Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neural Regen Res ; 17(12): 2589-2592, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35662186

RESUMO

Traumatic brain injury has a complex pathophysiology that produces both rapid and delayed brain damage. Rapid damage initiates immediately after injury. Treatment of traumatic brain injury is typically delayed many hours, thus only delayed damage can be targeted with drugs. Delayed traumatic brain injury includes neuroinflammation, oxidative damage, apoptosis, and glutamate toxicity. Both the speed and complexity of traumatic brain injury pathophysiology present large obstacles to drug development. Repurposing of Food and Drug Administration-approved drugs may be a highly efficient approach to get therapeutics to the clinic. This review examines the preclinical outcomes of minocycline and N-acetylcysteine as individual drugs and compares them to the minocycline plus N-acetylcysteine combination. Both minocycline and N-acetylcysteine are Food and Drug Administration-approved drugs with pleiotropic therapeutic effects. As individual drugs, minocycline and N-acetylcysteine are well tolerated, with known pharmacokinetics, and enter the brain through an intact blood-brain barrier. At concentrations greater than needed for anti-microbial action, minocycline is a potent anti-inflammatory minocycline, also acts as an antioxidant and inhibits multiple enzymes that promote brain injury including metalloproteases, caspases, and polyADP-ribose-polymerase-1. N-acetylcysteine alone is also an antioxidant. It increases brain glutathione, prevents lipid oxidation, and protects mitochondria. N-acetylcysteine also acts as an anti-inflammatory as well as increases extracellular glutamate by activating the Xc cystine-glutamate anti-transporter. These multiple actions of minocycline and N-acetylcysteine have made them attractive candidates to treat traumatic brain injury. When first dosed within the one hour after injury, either minocycline or N-acetylcysteine improves a diverse set of therapeutic outcome measures in multiple traumatic brain injury animal models. A small number of clinical trials for traumatic brain injury have established the safety of minocycline or N-acetylcysteine and suggested that either drug has some efficacy. Preclinical studies have shown that minocycline plus N-acetylcysteine have positive synergy resulting in therapeutic effects and a more prolonged therapeutic time window not seen with the individual drugs. This review compares the actions of minocycline and N-acetylcysteine, individually and in combination. Evidence supports that the combination has greater utility to treat traumatic brain injury than the individual drugs.

2.
Exp Neurol ; 345: 113816, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34310944

RESUMO

Multiple drugs to treat traumatic brain injury (TBI) have failed clinical trials. Most drugs lose efficacy as the time interval increases between injury and treatment onset. Insufficient therapeutic time window is a major reason underlying failure in clinical trials. Few drugs have been developed with therapeutic time windows sufficiently long enough to treat TBI because little is known about which brain functions can be targeted if therapy is delayed hours to days after injury. We identified multiple injury parameters that are improved by first initiating treatment with the drug combination minocycline (MINO) plus N-acetylcysteine (NAC) at 72 h after injury (MN72) in a mouse closed head injury (CHI) experimental TBI model. CHI produces spatial memory deficits resulting in impaired performance on Barnes maze, hippocampal neuronal loss, and bilateral damage to hippocampal neurons, dendrites, spines and synapses. MN72 treatment restores Barnes maze acquisition and retention, protects against hippocampal neuronal loss, limits damage to dendrites, spines and synapses, and accelerates recovery of microtubule associated protein 2 (MAP2) expression, a key protein in maintaining proper dendritic architecture and synapse density. These data show that in addition to the structural integrity of the dendritic arbor, spine and synapse density can be successfully targeted with drugs first dosed days after injury. Retention of substantial drug efficacy even when first dosed 72 h after injury makes MINO plus NAC a promising candidate to treat clinical TBI.


Assuntos
Acetilcisteína/administração & dosagem , Lesões Encefálicas Traumáticas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Minociclina/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Memória Espacial/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Esquema de Medicação , Quimioterapia Combinada , Sequestradores de Radicais Livres/administração & dosagem , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Memória Espacial/fisiologia
3.
Neurosci Lett ; 682: 16-20, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-29885452

RESUMO

The mouse closed head injury (CHI) model of traumatic brain injury (TBI) produces widespread demyelination. Myelin content is restored by minocycline (MINO) plus n-acetylcysteine (NAC) or MINO alone when first dosed at 12 h after CHI. In a rat controlled cortical impact model of TBl, a first dose of MINO plus NAC one h after injury protects resident oligodendrocytes that induce remyelination. In contrast, MINO less effectively protects oligodendrocytes and remyelination is mediated by oligodendrocyte precursor cell proliferation and differentiation. MINO plus NAC or MINO alone is hypothesized to work similarly in the CHI model as in the controlled cortical impact model even when first dosed at 12-h post-CHI. We tested this hypothesis by examining the time course of the changes in the oligodendrocyte antigenic markers CC1, 2',3'-Cyclic-nucleotide 3'-phosphodiesterase and phospholipid protein between 2 and 14 days post-CHI in mice treated with saline, NAC, MINO or MINO plus NAC. CHI produced a long-lasting loss of these markers that was not altered by NAC treatment. In contrast, oligodendrocyte marker expression was maintained by MINO plus NAC between 2 and 14 days post-injury. MINO alone did not prevent the early loss of oligodendrocyte markers, but marker expression significantly increased by 14-days post-injury. These data suggest that MINO plus NAC or MINO alone when first dosed 12 h after CHI increase myelin content using similar mechanisms seen when first dosed 1 h after closed head injury. These data also suggest that drugs protect oligodendrocytes with a clinically useful therapeutic time window.


Assuntos
Acetilcisteína/administração & dosagem , Traumatismos Cranianos Fechados/tratamento farmacológico , Minociclina/administração & dosagem , Neuroproteção/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Animais , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/patologia , Quimioterapia Combinada , Sequestradores de Radicais Livres/administração & dosagem , Traumatismos Cranianos Fechados/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção/fisiologia , Oligodendroglia/patologia , Fatores de Tempo
4.
J Cereb Blood Flow Metab ; 38(8): 1312-1326, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28685618

RESUMO

Mild traumatic brain injury afflicts over 2 million people annually and little can be done for the underlying injury. The Food and Drug Administration-approved drugs Minocycline plus N-acetylcysteine (MINO plus NAC) synergistically improved cognition and memory in a rat mild controlled cortical impact (mCCI) model of traumatic brain injury.3 The underlying cellular and molecular mechanisms of the drug combination are unknown. This study addressed the effect of the drug combination on white matter damage and neuroinflammation after mCCI. Brain tissue from mCCI rats given either sham-injury, saline, MINO alone, NAC alone, or MINO plus NAC was investigated via histology and qPCR at four time points (2, 4, 7, and 14 days post-injury) for markers of white matter damage and neuroinflammation. MINO plus NAC synergistically protected resident oligodendrocytes and decreased the number of oligodendrocyte precursor cells. Activation of microglia/macrophages (MP/MG) was synergistically increased in white matter two days post-injury after MINO plus NAC treatment. Patterns of M1 and M2 MP/MG were also altered after treatment. The modulation of neuroinflammation is a potential mechanism to promote remyelination and improve cognition and memory. These data also provide new and important insights into how drug treatments can induce repair after traumatic brain injury.


Assuntos
Acetilcisteína/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Lesões Encefálicas Traumáticas/tratamento farmacológico , Minociclina/uso terapêutico , Oligodendroglia/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Animais , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley
5.
Exp Neurol ; 249: 169-77, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24036416

RESUMO

Traumatic brain injury (TBI) differs in severity from severe to mild. This study examined whether a combination of the drugs minocycline (MINO) plus N-acetylcysteine (NAC) produces behavioral and histological improvements in a mild version of the controlled cortical impact model of TBI (mCCI). Following mCCI, rats acquired an active place avoidance task by learning the location of a stationary shock zone on a rotating arena. Rats acquired this task with a training protocol using a 10-minute intertrial interval. Mildly injured rats had an apparent deficit in long-term memory since they did not acquire the task when the intertrial interval was increased to 24 h. Mildly injured rats also had an apparent deficit in set shifting since, after successfully learning one shock zone location they did not learn the location of a second shock zone. MINO plus NAC synergistically limited these behavioral deficits in long-term memory and set shifting. mCCI also produced neuroinflammation at the impact site and at distal white matter tracts including the corpus callosum. At the impact site, MINO plus NAC attenuated CD68-expressing phagocytic microglia without altering neutrophil infiltration or astrocyte activation. The drugs had no effect on astrocyte activation in the corpus callosum or hippocampus. In the corpus callosum, MINO plus NAC decreased CD68 expression yet increased overall microglial activation as measured by Iba-1. MINO plus NAC acted synergistically to increase Iba-1 expression since MINO alone suppressed expression and NAC alone had no effect. Despite the known anti-inflammatory actions of the individual drugs, MINO plus NAC appeared to modulate, rather than suppress neuroinflammation. This modulation of neuroinflammation may underlie the synergistic improvement in memory and set-shifting by the drug combination after mCCI.


Assuntos
Acetilcisteína/administração & dosagem , Lesões Encefálicas/prevenção & controle , Transtornos Cognitivos/prevenção & controle , Modelos Animais de Doenças , Transtornos da Memória/prevenção & controle , Minociclina/administração & dosagem , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Sinergismo Farmacológico , Quimioterapia Combinada , Inflamação/patologia , Inflamação/prevenção & controle , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Sprague-Dawley
6.
PLoS One ; 5(8): e12490, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20824218

RESUMO

BACKGROUND: There are no drugs presently available to treat traumatic brain injury (TBI). A variety of single drugs have failed clinical trials suggesting a role for drug combinations. Drug combinations acting synergistically often provide the greatest combination of potency and safety. The drugs examined (minocycline (MINO), N-acetylcysteine (NAC), simvastatin, cyclosporine A, and progesterone) had FDA-approval for uses other than TBI and limited brain injury in experimental TBI models. METHODOLOGY/PRINCIPAL FINDINGS: Drugs were dosed one hour after injury using the controlled cortical impact (CCI) TBI model in adult rats. One week later, drugs were tested for efficacy and drug combinations tested for synergy on a hierarchy of behavioral tests that included active place avoidance testing. As monotherapy, only MINO improved acquisition of the massed version of active place avoidance that required memory lasting less than two hours. MINO-treated animals, however, were impaired during the spaced version of the same avoidance task that required 24-hour memory retention. Co-administration of NAC with MINO synergistically improved spaced learning. Examination of brain histology 2 weeks after injury suggested that MINO plus NAC preserved white, but not grey matter, since lesion volume was unaffected, yet myelin loss was attenuated. When dosed 3 hours before injury, MINO plus NAC as single drugs had no effect on interleukin-1 formation; together they synergistically lowered interleukin-1 levels. This effect on interleukin-1 was not observed when the drugs were dosed one hour after injury. CONCLUSIONS/SIGNIFICANCE: These observations suggest a potentially valuable role for MINO plus NAC to treat TBI.


Assuntos
Acetilcisteína/farmacologia , Lesões Encefálicas/fisiopatologia , Cognição/efeitos dos fármacos , Memória/efeitos dos fármacos , Minociclina/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Condicionamento Psicológico , Sinergismo Farmacológico , Interleucina-1beta/biossíntese , Bainha de Mielina/metabolismo , Percepção/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Análise e Desempenho de Tarefas , Fatores de Tempo
7.
Neurosci Lett ; 473(2): 67-71, 2010 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-19822191

RESUMO

The sodium-potassium ATPase (Na/K ATPase) is a major ionic transporter in the brain and is responsible for the maintenance of the Na(+) and K(+) gradients across the cell membrane. Cardiotonic steroids such as ouabain, digoxin and marinobufagenin are well-characterized inhibitors of the Na/K ATPase. Recently, cardiotonic steroids have been shown to have additional effects at concentrations below their IC(50) for pumping. The cardiotonic steroids ouabain, digoxin, and marinobufagenin all show an inverted U-shaped dose-response curve with inhibition of pumping at concentrations near their IC(50), while increasing Na/K ATPase activity at doses below their IC(50). This stimulatory effect of cardiotonic steroids was observed in vitro in hippocampal slice cultures as well as in the hippocampus in vivo. Increased Na/K ATPase activity has been shown to protect slice culture neurons from hypoxia-hypoglycemia. Ouabain protected slice culture neurons from experimental ischemia at concentrations that increased Na/K ATPase. This protective effect was observed when ouabain was dosed 30min before, or 2h following experimental ischemia. Ouabain no longer protected against experimental ischemia if the increase of Na/K ATPase was blocked. These data suggest that the protective effect of ouabain was due to increased Na/K ATPase activity. The demonstration of a neuroprotective effect of cardiotonic steroids could potentially assist in the treatment of stroke since digoxin, one of the cardiotonic steroids examined in this study, has approval by the Food and Drug Administration and can be safely administered at the concentrations that increase Na/K ATPase activity.


Assuntos
Isquemia Encefálica/prevenção & controle , Bufanolídeos/farmacologia , Cardiotônicos/farmacologia , Digoxina/farmacologia , Hipocampo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ouabaína/farmacologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Isquemia Encefálica/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Hipocampo/metabolismo , Técnicas In Vitro , Ratos
8.
Brain Res ; 1213: 127-39, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18455703

RESUMO

In ischemic preconditioning, a sublethal ischemic insult protects neurons from subsequent ischemia. In organotypic hippocampal slice cultures a sublethal 5-minute hypoxia-hypoglycemia treatment prevented neuronal loss after a 10-minute experimental ischemic (EI) treatment of hypoxia-hypoglycemia. Whereas preconditioning protected against EI given 24 h later, it did not protect when EI was given 2 h later, suggesting a slow development of neuroprotection. This model identified two regulators of ischemic preconditioning: the atypical protein kinase M zeta (PKMzeta), and the Na/K ATPase. Two hours following preconditioning, when there was no neuroprotection, Na/K ATPase activity was unchanged. In contrast, Na/K ATPase activity significantly increased 24 h after the preconditioning treatment. Elevated Na/K ATPase activity was accompanied by increased surface expression of the alpha1 and alpha2 isoforms of the Na/K ATPase. Similarly, active PKMzeta levels were increased at 24 h, but not 2 h, after preconditioning. PKMzeta overexpression by sindbis virus vectors also increased Na/K ATPase activity. To examine PKMzeta regulation of Na/K ATPase, occlusion experiments were performed using marinobufagenin to inhibit alpha1, dihydroouabain to inhibit alpha2/3 and a zeta-pseudosubstrate peptide to inhibit PKMzeta. These experiments showed that PKMzeta regulated both the activity and surface expression of the alpha1 isoform of the Na/K ATPase. Marinobufagenin, dihydroouabain, and zeta-pseudosubstrate peptide were used to determine if PKMzeta or the alpha1 and alpha2 Na/K ATPase isoforms protected neurons. All three compounds blocked neuroprotection following ischemic preconditioning. PKMzeta levels were elevated 3 days after ischemic preconditioning. These data indicate key roles of PKMzeta and Na/K ATPase in ischemic preconditioning.


Assuntos
Hipocampo/metabolismo , Hipoglicemia/prevenção & controle , Hipóxia/prevenção & controle , Precondicionamento Isquêmico/métodos , Proteína Quinase C/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Animais Recém-Nascidos , Biotinilação , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/fisiologia , Hipoglicemia/patologia , Hipoglicemia/fisiopatologia , Hipóxia/patologia , Hipóxia/fisiopatologia , Imunoprecipitação , Cloreto de Potássio/farmacologia , Ratos , Fatores de Tempo , Técnicas de Cultura de Tecidos
9.
Brain Res ; 1117(1): 213-23, 2006 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-16996036

RESUMO

Glucose is well accepted as the major fuel for neuronal activity, while it remains controversial whether lactate also supports neural activity. In hippocampal slice cultures, synaptic transmission supported by glucose was reversibly suppressed by lactate. To test whether lactate had a similar inhibitory effect in vivo, lactate was perfused into the hippocampi of unanesthetized rats while recording the firing of nearby pyramidal cells. Lactate perfusion suppressed pyramidal cell firing by 87.5+/-8.3% (n=6). Firing suppression was slow in onset and fully reversible and was associated with increased lactate concentration at the site of the recording electrode. In vivo suppression of neural activity by lactate occurred in the presence of glucose; therefore we tested whether suppression of neural firing was due to lactate interference with glucose metabolism. Competition between glucose and lactate was measured in hippocampal slice cultures. Lactate had no effect on glucose uptake. Lactate suppressed glucose oxidation when applied at an elevated, pathological concentration (10 mM), but not at its physiological concentration (1 mM). Pyruvate (10 mM) also inhibited glucose oxidation but was significantly less effective than lactate. The greater suppressive effect of lactate as compared to pyruvate suggests that alteration of the NAD(+)/NADH ratio underlies the suppression of glucose oxidation by lactate. ATP in slice culture was unchanged in glucose (1 mM), but significantly reduced in lactate (1 mM). ATP in slice culture was significantly increased by combination of glucose (1 mM) and lactate (1 mM). These data suggest that alteration of redox ratio underlies the suppression of neural discharge and glucose metabolism by lactate.


Assuntos
Potenciais de Ação/fisiologia , Metabolismo Energético/fisiologia , Glucose/metabolismo , Hipocampo/metabolismo , Ácido Láctico/metabolismo , Neurônios/metabolismo , Potenciais de Ação/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Interações Medicamentosas/fisiologia , Metabolismo Energético/efeitos dos fármacos , Glucose/farmacologia , Hipocampo/efeitos dos fármacos , Ácido Láctico/farmacologia , Masculino , NAD/metabolismo , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Oxirredução , Fosforilação Oxidativa/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacologia , Ratos , Ratos Long-Evans , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
Brain Res Brain Res Protoc ; 13(3): 135-43, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15296850

RESUMO

In ischemic preconditioning (IPC), brief sublethal ischemia protects neurons from a subsequent lethal ischemia. In vivo models faithfully display preconditioning, yet, these models are technically challenging, time-consuming and expensive. In vitro models of preconditioning have also been developed that are technically easier and less expensive. A drawback of pre-existing in vitro models is that since susceptibility to ischemic injury is age-dependent; neuroprotection is being studied in neurons that have intrinsic resistance to oxygen-glucose deprivation (OGD). This study introduces a new in vitro model of ischemic preconditioning in hippocampal slice cultures isolated from 20-30-day-old rats. Slice cultures show a high susceptibility and sharp thresholds toward ischemia that is comparable to that found in vivo. A 5-min OGD treatment was not neurotoxic to young adult slice cultures, while a 10-min OGD treatment was neurotoxic. In addition, the sublethal 5-min OGD treatment protected against a 10-min OGD treatment that was delivered 24 h later. Neuroprotection was seen in preconditioned slice cultures stained with propidium iodide (PI) or with antisera against the neuron-specific antigen NeuN. Energy failure is hypothesized to trigger ischemic preconditioning and a 5-min OGD treatment induced transient energy failure in young adult slice cultures. This model may assist in the search for new therapeutics for the prevention and/or treatment of stroke.


Assuntos
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Precondicionamento Isquêmico/métodos , Trifosfato de Adenosina/metabolismo , Animais , Anticorpos/farmacologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/fisiopatologia , Proteínas de Ligação a DNA , Cultura em Câmaras de Difusão , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Glucose/deficiência , Hipocampo/irrigação sanguínea , Hipocampo/fisiopatologia , Hipóxia Encefálica/tratamento farmacológico , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/fisiopatologia , Modelos Biológicos , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Fármacos Neuroprotetores/farmacologia , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Técnicas de Cultura de Órgãos/métodos , Propídio/farmacologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA