Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Atherosclerosis ; 284: 153-159, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30913515

RESUMO

BACKGROUND AND AIMS: Atherosclerosis is characterized by lipid deposition, monocyte infiltration and foam cell formation in the artery wall. Translocator protein (TSPO) is abundantly expressed in lipid rich tissues. Recently, TSPO has been identified as a potential diagnostic tool in cardiovascular disease. The purpose of this study was to determine if the TSPO ligand, 18F-PBR111, can identify early atherosclerotic lesions and if TSPO expression can be used to identify distinct macrophage populations during lesion progression. METHODS: ApoE-/- mice were maintained on a high-fat diet for 3 or 12 weeks. C57BL/6J mice maintained on chow diet served as controls. Mice were administered 18F-PBR111 intravenously and PET/CT imaged. After euthanasia, aortas were isolated, fixed and optically cleared. Cleared aortas were immunostained with DAPI, and fluorescently labelled with antibodies to TSPO, the tissue resident macrophage marker F4/80 and the monocyte-derived macrophage marker CD11b. TSPO expression and the macrophage markers were visualised in fatty streaks and established plaques by light sheet microscopy. RESULTS: While tissue resident F4/80 + macrophages were evident in the arteries of animals without atherosclerosis, no CD11b + macrophages were observed in these animals. In contrast, established plaques had high CD11b and low F4/80 expression. A ∼3-fold increase in the uptake of 18F-PBR111 was observed in the aortas of atherosclerotic mice relative to controls. CONCLUSIONS: Imaging of TSPO expression is a new approach for studying atherosclerotic lesion progression and inflammatory cell infiltration. The TSPO ligand, 18F-PBR111, is a potential clinical diagnostic tool for the detection and quantification of atherosclerotic lesion progression in humans.


Assuntos
Aterosclerose/sangue , Aterosclerose/diagnóstico , Antígeno CD11b/fisiologia , Macrófagos , Receptores de GABA/fisiologia , Animais , Antígeno CD11b/biossíntese , Progressão da Doença , Diagnóstico Precoce , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/administração & dosagem , Receptores de GABA/biossíntese
2.
Neurobiol Dis ; 124: 416-427, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30590180

RESUMO

Neuropathological findings in the amygdala obtained from patients with mesial temporal lobe epilepsy (MTLE) indicate varying degrees of histopathological alterations, such as neuronal loss and gliosis. The mechanisms underlying cellular damage in the amygdala of patients with MTLE have not been fully elucidated. In the present study, we assess cellular damage, determine the receptor expression of major inhibitory and excitatory neurotransmitters, and evaluate the correlation between the expression of various receptors and cell damage in the basolateral complex and the centromedial areas in the amygdala specimens resected during brain surgery on 30 patients with medically intractable MTLE. Our data reveal an increased rate of cell damage and apoptosis as well as decreased expression levels of several GABAergic receptor subunits (GABAARα1, GABAARß3, and GABABR1) and GAD65 in the amygdalae obtained during epilepsy surgery compared to autopsy specimens. Analyses of the expression of glutamate excitatory receptor subunits (NR1, NR2B, mGluR1α, GluR1, and GluR2) reveal no significant differences between the epileptic amygdalae and autopsy control tissues. Furthermore, the increased occurrence of apoptotic cells in the amygdala is negatively correlated with the reduced expression of the studied GABAergic receptor subunits and GAD65 but is not correlated with the expression of excitatory receptors. The present data point to the importance of GABAergic neurotransmission in seizure-induced cell injury in the amygdala of patients with MTLE and suggest several GABA receptor subunits as potential druggable target structures to control epilepsy and its comorbid disorders, such as anxiety.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Epilepsia do Lobo Temporal/fisiopatologia , Receptores de GABA/biossíntese , Adolescente , Adulto , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Apoptose/fisiologia , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transmissão Sináptica/fisiologia , Adulto Jovem
3.
Stroke ; 48(12): 3366-3374, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29146879

RESUMO

BACKGROUND AND PURPOSE: Focal cortical infarction causes neuronal apoptosis in the ipsilateral nonischemic thalamus and hippocampus, which is potentially associated with poststroke cognitive deficits. TSPO (translocator protein) is critical in regulating mitochondrial apoptosis pathways. We examined the effects of the novel TSPO ligand 2-(2-chlorophenyl) quinazolin-4-yl dimethylcarbamate (2-Cl-MGV-1) on poststroke cognitive deficits, neuronal mitochondrial apoptosis, and secondary damage in the ipsilateral thalamus and hippocampus after cortical infarction. METHODS: One hundred fourteen hypertensive rats underwent successful distal middle cerebral artery occlusion (n=76) or sham procedures (n=38). 2-Cl-MGV-1 or dimethyl sulfoxide as vehicle was administrated 2 hours after distal middle cerebral artery occlusion and then for 6 or 13 days (n=19 per group). Spatial learning and memory were tested using the Morris water maze. Secondary degeneration and mitochondrial apoptosis in the thalamus and hippocampus were assessed using Nissl staining, immunohistochemistry, terminal deoxynucleotidyl transferase dUTP nick end labeling, JC-1 staining, and immunoblotting 7 and 14 days after surgery. RESULTS: Infarct volumes did not significantly differ between the vehicle and 2-Cl-MGV-1 groups. There were more neurons and fewer glia in the ipsilateral thalamus and hippocampus in the vehicle groups than in the sham-operated group 7 and 14 days post-distal middle cerebral artery occlusion. 2-Cl-MGV-1 significantly ameliorated spatial cognitive impairment and decreased neuronal death and glial activation when compared with vehicle treatment (P<0.05). The collapse of mitochondrial transmembrane potential and cytoplasmic release of apoptosis-inducing factors and cytochrome c was prevented within the thalamus. Caspase cleavage and the numbers of terminal deoxynucleotidyl transferase dUTP nick end labeling+ or Nissl atrophic cells were reduced within the thalamus and hippocampus. This was accompanied by upregulation of B-cell lymphoma 2 and downregulation of Bax (P<0.05). CONCLUSIONS: 2-Cl-MGV-1 reduces neuronal apoptosis via mitochondrial-dependent pathways and attenuates secondary damage in the nonischemic thalamus and hippocampus, potentially contributing to ameliorated cognitive deficits after cortical infarction.


Assuntos
Apoptose/efeitos dos fármacos , Carbamatos/uso terapêutico , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/psicologia , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/psicologia , Hipocampo/patologia , Fármacos Neuroprotetores/uso terapêutico , Quinazolinas/uso terapêutico , Tálamo/patologia , Animais , Infarto Cerebral/patologia , Disfunção Cognitiva/etiologia , Hipocampo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Memória/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Neurônios/patologia , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley , Receptores de GABA/biossíntese , Receptores de GABA/genética , Tálamo/efeitos dos fármacos
4.
PLoS One ; 11(4): e0152919, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27054921

RESUMO

UNLABELLED: Chronic inflammation of the gastrointestinal tract increasing the risk of cancer has been described to be linked to the high expression of the mitochondrial translocator protein (18 kDa; TSPO). Accordingly, TSPO drug ligands have been shown to regulate cytokine production and to improve tissue reconstruction. We used HT-29 human colon carcinoma cells to evaluate the role of TSPO and its drug ligands in tumor necrosis factor (TNF)-induced inflammation. TNF-induced interleukin (IL)-8 expression, coupled to reactive oxygen species (ROS) production, was followed by TSPO overexpression. TNF also destabilized mitochondrial ultrastructure, inducing cell death by apoptosis. Treatment with the TSPO drug ligand PK 11195 maintained the mitochondrial ultrastructure, reducing IL-8 and ROS production and cell death. TSPO silencing and overexpression studies demonstrated that the presence of TSPO is essential to control IL-8 and ROS production, so as to maintain mitochondrial ultrastructure and to prevent cell death. Taken together, our data indicate that inflammation results in the disruption of mitochondrial complexes containing TSPO, leading to cell death and epithelia disruption. SIGNIFICANCE: This work implicates TSPO in the maintenance of mitochondrial membrane integrity and in the control of mitochondrial ROS production, ultimately favoring tissue regeneration.


Assuntos
Colo/metabolismo , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Mitocôndrias/metabolismo , Proteínas de Neoplasias/biossíntese , Receptores de GABA/biossíntese , Estresse Fisiológico , Morte Celular , Linhagem Celular Tumoral , Colo/patologia , Neoplasias do Colo/patologia , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Interleucina-8/biossíntese , Mitocôndrias/patologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/patologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/efeitos adversos , Fator de Necrose Tumoral alfa/farmacologia
5.
J Neuroinflammation ; 12: 195, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26511444

RESUMO

BACKGROUND: There are no specific treatments for the neurological alterations of cirrhotic patients with minimal hepatic encephalopathy (MHE). Rats with MHE due to portacaval shunt (PCS) show impaired spatial learning. The underlying mechanisms remain unknown. The aims of this work were to assess: (a) whether PCS rats show neuroinflammation in hippocampus, (b) whether treatment with sildenafil reduces neuroinflammation and restores spatial learning in PCS rats, and (c) analyze the underlying mechanisms. METHODS: Neuroinflammation was assessed by determining inflammatory markers by Western blot. Phosphorylation of MAP-kinase p38 was assessed by immunohistochemistry. Membrane expression of GABA and glutamate receptors was analyzed using BS3 cross-linker. Spatial learning was analyzed using the radial and Morris water mazes. To assess if sildenafil reverses the alterations, rats were treated with sildenafil in the drinking water. RESULTS: PCS rats show increased IL-1ß and TNF-α levels and phosphorylation (activity) of p38 in hippocampus. Membrane expression of subunits α1 of GABAA receptor and GluR2 of AMPA receptor are increased in PCS rats, while subunits GluR1 of AMPA receptors and NR1 and NR2a of NMDA receptors are reduced. PCS rats show reduced spatial learning in the radial and Morris water mazes. Sildenafil treatment normalizes IL-1ß and TNF-α levels, p38 phosphorylation, and membrane expression of GABAA, AMPA, and NMDA receptors and restores spatial learning. CONCLUSIONS: Increased IL-1ß alters GABAergic and glutamatergic neurotransmission in hippocampus and impairs spatial learning in rats with MHE. Sildenafil reduces neuroinflammation and restores learning. Phosphodiesterase-5 inhibitors may be useful to improve cognitive function in patients with MHE.


Assuntos
Encefalopatia Hepática/tratamento farmacológico , Encefalopatia Hepática/psicologia , Inflamação/tratamento farmacológico , Aprendizagem em Labirinto/efeitos dos fármacos , Citrato de Sildenafila/uso terapêutico , Vasodilatadores/uso terapêutico , Animais , Encefalopatia Hepática/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Inflamação/patologia , Interleucina-1beta/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Microglia/efeitos dos fármacos , Derivação Portocava Cirúrgica , Ratos , Ratos Wistar , Receptores de GABA/biossíntese , Receptores de Glutamato/biossíntese , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
J Cell Biochem ; 116(12): 2786-92, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25968977

RESUMO

Lung cancer is prevalent in cigarette smokers. The mitochondrial membrane translocator protein (TSPO), is thought to protect cells from free radical damage. We examined the effect of cigarette smoke (CS) (containing free radicals) alone and in the presence of saliva (containing redox active free iron), on survival of H1299 lung cancer cells and on their mitochondrial characteristics, and whether TSPO binding was influenced by CS and by saliva. We exposed H1299 cells to CS in the presence/absence of saliva and also characterized TSPO binding in the cells using [3H]PK 11195 as a radioligand. CS induced a significant drop in mitochondrial potential (ΔΨm), while addition of saliva did not lead to further loss of ΔΨm (42.5% vs. 39.85%). Scatchard analysis of the saturation curve of [3H]PK 11195 binding (0.2-6 nM final concentration) yielded a straight-line plot (R = 0.9). Average Bmax value was 3274 ± 787 fmol/mg of protein, and average Kd value was 9.2 ± 1.3 nM. Benzodiazepine diazepam partially prevented decrease in cell survival following exposure to CS and redox active iron containing media (saliva) while benzodiazepine clonazepam did not, indicating that this effect is TSPO-specific. Exposure of cells to CS resulted in alternation of biomolecules expressed by CLs peroxidation, reduction of TSPO binding, and depletion of the mitochondrial potential. This irreversible damage was enhanced in the presence of saliva. All these modulations may result in cellular death increase following CS exposure, enhanced in the presence of saliva.


Assuntos
Neoplasias Pulmonares/genética , Nicotiana/efeitos adversos , Receptores de GABA/biossíntese , Fumar/efeitos adversos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Oxirredução/efeitos dos fármacos , Receptores de GABA/genética , Saliva/efeitos dos fármacos , Saliva/metabolismo
7.
Bioorg Med Chem Lett ; 24(18): 4466-4471, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25172419

RESUMO

A novel synthesis of the translocator protein (TSPO) ligand 7-chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (SSR180575, 3) was achieved in four steps from commercially available starting materials. Focused structure-activity relationship development about the pyridazinoindole ring at the N3 position led to the discovery of 7-chloro-N,N,5-trimethyl-4-oxo-3(6-fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (14), a novel ligand of comparable affinity. Radiolabeling with fluorine-18 ((18)F) yielded 7-chloro-N,N,5-trimethyl-4-oxo-3(6-[(18)F]fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide ([(18)F]-14) in high radiochemical yield and specific activity. In vivo studies of [(18)F]-14 revealed this agent as a promising probe for molecular imaging of glioma.


Assuntos
Acetamidas/síntese química , Descoberta de Drogas , Glioma/diagnóstico , Indóis/síntese química , Imagem Molecular , Tomografia por Emissão de Pósitrons , Receptores de GABA/análise , Acetamidas/química , Acetamidas/farmacologia , Animais , Humanos , Indóis/química , Indóis/farmacologia , Ligantes , Masculino , Estrutura Molecular , Ratos , Ratos Wistar , Receptores de GABA/biossíntese
8.
J Nucl Med ; 54(2): 291-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23321458

RESUMO

UNLABELLED: Glial neuroinflammation is associated with the development and progression of multiple sclerosis. PET imaging offers a unique opportunity to evaluate neuroinflammatory processes longitudinally in a noninvasive and clinically translational manner. (18)F-PBR111 is a newly developed PET radiopharmaceutical with high affinity and selectivity for the translocator protein (TSPO), expressed on activated glia. This study aimed to investigate neuroinflammation at different phases of relapsing-remitting (RR) experimental autoimmune encephalomyelitis (EAE) in the brains of SJL/J mice by postmortem histologic analysis and in vivo by PET imaging with (18)F-PBR111. METHODS: RR EAE was induced by immunization with PLP(139-151) peptide in complete Freund's adjuvant. Naive female SJL/J mice and mice immunized with saline-complete Freund's adjuvant were used as controls. The biodistribution of (18)F-PBR111 was measured in 13 areas of the central nervous system and compared with PET imaging results during different phases of RR EAE. The extents of TSPO expression and glial activation were assessed with immunohistochemistry, immunofluorescence, and a real-time polymerase chain reaction. RESULTS: There was significant TSPO expression in all of the central nervous system areas studied at the peak of the first clinical episode and, importantly, at the preclinical stage. In contrast, only a few TSPO-positive cells were observed at the second episode. At the third episode, there was again an increase in TSPO expression. TSPO expression was associated with microglial cells or macrophages without obvious astrocyte labeling. The dynamics of (18)F-PBR111 uptake in the brain, as measured by in vivo PET imaging and biodistribution, followed the pattern of TSPO expression during RR EAE. CONCLUSION: PET imaging with the TSPO ligand (18)F-PBR111 clearly reflected the dynamics of microglial activation in the SJL/J mouse model of RR EAE. The results are the first to highlight the discrepancy between the clinical symptoms of EAE and TSPO expression in the brain, as measured by PET imaging at the peaks of various EAE episodes. The results suggest a significant role for PET imaging investigations of neuroinflammation in multiple sclerosis and allow for in vivo follow-up of antiinflammatory treatment strategies.


Assuntos
Sistema Nervoso Central/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Tomografia por Emissão de Pósitrons/métodos , Receptores de GABA/biossíntese , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/fisiopatologia , Feminino , Imuno-Histoquímica/métodos , Inflamação , Macrófagos/citologia , Camundongos , Microscopia de Fluorescência/métodos , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/fisiopatologia , Transporte Proteico , Reação em Cadeia da Polimerase em Tempo Real/métodos
9.
Cell Tissue Res ; 350(2): 261-75, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22868914

RESUMO

Translocator protein (TSPO), previously known as the peripheral-type benzodiazepine receptor, is a ubiquitous drug- and cholesterol-binding protein primarily found in the outer mitochondrial membrane as part of a mitochondrial cholesterol transport complex. TSPO is present at higher levels in steroid-synthesizing and rapidly proliferating tissues and its biological role has been mainly linked to mitochondrial function, steroidogenesis and cell proliferation/apoptosis. Aberrant TSPO levels have been linked to multiple diseases, including cancer, endocrine disorders, brain injury, neurodegeneration, ischemia-reperfusion injury and inflammatory diseases. Investigation of the functions of this protein in vitro and in vivo have been mainly carried out using high-affinity drug ligands, such as isoquinoline carboxamides and benzodiazepines and more recently, gene silencing methods. To establish a model to study the regulation of Tspo transcription in vivo, we generated a transgenic mouse model expressing green fluorescent protein (GFP) from Aequorea coerulescens under control of the Tspo promoter region (Tspo-AcGFP). The expression profiles of Tspo-AcGFP, endogenous TSPO and Tspo mRNA were found to be well-correlated. Tspo-AcGFP synthesis in the transgenic mice was seen in almost every tissue examined and as with TSPO in wild-type mice, Tspo-AcGFP was highly expressed in steroidogenic cells of the endocrine and reproductive systems, epithelial cells of the digestive system, skeletal muscle and other organs. In summary, this transgenic Tspo-AcGFP mouse model recapitulates endogenous Tspo expression patterns and could be a useful, tractable tool for monitoring the transcriptional regulation and function of Tspo in live animal experiments.


Assuntos
Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Receptores de GABA/biossíntese , Receptores de GABA/genética , Animais , Processos de Crescimento Celular/fisiologia , Modelos Animais de Doenças , Feminino , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de GABA/metabolismo , Transcrição Gênica , Transcriptoma
10.
Patol Fiziol Eksp Ter ; (1): 40-3, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22629859

RESUMO

Phorbol esters are known to modulate protein kinase C activity - enzyme involved in cell proliferation, differentiation and apoptosis regulation in skin cells. Besides it phorbol-12-myristate 13-acetate was shown possible to modulate promoter activity of TsPO - protein that is involved in steroidogenesis and cell proliferation regulation. Caspase-3 and TsPO expression was measured in squamous cell carcinoma cells after incubation with phorbol-12-myristate 13-acetate and ultraviolet radiation. Following alterations of TsPO and caspase-3 levels are explained by different mechanisms of regulation.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/farmacologia , Carcinoma de Células Escamosas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Receptores de GABA/biossíntese , Neoplasias Cutâneas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Carcinoma de Células Escamosas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Proteína Quinase C/metabolismo , Neoplasias Cutâneas/patologia
11.
PLoS One ; 7(5): e37041, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615883

RESUMO

GABA (γ-aminobutyric acid) is the main inhibitory neurotransmitter in the CNS and is present in high concentrations in presynaptic terminals of neuronal cells. More recently, GABA has been ascribed a more widespread role in the control of cell proliferation during development where low concentrations of extrasynaptic GABA induce a tonic activation of GABA receptors. The GABA-A receptor consists of a ligand-gated chloride channel, formed by five subunits that are selected from 19 different subunit isoforms. The functional and pharmacological properties of the GABA-A channels are dictated by their subunit composition. Here we used qRT-PCR to compare mRNA levels of all 19 GABA-A channel subunits in samples of human glioma (n = 29) and peri-tumoral tissue (n = 5). All subunits except the ρ1 and ρ3 subunit were consistently detected. Lowest mRNA levels were found in glioblastoma compared to gliomas of lower malignancy, except for the θ subunit. The expression and cellular distribution of the α1, γ1, ρ2 and θ subunit proteins was investigated by immunohistochemistry on tissue microarrays containing 87 gliomas grade II. We found a strong co-expression of ρ2 and θ subunits in both astrocytomas (r = 0.86, p<0.0001) and oligodendroglial tumors (r = 0.66, p<0.0001). Kaplan-Meier analysis and Cox proportional hazards modeling to estimate the impact of GABA-A channel subunit expression on survival identified the ρ2 subunit (p = 0.043) but not the θ subunit (p = 0.64) as an independent predictor of improved survival in astrocytomas, together with established prognostic factors. Our data give support for the presence of distinct GABA-A channel subtypes in gliomas and provide the first link between specific composition of the A-channel and patient survival.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioma/metabolismo , Glioma/patologia , Receptores de GABA/biossíntese , Ácido gama-Aminobutírico/biossíntese , Adulto , Neoplasias Encefálicas/genética , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Feminino , Glioma/genética , Humanos , Estimativa de Kaplan-Meier , Masculino , Estadiamento de Neoplasias , Prognóstico , Isoformas de Proteínas , Subunidades Proteicas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de GABA/genética , Estudos Retrospectivos , Taxa de Sobrevida , Ácido gama-Aminobutírico/genética
12.
Ann Neurol ; 71(4): 539-51, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22447678

RESUMO

OBJECTIVE: The most common neurological symptom of tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) is early life refractory epilepsy. As previous studies have shown enhanced excitatory glutamatergic neurotransmission in TSC and FCD brains, we hypothesized that neurons associated with these lesions may also express altered γ-aminobutyric acid (GABA)(A) receptor (GABA(A)R)-mediated inhibition. METHODS: Expression of the GABA(A)R subunits α1 and α4, and the Na(+)-K(+)-2Cl(-) (NKCC1) and the K(+)-Cl(-) (KCC2) transporters, in human TSC and FCD type II specimens were analyzed by Western blot and double label immunocytochemistry. GABA(A) R responses in dysplastic neurons from a single case of TSC were measured by perforated patch recording and compared to normal-appearing cortical neurons from a non-TSC epilepsy case. RESULTS: TSC and FCD type IIb lesions demonstrated decreased expression of GABA(A)R α1, and increased NKCC1 and decreased KCC2 levels. In contrast, FCD type IIa lesions showed decreased α4, and increased expression of both NKCC1 and KCC2 transporters. Patch clamp recordings from dysplastic neurons in acute slices from TSC tubers demonstrated excitatory GABA(A)R responses that were significantly attenuated by the NKCC1 inhibitor bumetanide, in contrast to hyperpolarizing GABA(A)R-mediated currents in normal neurons from non-TSC cortical slices. INTERPRETATION: Expression and function of GABA(A)Rs in TSC and FCD type IIb suggest the relative benzodiazepine insensitivity and more excitatory action of GABA compared to FCD type IIa. These factors may contribute to resistance of seizure activity to anticonvulsants that increase GABAergic function, and may justify add-on trials of the NKCC1 inhibitor bumetanide for the treatment of TSC and FCD type IIb-related epilepsy.


Assuntos
Encefalopatias/metabolismo , Malformações do Desenvolvimento Cortical/metabolismo , Inibição Neural/fisiologia , Neurônios/metabolismo , Esclerose Tuberosa/metabolismo , Adolescente , Adulto , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Encefalopatias/complicações , Encefalopatias/patologia , Criança , Pré-Escolar , Epilepsia/etiologia , Epilepsia/metabolismo , Epilepsia/patologia , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Malformações do Desenvolvimento Cortical/complicações , Malformações do Desenvolvimento Cortical/patologia , Malformações do Desenvolvimento Cortical do Grupo I , Neurônios/patologia , Técnicas de Patch-Clamp , Receptores de GABA/biossíntese , Simportadores de Cloreto de Sódio-Potássio/biossíntese , Membro 2 da Família 12 de Carreador de Soluto , Simportadores/biossíntese , Esclerose Tuberosa/complicações , Esclerose Tuberosa/patologia , Adulto Jovem , Cotransportadores de K e Cl-
13.
J Bioenerg Biomembr ; 43(6): 739-46, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22127435

RESUMO

The role of the TSPO in metabolism of human osteoblasts is unknown. We hypothesized that human osteoblast metabolism may be modulated by the TSPO. Therefore we evaluated the presence of TSPO in human osteoblast-like cells and the effect of its synthetic ligand PK 11195 on these cells. The presence of TSPO was determined by [(3)H]PK 11195 binding using Scatchard analysis: Bmax 7682 fmol/mg, Kd 9.24 nM. PK 11195 did not affect significantly cell proliferation, cell death, cellular viability, maturation, [(18)F]-FDG incorporation and hexokinase 2 gene expression or protein levels. PK 11195 exerted a suppressive effect on VDAC1 and caused an increase in TSPO gene expression or protein levels. In parallel there was an increase in mitochondrial mass, mitochondrial ATP content and a reduction in ΔΨm collapse. Thus, it appears that PK11195 (10(-5) M) stimulates mitochondrial activity in human osteoblast-like cells without affecting glycolytic activity and cell death.


Assuntos
Antineoplásicos/farmacologia , Isoquinolinas/farmacologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/biossíntese , Receptores de GABA/biossíntese , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Hexoquinase/biossíntese , Humanos , Osteoblastos , Canal de Ânion 1 Dependente de Voltagem/biossíntese
14.
Neuron Glia Biol ; 7(2-4): 187-97, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-22874716

RESUMO

Vinpocetine has long been used for cerebrovascular disorders and cognitive impairment. Based on the evidence that the translocator protein (TSPO, 18 kDa) was expressed in activated microglia, while Vinpocetine was able to bind TSPO, we explored the role of Vinpocetine on microglia treated with lipopolysaccharide (LPS) and oxygen-glucose deprivation (OGD) in vitro. Our results show that both LPS and OGD induced the up-regulation of TSPO expression on BV-2 microglia by RT-PCR, western blot and immunocytochemistry. Vinpocetine inhibited the production of nitrite oxide and inflammatory factors such as interleukin-1ß (IL-1ß), IL-6 and tumour necrosis factor-α (TNF-α) in BV-2 microglia, in which cells were treated with LPS or exposed to OGD, regardless of the time Vinpocetine was added. Next, we measured cell death-related molecules Akt, Junk and p38 as well as inflammation-related molecules nuclear factor-κB (NF-κB) and activator protein-1 (AP-1). Vinpocetine did not change cell death-related molecules, but inhibited the expression of NF-κB and AP-1 in LPS-stimulated microglia, indicating that Vinpocetine has an anti-inflammatory effect by partly targeting NF-κB/AP-1. Next, conditioned medium from Vinpocetine-treated microglia protected from primary neurons. As compared with in vitro, the administration of Vinpocetine in hypoxic mice also inhibited inflammatory molecules, indicating that Vinpocetine as a unique anti-inflammatory agent may be beneficial for the treatment of neuroinflammatory diseases.


Assuntos
Córtex Cerebral/metabolismo , Regulação para Baixo/fisiologia , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Receptores de GABA/biossíntese , Alcaloides de Vinca/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Transformada , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Fármacos Neuroprotetores/uso terapêutico , Regulação para Cima/fisiologia , Alcaloides de Vinca/farmacologia
15.
Biochemistry ; 49(23): 4766-78, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20469933

RESUMO

Translocator protein TSPO is an 18 kDa protein implicated in numerous cell functions and is highly expressed in secretory and glandular tissues, especially in steroidogenic cells. TSPO expression is altered in pathological conditions such as certain cancers and neurological diseases. In search of the factors regulating Tspo expression, we recently showed that high levels of TSPO in steroidogenic cells may be due to high constitutive expression of protein kinase Cepsilon (PKCepsilon), while phorbol 12-myristate 13-acetate (PMA) activation of PKCepsilon drives inducible TSPO expression in nonsteroidogenic cells, likely through activator protein 1 (AP1). In this study, we aimed to identify the signal transduction pathway through which PKCepsilon regulates Tspo gene expression. The MEK1/2 specific inhibitor U0126, but not NFkappaB inhibitors, reduced basal Tspo promoter activity in TSPO-rich steroidogenic cells (MA-10 Leydig), as well as basal and PMA-induced Tspo promoter levels in TSPO-poor nonsteroidogenic cells (NIH-3T3 fibroblasts). AP1 and signal transducer and activation of transcription 3 (STAT3) have binding sites in the Tspo promoter and are downstream targets of PKCepsilon and MAPK (Raf-1-ERK1/2) pathways. PKCepsilon overexpression induced STAT3 phosphorylation in NIH-3T3 cells, while PKCepsilon knockdown reduced STAT3 and c-Jun phosphorylation in Leydig cells. MEK1/2, ERK2, c-Jun, and STAT3 knockdown reduced Tspo mRNA and protein levels in Leydig cells. Additionally, Raf-1 reduced Tspo mRNA levels in the same cells. MEK1/2, c-Jun, and STAT3 knockdown also reduced basal as well as PMA-induced Tspo mRNA levels in NIH-3T3 cells. Together, these results demonstrate that PKCepsilon regulates Tspo gene expression through a MAPK (Raf-1-MEK1/2-ERK1/2) signal transduction pathway, acting at least in part through c-Jun and STAT3 transcription factors.


Assuntos
Regulação Enzimológica da Expressão Gênica , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase C-épsilon/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores de GABA/biossíntese , Receptores de GABA/genética , Fator de Transcrição STAT3/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Marcação de Genes/métodos , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Camundongos , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Receptores de GABA/deficiência , Fator de Transcrição STAT3/deficiência , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia
16.
Anesthesiology ; 110(1): 106-12, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19104177

RESUMO

BACKGROUND: Midazolam is widely used as an intravenous sedative. However, the role of midazolam on vascular endothelial activation is still unknown. The present study explores the action of midazolam on endothelial activation and its role to peripheral benzodiazepine receptor (PBR) in cultured human umbilical vein endothelial cells. METHODS: Intracellular localization of PBR in human umbilical vein endothelial cells was visualized with immunofluorescent staining. Monocyte adhesion and vascular cell adhesion molecule-1 expression were measured with monocyte adhesion assay and Western blot analysis. Involvement of PBR was assessed by using specific antagonists and small interfering RNA against PBR. RESULTS: PBR was localized in the mitochondria of human umbilical vein endothelial cells. Midazolam significantly inhibited tumor necrosis factor-alpha-induced vascular cell adhesion molecule-1 and monocyte adhesion in a dose-dependent manner (1-30 microM). The midazolam-mediated suppression on the tumor necrosis factor-alpha-induced vascular cell adhesion molecule-1 expression and monocyte adhesion were inhibited by the pretreatment of PK11195 and not inhibited by the flumazenil. Transfection of small interfering RNA for PBR decreased the expression of PBR (18 kDa) in human umbilical vein endothelial cells. Midazolam-mediated suppression on the tumor necrosis factor-alpha-induced vascular cell adhesion molecule-1 expression was abrogated by the transfection of small interfering RNA for PBR. CONCLUSION: These results suggest that midazolam has an inhibitory action on the endothelial activation and that its action is related to the activation of peripheral benzodiazepine receptor localized in mitochondria of the endothelial cells.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Midazolam/farmacologia , Receptores de GABA/biossíntese , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Células Cultivadas , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Receptores de GABA/análise , Fator de Necrose Tumoral alfa/metabolismo , Veias Umbilicais/efeitos dos fármacos , Veias Umbilicais/metabolismo , Molécula 1 de Adesão de Célula Vascular/biossíntese
17.
J Pharmacol Exp Ther ; 323(1): 236-47, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17636008

RESUMO

The gamma-aminobutyric acid type C (GABAC) receptor is structurally related to the GABAA receptors, yet quite distinct physiologically and pharmacologically. Neuroactive steroids are known to be potent and efficacious modulators of the GABAA receptor, but they are less well characterized in their actions on the GABAC receptor. We have examined the actions of neuroactive steroids and analogs on rho1 (GABAC) receptors expressed in Xenopus laevis oocytes, with two goals in mind. First, we tested a larger number of endogenous steroids, to determine whether particularly potent steroids could be found. Second, we examined the structure-activity relationship for steroid actions, and some mechanistic features, to determine the possible numbers of steroid binding sites and mechanisms of action. In total, 41 compounds were examined. Estradiols are inhibitors, essentially equipotent with picrotoxinin. No endogenous steroid tested was highly efficacious at potentiation. The results of the structure-activity studies and the effects of two mutations to the second transmembrane region of the rho1 GABAC receptor indicate that there are several mechanisms by which steroids can inhibit the receptor. Surprisingly, estradiol shares some features with picrotoxin. Inhibition by negatively charged compounds was not sensitive to membrane potential, and inhibition by all compounds tested was reduced at higher concentrations of GABA. The data indicate that the binding sites mediating potentiation and inhibition differ from each other and that there are several (three or more) mechanisms for producing inhibition.


Assuntos
Antagonistas GABAérgicos/farmacologia , Proteínas Recombinantes/antagonistas & inibidores , Esteroides/farmacologia , Animais , Antagonistas GABAérgicos/química , Humanos , Estrutura Molecular , Mutação , Oócitos/metabolismo , Técnicas de Patch-Clamp , Receptores de GABA/biossíntese , Receptores de GABA/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Estereoisomerismo , Esteroides/química , Relação Estrutura-Atividade , Xenopus laevis
18.
Eur Surg Res ; 39(6): 359-63, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17652962

RESUMO

BACKGROUND: The peripheral-type benzodiazepine receptor or translocator protein (TSPO) is an 18-kDa protein involved in cell proliferation and apoptosis. TSPO was shown to be overexpressed in malignant tumors and cancer cell lines, correlating with enhanced malignant behavior. The present study analyzed the role of TSPO in patients with colorectal carcinomas. METHODS: Tumor tissues and corresponding normal mucosa from 55 patients who underwent resection for colorectal carcinomas were analyzed for TSPO expression in correlation to GAPDH expression(glyceraldehyde-3-phosphate dehydrogenase) using a multiplex RT-PCR assay. RESULTS: TSPO was overexpressed in 67% of the tumors in comparison to corresponding normal mucosa, and positivity as well as expression levels in colon carcinomas were significantly higher than in the rectum carcinomas. In contrast, TSPO expression was not different in intermediate versus high-grade tumors or in lymph node-positive versus -negative patients. CONCLUSION: The differences in TSPO expression between colon and rectum carcinoma may imply that these tumors are of different biological behavior.


Assuntos
Neoplasias do Colo/metabolismo , Receptores de GABA/biossíntese , Neoplasias Retais/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias do Colo/patologia , Humanos , Intestino Grosso/metabolismo , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Mensageiro , Neoplasias Retais/patologia
19.
Biol Cell ; 99(11): 639-47, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17561806

RESUMO

BACKGROUND INFORMATION: TSPO (translocator protein), known previously as PBR (peripheral-type benzodiazepine receptor), is a 18 kDa protein expressed in the mitochondrial membrane of a variety of tissues. TSPO has been reported to be over-expressed in human colorectal tumours and cancer cell lines, but its function is not well characterized. RESULTS: We investigated the expression and function of TSPO in the human colon cancer cells HT-29. Immunohistochemical studies revealed that TSPO is localized in mitochondria, and its endogenous ligand, the polypeptide diazepam-binding inhibitor, in the cytosol. Radioligand binding studies using the specific high-affinity drug ligand [(3)H]PK 11195 and membrane fraction demonstrated saturable binding, with K(d) and B(max) values of 13.5+/-1.5 nM and 10.1+/-1.0 pmol/mg respectively. PK 11195 induced a rapid and transient dose-dependent rise in intracellular [Ca(2+)], which was unaffected by extracellular Ca(2+), but was blocked by the PTP (permeability transition pore) inhibitor, cyclosporin A, and by the TSPO partial agonist, flunitrazepam. Using HT-29 clone 19A cell line, which forms cell monolayers, we demonstrated that TSPO ligand stimulated a Ca(2+)-dependent transepithelial Cl(-) secretion. This secretion was inhibited: (i) after removal of extracellular Cl(-); (ii) by apical addition of the Cl(-) channel blocker NPPB [5-nitro-2-(3-phenylpropylamino)-benzoate]; and (iii) by basolateral addition of the Na(+)-K(+)-2Cl(-) co-transporter inhibitor bumetanide. Furthermore, the intracellular Ca(2+) chelator BAPTA/AM [bis-(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis(acetoxymethyl ester)] and cyclosporin A abolished the rise in PK 11195-induced Cl(-) secretion. CONCLUSIONS: These findings indicate that TSPO is located in mitochondrial membranes of HT-29 and reveal that its activation induces a rise in cytosolic Ca(2+), leading to the stimulation of Cl(-) secretion.


Assuntos
Antineoplásicos/farmacologia , Cálcio/metabolismo , Cloretos/metabolismo , Isoquinolinas/farmacologia , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/biossíntese , Proteínas de Neoplasias/biossíntese , Receptores de GABA/biossíntese , Inibidores da Angiogênese/farmacologia , Linhagem Celular Tumoral , Neoplasias do Colo , Ciclosporina/farmacologia , Inibidores Enzimáticos/farmacologia , Flunitrazepam/farmacologia , Moduladores GABAérgicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Mitocôndrias/metabolismo , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Nitrobenzoatos/farmacologia , Simportadores de Cloreto de Sódio-Potássio
20.
Anticancer Res ; 26(1A): 9-22, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16475673

RESUMO

The peripheral-type benzodiazepine receptor (PBR) is an 18-kDa high affinity drug- and cholesterol-binding protein that is involved in various cell functions, including cell proliferation and apoptosis. PBR was shown to be overexpressed in certain types of malignant human tumors and cancer cell lines, correlating with enhanced tumorigenicity and cell proliferation rates. The present study was conducted in order to further define the role of PBR in cancer and to extend our recent findings regarding the possible anticancer effects of the standardized Ginkgo biloba extract EGb 761. Treatment with EGb 761 decreased PBR mRNA levels and inhibited the proliferation of breast, glioma and hepatocarcinoma cell lines, further corroborating our previous contention that its mechanism of action is through the modification of PBR expression. In vivo treatment with Ginkgo biloba extract led to dose-dependent decreases in xenograft growth of both MDA-MB-231 breast cancer and U-87 glioma cell lines in nude mice, although the effects were not maintained after 50 days of treatment in the latter. The results obtained in MDA-MB-231 xenografts indicated pronounced inhibition of tumor growth, verified by MRI imaging. These results were obtained using a modified experimental protocol where the animals were treated with the extract before cell inoculation. Although an exact role for PBR in relation to the initiation and progression of various types of cancer remains to be defined, our results indicate that PBR overexpression in certain cancer cells is related to an aggressive phenotype. Since EGb 761 treatment opposes this aggressive phenotype by decreasing PBR overexpression, it could be useful in preventing or treating cancer invasiveness and metastasis.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Extratos Vegetais/farmacologia , Receptores de GABA/biossíntese , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Receptor alfa de Estrogênio/biossíntese , Receptor beta de Estrogênio/biossíntese , Feminino , Ginkgo biloba , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Neoplasias/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ensaio Radioligante , Receptores de GABA/genética , Receptores de GABA/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA