Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Metab ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902331

RESUMO

Alcohol use disorder (AUD) affects millions of people worldwide, causing extensive morbidity and mortality with limited pharmacological treatments. The liver is considered as the principal site for the detoxification of ethanol metabolite, acetaldehyde (AcH), by aldehyde dehydrogenase 2 (ALDH2) and as a target for AUD treatment, however, our recent data indicate that the liver only plays a partial role in clearing systemic AcH. Here we show that a liver-gut axis, rather than liver alone, synergistically drives systemic AcH clearance and voluntary alcohol drinking. Mechanistically, we find that after ethanol intake, a substantial proportion of AcH generated in the liver is excreted via the bile into the gastrointestinal tract where AcH is further metabolized by gut ALDH2. Modulating bile flow significantly affects serum AcH level and drinking behaviour. Thus, combined targeting of liver and gut ALDH2, and manipulation of bile flow and secretion are potential therapeutic strategies to treat AUD.

2.
J Clin Invest ; 133(15)2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37338984

RESUMO

The liver can fully regenerate after partial resection, and its underlying mechanisms have been extensively studied. The liver can also rapidly regenerate after injury, with most studies focusing on hepatocyte proliferation; however, how hepatic necrotic lesions during acute or chronic liver diseases are eliminated and repaired remains obscure. Here, we demonstrate that monocyte-derived macrophages (MoMFs) were rapidly recruited to and encapsulated necrotic areas during immune-mediated liver injury and that this feature was essential in repairing necrotic lesions. At the early stage of injury, infiltrating MoMFs activated the Jagged1/notch homolog protein 2 (JAG1/NOTCH2) axis to induce cell death-resistant SRY-box transcription factor 9+ (SOX9+) hepatocytes near the necrotic lesions, which acted as a barrier from further injury. Subsequently, necrotic environment (hypoxia and dead cells) induced a cluster of complement 1q-positive (C1q+) MoMFs that promoted necrotic removal and liver repair, while Pdgfb+ MoMFs activated hepatic stellate cells (HSCs) to express α-smooth muscle actin and induce a strong contraction signal (YAP, pMLC) to squeeze and finally eliminate the necrotic lesions. In conclusion, MoMFs play a key role in repairing the necrotic lesions, not only by removing necrotic tissues, but also by inducing cell death-resistant hepatocytes to form a perinecrotic capsule and by activating α-smooth muscle actin-expressing HSCs to facilitate necrotic lesion resolution.


Assuntos
Actinas , Neoplasias Hepáticas , Humanos , Actinas/metabolismo , Fígado/metabolismo , Hepatócitos/metabolismo , Macrófagos/metabolismo , Células Estreladas do Fígado/metabolismo , Necrose/metabolismo , Necrose/patologia , Neoplasias Hepáticas/metabolismo
3.
Exp Biol Med (Maywood) ; 248(7): 596-604, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37208920

RESUMO

Hyperexcitability is a major mechanism implicated in several neuropsychiatric disorders, such as organophosphate-induced status epilepticus (SE), primary epilepsy, stroke, spinal cord injury, traumatic brain injury, schizophrenia, and autism spectrum disorders. Underlying mechanisms are diverse, but a functional impairment and loss of GABAergic inhibitory neurons are common features in many of these disorders. While novel therapies abound to correct for the loss of GABAergic inhibitory neurons, it has been difficult at best to improve the activities of daily living for the majority of patients. Alpha-linolenic acid (ALA) is an essential omega-3 polyunsaturated fatty acid found in plants. ALA exerts pleiotropic effects in the brain that attenuate injury in chronic and acute brain disease models. However, the effect of ALA on GABAergic neurotransmission in hyperexcitable brain regions involved in neuropsychiatric disorders, such as the basolateral amygdala (BLA) and CA1 subfield of the hippocampus, is unknown. Administration of a single dose of ALA (1500 nmol/kg) subcutaneously increased the charge transfer of inhibitory postsynaptic potential currents mediated by GABAA receptors in pyramidal neurons by 52% in the BLA and by 92% in the CA1 compared to vehicle animals a day later. Similar results were obtained in pyramidal neurons from the BLA and CA1 when ALA was bath-applied in slices from naïve animals. Importantly, pretreatment with the high-affinity, selective TrkB inhibitor, k252, completely abolished the ALA-induced increase in GABAergic neurotransmission in the BLA and CA1, suggesting a brain-derived neurotrophic factor (BDNF)-mediated mechanism. Addition of mature BDNF (20 ng/mL) significantly increased GABAA receptor inhibitory activity in the BLA and CA1 pyramidal neurons similar to the results obtained with ALA. ALA may be an effective treatment for neuropsychiatric disorders where hyperexcitability is a major feature.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Ratos , Humanos , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Ácido alfa-Linolênico/farmacologia , Fator Neurotrófico Derivado do Encéfalo , Ratos Sprague-Dawley , Atividades Cotidianas , Transmissão Sináptica/fisiologia , Receptores de GABA-A/metabolismo
4.
Gut ; 72(10): 1942-1958, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36593103

RESUMO

OBJECTIVE: The current treatment for hepatocellular carcinoma (HCC) to block angiogenesis and immunosuppression provides some benefits only for a subset of patients with HCC, thus optimised therapeutic regimens are unmet needs, which require a thorough understanding of the underlying mechanisms by which tumour cells orchestrate an inflamed tumour microenvironment with significant myeloid cell infiltration. MicroRNA-223 (miR-223) is highly expressed in myeloid cells but its role in regulating tumour microenvironment remains unknown. DESIGN: Wild-type and miR-223 knockout mice were subjected to two mouse models of inflammation-associated HCC induced by injection of diethylnitrosamine (DEN) or orthotopic HCC cell implantation in chronic carbon tetrachloride (CCl4)-treated mice. RESULTS: Genetic deletion of miR-223 markedly exacerbated tumourigenesis in inflammation-associated HCC. Compared with wild-type mice, miR-223 knockout mice had more infiltrated programmed cell death 1 (PD-1+) T cells and programmed cell death ligand 1 (PD-L1+) macrophages after DEN+CCl4 administration. Bioinformatic analyses of RNA sequencing data revealed a strong correlation between miR-223 levels and tumour hypoxia, a condition that is well-documented to regulate PD-1/PD-L1. In vivo and in vitro mechanistic studies demonstrated that miR-223 did not directly target PD-1 and PD-L1 in immune cells rather than indirectly downregulated them by modulating tumour microenvironment via the suppression of hypoxia-inducible factor 1α-driven CD39/CD73-adenosine pathway in HCC. Moreover, gene delivery of miR-223 via adenovirus inhibited angiogenesis and hypoxia-mediated PD-1/PD-L1 activation in both HCC models, thereby hindering HCC progression. CONCLUSION: The miR-223 plays a critical role in modulating hypoxia-induced tumour immunosuppression and angiogenesis, which may serve as a novel therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , Camundongos , Animais , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Antígeno B7-H1 , Receptor de Morte Celular Programada 1 , Terapia de Imunossupressão , Carcinogênese , Camundongos Knockout , MicroRNAs/genética , Inflamação , Hipóxia , Microambiente Tumoral
5.
iScience ; 25(7): 104519, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35754718

RESUMO

Aging is an intricate process characterized by multiple hallmarks including stem cell exhaustion, genome instability, epigenome alteration, impaired proteostasis, and cellular senescence. Whereas each of these traits is detrimental at the cellular level, it remains unclear how they are interconnected to cause systemic organ deterioration. Here we show that abrogating Brap, a BRCA1-associated protein essential for neurogenesis, results in persistent DNA double-strand breaks and elevation of histone H2A mono- and poly-ubiquitination (H2Aub). These defects extend to cellular senescence and proteasome-mediated histone H2A proteolysis with alterations in cells' proteomic and epigenetic states. Brap deletion in the mouse brain causes neuroinflammation, impaired proteostasis, accelerated neurodegeneration, and substantially shortened the lifespan. We further show the elevation of H2Aub also occurs in human brain tissues with Alzheimer's disease. These data together suggest that chromatin aberrations mediated by H2Aub may act as a nexus of multiple aging hallmarks and promote tissue-wide degeneration.

6.
iScience ; 25(6): 104354, 2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35601919

RESUMO

The NDE1 gene encodes a scaffold protein essential for brain development. Although biallelic NDE1 loss of function (LOF) causes microcephaly with profound mental retardation, NDE1 missense mutations and copy number variations are associated with multiple neuropsychiatric disorders. However, the etiology of the diverse phenotypes resulting from NDE1 aberrations remains elusive. Here we demonstrate Nde1 controls neurogenesis through facilitating H4K20 trimethylation-mediated heterochromatin compaction. This mechanism patterns diverse chromatin landscapes and stabilizes constitutive heterochromatin of neocortical neurons. We demonstrate that NDE1 can undergo dynamic liquid-liquid phase separation, partitioning to the nucleus and interacting with pericentromeric and centromeric satellite repeats. Nde1 LOF results in nuclear architecture aberrations and DNA double-strand breaks, as well as instability and derepression of pericentromeric satellite repeats in neocortical neurons. These findings uncover a pivotal role of NDE1/Nde1 in establishing and protecting neuronal heterochromatin. They suggest that heterochromatin instability predisposes a wide range of brain dysfunction.

7.
Sci Rep ; 10(1): 17446, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060648

RESUMO

Traumatic brain injury generated by blast may induce long-term neurological and psychiatric sequelae. We aimed to identify molecular, histopathological, and behavioral changes in rats 2 weeks after explosive-driven double-blast exposure. Rats received two 30-psi (~ 207-kPa) blasts 24 h apart or were handled identically without blast. All rats were behaviorally assessed over 2 weeks. At Day 15, rats were euthanized, and brains removed. Brains were dissected into frontal cortex, hippocampus, cerebellum, and brainstem. Western blotting was performed to measure levels of total-Tau, phosphorylated-Tau (pTau), amyloid precursor protein (APP), GFAP, Iba1, αII-spectrin, and spectrin breakdown products (SBDP). Kinases and phosphatases, correlated with tau phosphorylation were also measured. Immunohistochemistry for pTau, APP, GFAP, and Iba1 was performed. pTau protein level was greater in the hippocampus, cerebellum, and brainstem and APP protein level was greater in cerebellum of blast vs control rats (p < 0.05). GFAP, Iba1, αII-spectrin, and SBDP remained unchanged. No immunohistochemical or neurobehavioral changes were observed. The dissociation between increased pTau and APP in different regions in the absence of neurobehavioral changes 2 weeks after double blast exposure is a relevant finding, consistent with human data showing that battlefield blasts might be associated with molecular changes before signs of neurological and psychiatric disorders manifest.


Assuntos
Traumatismos por Explosões/patologia , Lesões Encefálicas/patologia , Explosões , Transtornos Mentais/etiologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Comportamento Animal , Modelos Animais de Doenças , Masculino , Teste do Labirinto Aquático de Morris , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteínas tau/metabolismo
8.
Neuroimage ; 188: 419-426, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30576849

RESUMO

Traumatic Brain Injury (TBI) affects approximately 2.5 million people in the United States, of which 80% are considered to be mild (mTBI). Previous studies have shown that cerebral glucose uptake and metabolism are altered after brain trauma and functional metabolic deficits observed following mTBI are associated with changes in cognitive performance. Imaging of glucose uptake using [18F] Fluorodeoxyglucose (FDG) based Positron Emission Tomography (PET) with anesthesia during the uptake period demonstrated limited variability in results, but may have depressed uptake. Anesthesia has been found to interfere with blood glucose levels, and hence, FDG uptake. Conversely, forced cognitive testing during uptake may increase glucose demand in targeted regions, such as hippocampus, allowing for better differentiation of outcomes. Therefore, the objective of this study was to investigate the influence of a directed cognitive function task during the FDG uptake period on uptake measurements both in naïve rats and at 2 days after mild lateral fluid percussion (mLFP) TBI. Adult male Sprague Dawley rats underwent FDG uptake with either cognitive testing with the Novel Object Recognition (NOR) test or No Novel Object (NNO), followed by PET scans at baseline (prior to injury) and at 2days post mLFP. At baseline, FDG uptake in the right hippocampus was elevated in rats completing the NOR in comparison to the NNO (control group). Further, the NNO group rats demonstrated a greater fold change in the FDG uptake between baseline and post injury scans than the NOR group. Overall, these data suggest that cognitive activity during FDG uptake affects the regional uptake pattern in the brain, increasing uptake at baseline and suppressing the effects of injury.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Lateralidade Funcional/fisiologia , Hipocampo/fisiopatologia , Reconhecimento Psicológico/fisiologia , Animais , Comportamento Animal/fisiologia , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/metabolismo , Fluordesoxiglucose F18 , Hipocampo/diagnóstico por imagem , Hipocampo/metabolismo , Humanos , Masculino , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley
9.
Transl Psychiatry ; 8(1): 263, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30504810

RESUMO

Ketamine is a multimodal dissociative anesthetic, which provides powerful analgesia for victims with traumatic injury. However, the impact of ketamine administration in the peri-trauma period on the development of post-traumatic stress disorder (PTSD) remains controversial. Moreover, there is a major gap between preclinical and clinical studies because they utilize different doses and routes of ketamine administration. Here, we investigated the effects of sub-anesthetic doses of intravenous (IV) ketamine infusion on fear memory and brain glucose metabolism (BGluM) in rats. Male Sprague-Dawley rats received an IV ketamine infusion (0, 2, 10, and 20 mg/kg, 2 h) or an intraperitoneal (IP) injection (0 and 10 mg/kg) following an auditory fear conditioning (3 pairings of tone and foot shock [0.6 mA, 1 s]) on day 0. Fear memory retrieval, fear extinction, and fear recall were tested on days 2, 3, and 4, respectively. The effects of IV ketamine infusion (0 and 10 mg/kg) on BGluM were measured using 18F-fluoro-deoxyglucose positron emission tomography (FDG-PET) and computed tomography (CT). The IV ketamine infusion dose-dependently enhanced fear memory retrieval, delayed fear extinction, and increased fear recall in rats. The IV ketamine (10 mg/kg) increased BGluM in the hippocampus, amygdala, and hypothalamus, while decreasing it in the cerebellum. On the contrary, a single ketamine injection (10 mg/kg, IP) after fear conditioning facilitated fear memory extinction in rats. The current findings suggest that ketamine may produce differential effects on fear memory depending on the route and duration of ketamine administration.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Medo , Glucose/metabolismo , Ketamina/administração & dosagem , Memória/efeitos dos fármacos , Administração Intravenosa , Animais , Condicionamento Clássico , Extinção Psicológica/efeitos dos fármacos , Fluordesoxiglucose F18 , Locomoção/efeitos dos fármacos , Masculino , Rememoração Mental/efeitos dos fármacos , Tomografia por Emissão de Pósitrons , Ratos Sprague-Dawley
10.
Mol Neurobiol ; 55(1): 187-200, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28844093

RESUMO

Approximately, 1.7 million Americans suffer a TBI annually and TBI is a major cause of death and disability. The majority of the TBI cases are of the mild type and while most patients recover completely from mild TBI (mTBI) about 10% result in persistent symptoms and some result in lifelong disability. Anxiety disorders are the second most common diagnosis post-TBI. Of note, TBI-induced anxiety disorders are difficult to treat and remain a chronic condition suggesting that new therapies are needed. Previous work from our laboratory demonstrated that a mild TBI induced an anxiety-like phenotype, a key feature of the human condition, associated with loss of GABAergic interneurons and hyperexcitability in the basolateral amygdala (BLA) in rodents 7 and 30 days after a controlled cortical impact (CCI) injury. We now confirm that animals display significantly increased anxiety-like behavior 30 days after CCI. The anxiety-like behavior was associated with a significant loss of GABAergic interneurons and significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor-mediated inhibitory postsynaptic currents (IPSCs) in the BLA. Significantly, subchronic treatment with alpha-linolenic acid (ALA) after CCI prevents the development of anxiety-like behavior, the loss of GABAergic interneurons, hyperexcitability in the BLA and reduces the impact injury. Taken together, administration of ALA after CCI is a potent therapy against the neuropathology and pathophysiological effects of mTBI in the BLA.


Assuntos
Ansiedade/prevenção & controle , Lesões Encefálicas Traumáticas/tratamento farmacológico , Contusões/tratamento farmacológico , Ácido alfa-Linolênico/uso terapêutico , Animais , Ansiedade/etiologia , Ansiedade/fisiopatologia , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/fisiopatologia , Contusões/etiologia , Contusões/fisiopatologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Ácido alfa-Linolênico/farmacologia
11.
Molecules ; 20(11): 20355-80, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26569216

RESUMO

α-Linolenic acid (ALA) is a nutraceutical found in vegetable products such as flax and walnuts. The pleiotropic properties of ALA target endogenous neuroprotective and neurorestorative pathways in brain and involve the transcription factor nuclear factor kappa B (NF-κB), brain-derived neurotrophic factor (BDNF), a major neuroprotective protein in brain, and downstream signaling pathways likely mediated via activation of TrkB, the cognate receptor of BDNF. In this review, we discuss possible mechanisms of ALA efficacy against the highly toxic OP nerve agent soman. Organophosphate (OP) nerve agents are highly toxic chemical warfare agents and a threat to military and civilian populations. Once considered only for battlefield use, these agents are now used by terrorists to inflict mass casualties. OP nerve agents inhibit the critical enzyme acetylcholinesterase (AChE) that rapidly leads to a cholinergic crisis involving multiple organs. Status epilepticus results from the excessive accumulation of synaptic acetylcholine which in turn leads to the overactivation of muscarinic receptors; prolonged seizures cause the neuropathology and long-term consequences in survivors. Current countermeasures mitigate symptoms and signs as well as reduce brain damage, but must be given within minutes after exposure to OP nerve agents supporting interest in newer and more effective therapies. The pleiotropic properties of ALA result in a coordinated molecular and cellular program to restore neuronal networks and improve cognitive function in soman-exposed animals. Collectively, ALA should be brought to the clinic to treat the long-term consequences of nerve agents in survivors. ALA may be an effective therapy for other acute and chronic neurodegenerative disorders.


Assuntos
Suplementos Nutricionais , Agentes Neurotóxicos/efeitos adversos , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Organofosfatos/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Ácido alfa-Linolênico/farmacologia , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Humanos , Modelos Animais , Doenças do Sistema Nervoso/induzido quimicamente , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Neuropatologia , Fármacos Neuroprotetores/uso terapêutico , Receptores de N-Metil-D-Aspartato/metabolismo , Ácido alfa-Linolênico/uso terapêutico
12.
Neurotoxicology ; 51: 38-50, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26386148

RESUMO

Exposure to nerve agents results in severe seizures or status epilepticus caused by the inhibition of acetylcholinesterase, a critical enzyme that breaks down acetylcholine to terminate neurotransmission. Prolonged seizures cause brain damage and can lead to long-term consequences. Current countermeasures are only modestly effective against the brain damage supporting interest in the evaluation of new and efficacious therapies. The nutraceutical alpha-linolenic acid (LIN) is an essential omega-3 polyunsaturated fatty acid that has a wide safety margin. Previous work showed that a single intravenous injection of alpha-linolenic acid (500 nmol/kg) administered before or after soman significantly protected against soman-induced brain damage when analyzed 24h after exposure. Here, we show that administration of three intravenous injections of alpha-linolenic acid over a 7 day period after soman significantly improved motor performance on the rotarod, enhanced memory retention, exerted an anti-depressant-like activity and increased animal survival. This dosing schedule significantly reduced soman-induced neuronal degeneration in four major vulnerable brain regions up to 21 days. Taken together, alpha-linolenic acid reduces the profound behavioral deficits induced by soman possibly by decreasing neuronal cell death, and increases animal survival.


Assuntos
Antidepressivos/administração & dosagem , Cognição/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Soman/toxicidade , Ácido alfa-Linolênico/administração & dosagem , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Suplementos Nutricionais , Masculino , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Teste de Desempenho do Rota-Rod
13.
Neuromolecular Med ; 17(3): 251-69, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25920465

RESUMO

Exposure to organophosphorous (OP) nerve agents such as soman inhibits the critical enzyme acetylcholinesterase (AChE) leading to excessive acetylcholine accumulation in synapses, resulting in cholinergic crisis, status epilepticus and brain damage in survivors. The hippocampus is profoundly damaged after soman exposure leading to long-term memory deficits. We have previously shown that treatment with three sequential doses of alpha-linolenic acid, an essential omega-3 polyunsaturated fatty acid, increases brain plasticity in naïve animals. However, the effects of this dosing schedule administered after a brain insult and the underlying molecular mechanisms in the hippocampus are unknown. We now show that injection of three sequential doses of alpha-linolenic acid after soman exposure increases the endogenous expression of mature BDNF, activates Akt and the mammalian target of rapamycin complex 1 (mTORC1), increases neurogenesis in the subgranular zone of the dentate gyrus, increases retention latency in the passive avoidance task and increases animal survival. In sharp contrast, while soman exposure also increases mature BDNF, this increase did not activate downstream signaling pathways or neurogenesis. Administration of the inhibitor of mTORC1, rapamycin, blocked the alpha-linolenic acid-induced neurogenesis and the enhanced retention latency but did not affect animal survival. Our results suggest that alpha-linolenic acid induces a long-lasting neurorestorative effect that involves activation of mTORC1 possibly via a BDNF-TrkB-mediated mechanism.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Soman/toxicidade , Ácido alfa-Linolênico/farmacologia , Animais , Antígenos Nucleares/biossíntese , Antígenos Nucleares/genética , Derivados da Atropina/uso terapêutico , Aprendizagem da Esquiva/fisiologia , Dano Encefálico Crônico/etiologia , Dano Encefálico Crônico/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/genética , Replicação do DNA/efeitos dos fármacos , Diazepam/uso terapêutico , Proteínas do Domínio Duplacortina , Eletrochoque , Comportamento Exploratório/efeitos dos fármacos , Hipocampo/fisiopatologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/biossíntese , Complexos Multiproteicos/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/biossíntese , Neuropeptídeos/genética , Fármacos Neuroprotetores/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/metabolismo , Oximas/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética , Compostos de Piridínio/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptor trkB/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/complicações , Estado Epiléptico/tratamento farmacológico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/biossíntese , Serina-Treonina Quinases TOR/genética , Ácido alfa-Linolênico/antagonistas & inibidores , Ácido alfa-Linolênico/uso terapêutico
14.
PLoS One ; 9(6): e99818, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24955574

RESUMO

Organophosphorus (OP) nerve agents are deadly chemical weapons that pose an alarming threat to military and civilian populations. The irreversible inhibition of the critical cholinergic degradative enzyme acetylcholinesterase (AChE) by OP nerve agents leads to cholinergic crisis. Resulting excessive synaptic acetylcholine levels leads to status epilepticus that, in turn, results in brain damage. Current countermeasures are only modestly effective in protecting against OP-induced brain damage, supporting interest for evaluation of new ones. (-)-Phenserine is a reversible AChE inhibitor possessing neuroprotective and amyloid precursor protein lowering actions that reached Phase III clinical trials for Alzheimer's Disease where it exhibited a wide safety margin. This compound preferentially enters the CNS and has potential to impede soman binding to the active site of AChE to, thereby, serve in a protective capacity. Herein, we demonstrate that (-)-phenserine protects neurons against soman-induced neuronal cell death in rats when administered either as a pretreatment or post-treatment paradigm, improves motoric movement in soman-exposed animals and reduces mortality when given as a pretreatment. Gene expression analysis, undertaken to elucidate mechanism, showed that (-)-phenserine pretreatment increased select neuroprotective genes and reversed a Homer1 expression elevation induced by soman exposure. These studies suggest that (-)-phenserine warrants further evaluation as an OP nerve agent protective strategy.


Assuntos
Substâncias para a Guerra Química/toxicidade , Inibidores da Colinesterase/farmacologia , Fisostigmina/análogos & derivados , Soman/toxicidade , Estado Epiléptico , Animais , Proteínas de Transporte/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Arcabouço Homer , Masculino , Fisostigmina/farmacologia , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/metabolismo
15.
Neurotoxicology ; 33(5): 1219-29, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22884490

RESUMO

Nerve agents are deadly threats to military and civilian populations around the world. Nerve agents cause toxicity to peripheral and central sites through the irreversible inhibition of acetylcholinesterase, the enzyme that metabolizes acetylcholine. Excessive acetylcholine accumulation in synapses results in status epilepticus in the central nervous system. Prolonged status epilepticus leads to brain damage, neurological dysfunction and poor outcome. Anticonvulsants are effective but must be given rapidly following exposure. Because these agents cause mass casualties, effective neuroprotective agents are needed to reduce brain damage and improve cognitive outcome. α-Linolenic acid is an omega-3 fatty acid that is found in vegetable products and has no known side effects. α-Linolenic acid is neuroprotective against kainic acid-induced brain damage in vivo, but its neuroprotective efficacy against nerve agents is unknown. α-Linolenic acid also exerts anti-depressant and anti-inflammatory activities and enhances synaptic plasticity in vivo. These properties make this polyunsaturated fatty acid (PUFA) a potential candidate against nerve agent-induced neuropathology. Here we show that α-linolenic acid is neuroprotective against soman-induced neuropathology in either a pretreatment or post-treatment paradigm. We also show that subcutaneous injection of α-linolenic acid shows greater neuroprotective efficacy compared with intravenous injection in a brain region-specific manner.


Assuntos
Inibidores da Colinesterase/toxicidade , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Ácido alfa-Linolênico/uso terapêutico , Análise de Variância , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Ensaio de Desvio de Mobilidade Eletroforética , Fluoresceínas , Masculino , NF-kappa B/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/prevenção & controle , Síndromes Neurotóxicas/complicações , Compostos Orgânicos , Ratos , Ratos Sprague-Dawley , Soman/toxicidade
16.
Dose Response ; 9(3): 416-33, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22013403

RESUMO

Synaptic function is critical for the brain to process experiences dictated by the environment requiring change over the lifetime of the organism. Experience-driven adaptation requires that receptors, signal transduction pathways, transcription and translational mechanisms within neurons respond rapidly over its lifetime. Adaptive responses communicated through the rapid firing of neurons are dependent upon the integrity and function of synapses. These rapid responses via adaptation underlie the organism's ability to perceive, learn, remember, calculate and plan. Glutamate, the endogenous neurotransmitter required for physiological excitation in the brain, is critically involved in neuronal adaptive responses and in the pathophysiology of neurodegenerative disorders. Using neuronal experimental systems, we will discuss how compounds with low dose effects mediated via glutamate receptors can result either in a neuroprotective or neurotoxic response. Because the brain has evolved to respond rapidly to environmental cues, exposure of neurons to stressful stimuli can result in a pivotal response toward either synaptic adaptation or dysfunction and neuronal cell death. Understanding how neurons adapt to stressful stimuli will provide important clues toward the development of strategies to protect the brain against neurodegeneration.

17.
Neuropsychopharmacology ; 34(12): 2548-59, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19641487

RESUMO

Omega-3 polyunsaturated fatty acids are known to have therapeutic potential in several neurological and psychiatric disorders. However, the molecular mechanisms of action underlying these effects are not well elucidated. We previously showed that alpha-linolenic acid (ALA) reduced ischemic brain damage after a single treatment. To follow-up this finding, we investigated whether subchronic ALA treatment promoted neuronal plasticity. Three sequential injections with a neuroprotective dose of ALA increased neurogenesis and expression of key proteins involved in synaptic functions, namely, synaptophysin-1, VAMP-2, and SNAP-25, as well as proteins supporting glutamatergic neurotransmission, namely, V-GLUT1 and V-GLUT2. These effects were correlated with an increase in brain-derived neurotrophic factor (BDNF) protein levels, both in vitro using neural stem cells and hippocampal cultures and in vivo, after subchronic ALA treatment. Given that BDNF has antidepressant activity, this led us to test whether subchronic ALA treatment could produce antidepressant-like behavior. ALA-treated mice had significantly reduced measures of depressive-like behavior compared with vehicle-treated animals, suggesting another aspect of ALA treatment that could stimulate functional stroke recovery by potentially combining acute neuroprotection with long-term repair/compensatory plasticity. Indeed, three sequential injections of ALA enhanced protection, either as a pretreatment, wherein it reduced post-ischemic infarct volume 24 h after a 1-hour occlusion of the middle cerebral artery or as post-treatment therapy, wherein it augmented animal survival rates by threefold 10 days after ischemia.


Assuntos
Encéfalo/efeitos dos fármacos , Depressão/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ácido alfa-Linolênico/farmacologia , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Células Cultivadas , Depressão/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/fisiologia
18.
J Neurochem ; 109(5): 1375-88, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19476549

RESUMO

To determine the epigenetic events associated with NMDA receptor-mediated activation of brain-derived neurotrophic factor gene (Bdnf) promoter 1 by hippocampal neurons in culture, we screened 12 loci across 4.5 kb of genomic DNA 5' of the transcription start site (TSS) of rat Bdnf for specific changes in histone modification and transcription factor binding following NMDA receptor stimulation. Chromatin immunoprecipitation (ChIP) assays showed that NMDA receptor stimulation produced a durable, time-dependent decrease in histone H3 at lysine 9 dimethylation (H3K9me2), within 3 h after NMDA treatment across multiple loci. Concomitant increases in H3K4me2 and H3K9/14 acetylation (H3AcK9/14) were associated with transcriptional activation, but occurred at fewer sites within the promoter. The decrease in H3K9me2 was associated with release of HDAC1, MBD1, MeCP2, and REST from specific locations within promoter 1, although with different kinetics. In addition, occupancy of sites proximal to and distal to the TSS by the transcription factors NF-kappaB, CREB-binding protein (CBP), and cAMP-response element-binding protein were correlated with increased occupancy of RNA polymerase II at two loci proximal to the TSS following NMDA receptor stimulation. These temporal changes in promoter occupancy could occur thousands of base pairs 5' of the TSS, suggesting a mechanism that produces waves of Bdnf transcription.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Montagem e Desmontagem da Cromatina/fisiologia , Hipocampo/citologia , Neurônios/metabolismo , Regiões Promotoras Genéticas/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Acetilação/efeitos dos fármacos , Análise de Variância , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Imunoprecipitação da Cromatina/métodos , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Éxons/fisiologia , Histona Desacetilase 1 , Histona Desacetilases/metabolismo , Histonas/metabolismo , Ácidos Hidroxâmicos/farmacologia , Técnicas In Vitro , Lisina/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Metilação/efeitos dos fármacos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo
19.
CNS Neurol Disord Drug Targets ; 7(4): 382-90, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18991667

RESUMO

A variety of sublethal or stressful stimuli induce a phenomenon in the brain known as tolerance, an adaptive response that protects the brain against the same stress, or against a different stress (cross-tolerance). Understanding the molecular mechanisms of brain preconditioning holds promise in developing innovative therapies to prevent and treat neurodegenerative disorders, particularly ischemic stroke. Many of the detailed steps involved in tolerance and cross-tolerance are unknown. It is also likely that different stressors differentially regulate sets of genes, transcription factors, and signal transduction pathways that depend upon the molecules that are released in response to the stressor, activation of particular receptors, and the surrounding milieu. The focus of this review is to highlight a few examples of stimuli that induce tolerance: 1) cortical spreading depression; 2) 3-nitropropionic acid; and 3) 2-deoxy-D-glucose. We will summarize by discussing one pathway where intracellular mediators may converge to upregulate intrinsic neuronal survival pathways to promote survival by resisting damage. This mechanism, activation of N-methyl-D-aspartate receptors and its integral relationship with brain-derived neurotrophic factor, may be a critical and general mechanism developed in brain to respond to stressful stimuli.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Precondicionamento Isquêmico/métodos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Desoxiglucose/farmacologia , Desoxiglucose/uso terapêutico , Humanos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Nitrocompostos/farmacologia , Nitrocompostos/uso terapêutico , Propionatos/farmacologia , Propionatos/uso terapêutico , Receptores de N-Metil-D-Aspartato/metabolismo , Estresse Fisiológico/fisiologia
20.
J Neurosci ; 28(5): 1118-30, 2008 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-18234890

RESUMO

Brain-derived neurotrophic factor (BDNF), via activation of TrkB receptors, mediates vital physiological functions in the brain, ranging from neuronal survival to synaptic plasticity, and has been implicated in the pathophysiology of neurodegenerative disorders. Although transcriptional regulation of the BDNF gene (Bdnf) has been extensively studied, much remains to be understood. We discovered a sequence within Bdnf promoter 4 that binds the basic helix-loop-helix protein BHLHB2 and is a target for BHLHB2-mediated transcriptional repression. NMDA receptor activation de-repressed promoter 4-mediated transcription and correlated with reduced occupancy of the promoter by BHLHB2 in cultured hippocampal neurons. Bhlhb2 gene -/- mice showed increased hippocampal exon 4-specific Bdnf mRNA levels compared with +/+ littermates under basal and activity-dependent conditions. Bhlhb2 knock-out mice also showed increased status epilepticus susceptibility, suggesting that BHLHB2 alters neuronal excitability. Together, these results support a role for BHLHB2 as a new modulator of Bdnf transcription and neuronal excitability.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Homeodomínio/fisiologia , Neurônios/fisiologia , Regiões Promotoras Genéticas/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Células Cultivadas , Feminino , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células NIH 3T3 , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...