance amongst popular brain tonics. The present study is aimed to examine the outcome of EA pre-treatment in 3-nitropropionic acid (3-NP) rat prototype of HD. Separate rat groups were pre-treated with EA (25, 50, and 100 mg/kg, p.o.) for 21 days and 3-NP (10 mg/kg, i.p.) was given for 14 days alongside to induce symptoms of HD. The physical/motor functions (narrow beam paradigm, footprint study, hanging-wire assessment) and cognitive abilities using elevated plus maze and novel object recognition task were evaluated. Entire brain was isolated and succinate dehydrogenase activity and parameters of oxido-nitrosative stress were assessed in mitochondrial fraction. 3-NP accrued oxido-nitrosative stress and significant decrease in succinate dehydrogenase activity caused motor and cognitive deficits in rats. EA pre-treatment resurrected succinate dehydrogenase activity in 3-NP treated rats that indicated preservation of mitochondrial function. A significant decrease in thiobarbituric acid reactive substances and nitrite levels and increase in glutathione and catalase activity by EA in 3-NP treated rats was noted. EA protected the rats against 3-NP triggered cognitive insufficiency and motor disturbances. It can be inferred that ellagic acid protects against 3-NP induced mitochondrial dysfunction and oxido-nitrosative stress in the brain. EA supplements or nutraceuticals might possess protective potential against symptoms of HD.


Asunto(s)
Trastornos del Conocimiento/prevención & control , Ácido Elágico/farmacología , Enfermedad de Huntington/prevención & control , Fármacos Neuroprotectores/farmacología , Animales , Catalasa/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ácido Elágico/administración & dosificación , Glutatión/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Nitrocompuestos , Propionatos , Ratas , Ratas Wistar
7.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33947817

RESUMEN

DNA damage plays a central role in the cellular pathogenesis of polyglutamine (polyQ) diseases, including Huntington's disease (HD). In this study, we showed that the expression of untranslatable expanded CAG RNA per se induced the cellular DNA damage response pathway. By means of RNA sequencing (RNA-seq), we found that expression of the Nudix hydrolase 16 (NUDT16) gene was down-regulated in mutant CAG RNA-expressing cells. The loss of NUDT16 function results in a misincorporation of damaging nucleotides into DNAs and leads to DNA damage. We showed that small CAG (sCAG) RNAs, species generated from expanded CAG transcripts, hybridize with CUG-containing NUDT16 mRNA and form a CAG-CUG RNA heteroduplex, resulting in gene silencing of NUDT16 and leading to the DNA damage and cellular apoptosis. These results were further validated using expanded CAG RNA-expressing mouse primary neurons and in vivo R6/2 HD transgenic mice. Moreover, we identified a bisamidinium compound, DB213, that interacts specifically with the major groove of the CAG RNA homoduplex and disfavors the CAG-CUG heteroduplex formation. This action subsequently mitigated RNA-induced silencing complex (RISC)-dependent NUDT16 silencing in both in vitro cell and in vivo mouse disease models. After DB213 treatment, DNA damage, apoptosis, and locomotor defects were rescued in HD mice. This work establishes NUDT16 deficiency by CAG repeat RNAs as a pathogenic mechanism of polyQ diseases and as a potential therapeutic direction for HD and other polyQ diseases.


Asunto(s)
Apoptosis/genética , Daño del ADN , Enfermedad de Huntington/genética , Péptidos/genética , Pirofosfatasas/genética , ARN/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Apoptosis/efectos de los fármacos , Benzamidinas/metabolismo , Benzamidinas/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/prevención & control , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación de Dinámica Molecular , Pirofosfatasas/metabolismo , ARN/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-33991587

RESUMEN

There is currently no effective treatment either for neurological illnesses (ischemia and neurodegenerative diseases) or psychiatric disorders (depression), in which the Glu/GABA balance is disturbed and accompanied by significant excitotoxicity. Therefore, the search for new and effective therapeutic strategies is imperative for these disorders. Studies conducted over the last several years indicate that the neuropeptide Y (NPY)-ergic system may be a potential therapeutic target for neuroprotective or antidepressant compounds. This review focuses on the neuroprotective roles of Y2 and Y5 receptors (YRs) in neurological disorders such as ischemia, Alzheimer's disease, Parkinson's disease, Huntington's disease, and in psychiatric disorders such as depression. It summarizes current knowledge on the possible mechanisms underlying the neuroprotective or antidepressant-like actions of Y2R and Y5R ligands. The review also discusses ligands acting at Y2R and Y5R and their limitations as in vivo pharmacological tools. The results from the preclinical studies discussed here may be useful in developing effective therapeutic strategies to treat neurological diseases on the one hand and psychiatric disorders on the other, and may pave the way for the development of novel Y2R and Y5R ligands as candidate drugs for the treatment of these diseases.


Asunto(s)
Descubrimiento de Drogas , Neuropéptido Y/farmacología , Neuroprotección/efectos de los fármacos , Receptores de Neuropéptido Y/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Animales , Depresión/tratamiento farmacológico , Depresión/prevención & control , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/prevención & control , Isquemia/tratamiento farmacológico , Isquemia/prevención & control , Ratones , Sinapsis/metabolismo
9.
ASN Neuro ; 13: 17590914211009857, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33906482

RESUMEN

Huntington's disease (HD) is a genetic disorder marked by transcriptional alterations that result in neuronal impairment and death. MicroRNAs (miRNAs) are non-coding RNAs involved in post-transcriptional regulation and fine-tuning of gene expression. Several studies identified altered miRNA expression in HD and other neurodegenerative diseases, however their roles in early stages of HD remain elusive. Here, we deep-sequenced miRNAs from the striatum of the HD mouse model, BACHD, at the age of 2 and 8 months, representing the pre-symptomatic and symptomatic stages of the disease. Our results show that 44 and 26 miRNAs were differentially expressed in 2- and 8-month-old BACHD mice, respectively, as compared to wild-type controls. Over-representation analysis suggested that miRNAs up-regulated in 2-month-old mice control the expression of genes crucial for PI3K-Akt and mTOR cell signaling pathways. Conversely, miRNAs regulating genes involved in neuronal disorders were down-regulated in 2-month-old BACHD mice. Interestingly, primary striatal neurons treated with anti-miRs targeting two up-regulated miRNAs, miR-449c-5p and miR-146b-5p, showed higher levels of cell death. Therefore, our results suggest that the miRNAs altered in 2-month-old BACHD mice regulate genes involved in the promotion of cell survival. Notably, over-representation suggested that targets of differentially expressed miRNAs at the age of 8 months were not significantly enriched for the same pathways. Together, our data shed light on the role of miRNAs in the initial stages of HD, suggesting a neuroprotective role as an attempt to maintain or reestablish cellular homeostasis.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Enfermedad de Huntington/genética , MicroARNs/biosíntesis , MicroARNs/genética , Neuroprotección/fisiología , Síntomas Prodrómicos , Animales , Células Cultivadas , Femenino , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Secuencia de ARN/métodos , Regulación hacia Arriba/fisiología
10.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998479

RESUMEN

With aging, the nervous system gradually undergoes degeneration. Increased oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, and cell death are considered to be common pathophysiological mechanisms of various neurodegenerative diseases (NDDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), organophosphate-induced delayed neuropathy (OPIDN), and amyotrophic lateral sclerosis (ALS). Autophagy is a cellular basic metabolic process that degrades the aggregated or misfolded proteins and abnormal organelles in cells. The abnormal regulation of neuronal autophagy is accompanied by the accumulation and deposition of irregular proteins, leading to changes in neuron homeostasis and neurodegeneration. Autophagy exhibits both a protective mechanism and a damage pathway related to programmed cell death. Because of its "double-edged sword", autophagy plays an important role in neurological damage and NDDs including AD, PD, HD, OPIDN, and ALS. Melatonin is a neuroendocrine hormone mainly synthesized in the pineal gland and exhibits a wide range of biological functions, such as sleep control, regulating circadian rhythm, immune enhancement, metabolism regulation, antioxidant, anti-aging, and anti-tumor effects. It can prevent cell death, reduce inflammation, block calcium channels, etc. In this review, we briefly discuss the neuroprotective role of melatonin against various NDDs via regulating autophagy, which could be a new field for future translational research and clinical studies to discover preventive or therapeutic agents for many NDDs.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/prevención & control , Esclerosis Amiotrófica Lateral/prevención & control , Enfermedad de Huntington/prevención & control , Melatonina/farmacología , Enfermedad de Parkinson/prevención & control , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Relacionadas con la Autofagia/agonistas , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Ritmo Circadiano/fisiología , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Melatonina/biosíntesis , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/metabolismo , Sistema Nervioso/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Glándula Pineal/fisiología
11.
Neurobiol Dis ; 141: 104943, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32407769

RESUMEN

Huntington's disease (HD) is caused by a highly polymorphic CAG trinucleotide expansion in the gene encoding for the huntingtin protein (HTT). The resulting mutant huntingtin protein (mutHTT) is ubiquitously expressed but also exhibits the ability to propagate from cell-to-cell to disseminate pathology; a property which may serve as a new therapeutic focus. Accordingly, we set out to develop a monoclonal antibody (mAB) targeting a particularly exposed region close to the aa586 caspase-6 cleavage site of the HTT protein. This monoclonal antibody, designated C6-17, effectively binds mutHTT and is able to deplete the protein from cell culture supernatants. Using cell-based assays, we demonstrate that extracellular secretion of mutHTT into cell culture media and its subsequent uptake in recipient HeLa cells can be almost entirely blocked by mAB C6-17. Immunohistochemical stainings of post-mortem HD brain tissue confirmed the specificity of mAB C6-17 to human mutHTT aggregates. These findings demonstrate that mAB C6-17 not only successfully engages with its target, mutHTT, but also inhibits cell uptake suggesting that this antibody could interfere with the pathological processes of mutHTT spreading in vivo.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/inmunología , Enfermedad de Huntington/metabolismo , Animales , Transporte Biológico , Femenino , Células HEK293 , Células HeLa , Humanos , Enfermedad de Huntington/prevención & control , Ratones Endogámicos BALB C , Mutación , Agregación Patológica de Proteínas/inmunología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/prevención & control
12.
Food Chem Toxicol ; 141: 111323, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32278002

RESUMEN

Huntington's disease (HD) is inherited neurodegenerative disease, it is characterized by excessive motor movements and cognitive and emotional deficits. HD is caused by an abnormally long polyglutamine (polyQ) expansion in the huntingtin (Htt) protein, which confers toxic functions to mutant Htt leading to neurodegeneration. Rutin is a flavonoid found in plants, buckwheat, some teas and also in apples. Although previous studies have already indicated that rutin has some protective effects in HD's models, the underlying mechanisms are still unknown. In our study, we investigated the effects of rutin in Caenorhabditis elegans model of HD. We assessed polyQ aggregation, oxidative damage, neurodegeneration level and lifespan, and investigated the possible role of autophagy and insulin/IGF1 (IIS) signaling pathways in the beneficial effects induced by rutin. Overall, our data demonstrate that chronic rutin treatment reduced polyglutamine (polyQ) protein aggregation in muscle, reduced polyQ-mediated neuronal death in ASH sensory neurons, and extended lifespan. The possible mechanisms involved are antioxidant activity, activation of protein degradation (autophagy) and insulin/IGF1 (IIS) signaling pathways. These findings indicate that rutin consumption might be helpful in preventing HD and also provide possible pathways to be explored to search for new therapies against proteinopathies related to aging.


Asunto(s)
Autofagia/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Enfermedad de Huntington/prevención & control , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Rutina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Conducta Animal/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Longevidad , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
13.
Neurotox Res ; 37(1): 77-92, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31332714

RESUMEN

Systemic administration of 3-nitropropionic acid (3-NPA) is commonly used to induce Huntington's disease (HD)-like symptoms in experimental animals. Here, the potential neuroprotective efficiency of rutin and selenium (RSe) co-administration on 3-NPA-induced HD-like symptoms model in mice was investigated. 3-NPA injection evoked severe alterations in redox status, as indicated via increased striatal malondialdehyde and nitric oxide levels, accompanied by a decrease in levels of antioxidant molecules including glutathione, glutathione peroxidase, glutathione reductase, superoxide dismutase, and catalase. Moreover, 3-NPA potentiated inflammatory status by enhancing the production of interleukin-1ß, tumor necrosis factor-α, and myeloperoxidase activity. Pro-apoptotic cascade was also recorded in the striatum as evidenced through upregulation of cleaved caspase-3 and Bax, and downregulation of Bcl-2. 3-NPA activated astrocytes as indicated by the upregulated glial fibrillary acidic protein and inhibited brain-derived neurotrophic factor. Furthermore, perturbations in cholinergic and monoaminergic systems were observed. RSe provided neuroprotective effects by preventing body weight loss, oxidative stress, neuroinflammation, and the apoptotic cascade. RSe inhibited the activation of astrocytes, increased brain-derived neurotrophic factor, and improved cholinergic and monoaminergic transmission following 3-NPA intoxication. Taken together, RSe co-administration may prevent or delay the progression of HD and its associated impairments through its antioxidant, anti-inflammatory, anti-apoptotic, and neuromodulatory effects.


Asunto(s)
Enfermedad de Huntington/prevención & control , Estrés Oxidativo/efectos de los fármacos , Rutina/farmacología , Selenio/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Caspasa 3 , Catalasa/metabolismo , Cuerpo Estriado/metabolismo , Regulación hacia Abajo , Sinergismo Farmacológico , Proteína Ácida Fibrilar de la Glía/biosíntesis , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/metabolismo , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/metabolismo , Interleucina-1beta/biosíntesis , Masculino , Malondialdehído/metabolismo , Ratones , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Nitrocompuestos , Peroxidasa/metabolismo , Propionatos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Superóxido Dismutasa/metabolismo , Transmisión Sináptica/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba , Proteína X Asociada a bcl-2/biosíntesis
14.
ACS Chem Neurosci ; 11(3): 328-343, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31880908

RESUMEN

Huntington's disease (HD), a genetic neurodegenerative disease, is caused by an expanded polyglutamine (polyQ) domain in the first exon of the huntingtin protein (htt). PolyQ expansion destabilizes protein structure, resulting in aggregation into a variety of oligomers, protofibrils, and fibrils. Beyond the polyQ domain, adjacent protein sequences influence the aggregation process. Specifically, the first 17 N-terminal amino acids (Nt17) directly preceding the polyQ domain promote the formation of α-helix-rich oligomers that represent intermediate species associated with fibrillization. Due to its propensity to form an amphipathic α-helix, Nt17 also facilitates lipid binding. Three lysine residues (K6, K9, and K15) within Nt17 can be SUMOylated, which modifies htt's accumulation and toxicity within cells in a variety of HD models. The impact of SUMOylation on htt aggregation and direct interaction with lipid membranes was investigated. SUMOylation of htt-exon1 inhibited fibril formation while promoting larger, amorphous aggregate species. These amorphous aggregates were SDS soluble but nonetheless exhibited levels of ß-sheet structure similar to that of htt-exon1 fibrils. In addition, SUMOylation prevented htt binding, aggregation, and accumulation on model lipid bilayers comprised of total brain lipid extract. Collectively, these observations demonstrate that SUMOylation promotes a distinct htt aggregation pathway that may affect htt toxicity.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/prevención & control , Lípidos de la Membrana/metabolismo , Amiloide/metabolismo , Humanos , Proteína Huntingtina/genética , Proteínas del Tejido Nervioso/metabolismo , Sumoilación
15.
Pharmacol Res ; 150: 104371, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31415915

RESUMEN

This paper assesses in vivo cytotoxicity models of Huntington's disease (HD). Nearly 150 agents were found to be moderately to highly effective in mitigating the pathological sequelae of cytotoxic induction of HD features in multiple rodent models. Typically, rodents are treated with a prospective HD-protective agent before, during, or after the application of a chemical or transgenic process for inducing histopathological and behavioral symptoms of HD. Although transgenic and knockout rodent models (1) display relatively high construct and face validity, and (2) are ever more routinely employed to mimic genetic-to-phenotypic expression of HD features, toxicant models are also often employed, and have served as valuable test beds for the elucidation of biochemical processes and discovery of therapeutic targets in HD. Literature searches of the toxicant HD rodent models yielded nearly 150 agents that were moderately to highly effective in mitigating pathological sequelae in multiple mouse and rat HD models. Experimental models, study designs, and exposure protocols (e.g., pre- and post-conditioning) used in testing these agents were assessed, including dosing strategies, endpoints, and dose-response features. Hormetic-like biphasic dose responses, chemoprotective mechanisms, and the translational relevance of the preclinical studies and their therapeutic implications are critically analyzed in the present report. Notably, not one of the 150 agents that successfully delayed onset and progression of HD in the experimental models has been successfully translated to the treatment of humans in a clinical setting. Potential reasons for these translational failures are (1) the inadequacy of dose-response analyses and subsequent lack of useful dosing data; (2) effective rodent doses that are too high for safe human application; (3) key differences between the experimental models and humans in pharmacokinetic/pharmacodynamic features, ages and routes of agent administration; (4) lack of robust pharmacokinetic, mechanistic or systematic approaches to probe novel treatment strategies; and (5) inadequacies of the chemically induced HD model in rats to mimic accurately the complex genetic and developmental origin and progression of HD in humans. These deficiencies need to be urgently addressed if pharmaceutical agents for the treatment of HD are going to be successfully developed in experimental models and translated with fidelity to the clinic.


Asunto(s)
Hormesis , Enfermedad de Huntington/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Investigación Biomédica Traslacional/métodos , Animales , Humanos , Enfermedad de Huntington/patología , Investigación Biomédica Traslacional/estadística & datos numéricos
17.
Oxid Med Cell Longev ; 2019: 4032428, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31049134

RESUMEN

Tolfenamic acid is a nonsteroidal anti-inflammatory drug with neuroprotective properties, and it alleviates learning and memory deficits in the APP transgenic mouse model of Alzheimer's disease. However, whether tolfenamic acid can prevent motor and memory dysfunction in transgenic animal models of Huntington's disease (HD) remains unclear. To this end, tolfenamic acid was orally administered to transgenic R6/1 mice from 10 to 20 weeks of age, followed by several behavioral tests to evaluate motor and memory function. Tolfenamic acid improved motor coordination in R6/1 mice as tested by rotarod, grip strength, and locomotor behavior tests and attenuated memory dysfunction as analyzed using the novel object recognition test and passive avoidance test. Tolfenamic acid decreased the expression of mutant huntingtin in the striatum of 20-week-old R6/1 mice by inhibiting specificity protein 1 expression and enhancing autophagic function. Furthermore, tolfenamic acid exhibited antioxidant effects in both R6/1 mice and PC12 cell models. Collectively, these results suggest that tolfenamic acid has a good therapeutic effect on R6/1 mice, and may be a potentially useful agent in the treatment of HD.


Asunto(s)
Antioxidantes/farmacología , Conducta Animal/efectos de los fármacos , Enfermedad de Huntington , Trastornos de la Memoria , Desempeño Psicomotor/efectos de los fármacos , ortoaminobenzoatos/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/prevención & control , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Trastornos de la Memoria/prevención & control , Ratones , Ratones Transgénicos , Mutación , Células PC12 , Ratas
18.
Biochimie ; 153: 70-79, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30107216

RESUMEN

Huntington's Disease (HD) is an autosomal dominant neurodegenerative disease caused by abnormal polyglutamine expansion in huntingtin (mHtt) protein leading to degeneration of striatal neurons. Excitotoxicity, consecutive to overstimulation of N-methyl d-aspartate receptors (NMDARs) has a pivotal role in many neurological disorders including HD. Mutant Htt causes enhanced NMDA sensitivity, alteration of NMDAR expression and localization in neurons. Excitotoxic events initiate neuronal death in numerous ways, including activation of apoptotic cascades. Among the NMDAR subunits involved in glutamatergic-mediated excitotoxicity, GluN2B has been extensively reported. In addition to excitotoxicity, alteration of cholesterol metabolism has been observed in HD, with a decrease of cholesterol precursor synthesis along with an increase of cholesterol accumulation, which is deleterious for neurons. Expression of Cholesterol Hydroxylase enzyme, CYP46A1, which converts cholesterol into 24 S-hydroxycholesterol is down-regulated in HD. We found that CYP46A1 overexpression is beneficial in HD neurons and mouse model, but the mechanisms involved still remain unclear. In this study we addressed the effect of CYP46A1 on NMDAR-mediated excitotoxicity in HD primary neurons and its role in modulating cholesterol and localization of GLUN2B in lipid rafts. We showed that CYP46A1 is protective against NMDAR-mediated excitotoxicity in two different HD neuronal cell models. Cholesterol as well as GluN2B level in lipid raft, are significantly increased by mHtt. Despite a clear effect of CYP46A1 in reducing cholesterol content in lipid raft extracts from wild type neurons, CYP46A1 overexpression in HD neurons could not normalize the increased cholesterol levels in lipid rafts. This study highlights the beneficial role of CYP46A1 against NMDAR-mediated excitotoxicity and gives further insights into the cellular mechanisms underlying CYP46A1-mediated neuroprotection.


Asunto(s)
Colesterol 24-Hidroxilasa/metabolismo , Enfermedad de Huntington/prevención & control , Microdominios de Membrana/metabolismo , N-Metilaspartato/toxicidad , Animales , Colesterol/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/enzimología , Femenino , Homeostasis , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , Masculino , Ratones , Mutación , Neuronas/enzimología , Receptores de N-Metil-D-Aspartato/metabolismo
19.
Metab Brain Dis ; 33(6): 1911-1921, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30054774

RESUMEN

Huntington's disease (HD) is characterized by cognitive and psychiatric impairment caused by neuronal degeneration in the brain. Several studies have supported the hypothesis that oxidative stress is the main pathogenic factor in HD. The current study aims to determine the possible neuroprotective effects of nicotinamide on 3-nitropropionic acid (3-NP) induced HD. Male Wistar albino rats were divided into six groups. Group I was the vehicle-treated control, group II received 3-NP (20 mg/kg, intraperitoneally (i.p.) for 4 days, group III received nicotinamide (500 mg/kg, i.p.). The remaining groups received a combination of 3-NP plus nicotinamide 100, 300 or 500 mg/kg, i.p. respectively for 8 days. Afterward, the motor function and hind paw activity in the limb withdrawal were tested; rats were then euthanized for biochemical and histopathological analyses. Treatment of rats with 3-NP altered the motor function, elevated oxidative stress and caused significant histopathological changes in the brain. The treatment of rats with nicotinamide (100, 300 and 500 mg/kg) improved the motor function tested by locomotor activity test, movement analysis, and limb withdrawal test, which was associated with decreased oxidative stress markers (malondialdehyde, nitrites) and increased antioxidant enzyme (glutathione) levels. In addition, nicotinamide treatment decreased lactate dehydrogenase and prevented neuronal death in the striatal region. Our study, therefore, concludes that antioxidant drugs like nicotinamide might slow progression of clinical HD and may improve the motor functions in HD patients. To the best of our knowledge, this study is the first to explore the neuroprotective effects of nicotinamide on 3-NP-induced HD.


Asunto(s)
Enfermedad de Huntington/metabolismo , Neuroprotección/efectos de los fármacos , Niacinamida/uso terapéutico , Nitrocompuestos/toxicidad , Poli(ADP-Ribosa) Polimerasas/metabolismo , Propionatos/toxicidad , Complejo Vitamínico B/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/prevención & control , Masculino , Neuroprotección/fisiología , Niacinamida/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar , Complejo Vitamínico B/farmacología
20.
J Neuropsychiatry Clin Neurosci ; 30(2): 115-121, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29183234

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disease involving motor, cognitive, and psychiatric/behavioral impairments that will eventually affect work role functioning. Few objective data exist regarding predictors of workplace disability in HD. The authors explored the predictors of work impairment and disability in a cross-sectional cohort of 656 employed, premanifest HD (preHD) individuals. In this cohort-the majority of whom were female, urban-dwelling, married/partnered, and working full-time, with minimal cognitive impairment, good function, minimal motor abnormality, and no indication of significant mental health issues-the number of participants who reported that they had missed work due to HD was low (2.4%). However, 12% of the study sample reported experiencing impairment while working due to preHD, 12.2% reported work-related activity impairment due to preHD, and 12.7% reported impairment in their overall work ability. Higher numbers of CAG repeats on the mutant allele and having more motor symptoms were associated with significantly higher odds of experiencing workplace impairment. Importantly, several modifiable factors were also found to predict workplace disability. Specifically, higher levels of anxiety symptoms were associated with significantly higher odds of experiencing workplace impairment. Good mental and physical health served as protective factors, where good physical health was associated with 6% lower odds of experiencing impairment or missing work time and good mental health was associated with of 10%-12% lower. The results provide important new knowledge for the development of future targeted intervention trials to support preHD individuals in maintaining their work roles as long as possible.


Asunto(s)
Empleo , Enfermedad de Huntington/prevención & control , Absentismo , Adulto , Estudios de Cohortes , Estudios Transversales , Evaluación de la Discapacidad , Femenino , Predicción , Indicadores de Salud , Humanos , Enfermedad de Huntington/genética , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Factores Socioeconómicos , Encuestas y Cuestionarios , Lugar de Trabajo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...