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.
Brain Res ; 1741: 146879, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32418890

RESUMO

The nitric oxide (NO) metabolome and the NO metabolite-based neurovascular protective pathways are dysregulated after stroke. The major NO metabolite S-nitrosoglutahione (GSNO) is essential for S-nitrosylation-based signaling events and the inhibition of S-nitrosoglutahione (GSNO)-metabolizing enzyme GSNO reductase (GSNOR) provides protective effects following cardiac ischemia. However, the role of GSNOR and GSNOR inhibition-mediated increased GSNO/S-nitrosylation is not understood in neurovascular diseases such as stroke. Because age is the major risk factor of stroke and recovery in aged stroke patients is low and slow, we investigated the efficacy of GSNOR inhibition using a GSNOR selective inhibitor N6022 in a clinically relevant middle-aged cerebral ischemia and reperfusion (IR) mouse model of stroke. N6022 (5 mg/kg; iv) treatment of IR mice at 2 h after reperfusion followed by the treatment of the same dose daily for 3 days reduced the infarct volume and decreased the neurological score. Daily treatment of IR animals with N6022 for 2 weeks significantly improved neurological score, brain infarctions/atrophy, survival rate, motor (measured by cylinder test) and cognitive (evaluated by novel object recognition test) functions which paralleled the decreased activity of GSNOR, reduced levels of peroxynitrite and decreased neurological score. These results are the first evidence of a new pathway for the treatment of stroke via the inhibition of GSNOR. Based on the efficacy of N6022 in the stroke animal model and its use in human therapeutic studies without toxicity, we submit that GSNOR is a druggable target, and N6022 is a promising drug candidate for human stroke therapy.


Assuntos
Envelhecimento/efeitos dos fármacos , Álcool Desidrogenase/antagonistas & inibidores , Benzamidas/administração & dosagem , Modelos Animais de Doenças , Pirróis/administração & dosagem , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Envelhecimento/metabolismo , Álcool Desidrogenase/metabolismo , Animais , Sistemas de Liberação de Medicamentos/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
2.
Immunobiology ; 223(10): 549-554, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29960806

RESUMO

In this study, we investigated IL-10 and IL-17 specific immunomodulatory potential of S-nitrosoglutathione (GSNO), a physiological nitric oxide carrier molecule, in experimental autoimmune encephalomyelitis (EAE). In active EAE model, GSNO treatment attenuated EAE severity and splenic CD4+ T cells isolated from these mice exhibited decreased IL-17 expression without affecting the IFN-γ expression compared to the cells from untreated EAE mice. Similarly, adoptive transfer of these cells to nave mice resulted in reduction in IL-17 expression in the spinal cords of recipient mice with milder EAE severity. CD4+ T cells isolated from GSNO treated EAE mice, as compared to untreated EAE mice, still expressed lower levels of IL-17 under TH17 skewing conditions, but expressed similar levels of IFN-γ under TH1 skewing condition. Interestingly, under both TH17 and TH1 skewing condition, CD4+ T cells isolated from GSNO treated EAE mice, as compared to untreated EAE mice, expressed higher levels of IL-10 and adoptive transfer of these TH17 and TH1 skewed cells seemingly exhibited milder EAE disease. In addition, adoptive transfer of CD4+ T cells from GSNO treated EAE mice to active EAE mice also ameliorated EAE disease with induction of spinal cord expression of IL-10 and reduction in of IL-17, thus suggesting the participation of IL-10 mechanism in GSNO mediated immunomodulation. GSNO treatment of mice passively immunized with CD4+ T cells either from GSNO treated EAE mice or untreated mice further ameliorated EAE disease, supporting efficacy of GSNO for prophylaxis and therapy in EAE. Overall, these data document a modulatory role of GSNO in IL-17/IL-10 axis of EAE and other autoimmune diseases.


Assuntos
Transferência Adotiva , Encefalomielite Autoimune Experimental/terapia , Imunomodulação , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Doadores de Óxido Nítrico/farmacologia , S-Nitrosoglutationa/farmacologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Encefalomielite Autoimune Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th1/transplante , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Células Th17/transplante
3.
Neuroscience ; 384: 41-53, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29782905

RESUMO

Alzheimer's disease (AD) is the most common form of dementia that is often accompanied by mood and emotional disturbances and seizures. There is growing body of evidence that neurons expressing γ-aminobutyric acid (GABA) play an important role in regulation of cognition, mood, and emotion as well as seizure susceptibility, but participation of GABAergic neuronal pathology in Alzheimer's disease (AD) is not understood well at present. Here, we report that transgenic mice expressing human amyloid precursor protein Swedish-Dutch-Iowa mutant (APPSweDI) exhibit early loss of neurons expressing GAD67, a GABA-synthesizing enzyme, in advance of the loss of pyramidal neurons in hippocampal CA1 region. The loss of GAD67+ neurons in APPSweDI mice accompanied with decreased spatial cognition as well as increased anxiety-like behaviors and kainic acid-induced seizure susceptibility at early phase. In the hippocampal CA1 region, GAD67+ neurons expressed high basal levels of neuronal nitric oxide synthase (nNOS) and nitrosative stress (nitrotyrosine). Similarly, GAD67+ neurons in primary cortical and hippocampal neuron cultures also expressed high basal levels of nNOS and degenerated in response to lower Aß concentrations due to their high basal levels of nitrosative stress. Given the role of GABAergic neurons in cognitive and neuropsychiatric functions, this study reports the role of nNOS-mediated nitrosative stress in dysfunction of GABAergic neurons and its potential participation in early development of cognitive and neuropsychiatric symptoms in AD.


Assuntos
Doença de Alzheimer/metabolismo , Neurônios GABAérgicos/metabolismo , Hipocampo/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Neurônios GABAérgicos/patologia , Hipocampo/patologia , Camundongos , Camundongos Transgênicos
4.
Free Radic Biol Med ; 106: 245-253, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28232202

RESUMO

Numerous reports suggest that aberrant activations of STAT3 and NF-κB promote survival and proliferation of multiple myeloma (MM) cells. In the present report, we demonstrate that a synthetic S-nitrosothiol compound, S-nitroso-N-acetylcysteine (SNAC), inhibits proliferation and survival of multiple MM cells via S-nitrosylation-dependent inhibition of STAT3 and NF-κB. In human MM cells (e.g. U266, H929, and IM-9 cells), SNAC treatment increased S-nitrosylation of STAT3 and NF-κB and inhibited their activities. Consequently, SNAC treatment resulted in MM cell cycle arrest at G1/S check point and inhibited their proliferation. SNAC also decreased the expression of cell survival factors and increased the activities of caspases, thus increased sensitivity of MM cells to melphalan, a chemotherapeutic agent for MM. In U266 xenografted mice, SNAC treatment decreased the activity of STAT3 and reduced the growth of human CD138 positive cells (U266 cells) in the bone marrow and also reduced their production of human IgE into the serum. Taken together, these data document the S-nitrosylation mediated inhibition of MM cell proliferation and cell survival via inhibition of STAT3 and NF-κB pathways and its efficacy in animal model of MM.


Assuntos
Acetilcisteína/análogos & derivados , Mieloma Múltiplo/metabolismo , NF-kappa B/genética , Fator de Transcrição STAT3/genética , Acetilcisteína/administração & dosagem , Acetilcisteína/síntese química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Neurosci Res ; 94(11): 990-1006, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27638584

RESUMO

Krabbe's disease (KD; also called globoid cell leukodystrophy) is a genetic disorder involving demyelination of the central (CNS) and peripheral (PNS) nervous systems. The disease may be subdivided into three types, an infantile form, which is the most common and severe; a juvenile form; and a rare adult form. KD is an autosomal recessive disorder caused by a deficiency of galactocerebrosidase activity in lysosomes, leading to accumulation of galactoceramide and neurotoxic galactosylsphingosine (psychosine [PSY]) in macrophages (globoid cells) as well as neural cells, especially in oligodendrocytes and Schwann cells. This ultimately results in damage to myelin in both CNS and PNS with associated morbidity and mortality. Accumulation of PSY, a lysolipid with detergent-like properties, over a threshold level could trigger membrane destabilization, leading to cell lysis. Moreover, subthreshold concentrations of PSY trigger cell signaling pathways that induce oxidative stress, mitochondrial dysfunction, apoptosis, inflammation, endothelial/vascular dysfunctions, and neuronal and axonal damage. From the time the "psychosine hypothesis" was proposed, considerable efforts have been made in search of an effective therapy for lowering PSY load with pharmacological, gene, and stem cell approaches to attenuate PSY-induced neurotoxicity. This Review focuses on the recent advances and prospective research for understanding disease mechanisms and therapeutic approaches for KD. © 2016 Wiley Periodicals, Inc.


Assuntos
Leucodistrofia de Células Globoides/patologia , Leucodistrofia de Células Globoides/terapia , Animais , Encéfalo/patologia , Doenças Desmielinizantes , Galactosilceramidase/deficiência , Humanos , Leucodistrofia de Células Globoides/classificação , Leucodistrofia de Células Globoides/genética , Lisossomos/enzimologia , Sistema Nervoso/patologia , Neurônios/metabolismo , Neurônios/patologia , Psicosina/metabolismo
6.
Brain Res ; 1624: 359-369, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26271717

RESUMO

We have previously reported that treatment of rats subjected to permanent bilateral common carotid artery occlusion (pBCCAO), a model of chronic cerebral hypoperfusion (CCH), with S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier, improved cognitive functions and decreased amyloid-ß accumulation in the brains. Since CCH has been implicated in tau hyperphosphorylation induced neurodegeneration, we investigated the role of GSNO in regulation of tau hyperphosphorylation in rat pBCCAO model. The rats subjected to pBCCAO had a significant increase in tau hyperphosphorylation with increased neuronal loss in hippocampal/cortical areas. GSNO treatment attenuated not only the tau hyperphosphorylation, but also the neurodegeneration in pBCCAO rat brains. The pBCCAO rat brains also showed increased activities of GSK-3ß and Cdk5 (major tau kinases) and GSNO treatment significantly attenuated their activities. GSNO attenuated the increased calpain activities and calpain-mediated cleavage of p35 leading to production of p25 and aberrant Cdk5 activation. In in vitro studies using purified calpain protein, GSNO treatment inhibited calpain activities while 3-morpholinosydnonimine (a donor of peroxynitrite) treatment increased its activities, suggesting the opposing role of GSNO vs. peroxynitrite in regulation of calpain activities. In pBCCAO rat brains, GSNO treatment attenuated the expression of inducible nitric oxide synthase (iNOS) expression and also reduced the brain levels of nitro-tyrosine formation, thereby indicating the protective role of GSNO in iNOS/nitrosative-stress mediated calpain/tau pathologies under CCH conditions. Taken together with our previous report, these data support the therapeutic potential of GSNO, a biological NO carrier, as a neuro- and cognitive-protective agent under conditions of CCH.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , S-Nitrosoglutationa/uso terapêutico , Proteínas tau/metabolismo , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Calpaína/metabolismo , Doença Crônica , Quinase 5 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
7.
Redox Biol ; 6: 41-50, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26177470

RESUMO

S-nitrosoglutathione (GSNO) is an endogenous nitric oxide (NO) carrier that plays a critical role in redox based NO signaling. Recent studies have reported that GSNO regulates the activities of STAT3 and NF-κB via S-nitrosylation dependent mechanisms. Since STAT3 and NF-κB are key transcription factors involved in tumor progression, chemoresistance, and metastasis of head and neck cancer, we investigated the effect of GSNO in cell culture and mouse xenograft models of head and neck squamous cell carcinoma (HNSCC). For the cell culture studies, three HNSCC cell lines were tested (SCC1, SCC14a and SCC22a). All three cell lines had constitutively activated (phosphorylated) STAT3 (Tyr(705)). GSNO treatment of these cell lines reversibly decreased the STAT3 phosphorylation in a concentration dependent manner. GSNO treatment also decreased the basal and cytokine-stimulated activation of NF-κB in SCC14a cells and reduced the basal low degree of nitrotyrosine by inhibition of inducible NO synthase (iNOS) expression. The reduced STAT3/NF-κB activity by GSNO treatment was correlated with the decreased cell proliferation and increased apoptosis of HNSCC cells. In HNSCC mouse xenograft model, the tumor growth was reduced by systemic treatment with GSNO and was further reduced when the treatment was combined with radiation and cisplatin. Accordingly, GSNO treatment also resulted in decreased levels of phosphorylated STAT3. In summary, these studies demonstrate that GSNO treatment blocks the NF-κB and STAT3 pathways which are responsible for cell survival, proliferation and that GSNO mediated mechanisms complement cispaltin and radiation therapy, and thus could potentiate the therapeutic effect in HNSCC.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/terapia , Cisplatino/farmacologia , Neoplasias de Cabeça e Pescoço/terapia , Doadores de Óxido Nítrico/farmacologia , S-Nitrosoglutationa/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quimioterapia Combinada , Raios gama/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Camundongos , Camundongos Nus , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biochem Biophys Res Commun ; 458(1): 214-9, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25640839

RESUMO

Hyperphosphorylation and polymerization of microtubule-associated protein tau into paired helical filaments (PHFs) is one of the hallmarks of Alzheimer's disease (AD). Here we report that neuronal tau hyperphosphorylation under AD conditions is regulated by S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier molecule. In cultured rat cortical primary neurons, we observed that GSNO treatment decreased the ß-amyloid (Aß25₋35)-induced pathological tau hyperphosphorylation (Ser396, Ser404, and Ser202/Thr205). The decreased tau hyperphosphorylation correlated with decreased activity of calpain and decreased p35 proteolysis into p25 and Cdk5 activation. GSNO treatment also attenuated the Aß25₋35-induced activation of GSK-3ß which is known to play critical role in tau hyperphosphorylation in addition to Cdk5. Consistent with above studies using cultured neurons, we also observed that systemic GSNO treatment of transgenic mouse model of AD (APPSw/PS1(dE9)) attenuated calpain-mediated p35 proteolysis and Cdk5/GSK-3ß activities as well as tau hyperphosphorylation. In addition, GSNO treatment provided neuro- and cognitive protection in APPSw/PS1(dE9) mice. This study describing the GSNO-mediated regulation of tau hyperphosphorylation and cognitive function, for the first time, suggests for therapeutic potential of GSNO as neuro- and cognitive-protective agent for AD.


Assuntos
Neurônios/metabolismo , S-Nitrosoglutationa/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Animais , Calpaína/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Fosforilação , Ratos Sprague-Dawley , S-Nitrosoglutationa/farmacologia
9.
Redox Biol ; 5: 416-417, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-28162277

RESUMO

In this study, we assessed S-nitrosylation-based regulation of Janus-activated kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway. Our studies show that STAT3 in stimulated microglia underwent two distinct redox-dependent modifications, S-nitrosylation and S-glutathionylation. STAT3 S-nitrosylation was associated with inducible nitric oxide synthase (iNOS)-produced nitric oxide (NO) and S-nitrosoglutathione (GSNO), whereas S-glutathionylation of STAT3 was associated with cellular oxidative stress. NO produced by iNOS or treatment of microglia with exogenous GSNO inhibited STAT3 activation via inhibiting STAT3 phosphorylation (Tyr705). Consequently, the interleukin-6 (IL-6)-induced microglial proliferation and associated gene expressions were also reduced. In cell-free kinase assay using purified JAK2 and STAT3, STAT3 phosphorylation was inhibited by its selective preincubation with GSNO, but not by preincubation of JAK2 with GSNO, indicating that GSNO-mediated mechanisms inhibit STAT3 phosphorylation through S-nitrosylation of STAT3 rather than JAK2. In this study, we identified that Cys259 was the target Cys residue of GSNO-mediated S-nitrosylation of STAT3. The replacement of Cys259 residue with Ala abolished the inhibitory role of GSNO in IL-6-induced STAT3 phosphorylation and transactivation, suggesting the role of Cys259S-nitrosylation in STAT3phosphorylation. Since STAT3 activation is involved in tumor progression and metastasis, we investigated the effect of GSNO in cell culture and mouse xenograft model of head and neck squamous cell carcinoma (HNSCC). GSNO treatment of HNSCCN cell lines reversibly decreases the activation (phosphorylation) of STAT3 in a concentration dependent manner. The reduced STAT3/NF-kB activity by GSNO correlated with decreased cell proliferation and increased apoptosis of HNSCC cells. In HNSCC mouse xenograft model, the tumor growth was reduced by systemic treatment with GSNO and was further reduced when the treatment combined with radiation and cisplatin. Accordingly, GSNO treatment also resulted in decreased levels of pSTAT3 and tumor growth regulators (ie. cyclin D2, VEGF and Bcl-2) in tumor tissue. In summary, these findings have implications for the development of new therapeutics targeting of STAT3 for treating diseases associated with inflammatory/immune responses and abnormal cell proliferation, including cancer.


Assuntos
Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Fator de Transcrição STAT3/metabolismo , Animais , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Proteínas de Neoplasias/genética , Neoplasias/genética , Óxido Nítrico/genética , Oxirredução , Fosforilação , Fator de Transcrição STAT3/genética
10.
Antioxid Redox Signal ; 20(16): 2514-27, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24063605

RESUMO

AIMS: S-nitrosylation and S-glutathionylation, redox-based modifications of protein thiols, are recently emerging as important signaling mechanisms. In this study, we assessed S-nitrosylation-based regulation of Janus-activated kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway that plays critical roles in immune/inflammatory responses and tumorigenesis. RESULTS: Our studies show that STAT3 in stimulated microglia underwent two distinct redox-dependent modifications, S-nitrosylation and S-glutathionylation. STAT3 S-nitrosylation was associated with inducible nitric oxide synthase (iNOS)-produced nitric oxide (NO) and S-nitrosoglutathione (GSNO), whereas S-glutathionylation of STAT3 was associated with cellular oxidative stress. NO produced by iNOS or treatment of microglia with exogenous GSNO inhibited STAT3 activation via inhibiting STAT3 phosphorylation (Tyr(705)). Consequently, the interleukin-6 (IL-6)-induced microglial proliferation and associated gene expressions were also reduced. In cell-free kinase assay using purified JAK2 and STAT3, STAT3 phosphorylation was inhibited by its selective preincubation with GSNO, but not by preincubation of JAK2 with GSNO, indicating that GSNO-mediated mechanisms inhibit STAT3 phosphorylation through S-nitrosylation of STAT3 rather than JAK2. In this study, we identified that Cys(259) was the target Cys residue of GSNO-mediated S-nitrosylation of STAT3. The replacement of Cys(259) residue with Ala abolished the inhibitory role of GSNO in IL-6-induced STAT3 phosphorylation and transactivation, suggesting the role of Cys(259) S-nitrosylation in STAT3 phosphorylation. INNOVATION: Microglial proliferation is regulated by NO via S-nitrosylation of STAT3 (Cys(259)) and inhibition of STAT3 (Tyr(705)) phosphorylation. CONCLUSION: Our results indicate the regulation of STAT3 by NO-based post-translational modification (S-nitrosylation). These findings have important implications for the development of new therapeutics targeting STAT3 for treating diseases associated with inflammatory/immune responses and abnormal cell proliferation, including cancer.


Assuntos
Inflamação/metabolismo , Óxido Nítrico/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Doença , Camundongos , Óxido Nítrico/farmacologia , Oxirredução , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética
11.
Brain Res ; 1508: 44-52, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23438514

RESUMO

Krabbe disease is a lethal, demyelinating condition caused by genetic deficiency of galactocerebrosidase (GALC) and resultant accumulation of its cytotoxic substrate, psychosine (galactosylsphingosine), primarily in oligodendrocytes (OLs). Psychosine is generated by galactosylation of sphingosine by UDP-galactose:ceramide galactosyltransferase (CGT), a galactosylceramide synthesizing enzyme which is primarily expressed in OLs. The expression of CGT and the synthesis of galactosyl-sphingolipids are associated with the terminal differentiation of OL, but little is known about the participation of endogenous psychosine accumulation in OL differentiation under GALC deficient conditions. In this study, we report that accumulation of endogenous psychosine under GALC deficient Krabbe conditions impedes OL differentiation process both by decreasing the expression of myelin lipids and protein and by inducing the cell death of maturating OLs. The psychosine pathology under GALC deficient conditions involves participation of secretory phospholipase A2 (sPLA2) activation and increase in its metabolites, as evidenced by attenuation of psychosine-induced pathology by treatment with pharmacological inhibitor of sPLA2 7,7-dimethyleicosadienoic acid (DEDA). These observations suggest for potential therapeutic efficacy of sPLA2 inhibitor in Krabbe disease.


Assuntos
Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/patologia , Oligodendroglia/efeitos dos fármacos , Psicosina/metabolismo , Animais , Western Blotting , Imunofluorescência , Galactosilceramidase/metabolismo , Galactosilceramidas/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Metabolismo dos Lipídeos/fisiologia , Microscopia Eletrônica , Microscopia de Fluorescência , Bainha de Mielina/metabolismo , Interferência de RNA , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
12.
J Alzheimers Dis ; 34(3): 621-35, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23254638

RESUMO

Chronic cerebral hypoperfusion (CCH), featuring in most of the Alzheimer's disease spectrum, plays a detrimental role in brain amyloid-ß (Aß) homeostasis, cerebrovascular morbidity, and cognitive decline; therefore, early management of cerebrovascular pathology is considered to be important for intervention in the impending cognitive decline. S-nitrosoglutathione (GSNO) is an endogenous nitric oxide carrier modulating endothelial function, inflammation, and neurotransmission. Therefore, the effect of GSNO treatment on CCH-associated neurocognitive pathologies was determined in vivo by using rats with permanent bilateral common carotid artery occlusion (BCCAO), a rat model of chronic cerebral hypoperfusion. We observed that rats subjected to permanent BCCAO showed a significant decrease in learning/memory performance and increases in brain levels of Aß and vascular inflammatory markers. GSNO treatment (50 µg/kg/day for 2 months) significantly improved learning and memory performance of BCCAO rats and reduced the Aß levels and ICAM-1/VCAM-1 expression in the brain. Further, in in vitro cell culture studies, GSNO treatment also decreased the cytokine-induced proinflammatory responses, such as activations of NFκB and STAT3 and expression of ICAM-1 and VCAM-1 in endothelial cells. In addition, GSNO treatment increased the endothelial and microglial Aß uptake. Additionally, GSNO treatment inhibited the ß-secretase activity in primary rat neuron cell culture, thus reducing secretion of Aß, suggesting GSNO mediated mechanisms in anti-inflammatory and anti-amyloidogenic activities. Taken together, these data document that systemic GSNO treatment is beneficial for improvement of cognitive decline under the conditions of chronic cerebral hypoperfusion and suggests a potential therapeutic use of GSNO for cerebral hypoperfusion associated mild cognitive impairment in Alzheimer's disease.


Assuntos
Transtornos Cognitivos/prevenção & controle , Modelos Animais de Doenças , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/uso terapêutico , S-Nitrosoglutationa/administração & dosagem , S-Nitrosoglutationa/uso terapêutico , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Linhagem Celular , Células Cultivadas , Doença Crônica , Transtornos Cognitivos/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
13.
Biochem Biophys Res Commun ; 399(4): 487-91, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20659426

RESUMO

AMP-activated-protein-kinase (AMPK) is a key sensor and regulator of cellular and whole-body energy metabolism and plays a key role in regulation of lipid metabolism. Since lipid metabolism has been implicated in neuronal amyloid-beta (Abeta) homeostasis and onset of Alzheimer's disease, we investigated the involvement of AMPK in neuronal lipid metabolism and Abeta production. We observed in cultured rat cortical neurons that Abeta production was significantly reduced when the neurons were stimulated with AMPK activator, 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR), but increased when AMPKalpha2 was knocked out, thus indicating the role of AMPK in amyloidogenesis. Although the detailed mechanisms by which AMPK regulates Abeta generation is not well understood, AMPK-mediated alterations in cholesterol and sphingomyelin homeostasis and in turn the altered distribution of Abeta precursor-protein (APP) in cholesterol and sphingomyelin rich membrane lipid rafts participate in Abeta generation. Taken together, this is the first report on the role of AMPK in regulation of neuronal amyloidogenesis.


Assuntos
Proteínas Quinases Ativadas por AMP/biossíntese , Peptídeos beta-Amiloides/antagonistas & inibidores , Microdomínios da Membrana/metabolismo , Neurônios/metabolismo , Esfingomielinas/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Colesterol/metabolismo , Ativação Enzimática , Técnicas de Inativação de Genes , Neurônios/enzimologia , Ratos , Ratos Sprague-Dawley
14.
Neurosci Lett ; 440(3): 260-4, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18583044

RESUMO

Statins are inhibitors of HMG-CoA reductase that have been recently recognized as anti-inflammatory and neuroprotective drugs. Herein, we investigated anti-excitotoxic and anti-seizure effects of statins by using kainic acid (KA)-rat seizure model, an animal model for temporal lobe epilepsy and excitotoxic neurodegeneration. We observed that pre-treatment with Lipitor (atorvastatin) efficiently reduced KA-induced seizure activities, hippocampal neuron death, monocyte infiltration and proinflammatory gene expression. In addition, we also observed that lovastatin treatment attenuated KA- or glutamate-induced excitotoxicity of cultured hippocampal neurons. These observations suggest a potential for use of statin treatment in modulation of seizures and other neurological diseases associated with excitotoxicity.


Assuntos
Ácidos Heptanoicos/uso terapêutico , Hipocampo/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Inflamação/tratamento farmacológico , Pirróis/uso terapêutico , Convulsões/tratamento farmacológico , Animais , Atorvastatina , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ectodisplasinas/metabolismo , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas/métodos , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-1beta/metabolismo , Ácido Caínico , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/patologia , Fator de Necrose Tumoral alfa/metabolismo
15.
J Neurochem ; 105(4): 1536-49, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18266936

RESUMO

Previous studies have described that statins (inhibitors of cholesterol and isoprenoid biosynthesis) inhibit the output of amyloid-beta (Abeta) in the animal model and thus decrease risk of Alzheimer's disease. However, their action mechanism(s) in Abeta precursor protein (APP) processing and Abeta generation is not fully understood. In this study, we report that lovastatin treatment reduced Abeta output in cultured hippocampal neurons as a result of reduced APP levels and beta-secretase activities in low density Lubrol WX (non-ionic detergent) extractable lipid rafts (LDLR). Rather than altering cholesterol levels in lipid raft fractions and thus disrupting lipid raft structure, lovastatin decreased Abeta generation through down-regulating geranylgeranyl-pyrophosphate dependent endocytosis pathway. The inhibition of APP endocytosis by treatment with lovastatin and reduction of APP levels in LDLR fractions by treatment with phenylarsine oxide (a general endocytosis inhibitor) support the involvement of APP endocytosis in APP distribution in LDLR fractions and subsequent APP beta-cleavage. Moreover, lovastatin-mediated down-regulation of endocytosis regulators, such as early endosomal antigen 1, dynamin-1, and phosphatidylinositol 3-kinase activity, indicates that lovastatin modulates APP endocytosis possibly through its pleiotropic effects on endocytic regulators. Collectively, these data report that lovastatin mediates inhibition of LDLR distribution and beta-cleavage of APP in a geranylgeranyl-pyrophosphate and endocytosis-dependent manner.


Assuntos
Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Lovastatina/farmacologia , Microdomínios da Membrana/metabolismo , Polietilenoglicóis/farmacologia , Precursor de Proteína beta-Amiloide/análise , Animais , Células Cultivadas , Microdomínios da Membrana/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
16.
J Neurochem ; 103 Suppl 1: 180-91, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17986153

RESUMO

Inflammatory disease plays a critical role in the pathogenesis of many neurological disorders. Astrogliosis and induction of pro-inflammatory mediators such as chemokines, cytokines and inducible nitric oxide synthase (iNOS) are the 'hallmarks' of inflammatory disease. Increased activity of lactosylceramide (LacCer) synthase and increased synthesis of LacCer during glial proliferation, and induction of pro-inflammatory cytokines and iNOS suggests a role for LacCer in these cellular signaling pathways. Studies using complementary techniques of inhibitors and antisense reported that inhibition of LacCer synthesis inhibits glial proliferation, as well as the induction of pro-inflammatory mediators (cytokines and iNOS). This inhibition was bypassed by exogenous LacCer, but not by other related lipids (e.g. glucosylceramide, galactocerebroside, GD1, GM1), indicating a role for LacCer in inflammatory signaling pathways. Furthermore, inhibition of glial proliferation and induction of inflammatory mediators by antisense to Ras GTPase, PI3Kinase and inhibitors of mitogen-activated protein kinase indicate the participation of the phosphoinositide 3-kinase (PI3Kinas)/Ras/mitogen-activated protein kinase/nuclear factor-kappaB (NF-kappaB) signaling pathways in LacCer-mediated inflammatory events thus exposing additional targets for therapeutics for inflammatory disease conditions.


Assuntos
Inflamação/complicações , Lactosilceramidas/metabolismo , Doenças do Sistema Nervoso/etiologia , Sistemas do Segundo Mensageiro/fisiologia , Animais , Humanos , Inflamação/metabolismo , Modelos Biológicos , Doenças do Sistema Nervoso/metabolismo
17.
Free Radic Biol Med ; 40(11): 1875-88, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16716889

RESUMO

Sphingolipids including ceramide and its derivatives such as ceramide-1-phosphate, glycosyl-ceramide, and sphinogosine (-1-phosphate) are now recognized as novel intracellular signal mediators for regulation of inflammation, apoptosis, proliferation, and differentiation. One of the important and regulated steps in these events is the generation of these sphingolipids via hydrolysis of sphingomyelin through the action of sphingomyelinases (SMase). Several lines of evidence suggest that reactive oxygen species (ROS; O2-, H2O2, and OH-,) and reactive nitrogen species (RNS; NO, and ONOO-) and cellular redox potential, which is mainly regulated by cellular glutathione (GSH), are tightly linked to the regulation of SMase activation. On the other hand, sphingolipids are also known to play an important role in maintaining cellular redox homeostasis through regulation of NADPH oxidase, mitochondrial integrity, and antioxidant enzymes. Therefore, this paper reviews the relationship between cellular redox and sphingolipid metabolism and its biological significance.


Assuntos
Transdução de Sinais , Esfingolipídeos/metabolismo , Animais , Humanos , Oxirredução , Estresse Oxidativo
18.
Glia ; 51(1): 13-21, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15779087

RESUMO

The present study underlines the importance of phospholipase A2 (PLA2)- and lipoxygenase (LO)-mediated signaling processes in the regulation of inducible nitric oxide synthase (iNOS) gene expression. In glial cells, lipopolysaccharide (LPS) induced the activities of PLA2 (calcium-independent PLA2; iPLA2 and cytosolic PLA2; cPLA2) as well as gene expression of iNOS. The inhibition of cPLA2 by methyl arachidonyl fluorophosphates (MAFP) or antisense oligomer against cPLA2 and inhibition of iPLA2 by bromoenol lactone reduced the LPS-induced iNOS gene expression and NFkappaB activation. In addition, the inhibition of LO by nordihydroguaiaretic acid (NDGA; general LO inhibitor) or MK886 (5-LO inhibitor), but not baicalein (12-LO inhibitor), completely abrogated the LPS-induced iNOS expression. Because NDGA could abrogate the LPS-induced activation of NFkappaB, while MK886 had no effect on it, LO-mediated inhibition of iNOS gene induction by LPS may involve an NFkappaB-dependent or -independent (by 5-LO) pathway. In contrast to LO, however, the cyclooxygenase (COX) may not be involved in the regulation of LPS-mediated induction of iNOS gene because COX inhibition by indomethacin (general COX inhibitor), SC560 (COX-1 inhibitor), and NS398 (COX-2 inhibitor) affected neither the LPS-induced iNOS expression nor activation of NFkappaB. These results indicate a role for cPLA2 and iPLA2 in LPS-mediated iNOS gene induction in glial cells and the involvement of LO in these reactions.


Assuntos
Lipopolissacarídeos/farmacologia , Lipoxigenase/metabolismo , Neuroglia/enzimologia , Óxido Nítrico Sintase/biossíntese , Fosfolipases A/biossíntese , Animais , Araquidonato 5-Lipoxigenase/metabolismo , Ácido Araquidônico/metabolismo , Northern Blotting , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/genética , Ensaio de Desvio de Mobilidade Eletroforética , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Isoenzimas/genética , NF-kappa B/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Fosfolipases A/antagonistas & inibidores , Fosfolipases A/genética , Fosfolipases A2 , RNA/análise , RNA/biossíntese , Ratos , Ativação Transcricional , Transfecção
19.
Neuropharmacology ; 48(1): 151-60, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15617735

RESUMO

The present study demonstrates the anti-inflammatory effect of adenosine kinase inhibitor (ADKI) in glial cells. Treatment of glial cells with IC51, an ADKI, stimulated the extracellular adenosine release and reduced the LPS/IFNgamma-mediated production of NO, and induction of iNOS and TNF-alpha gene expression. The recovery of IC51-mediated inhibition of iNOS expression by adenosine transport inhibitor, S-(4-nitrobenzyl)-6-thioinosine (NBTI), and the inhibition of LPS/IFNgamma-induced iNOS gene expression by exogenous adenosine indicate a role for adenosine release in IC51-mediated iNOS expression. The rescue of IC51-mediated inhibition of iNOS expression by adenosine receptor antagonist for A2A, 8-(3-chlorostyryl)caffeine (CSC) and alloxazine for A2B, further supports a role for interaction of adenosine and its receptors in anti-inflammatory activity. The IC51-mediated induction of cAMP levels, downstream target of A2A and A2B, and inhibition of LPS/IFNgamma-induced expression of iNOS by forskolin, a cAMP activator, document a role for cAMP mediated pathway in anti-inflammatory activity of IC51. Taken together, these studies document that IC51-mediated inhibition of iNOS expression is through activation of adenosine receptors, which activates A2A and A2B resulting in increased cAMP levels following LPS/IFNgamma stimulation. Moreover, the lack of effect of IC51 or adenosine on NFkappaB DNA binding activity and its transactivity indicates that the inhibition of iNOS expression mediated by IC51 may be through an NFkappaB independent pathway.


Assuntos
Adenosina Quinase/antagonistas & inibidores , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Prolina/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Adenosina/metabolismo , Análise de Variância , Animais , Northern Blotting/métodos , Western Blotting/métodos , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Glioma , Interferon gama/farmacologia , L-Lactato Desidrogenase/metabolismo , Leupeptinas/metabolismo , Lipopolissacarídeos/farmacologia , Neuroglia/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Prolina/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Tiocarbamatos/farmacologia , Transfecção/métodos , Fator de Necrose Tumoral alfa/metabolismo , Quinase Induzida por NF-kappaB
20.
Free Radic Biol Med ; 37(11): 1834-44, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15528042

RESUMO

We reported previously that cAMP analogues or cAMP synthesis activator (forskolin; FSK) inhibit lipopolysaccharide (LPS)-induced inducible nitric-oxide systase (iNOS) gene expression in astrocytes, while they enhance that in macrophages. Here, we report that the FSK-mediated inhibition of iNOS expression in C6 glial cells is due to its reduced transcriptional activity, while the FSK-mediated enhancement of iNOS expression in RAW264.7 macrophages is a result of increased stability of iNOS protein without transcriptional enhancement. The LPS/interferon-gamma (IFN)-induced iNOS transcription was inhibited by FSK via inhibition of p38-MAPK/ATF-2 activity in glial cells while it was not affected in macrophages. In both cell types, proteasome activities were required for the spontaneous degradation of iNOS protein, and the inhibition of proteasome activity by MG132 after maximum increase of iNOS protein levels further enhanced iNOS protein induction by LPS/IFN, suggesting the involvement of proteasome in iNOS degradation. More importantly, the iNOS protein levels were equalized by the MG132 posttreatment in macrophages treated with LPS/IFN alone and along with FSK, and ubiquitinated iNOS protein levels were reduced by FSK posttreatment, suggesting that the FSK-mediated inhibition of ubiquitination of iNOS protein and the following increased stability of iNOS protein are one of the mechanisms of cAMP-pathway-mediated enhancement of iNOS gene expression in macrophages. To our knowledge, this is the first evidence that cAMP regulates iNOS expression at the posttranslational level in macrophages.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , AMP Cíclico/fisiologia , Macrófagos/enzimologia , Neuroglia/enzimologia , Óxido Nítrico Sintase/metabolismo , Fatores de Transcrição/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Fator 2 Ativador da Transcrição , Animais , Linhagem Celular Tumoral , Colforsina/farmacologia , Expressão Gênica/efeitos dos fármacos , Leupeptinas/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...