Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008791

RESUMO

Methamphetamine (METH) is a highly abused psychostimulant that is neurotoxic to dopaminergic (DAergic) nerve terminals in the striatum and increases the risk of developing Parkinson's disease (PD). In vivo, METH-mediated DA release, followed by DA-mediated oxidative stress and mitochondrial dysfunction in pre- and postsynaptic neurons, mediates METH neurotoxicity. METH-triggered oxidative stress damages parkin, a neuroprotective protein involved in PD etiology via its involvement in the maintenance of mitochondria. It is not known whether METH itself contributes to mitochondrial dysfunction and whether parkin regulates complex I, an enzymatic complex downregulated in PD. To determine this, we separately assessed the effects of METH or DA alone on electron transport chain (ETC) complexes and the protein parkin in isolated striatal mitochondria. We show that METH decreases the levels of selected complex I, II, and III subunits (NDUFS3, SDHA, and UQCRC2, respectively), whereas DA decreases the levels only of the NDUFS3 subunit in our preparations. We also show that the selected subunits are not decreased in synaptosomal mitochondria under similar experimental conditions. Finally, we found that parkin overexpression does not influence the levels of the NDUFS3 subunit in rat striatum. The presented results indicate that METH itself is a factor promoting dysfunction of striatal mitochondria; therefore, it is a potential drug target against METH neurotoxicity. The observed decreases in ETC complex subunits suggest that DA and METH decrease activities of the ETC complexes via oxidative damage to their subunits and that synaptosomal mitochondria may be somewhat "resistant" to DA- and METH-induced disruption in mitochondrial ETC complexes than perikaryal mitochondria. The results also suggest that parkin does not regulate NDUFS3 turnover in rat striatum.


Assuntos
Corpo Estriado/metabolismo , Dopamina/farmacologia , Metanfetamina/toxicidade , Neurotoxinas/toxicidade , Ubiquitina-Proteína Ligases/metabolismo , Animais , Soluções Tampão , Corpo Estriado/efeitos dos fármacos , Transporte de Elétrons/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , NADH Desidrogenase/metabolismo , Subunidades Proteicas/metabolismo , Ratos , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
2.
Eur J Neurosci ; 46(3): 1918-1932, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28661099

RESUMO

Methamphetamine (METH) is a highly abused psychostimulant that is associated with an increased risk for developing Parkinson's disease (PD). This enhanced vulnerability likely relates to the toxic effects of METH that overlap with PD pathology, for example, aberrant functioning of α-synuclein and parkin. In PD, peripheral factors are thought to contribute to central nervous system (CNS) degeneration. For example, α-synuclein levels in the enteric nervous system (ENS) are elevated, and this precedes the onset of motor symptoms. It remains unclear whether neurons of the ENS, particularly catecholaminergic neurons, exhibit signs of METH-induced toxicity as seen in the CNS. The aim of this study was to determine whether self-administered METH altered the levels of α-synuclein, parkin, tyrosine hydroxylase (TH), and dopamine-ß-hydroxylase (DßH) in the myenteric plexus of the distal colon ENS. Young adult male Sprague-Dawley rats self-administered METH for 3 h per day for 14 days and controls were saline-yoked. Distal colon tissue was collected at 1, 14, or 56 days after the last operant session. Levels of α-synuclein were increased, while levels of parkin, TH, and DßH were decreased in the myenteric plexus in the METH-exposed rats at 1 day following the last operant session and returned to the control levels after 14 or 56 days of forced abstinence. The changes were not confined to neurofilament-positive neurons. These results suggest that colon biomarkers may provide early indications of METH-induced neurotoxicity, particularly in young chronic METH users who may be more susceptible to progression to PD later in life.


Assuntos
Colo/efeitos dos fármacos , Metanfetamina/toxicidade , Plexo Mientérico/efeitos dos fármacos , Autoadministração , Animais , Biomarcadores/metabolismo , Colo/inervação , Colo/metabolismo , Condicionamento Operante , Dopamina beta-Hidroxilase/genética , Dopamina beta-Hidroxilase/metabolismo , Masculino , Metanfetamina/administração & dosagem , Plexo Mientérico/metabolismo , Ratos , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
3.
J Pharmacol Exp Ther ; 362(3): 474-488, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28630283

RESUMO

Understanding the relationship between the molecular mechanisms underlying neurotoxicity of high-dose methamphetamine (METH) and related clinical manifestations is imperative for providing more effective treatments for human METH users. This article provides an overview of clinical manifestations of METH neurotoxicity to the central nervous system and neurobiology underlying the consequences of administration of neurotoxic METH doses, and discusses implications of METH neurotoxicity for treatment of human abusers of the drug.


Assuntos
Metanfetamina/toxicidade , Síndromes Neurotóxicas/complicações , Transtornos Relacionados ao Uso de Anfetaminas/tratamento farmacológico , Animais , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Neurônios Dopaminérgicos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Metanfetamina/farmacologia , Vias Neurais/efeitos dos fármacos , Síndromes Neurotóxicas/tratamento farmacológico , Neurônios Serotoninérgicos/efeitos dos fármacos
4.
Nanotechnology ; 28(22): 224001, 2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28488596

RESUMO

The triblock copolymer polyethylenimine-polycaprolactone-polyethylene glycol (PEI-PCL-PEG) has been shown to spontaneously assemble into nano-sized particulate carriers capable of complexing with nucleic acids for gene delivery. The objective of this study was to investigate micelleplex characteristics, their in vitro and in vivo fate following microfluidic preparation of siRNA nanoparticles compared to the routinely used batch reactor mixing technique. Herein, PEI-PCL-PEG nanoparticles were prepared with batch reactor or microfluidic mixing techniques and characterized by various biochemical assays and in cell culture. Microfluidic nanoparticles showed a reduction of overall particle size as well as a more uniform size distribution when compared to batch reactor pipette mixing. Confocal microscopy, flow cytometry and qRT-PCR displayed the subcellular delivery of the microfluidic formulation and confirmed the ability to achieve mRNA knockdown. Intratracheal instillation of microfluidic formulation resulted in a significantly more efficient (p < 0.05) knockdown of GAPDH compared to treatment with the batch reactor formulation. The use of microfluidic mixing techniques yields an overall smaller and more uniform PEG-PCL-PEI nanoparticle that is able to more efficiently deliver siRNA in vivo. This preparation method may prove to be useful when a scaled up production of well-defined polyplexes is required.


Assuntos
Nanopartículas/química , Poliésteres/química , Polietilenoglicóis/química , Polietilenoimina/análogos & derivados , Polietilenoimina/química , Polímeros/química , Células A549 , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Portadores de Fármacos/química , Inativação Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Humanos , Microfluídica/métodos , Tamanho da Partícula , RNA Interferente Pequeno/química
5.
J Neurochem ; 136(3): 510-25, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26465779

RESUMO

Exposure to binge methamphetamine (METH) can result in a permanent or transient loss of dopaminergic (DAergic) markers such as dopamine (DA), dopamine transporter, and tyrosine hydroxylase (TH) in the striatum. We hypothesized that the METH-induced loss of striatal DAergic markers was, in part, due to a destabilization of microtubules (MTs) in the nigrostriatal DA pathway that ultimately impedes anterograde axonal transport of these markers. To test this hypothesis, adult male Sprague-Dawley rats were treated with binge METH or saline in the presence or absence of epothilone D (EpoD), a MT-stabilizing compound, and assessed 3 days after the treatments for the levels of several DAergic markers as well as for the levels of tubulins and their post-translational modifications (PMTs). Binge METH induced a loss of stable long-lived MTs within the striatum but not within the substantia nigra pars compacta (SNpc). Treatment with a low dose of EpoD increased the levels of markers of stable MTs and prevented METH-mediated deficits in several DAergic markers in the striatum. In contrast, administration of a high dose of EpoD appeared to destabilize MTs and potentiated the METH-induced deficits in several DAergic markers. The low-dose EpoD also prevented the METH-induced increase in striatal DA turnover and increased behavioral stereotypy during METH treatment. Together, these results demonstrate that MT dynamics plays a role in the development of METH-induced losses of several DAergic markers in the striatum and may mediate METH-induced degeneration of terminals in the nigrostriatal DA pathway. Our study also demonstrates that MT-stabilizing drugs such as EpoD have a potential to serve as useful therapeutic agents to restore function of DAergic nerve terminals following METH exposure when administered at low doses. Administration of binge methamphetamine (METH) negatively impacts neurotransmission in the nigrostriatal dopamine (DA) system. The effects of METH include decreasing the levels of DAergic markers in the striatum. We have determined that high-dose METH destabilizes microtubules in this pathway, which is manifested by decreased levels of acetylated (Acetyl) and detyrosinated (Detyr) α-tubulin (I). A microtubule stabilizing agent epothilone D protects striatal microtubules form the METH-induced loss of DAergic markers (II). These findings provide a new strategy for protection form METH - restoration of proper axonal transport.


Assuntos
Estimulantes do Sistema Nervoso Central/administração & dosagem , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Epotilonas/farmacologia , Metanfetamina/administração & dosagem , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Ácido Homovanílico/metabolismo , Masculino , Microtúbulos/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Anal Chem ; 88(7): 4071-84, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26937787

RESUMO

The aberrant aggregation of α-synuclein in the brain is a hallmark of Parkinson's disease (PD). In vivo soluble α-synuclein occurs as a monomer and several multimers, the latter of which may be important for the biological function of α-synuclein. Currently, there is a lack of reproducible methods to compare α-synuclein multimer abundance between complex biological samples. Here we developed a method, termed "multimer-PAGE," that combines in-gel chemical cross-linking with several common electrophoretic techniques to measure the stoichiometry of soluble α-synuclein multimers in brain tissue lysates. Results show that soluble α-synuclein from the rat brain exists as several high molecular weight species of approximately 56 kDa (αS56), 80 kDa (αS80), and 100 kDa (αS100) that comigrate with endogenous lipids, detergents, and/or micelles during blue native gel electrophoresis (BN-PAGE). Co-extraction of endogenous lipids with α-synuclein was essential for the detection of soluble α-synuclein multimers. Homogenization of brain tissue in small buffer volumes (>50 mg tissue per 1 mL buffer) increased relative lipid extraction and subsequently resulted in abundant soluble multimer detection via multimer-PAGE. α-Synuclein multimers captured by directly cross-linking soluble lysates resembled those observed following multimer-PAGE. The ratio of multimer (αS80) to monomer (αS17) increased linearly with protein input into multimer-PAGE, suggesting to some extent, multimers were also formed during electrophoresis. Overall, soluble α-synuclein maintains lipid interactions following tissue disruption and readily forms multimers when this lipid-protein complex is preserved. Once the multimer-PAGE technique was validated, relative stoichiometric comparisons could be conducted simultaneously between 14 biological samples. Multimer-PAGE provides a simple inexpensive biochemical technique to study the molecular factors influencing α-synuclein multimerization.


Assuntos
Eletroforese em Gel de Poliacrilamida/métodos , alfa-Sinucleína/análise , Animais , Encéfalo/metabolismo , Masculino , Multimerização Proteica , Ratos , Ratos Sprague-Dawley , alfa-Sinucleína/metabolismo
7.
Molecules ; 21(10)2016 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-27735873

RESUMO

The use of small interference RNA (siRNA) to target oncogenes is a promising treatment approach for cancer. However, siRNA cancer therapies are hindered by poor delivery of siRNA to cancer cells. Transferrin receptor (TfR) is overexpressed in many types of tumor cells and therefore is a potential target for the selective delivery of siRNA to cancer cells. Here, we used the TfR binding peptide HAIYPRH (HAI peptide) conjugated to cationic polymer branched polyethylenimine (bPEI), optimized the coupling strategy, and the TfR selective delivery of siRNA was evaluated in cells with high (H1299) and low TfR expression (A549 and H460). The HAI-bPEI conjugate exhibited chemico-physical properties in terms of size, zeta-potential, and siRNA condensation efficiency similar to unmodified bPEI. Confocal microscopy and flow cytometry results revealed that HAI-bPEI selectively delivered siRNA to H1299 cells compared with A549 or H460 cells. Moreover, HAI-bPEI achieved more efficient glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene knockdown in H1299 cells compared with bPEI alone. However, despite optimization of the targeting peptide and coupling strategy, HAI-bPEI can only silence reporter gene enhanced green fluorescent protein (eGFP) at the protein level when chloroquine is present, indicating that further optimization of the conjugate is required. In conclusion, the HAI peptide may be useful to target TfR overexpressing tumors in targeted gene and siRNA delivery approaches.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Peptídeos , Polietilenoimina , RNA Interferente Pequeno/genética , Receptores da Transferrina/genética , Transfecção , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Genes Reporter , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Humanos , Microscopia Confocal , Peptídeos/química , Polietilenoimina/química , Transfecção/métodos
8.
Mol Pharm ; 12(2): 362-74, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25437915

RESUMO

Amphiphilic nucleic acid carriers have attracted strong interest. Three groups of nylon-3 copolymers (poly-ß-peptides) possessing different cationic/hydrophobic content were evaluated as siRNA delivery agents in this study. Their ability to condense siRNA was determined in SYBR Gold assays. Their cytotoxicity was tested by MTT assays, their efficiency of delivering Alexa Fluor-488-labeled siRNA intracellularly in the presence and absence of uptake inhibitors was assessed by flow cytometry, and their transfection efficacies were studied by luciferase knockdown in a cell line stably expressing luciferase (H1299/Luc). Endosomal release was determined by confocal laser scanning microscopy and colocalization with lysotracker. All polymers efficiently condensed siRNA at nitrogen-to-phosphate (N/P) ratios of 5 or lower, as reflected in hydrodynamic diameters smaller than that at N/P 1. Although several formulations had negative zeta potentials at N/P 1, G2C and G2D polyplexes yielded >80% uptake in H1299/Luc cells, as determined by flow cytometry. Luciferase knockdown (20-65%) was observed after transfection with polyplexes made of the high molecular weight polymers that were the most hydrophobic. The ability of nylon-3 polymers to deliver siRNA intracellularly even at negative zeta potential implies that they mediate transport across cell membranes based on their amphiphilicity. The cellular uptake route was determined to strongly depend on the presence of cholesterol in the cell membrane. These polymers are, therefore, very promising for siRNA delivery at reduced surface charge and toxicity. Our study identified nylon-3 formulations at low N/P ratios for effective gene knockdown, indicating that nylon-3 polymers are a new, promising type of gene delivery agent.


Assuntos
Cátions/química , Técnicas de Transferência de Genes , Nylons/química , Polímeros/química , RNA Interferente Pequeno/química , Transfecção/métodos , Estrutura Molecular
9.
J Neurochem ; 128(5): 764-75, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24151877

RESUMO

Psychostimulant methamphetamine (METH) is toxic to striatal dopaminergic and serotonergic nerve terminals in adult, but not in the adolescent, brain. Betulinic acid (BA) and its derivatives are promising anti-HIV agents with some toxic properties. Many METH users, particularly young men, are HIV-positive; therefore, they might be treated with BA or its derivative for HIV infection. It is not known whether BA, or any of its derivatives, are neurotoxic in combination with METH in the adolescent brain. The present study investigated the effects of BA and binge METH in the striatum of late adolescent rats. BA or METH alone did not decrease the levels of dopaminergic or serotonergic markers in the striatum whereas BA and METH together decreased these markers in a BA dose-dependent manner. BA+METH also caused decreases in the levels of mitochondrial complex I in the same manner; BA alone only slightly decreased the levels of this enzyme in striatal synaptosomes. BA or METH alone increased cytochrome c. METH alone decreased parkin, increased complex II and striatal BA levels. These results suggest that METH in combination with BA can be neurotoxic to striatal dopaminergic and serotonergic nerve terminals in the late adolescent brain via mitochondrial dysfunction and parkin deficit. We report a synergistic neurotoxicity of betulinic acid (BA) and methamphetamine (METH) to monoaminergic terminals in the striatum of male late adolescent rats. BA contribution to the neurotoxicity is decreasing mitochondrial complex I whereas METH contribution is decreasing parkin and increasing brain concentration of BA. We propose that clinical use of BA in young male METH users can be neurotoxic.


Assuntos
Fármacos Anti-HIV/toxicidade , Dopaminérgicos/toxicidade , Neurônios Dopaminérgicos/efeitos dos fármacos , Metanfetamina/toxicidade , Neostriado/patologia , Terminações Nervosas/efeitos dos fármacos , Neurônios Serotoninérgicos/efeitos dos fármacos , Triterpenos/toxicidade , Animais , Western Blotting , Química Encefálica/efeitos dos fármacos , Caspase 3/metabolismo , Quimotripsina/metabolismo , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Complexo I de Transporte de Elétrons/efeitos dos fármacos , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/efeitos dos fármacos , Complexo II de Transporte de Elétrons/metabolismo , Febre/induzido quimicamente , Febre/fisiopatologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Neostriado/efeitos dos fármacos , Triterpenos Pentacíclicos , Ratos , Ratos Sprague-Dawley , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/patologia , Ubiquitina-Proteína Ligases/metabolismo , Ácido Betulínico
10.
Int J Mol Sci ; 15(4): 5884-906, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24717411

RESUMO

Methamphetamine (METH) is a central nervous system psychostimulant with a high potential for abuse. At high doses, METH causes a selective degeneration of dopaminergic terminals in the striatum. Dopamine D2 receptor antagonists and dopamine transporter (DAT) inhibitors protect against neurotoxicity of the drug by decreasing intracellular dopamine content and, consequently, dopamine autoxidation and production of reactive oxygen species. In vitro, amphetamines regulate D2 receptor and DAT functions via regulation of their intracellular trafficking. No data exists on axonal transport of both proteins and there is limited data on their interactions in vivo. The aim of the present investigation was to examine synaptosomal levels of presynaptic D2 autoreceptor and DAT after two different regimens of METH and to determine whether METH affects the D2 autoreceptor-DAT interaction in the rat striatum. We found that, as compared to saline controls, administration of single high-dose METH decreased D2 autoreceptor immunoreactivity and increased DAT immunoreactivity in rat striatal synaptosomes whereas binge high-dose METH increased immunoreactivity of D2 autoreceptor and had no effect on DAT immunoreactivity. Single METH had no effect on D2 autoreceptor-DAT interaction whereas binge METH increased the interaction between the two proteins in the striatum. Our results suggest that METH can affect axonal transport of both the D2 autoreceptor and DAT in an interaction-dependent and -independent manner.


Assuntos
Corpo Estriado/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/biossíntese , Inibidores da Captação de Dopamina/farmacologia , Metanfetamina/farmacologia , Receptores de Dopamina D2/biossíntese , Animais , Autorreceptores , Transporte Axonal/efeitos dos fármacos , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/imunologia , Febre/induzido quimicamente , Masculino , Metanfetamina/administração & dosagem , Síndromes Neurotóxicas , Ratos , Ratos Sprague-Dawley , Sinaptossomos/imunologia , Sinaptossomos/metabolismo
11.
Commun Biol ; 6(1): 991, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37758941

RESUMO

Psychostimulant methamphetamine (METH) is neurotoxic to the brain and, therefore, its misuse leads to neurological and psychiatric disorders. The gene regulatory network (GRN) response to neurotoxic METH binge remains unclear in most brain regions. Here we examined the effects of binge METH on the GRN in the nucleus accumbens, dentate gyrus, Ammon's horn, and subventricular zone in male rats. At 24 h after METH, ~16% of genes displayed altered expression and over a quarter of previously open chromatin regions - parts of the genome where genes are typically active - showed shifts in their accessibility. Intriguingly, most changes were unique to each area studied, and independent regulation between transcriptome and chromatin accessibility was observed. Unexpectedly, METH differentially impacted gene activity and chromatin accessibility within the dentate gyrus and Ammon's horn. Around 70% of the affected chromatin-accessible regions in the rat brain have conserved DNA sequences in the human genome. These regions frequently act as enhancers, ramping up the activity of nearby genes, and contain mutations linked to various neurological conditions. By sketching out the gene regulatory networks associated with binge METH in specific brain regions, our study offers fresh insights into how METH can trigger profound, region-specific molecular shifts.


Assuntos
Metanfetamina , Transcriptoma , Humanos , Masculino , Animais , Ratos , Metanfetamina/toxicidade , Encéfalo , Cromatina/genética , Epigênese Genética
12.
J Neurochem ; 116(6): 1005-17, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21166679

RESUMO

Methamphetamine (METH) is toxic to dopaminergic (DAergic) terminals in animals and humans. An early event in METH neurotoxicity is an oxidative stress followed by damage to proteins and lipids. The removal of damaged proteins is accomplished by the ubiquitin-proteasome system (UPS) and the impairment of this system can cause neurodegeneration. Whether dysfunction of the UPS contributes to METH toxicity to DAergic terminals has not been determined. The present investigation examined the effects of METH on functions of parkin and proteasome in rat striatal synaptosomes. METH rapidly modified parkin via conjugation with 4-hydroxy-2-nonenal (4-HNE) to decrease parkin levels and decreased the activity of the 26S proteasome while simultaneously increasing chymotrypsin-like activity and 20S proteasome levels. Prior injections of vitamin E diminished METH-induced changes to parkin and the 26S proteasome as well as long-term decreases in DA and its metabolites' concentrations in striatal tissue. These results suggest that METH causes lipid peroxidation-mediated damage to parkin and the 26S proteasome. As the changes in parkin and 26S occur before the sustained deficits in DAergic markers, an early loss of UPS function may be important in mediating the long-term degeneration of striatal DAergic terminals via toxic accumulation of parkin substrates and damaged proteins.


Assuntos
Inibidores da Captação de Dopamina/farmacologia , Metanfetamina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina-Proteína Ligases/efeitos dos fármacos , Aldeídos/farmacologia , Animais , Antioxidantes/administração & dosagem , Corpo Estriado/ultraestrutura , Cisteína/análogos & derivados , Cisteína/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Imunoprecipitação/métodos , Masculino , Ratos , Ratos Sprague-Dawley , S-Nitrosotióis/metabolismo , Estatísticas não Paramétricas , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Fatores de Tempo , Tubulina (Proteína)/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos dos fármacos , Vitamina E/administração & dosagem
13.
Curr Neuropharmacol ; 19(12): 2077-2091, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34344291

RESUMO

Almost two decades have passed since the last methamphetamine (METH) abuse epidemic. In recent years, METH abuse in the United States has been rapidly increasing and is currently one of the leading causes of death in our country. Available statistical data indicates reemergence of METH popularity and suggest an impending third epidemic of METH abuse. Alarmingly, there is no FDA-approved medication for METH use disorder (MUD). This disorder is currently treated with behavioral therapies; however, these therapies have limitations and would benefit from the addition of a MUD pharmacotherapy. Unfortunately, clinical trials have not yet found consistently effective pharmacotherapy for MUD. This review outlines the history of METH use, provides information on current prevalence of METH abuse and MUD, describes medications that have been in clinical trials for MUD, and addresses current as well as potential new treatments for MUD.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas , Estimulantes do Sistema Nervoso Central , Metanfetamina , Transtornos Relacionados ao Uso de Anfetaminas/tratamento farmacológico , Transtornos Relacionados ao Uso de Anfetaminas/terapia , Estimulantes do Sistema Nervoso Central/uso terapêutico , Humanos
14.
Exp Neurol ; 345: 113811, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34298012

RESUMO

Methamphetamine (METH) is a highly addictive and powerful central nervous system psychostimulant with no FDA-approved pharmacotherapy. Parkin is a neuroprotective protein and its loss of function contributes to Parkinson's disease. This study used 3-month-old homozygous parkin knockout (PKO) rats to determine whether loss of parkin protein potentiates neurotoxicity of chronic METH to the nigrostriatal dopamine pathway. PKO rats were chronically treated with 10 mg/kg METH for 10 consecutive days and assessed for neurotoxicity markers in the striatum on the 5th and 10th day of withdrawal from METH. The PKO rats showed higher METH-induced hyperthermia; however, they did not display augmented deficits in dopaminergic and serotonergic neurotoxicity markers, astrocyte activation or decreased mitochondrial enzyme levels as compared to wild-type (WT) rats. Interestingly, saline-treated PKO rats had lower levels of dopamine (DA) as well as mitochondrial complex I and II levels while having increased basal levels of glial fibrillary acidic protein (GFAP), a marker of gliosis. These results indicate PKO display a certain resistance to METH neurotoxicity, possibly mediated by lowered DA levels and downregulated mitochondria.


Assuntos
Estimulantes do Sistema Nervoso Central/toxicidade , Dopamina/metabolismo , Locomoção/efeitos dos fármacos , Metanfetamina/toxicidade , Ubiquitina-Proteína Ligases/deficiência , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Estimulantes do Sistema Nervoso Central/administração & dosagem , Dopamina/genética , Esquema de Medicação , Hipertermia Induzida/efeitos adversos , Hipertermia Induzida/métodos , Locomoção/fisiologia , Masculino , Metanfetamina/administração & dosagem , Ratos , Ratos Long-Evans , Ratos Transgênicos , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Ubiquitina-Proteína Ligases/genética
15.
Transl Psychiatry ; 11(1): 293, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001858

RESUMO

There is no FDA-approved medication for methamphetamine (METH) use disorder. New therapeutic approaches are needed, especially for people who use METH heavily and are at high risk for overdose. This study used genetically engineered rats to evaluate PARKIN as a potential target for METH use disorder. PARKIN knockout, PARKIN-overexpressing, and wild-type young adult male Long Evans rats were trained to self-administer high doses of METH using an extended-access METH self-administration paradigm. Reinforcing/rewarding properties of METH were assessed by quantifying drug-taking behavior and time spent in a METH-paired environment. PARKIN knockout rats self-administered more METH and spent more time in the METH-paired environment than wild-type rats. Wild-type rats overexpressing PARKIN self-administered less METH and spent less time in the METH-paired environment. PARKIN knockout rats overexpressing PARKIN self-administered less METH during the first half of drug self-administration days than PARKIN-deficient rats. The results indicate that rats with PARKIN excess or PARKIN deficit are useful models for studying neural substrates underlying "resilience" or vulnerability to METH use disorder and identify PARKIN as a novel potential drug target to treat heavy use of METH.


Assuntos
Estimulantes do Sistema Nervoso Central , Metanfetamina , Animais , Masculino , Ratos , Ratos Long-Evans , Autoadministração , Ubiquitina-Proteína Ligases/genética
16.
Genes (Basel) ; 11(4)2020 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-32231019

RESUMO

Methamphetamine (METH) is a widely abused psychostimulant with the potential to cause a broad range of severe cognitive deficits as well as neurobehavioral abnormalities when abused chronically, particularly at high doses. Cognitive deficits are related to METH neurotoxicity in the striatum and hippocampus. The activation of transposable Long INterspersed Nuclear Element 1 (LINE-1) is associated with several neurological diseases and drug abuse, but there are very limited data regarding the effects of high-dose METH on the activity of LINE-1 in the adult brain. Using real-time quantitative PCR, the present study demonstrates that the chronic administration of neurotoxic METH doses results in the increased expression of LINE-1-encoded Open Reading Frame 1 (ORF-1) in rat striatum shortly after the last dose of the drug and decreased ORF-1 expression during METH withdrawal, with dentate gyrus potentially developing "tolerance" to these METH effects. LINE-1 activation may be a new factor mediating the neurotoxic effects of chronic METH in the striatum and, therefore, a new drug target against METH-induced psychomotor impairments in chronic METH users.


Assuntos
Corpo Estriado/patologia , Giro Denteado/patologia , Elementos Nucleotídeos Longos e Dispersos , Metanfetamina/toxicidade , Síndromes Neurotóxicas/patologia , Fases de Leitura Aberta/genética , Córtex Pré-Frontal/patologia , Animais , Estimulantes do Sistema Nervoso Central/toxicidade , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Masculino , Síndromes Neurotóxicas/etiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley
17.
Sci Rep ; 10(1): 10150, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32576878

RESUMO

ATAC-seq is widely used to measure chromatin accessibility and identify open chromatin regions (OCRs). OCRs usually indicate active regulatory elements in the genome and are directly associated with the gene regulatory network. The identification of differential accessibility regions (DARs) between different biological conditions is critical in determining the differential activity of regulatory elements. Differential analysis of ATAC-seq shares many similarities with differential expression analysis of RNA-seq data. However, the distribution of ATAC-seq signal intensity is different from that of RNA-seq data, and higher sensitivity is required for DARs identification. Many different tools can be used to perform differential analysis of ATAC-seq data, but a comprehensive comparison and benchmarking of these methods is still lacking. Here, we used simulated datasets to systematically measure the sensitivity and specificity of six different methods. We further discussed the statistical and signal density cut-offs in the differential analysis of ATAC-seq by applying them to real data. Batch effects are very common in high-throughput sequencing experiments. We illustrated that batch-effect correction can dramatically improve sensitivity in the differential analysis of ATAC-seq data. Finally, we developed a user-friendly package, BeCorrect, to perform batch effect correction and visualization of corrected ATAC-seq signals in a genome browser.


Assuntos
Sequenciamento de Cromatina por Imunoprecipitação/métodos , Cromatina/genética , Redes Reguladoras de Genes/genética , Bases de Dados de Ácidos Nucleicos , Conjuntos de Dados como Assunto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Sensibilidade e Especificidade
18.
Sci Rep ; 8(1): 1517, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367643

RESUMO

Mutations in parkin gene (Park2) are linked to early-onset autosomal recessive Parkinson's disease (PD) and young-onset sporadic PD. Park2 knockout (PKO) rodents; however, do not display neurodegeneration of the nigrostriatal pathway, suggesting age-dependent compensatory changes. Our goal was to examine dopaminergic (DAergic) system in the striatum of 2 month-old PKO rats in order to characterize compensatory mechanisms that may have occurred within the system. The striata form wild type (WT) and PKO Long Evans male rats were assessed for the levels of DAergic markers, for monoamine oxidase (MAO) A and B activities and levels, and for the levels of their respective preferred substrates, serotonin (5-HT) and ß-phenylethylamine (ß-PEA). The PKO rats displayed lower activities of MAOs and higher levels of ß-PEA in the striatum than their WT counterparts. Decreased levels of ß-PEA receptor, trace amine-associated receptor 1 (TAAR-1), and postsynaptic DA D2 (D2L) receptor accompanied these alterations. Drug-naive PKO rats displayed normal locomotor activity; however, they displayed decreased locomotor response to a low dose of psychostimulant methamphetamine, suggesting altered DAergic neurotransmission in the striatum when challenged with an indirect agonist. Altogether, our findings suggest that 2 month-old PKO male rats have altered DAergic and trace aminergic signaling.


Assuntos
Corpo Estriado/patologia , Neurônios Dopaminérgicos/patologia , Ubiquitina-Proteína Ligases/deficiência , Animais , Técnicas de Inativação de Genes , Monoaminoxidase/análise , Fenetilaminas/análise , Ratos , Serotonina/análise
19.
Drug Alcohol Depend ; 190: 20-28, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29960919

RESUMO

BACKGROUND: Animal studies suggest that exposure to either of the two widely used drugs of abuse, heroin or cocaine, causes depletion of the antioxidant, reduced glutathione, a hallmark of oxidative stress, in the brain. However, the relevance of the animal findings to the human is uncertain and clinical trials with the antioxidant GSH precursor n-acetylcysteine have produced mixed results in cocaine dependence. METHODS: Our major objective was to compare glutathione levels, determined by an HPLC-coulometric procedure, in autopsied brain of chronic heroin (n = 11) and cocaine users (n = 9), who were positive for the drugs in the brain, to those of matched controls (n = 16). Six brain regions were examined, including caudate, hippocampus, thalamus and frontal, temporal and insular cortices. RESULTS: In contrast to experimental animal findings, we found no statistically significant difference between mean levels of reduced or oxidized glutathione in the drug user vs. control groups. Moreover, no correlation was found between levels of drugs in the brain and those of glutathione. CONCLUSIONS: Acknowledging the many generic limitations of an autopsied human brain study and the preliminary nature of the findings, our data nevertheless suggest that any oxidative stress caused by heroin or cocaine in chronic users of the drugs might not be sufficient to cause substantial loss of stores of glutathione in the human brain, at least during early withdrawal. These findings, requiring replication, might also have some relevance to future clinical trials employing glutathione supplement therapy as an anti-oxidative strategy in chronic users of the two abused drugs.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cocaína/administração & dosagem , Glutationa/metabolismo , Heroína/administração & dosagem , Adulto , Antioxidantes/metabolismo , Autopsia , Encéfalo/patologia , Cocaína/efeitos adversos , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/patologia , Feminino , Heroína/efeitos adversos , Dependência de Heroína/metabolismo , Dependência de Heroína/patologia , Humanos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
20.
J Control Release ; 276: 50-58, 2018 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-29474962

RESUMO

The block copolymer VIPER (virus-inspired polymer for endosomal release) has been reported to be a promising novel delivery system of DNA plasmids both in vitro and in vivo. VIPER is comprised of a polycation segment for condensation of nucleic acids as well as a pH-sensitive segment that exposes the membrane lytic peptide melittin in acidic environments to facilitate endosomal escape. The objective of this study was to investigate VIPER/siRNA polyplex characteristics, and compare their in vitro and in vivo performance with commercially available transfection reagents and a control version of VIPER lacking melittin. VIPER/siRNA polyplexes were formulated and characterized at various charge ratios and shown to be efficiently internalized in cultured cells. Target mRNA knockdown was confirmed by both flow cytometry and qRT-PCR and the kinetics of knockdown was monitored by live cell spinning disk microscopy, revealing knockdown starting by 4 h post-delivery. Intratracheal instillation of VIPER particles formulated with sequence specific siRNA to the lung of mice resulted in a significantly more efficient knockdown of GAPDH compared to treatment with VIPER particles formulated with scrambled sequence siRNA. We also demonstrated using pH-sensitive labels that VIPER particles experience less acidic environments compared to control polyplexes. In summary, VIPER/siRNA polyplexes efficiently deliver siRNA in vivo resulting in robust gene silencing (>75% knockdown) within the lung.


Assuntos
Pulmão/metabolismo , Polímeros/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Animais , Linhagem Celular Tumoral , Feminino , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Proteínas de Fluorescência Verde/genética , Humanos , Pulmão/citologia , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA