Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
J Crit Care ; 82: 154766, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38479298

RESUMO

BACKGROUND: Cytomegalovirus (CMV) infection is associated with poor outcome in ICU patients. However, data on immunocompromised patients are scarce. This study aims to describe characteristics and outcomes of critically ill hematological patients and CMV infection. CMV disease characteristics and relationship between CMV viral load, CMV disease, coinfections by other pathogens and outcomes are described. METHODS: Retrospective single center study (Jan 2010-Dec 2017). Adult patients, admitted to the ICU, having underlying hematological malignancy and CMV infection were included. Results are reported as median (interquartile) or n (%). Factors associated with hospital mortality or CMV disease were analysed using logistic regression. RESULTS: 178 patients were included (median age 55y [42-64], 69.1% male). Hospital mortality was 53% (n = 95). Median viral load was 2.7 Log [2.3-3.5]. CMV disease occurred in 44 (24.7%) patients. Coinfections concerned 159 patients (89.3%). After adjustment for confounders, need for vasopressors (OR 2.53; 95%CI 1.11-5.97) and viral load (OR 1.88 per Log; 95%CI 1.29-2.85) were associated with hospital mortality. However, neither CMV disease nor treatment were associated with outcomes. Allogeneic stem cell transplantation (OR 2.55; 95%CI 1.05-6.16), mechanical ventilation (OR 4.11; OR 1.77-10.54) and viral load (OR 1.77 per Log; 95%CI 1.23-2.61) were independently associated with CMV disease. Coinfections were not associated with CMV disease or hospital mortality. CONCLUSION: In critically-ill hematological patients, CMV viral load is independently associated with hospital mortality. Conversely, neither CMV disease nor treatment was associated with outcome suggesting viral load to be a surrogate for immune status rather than a cause of poor outcome.


Assuntos
Infecções por Citomegalovirus , Neoplasias Hematológicas , Mortalidade Hospitalar , Unidades de Terapia Intensiva , Carga Viral , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Infecções por Citomegalovirus/mortalidade , Infecções por Citomegalovirus/epidemiologia , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/mortalidade , Estudos Retrospectivos , Unidades de Terapia Intensiva/estatística & dados numéricos , Adulto , Estado Terminal , Hospedeiro Imunocomprometido , Coinfecção/epidemiologia , Citomegalovirus/isolamento & purificação
3.
Mol Neurobiol ; 55(8): 6834-6840, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29349578

RESUMO

Delivering peptide-based drugs to the brain is a major challenge because of the existence of the blood-brain barrier (BBB). To overcome this problem, cell-penetrating peptides derived from proteins that are able to cross biological membranes have been used as cell-permeable and brain-penetrant compounds. An example is the transactivator of transcription protein transduction domain (Tat) of the human immunodeficiency virus. The basic domain of Tat is formed of arginine and lysine amino acid residues. Tat has been used as brain-penetrant carrier also in therapies for Alzheimer disease (AD), the most common form of dementia characterized by extracellular cerebral deposits of amyloid made up of Aß peptide. The aim of our study was to assess whether Tat bind to amyloid deposits of AD and other amyloidoses. An in situ labeling using biotinylated Tat 48-57 peptide was employed in the brain tissue with amyloid deposits made up of Aß (patients with AD and transgenic AD mice), of prion protein (patients with Gerstmann-Straussler-Scheinker disease), and other amyloidosis, processed by different fixations and pretreatments of histological sections. Our results showed that Tat peptide binds amyloid deposits made up of Aß, PrP, and immunoglobulin lambda chains in the brain and other tissues processed by alcoholic fixatives but not in formalin-fixed tissue. The fact that biotinylated Tat peptide stains amyloid of different biochemical composition and the specific charge characteristics of the molecules suggests that Tat may bind to heparan sulfate glicosaminoglicans, that are present in amyloid deposits. Inhibition of the binding by Tat pre-incubation with protamine reinforces this hypothesis. Binding of Tat to amyloid deposits should be kept in mind in interpreting the results of studies employing this molecule as brain-penetrating compound for the treatment of cerebral amyloidoses. Our results also suggest that Tat may be helpful for the analysis of the mechanisms of amyloidogenesis, and in particular, the interactions between specific amyloid peptides and glicosaminoglicans.


Assuntos
Amiloide/metabolismo , Encéfalo/metabolismo , Peptídeos/metabolismo , Coloração e Rotulagem/métodos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Amiloidose/patologia , Animais , Cartilagem/patologia , Núcleo Celular/metabolismo , Condroma/patologia , Endodesoxirribonucleases/metabolismo , Endorribonucleases/metabolismo , Formaldeído , Camundongos Transgênicos , Protaminas/metabolismo
4.
J Infect ; 75(2): 155-159, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28551368

RESUMO

OBJECTIVES: We report the biological and clinical impacts possibly associated with HHV-6 reactivation in autologous hematopoietic stem cell transplant (AHSCT) recipients after intensive chemotherapy regimen for lymphoma. METHODS: We retrospectively reviewed clinical, biological, radiological, treatment and outcomes of patients with positive HHV-6 DNA in whole blood following autologous hematopoietic stem cell transplantation. RESULTS: Blood HHV-6 reactivation was reported in 27 (8.5%) patients among 316 AHSCT recipients after high dose therapy for lymphoma. Thirteen (4.1%) patients were symptomatic with fever (100%), diarrhea (61.5%), skin rash (46.1%), and pneumonia (23.1%). Antiviral treatment was administered in 9 (69%) patients and outcome was favorable in all cases. CONCLUSION: Our study suggests a possible pathogenic role of HHV-6 in AHSCT recipients and suggests an impact of antiviral treatments on viral replication and clinical signs resolution.


Assuntos
Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Herpesvirus Humano 6 , Infecções por Roseolovirus , Transplantados/estatística & dados numéricos , Adulto , Idoso , Feminino , Febre/etiologia , Humanos , Hospedeiro Imunocomprometido , Linfoma/cirurgia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Infecções por Roseolovirus/complicações , Infecções por Roseolovirus/virologia , Ativação Viral
5.
PLoS One ; 11(3): e0152465, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27023064

RESUMO

The activation of Nrf2 has been demonstrated to play a crucial role in cancer cell resistance to different anticancer therapies. The inhibition of proteasome activity has been proposed as a chemosensitizing therapy but the activation of Nrf2 could reduce its efficacy. Using the highly chemoresistant neuroblastoma cells HTLA-230, here we show that the strong reduction in proteasome activity, obtained by using low concentration of bortezomib (BTZ, 2.5 nM), fails in reducing cell viability. BTZ treatment favours the binding of Nrf2 to the ARE sequences in the promoter regions of target genes such as heme oxygenase 1 (HO-1), the modulatory subunit of γ-glutamylcysteine ligase (GCLM) and the transporter for cysteine (x-CT), enabling their transcription. GSH level is also increased after BTZ treatment. The up-regulation of Nrf2 target genes is responsible for cell resistance since HO-1 silencing and GSH depletion synergistically decrease BTZ-treated cell viability. Moreover, cell exposure to all-trans-Retinoic acid (ATRA, 3 µM) reduces the binding of Nrf2 to the ARE sequences, decreases HO-1 induction and lowers GSH level increasing the efficacy of bortezomib. These data suggest the role of Nrf2, HO-1 and GSH as molecular targets to improve the efficacy of low doses of bortezomib in the treatment of malignant neuroblastoma.


Assuntos
Bortezomib/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glutationa/metabolismo , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neuroblastoma/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Elementos de Resposta Antioxidante/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/metabolismo , Heme Oxigenase-1/genética , Humanos , Neuroblastoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Tretinoína/farmacologia , Regulação para Cima/efeitos dos fármacos
6.
Nanotechnology ; 25(33): 335706, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25074670

RESUMO

One of the major drawbacks that limits the clinical application of nanoparticles is the lack of preliminary investigations related to their biocompatibility, biodegradability and biodistribution. In this work, biodegradable PEGylated polymer nanoparticles (NPs) have been synthesized by using macromonomers based on poly(ε-caprolaconte) oligomers. More in detail, NPs have been produced by adopting a surfactant-free semibatch emulsion polymerization process using PEG chains as a stabilizing agent. The NPs were also labeled with rhodamine B covalently bound to the NPs to quantitatively study their biodistribution in vivo. NPs were investigated in both in vitro and in vivo preclinical systems to study their biodistribution in mice bearing B16/F10 melanoma, as well as their biocompatibility and biodegradability. The NP concentration was evaluated in different tissues at several times after intravenous injection. The disappearance of the NPs from the plasma was biphasic, with distribution and elimination half-lives of 30 min and 15 h, respectively. NPs were retained in tumors and in filter organs for a long time, were still detectable after 7 d and maintained a steady concentration in the tumor for 120 h. 48 h after injection, 70 ± 15% of the inoculated NPs were excreted in the feces. The favorable tumor uptake, fast excretion and absence of cytotoxicity foster the further development of produced NPs as drug delivery carriers.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas/química , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Polímeros , Rodaminas/química , Rodaminas/farmacocinética , Distribuição Tecidual
7.
Cell Death Dis ; 5: e1019, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24457963

RESUMO

Altered synaptic function is considered one of the first features of Alzheimer disease (AD). Currently, no treatment is available to prevent the dysfunction of excitatory synapses in AD. Identification of the key modulators of synaptopathy is of particular significance in the treatment of AD. We here characterized the pathways leading to synaptopathy in TgCRND8 mice and showed that c-Jun N-terminal kinase (JNK) is activated at the spine prior to the onset of cognitive impairment. The specific inhibition of JNK, with its specific inhibiting peptide D-JNKI1, prevented synaptic dysfunction in TgCRND8 mice. D-JNKI1 avoided both the loss of postsynaptic proteins and glutamate receptors from the postsynaptic density and the reduction in size of excitatory synapses, reverting their dysfunction. This set of data reveals that JNK is a key signaling pathway in AD synaptic injury and that its specific inhibition offers an innovative therapeutic strategy to prevent spine degeneration in AD.


Assuntos
Doença de Alzheimer/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sinapses/enzimologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Masculino , Camundongos , Camundongos Transgênicos , Peptídeos/administração & dosagem , Transdução de Sinais
8.
Neuroscience ; 159(1): 94-103, 2009 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-19135136

RESUMO

The c-Jun N-terminal kinase (JNK) is a mitogen-activated protein kinase (MAPK) activated by stress-signals and involved in many different diseases. Previous results proved the powerful effect of the cell permeable peptide inhibitor d-JNKI1 (d-retro-inverso form of c-Jun N-terminal kinase-inhibitor) against neuronal death in CNS diseases, but the precise features of this neuroprotection remain unclear. We here performed cell-free and in vitro experiments for a deeper characterization of d-JNKI1 features in physiological conditions. This peptide works by preventing JNK interaction with its c-Jun N-terminal kinase-binding domain (JBD) dependent targets. We here focused on the two JNK upstream MAPKKs, mitogen-activated protein kinase kinase 4 (MKK4) and mitogen-activated protein kinase kinase 7 (MKK7), because they contain a JBD homology domain. We proved that d-JNKI1 prevents MKK4 and MKK7 activity in cell-free and in vitro experiments: these MAPKK could be considered not only activators but also substrates of JNK. This means that d-JNKI1 can interrupt downstream but also upstream events along the JNK cascade, highlighting a new remarkable feature of this peptide. We also showed the lack of any direct effect of the peptide on p38, MEK1, and extracellular signal-regulated kinase (ERK) in cell free, while in rat primary cortical neurons JNK inhibition activates the MEK1-ERK-Ets1/c-Fos cascade. JNK inhibition induces a compensatory effect and leads to ERK activation via MEK1, resulting in an activation of the survival pathway-(MEK1/ERK) as a consequence of the death pathway-(JNK) inhibition. This study should hold as an important step to clarify the strong neuroprotective effect of d-JNKI1.


Assuntos
Córtex Cerebral/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase 7/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Fator 2 Ativador da Transcrição/metabolismo , Sequência de Aminoácidos , Análise de Variância , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular , L-Lactato Desidrogenase/metabolismo , Peptídeos/farmacologia , Fosforilação , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas Elk-1 do Domínio ets/metabolismo
9.
J Virol ; 77(15): 8462-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12857915

RESUMO

Based on in vitro observations in scrapie-infected neuroblastoma cells, quinacrine has recently been proposed as a treatment for Creutzfeldt-Jakob disease (CJD), including a new variant CJD which is linked to contamination of food by the bovine spongiform encephalopathy (BSE) agent. The present study investigated possible mechanisms of action of quinacrine on prions. The ability of quinacrine to interact with and to reduce the protease resistance of PrP peptide aggregates and PrPres of human and animal origin were analyzed, together with its ability to inhibit the in vitro conversion of the normal prion protein (PrPc) to the abnormal form (PrPres). Furthermore, the efficiencies of quinacrine and chlorpromazine, another tricyclic compound, were examined in different in vitro models and in an experimental murine model of BSE. Quinacrine efficiently hampered de novo generation of fibrillogenic prion protein and PrPres accumulation in ScN2a cells. However, it was unable to affect the protease resistance of preexisting PrP fibrils and PrPres from brain homogenates, and a "curing" effect was obtained in ScGT1 cells only after lengthy treatment. In vivo, no detectable effect was observed in the animal model used, consistent with other recent studies and preliminary observations in humans. Despite its ability to cross the blood-brain barrier, the use of quinacrine for the treatment of CJD is questionable, at least as a monotherapy. The multistep experimental approach employed here could be used to test new therapeutic regimes before their use in human trials.


Assuntos
Doenças Priônicas/tratamento farmacológico , Príons/efeitos dos fármacos , Quinacrina/uso terapêutico , Animais , Clorpromazina/farmacologia , Clorpromazina/uso terapêutico , Cricetinae , Resistência a Medicamentos , Endopeptidase K/farmacologia , Humanos , Melatonina/farmacologia , Melatonina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/síntese química , Peptídeos/metabolismo , Proteínas PrPC/efeitos dos fármacos , Proteínas PrPC/metabolismo , Proteínas PrPSc/efeitos dos fármacos , Proteínas PrPSc/metabolismo , Príons/química , Quinacrina/farmacologia , Células Tumorais Cultivadas
10.
J Biol Chem ; 276(49): 46347-63, 2001 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-11562361

RESUMO

We report the cloning of the AOH1 and AOH2 genes, which encode two novel mammalian molybdo-flavoproteins. We have purified the AOH1 protein to homogeneity in its catalytically active form from mouse liver. Twenty tryptic peptides, identified or directly sequenced by mass spectrometry, confirm the primary structure of the polypeptide deduced from the AOH1 gene. The enzyme contains one molecule of FAD, one atom of molybdenum, and four atoms of iron per subunit and shows spectroscopic features similar to those of the prototypic molybdo-flavoprotein xanthine oxidoreductase. The AOH1 and AOH2 genes are 98 and 60 kilobases long, respectively, and consist of 35 coding exons. The AOH1 gene has the potential to transcribe an extra leader non-coding exon, which is located downstream of exon 26, and is transcribed in the opposite orientation relative to all the other exons. AOH1 and AOH2 map to chromosome 1 in close proximity to each other and to the aldehyde oxidase gene, forming a molybdo-flavoenzyme gene cluster. Conservation in the position of exon/intron junctions among the mouse AOH1, AOH2, aldehyde oxidase, and xanthine oxidoreductase loci indicates that these genes are derived from the duplication of an ancestral precursor.


Assuntos
Aldeído Oxirredutases/isolamento & purificação , Mapeamento Cromossômico , Flavoproteínas/genética , Família Multigênica , Aldeído Oxirredutases/química , Aldeído Oxirredutases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Cromatografia por Troca Iônica , Clonagem Molecular , DNA Complementar , Eletroforese em Gel de Poliacrilamida , Fígado/enzimologia , Camundongos , Dados de Sequência Molecular , Mapeamento de Peptídeos , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
11.
Arterioscler Thromb Vasc Biol ; 21(8): 1327-32, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11498461

RESUMO

This study set out to clarify whether the inhibition of sterol or nonsterol derivatives arising from mevalonate biotransformation plays a major role in the in vivo anti-inflammatory action of statins. Hepatic synthesis of all these derivatives was inhibited in mice by administered statins, whereas squalestatin inhibited only sterol derivatives. Using a short-term treatment schedule, we found that statins reduced the hepatic activity of 3-hydroxy-3-methylglutaryl coenzyme A reductase without affecting blood cholesterol. This treatment inhibited lipopolysaccharide- and carrageenan-induced pouch leukocyte recruitment and the exudate production of interleukin-6, monocyte chemotactic protein-1, and RANTES. Coadministration of mevalonate reversed the effect of statin on leukocyte recruitment. The inhibition of sterol synthesis by squalestatin did not have any anti-inflammatory effect, indicating that the biosynthesis of nonsterol compounds arising from mevalonate is crucial for the in vivo regulation of cytokine and chemokine production by statins. Their inhibition by statins may account for the reported anti-inflammatory effects of these drugs and may provide a biochemical basis for the recently reported effects of statins in the prevention of cardiovascular disease and mortality.


Assuntos
Anti-Inflamatórios/farmacologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipolipemiantes/farmacologia , Ácido Mevalônico/metabolismo , Animais , Permeabilidade Capilar/efeitos dos fármacos , Carragenina/farmacologia , Inibição de Migração Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Hidroximetilglutaril-CoA Redutases/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Modelos Animais
12.
J Biol Chem ; 276(28): 25692-6, 2001 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-11316802

RESUMO

A synthetic peptide consisting of amino acid residues 106-126 of the human prion protein (PrP-(106--126)) has been previously demonstrated to be neurotoxic and to induce microglial activation. The present study investigated the expression of the inducible form of the nitric-oxide synthase (NOS-II) in human microglial cells treated with PrP-(106--126). Using reverse transcriptase-polymerase chain reaction, we found that PrP-(106--126) induces NOS-II gene expression after 24 h of treatment and that this effect is accompanied by a peak of nuclear factor kappa B (NF-kappa B) binding at 30 min as evaluated by electrophoretic mobility shift assay. Since our previous data demonstrated tumor necrosis factor-alpha (TNF-alpha) to be a potent inducer of NOS-II in these cells, we analyzed the expression of this cytokine in PrP-(106--126)-treated microglia. PrP-(106--126) caused the release of TNF-alpha as detected by enzyme-linked immunosorbent assay, and a blocking antibody, anti-TNF-alpha, abolished NOS-II induction elicited by this peptide. Moreover, PrP-(106-126) activates p38 mitogen-activated protein kinase, and the inhibition of this pathway determines the ablation of NF-kappa B binding induced by this fragment peptide.


Assuntos
Microglia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico Sintase/biossíntese , Fragmentos de Peptídeos/farmacologia , Príons/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Quinases p38 Ativadas por Mitógeno
13.
J Biol Chem ; 276(8): 6009-15, 2001 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-11087738

RESUMO

Gerstmann-Sträussler-Scheinker disease (GSS) is a cerebral amyloidosis associated with mutations in the prion protein (PrP) gene (PRNP). The aim of this study was to characterize amyloid peptides purified from brain tissue of a patient with the A117V mutation who was Met/Val heterozygous at codon 129, Val(129) being in coupling phase with mutant Val117. The major peptide extracted from amyloid fibrils was a approximately 7-kDa PrP fragment. Sequence analysis and mass spectrometry showed that this fragment had ragged N and C termini, starting mainly at Gly88 and Gly90 and ending with Arg148, Glu152, or Asn153. Only Val was present at positions 117 and 129, indicating that the amyloid protein originated from mutant PrP molecules. In addition to the approximately 7-kDa peptides, the amyloid fraction contained N- and C-terminal PrP fragments corresponding to residues 23-41, 191-205, and 217-228. Fibrillogenesis in vitro with synthetic peptides corresponding to PrP fragments extracted from brain tissue showed that peptide PrP-(85-148) readily assembled into amyloid fibrils. Peptide PrP-(191-205) also formed fibrillary structures although with different morphology, whereas peptides PrP-(23-41) and PrP-(217-228) did not. These findings suggest that the processing of mutant PrP isoforms associated with Gerstmann-Sträussler-Scheinker disease may occur extracellularly. It is conceivable that full-length PrP and/or large PrP peptides are deposited in the extracellular compartment, partially degraded by proteases and further digested by tissue endopeptidases, originating a approximately 7-kDa protease-resistant core that is similar in patients with different mutations. Furthermore, the present data suggest that C-terminal fragments of PrP may participate in amyloid formation.


Assuntos
Amiloide/genética , Doença de Gerstmann-Straussler-Scheinker/etiologia , Fragmentos de Peptídeos/isolamento & purificação , Príons/patogenicidade , Precursores de Proteínas/genética , Adulto , Alelos , Córtex Cerebral/patologia , Doença de Gerstmann-Straussler-Scheinker/genética , Heterozigoto , Humanos , Masculino , Metionina/genética , Proteínas Priônicas , Príons/isolamento & purificação , Análise de Sequência de Proteína , Síndrome , Valina/genética
14.
Eur Cytokine Netw ; 12(4): 625-30, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11781189

RESUMO

Endotoxin (LPS), by raising the levels of cytokines, markedly influences lipid metabolism. To clarify the molecular mechanism of this effect, we examined the action of endotoxin in vitro and in vivo on the regulation of sterol regulatory element binding protein-1 (SREBP-1). In HepG2 cells stimulated with LPS, a dose-dependent increase in the level of the mature form of SREBP-1 was observed. For in vivo studies, endotoxin was administered intraperitoneally to CD1 mice fed with a standard or a cholesterol-enriched diet to increase the basal levels of circulating and liver cholesterol. Endotoxin raised cholesterol levels and stimulated the maturation of hepatic SREBP-1 in both normal and cholesterol-fed mice, indicating that the lipogenic effect of LPS was independent of endogenous sterol levels. To assess whether the lipogenic effect of endotoxin was linked to cytokine production, we administered LPS to C57Bl/6J endotoxin-sensitive and to C3H/HeJ endotoxin-resistant mice, which do not produce tumor necrosis factor in response to LPS. Significant induction of cholesterol levels and SREBP-1 activation was observed only in C57Bl/6J mice, indicating that cytokine production is crucial for the regulation of SREBP-1, and that the transcriptional activation of cholesterol biosynthesis may be part of the acute-phase response.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Citocinas/biossíntese , Proteínas de Ligação a DNA/metabolismo , Lipopolissacarídeos/farmacologia , Fatores de Transcrição , Animais , Western Blotting , Linhagem Celular , Colesterol/biossíntese , Colesterol/sangue , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteína de Ligação a Elemento Regulador de Esterol 1 , Ativação Transcricional
15.
J Biol Chem ; 275(39): 30690-700, 2000 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-10893244

RESUMO

The cDNAs coding for two novel mouse molybdo-flavoproteins, AOH1 and AOH2 (aldehyde oxidase homolog 1 and 2), were isolated. The AOH1 and AOH2 cDNAs code for polypeptides of 1336 amino acids. The two proteins have similar primary structure and show striking amino acid identity with aldehyde oxidase and xanthine oxidoreductase, two other molybdo-flavoenzymes. AOH1 and AOH2 contain consensus sequences for a molybdopterin-binding site and two distinct 2Fe-2S redox centers. In its native conformation, AOH1 has a molecular weight consistent with a homotetrameric structure. Transfection of the AOH1 and AOH2 cDNAs results in the production of proteins with phenanthridine but not hypoxanthine oxidizing activity. Furthermore, the AOH1 protein has benzaldehyde oxidizing activity with electrophoretic characteristics identical to those of a previously identified aldehyde oxidase isoenzyme (Holmes, R. S. (1979) Biochem. Genet. 17, 517-528). The AOH1 transcript is expressed in the hepatocytes of the adult and fetal liver and in spermatogonia. In liver, the AOH1 protein is synthesized in a gender-specific fashion. The expression of AOH2 is limited to keratinized epithelia and the basal layer of the epidermis and hair folliculi. The selective cell and tissue distribution of AOH1 and AOH2 mRNAs is consistent with the localization of the respective protein products.


Assuntos
Coenzimas , Molibdênio , Oxirredutases/genética , Aldeído Oxidase , Aldeído Oxirredutases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Clonagem Molecular , Sequência Consenso , DNA Complementar , Desulfovibrio/enzimologia , Evolução Molecular , Feminino , Humanos , Masculino , Metaloproteínas/metabolismo , Camundongos , Proteínas Mitocondriais , Dados de Sequência Molecular , Cofatores de Molibdênio , Oxirredutases/metabolismo , Filogenia , Proteínas de Plantas/genética , Pteridinas/metabolismo , Proteínas Recombinantes/biossíntese , Homologia de Sequência de Aminoácidos , Caracteres Sexuais , Distribuição Tecidual , Xantina Oxidase/genética
16.
J Biol Chem ; 274(25): 17705-10, 1999 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-10364211

RESUMO

Brefeldin A (BFA) is a fungal metabolite that disassembles the Golgi apparatus into tubular networks and causes the dissociation of coatomer proteins from Golgi membranes. We have previously shown that an additional effect of BFA is to stimulate the ADP-ribosylation of two cytosolic proteins of 38 and 50 kDa (brefeldin A-ADP-riboslyated substrate (BARS)) and that this effect greatly facilitates the Golgi-disassembling activity of the toxin. In this study, BARS has been purified from rat brain cytosol and microsequenced, and the BARS cDNA has been cloned. BARS shares high homology with two known proteins, C-terminal-binding protein 1 (CtBP1) and CtBP2. It is therefore a third member of the CtBP family. The role of BARS in Golgi disassembly by BFA was verified in permeabilized cells. In the presence of dialyzed cytosol that had been previously depleted of BARS or treated with an anti-BARS antibody, BFA potently disassembled the Golgi. However, in cytosol complemented with purified BARS, or even in control cytosols containing physiological levels of BARS, the action of BFA on Golgi disassembly was strongly inhibited. These results suggest that BARS exerts a negative control on Golgi tubulation, with important consequences for the structure and function of the Golgi complex.


Assuntos
Brefeldina A/farmacologia , Proteínas de Transporte/genética , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/efeitos dos fármacos , Fatores de Transcrição , Fatores de Ribosilação do ADP , Oxirredutases do Álcool , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Células COS , Proteínas de Transporte/química , Clonagem Molecular , Proteínas de Ligação a DNA/química , Pulmão/metabolismo , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Fosfoproteínas/química , Ratos , Ratos Sprague-Dawley , Alinhamento de Sequência , Baço/metabolismo , Transfecção , Células Tumorais Cultivadas
17.
Neuroreport ; 10(4): 723-9, 1999 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-10208538

RESUMO

The scrapie isoform of the prion protein (PrPres) induces neurodegeneration and gliosis in the central nervous system. These features may be reproduced in vitro on exposure of neuronal and glial cultures to PrPres and the peptide HuPr P106-126. In the present study, we investigated the role of microglial cells and astrocytes in the pathological process by studying their molecular response to PrP 106-126 exposure. PrP 106-126 elicited a specific overproduction of pro-inflammatory cytokines IL1beta and IL6 in microglial cells (but not increased expression of TNFalpha, IL10, and TGFbeta1) and over-expression of GFAP in astrocytes. These effects were strictly dependent on the ability of the peptide to form amyloid fibrils. These data strongly suggest that microglial cells contribute to prion-related neurodegenerative processes by producing proinflammatory cytokines in the brain areas of amyloid PrP deposition.


Assuntos
Citocinas/biossíntese , Microglia/metabolismo , Fragmentos de Peptídeos/farmacologia , Príons/farmacologia , Animais , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Humanos , Inflamação/metabolismo , Interleucina-1/biossíntese , Interleucina-6/biossíntese , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Degeneração Neural/patologia , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/biossíntese
18.
Eur J Immunol ; 29(3): 781-8, 1999 03.
Artigo em Inglês | MEDLINE | ID: mdl-10092080

RESUMO

Three molecular forms of the IL-1 receptor antagonist (IL-1ra) have been identified and cloned. Secreted IL-1ra (sIL-1ra or IL-1ra1) contains a classical leader peptide giving a released mature protein. Two intracellular isoforms, icIL-1ra type I (IL-1ra2) and icIL-1ra type II (IL-1ra3), have no leader sequence, thus predicting that these proteins remain intracellular. In an effort to define its biological role, we structurally and functionally characterized IL-1ra3. Endogenous immunoreactive IL-1ra3 was detected in a variety of inflammatory cells and tissues. We used a gene transfer strategy to explore the possible intracellular functions of IL-1ra3 (and IL-1ra2) and the cell-associated agonist IL-1alpha. The intracellular IL-1ra3 isoform, as well as IL-1ra2, does not block the action of exogenous and endogenous IL-1 under these conditions. Intact IL-1ra3 was released from the cells killed by NK effectors. The intracellular isoforms may represent a reservoir of IL-1ra, released upon cell death, whose function is to limit the pro-inflammatory action of cell debris.


Assuntos
Sialoglicoproteínas/metabolismo , Animais , Éxons , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/metabolismo , Interleucina-1/farmacologia , Camundongos , Isoformas de Proteínas , Sialoglicoproteínas/genética , Células Tumorais Cultivadas
19.
J Neurosci Res ; 54(3): 341-52, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9819139

RESUMO

The prion diseases are transmissible neurodegenerative pathologies characterized by the accumulation of altered forms of the prion protein (PrP), termed PrP(Sc), in the brain. Previous studies have shown that a synthetic peptide homologous to residues 106-126 of PrP (PrP 106-126) maintains many characteristics of PrP(Sc), i.e., the ability to form amyloid fibrils and to induce apoptosis in neurons. We have investigated the intracellular mechanisms involved in the cellular degeneration induced by PrP 106-126, using the GH3 cells as a model of excitable cells. When assayed in serum-deprived conditions (48 hr), PrP 106-126 (50 microM) induced cell death time-dependently, and this process showed the characteristics of the apoptosis. This effect was specific because a peptide with a scrambled sequence of PrP 106-126 was not effective. Then we performed microfluorimetric analysis of single cells to monitor intracellular calcium concentrations and showed that PrP 106-126 caused a complete blockade of the increase in the cytosolic calcium levels induced by K+ (40 mM) depolarization. Conversely, the scrambled peptide was ineffective. The L-type voltage-sensitive calcium channel blocker nicardipine (1 microM) also induced apoptosis in GH3 cells, suggesting that the blockade of Ca2+ entry through this class of calcium channels may cause GH3 apoptotic cell death. We thus analyzed, by means of electrophysiological studies, whether Prp 106-126 modulate L-type calcium channels activity and demonstrated that the apoptotic effect of PrP 106-126 was due to a dose-dependent inactivation of the L-type calcium channels. These data demonstrate that the prion protein fragment 106-126 induces a GH3 apoptotic cell death inducing a selective inhibition of the activity of the L-type voltage-sensitive calcium channels.


Assuntos
Apoptose , Canais de Cálcio/metabolismo , Fragmentos de Peptídeos/farmacologia , Príons/farmacologia , Sequência de Aminoácidos , Animais , Cádmio/farmacologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Formazans , Marcação In Situ das Extremidades Cortadas , Potenciais da Membrana , Dados de Sequência Molecular , Nicardipino/farmacologia , Hipófise , Potássio/farmacologia , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio , Células Tumorais Cultivadas
20.
Cancer Res ; 58(16): 3654-9, 1998 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9721875

RESUMO

c-Myc is a nuclear protein with important roles in cell transformation, cell proliferation, and gene transcription. It has been previously shown that a 14-amino acid (aa) modified peptide (H1-S6A,F8A) derived from the helix 1 (H1) carboxylic region of c-Myc can interfere in vitro with specific c-Myc DNA binding. Here, we have linked the above Myc-derived 14-aa peptide to a 16-aa sequence from the third helix of Antennapedia (Int). It has been repeatedly reported that this 16-aa Antennapedia peptide is able to cross mammalian cell membranes and to work as a vector for short peptides. Using fluorescent (dansylated or rhodaminated) peptides, we have shown that the fusion peptide with the Antennapedia fragment (Int-H1-S6A,F8A) but not the c-Myc derived fragment alone (H1-S6A,F8A) was capable of internalization inside MCF-7 human breast cancer cells. Int-H1-S6A,F8A and H1-S6A,F8A were the only two peptides capable of inhibiting coimmunoprecipitation of the c-Myc/Max heterodimer in vitro. We have treated (continuously for 10-11 days) MCF-7 cells with four different peptides: Int, H1-S6A,F8A, Int-H1-S6A,F8A, and Int-H1wt [a peptide differing from Int-H1-S6A,F8A by 2 aa (S6 and F8) in the H1 region]. In intact MCF-7 cells, Int-H1-S6A,F8A was the only active peptide capable of inducing the following biological effects: (a) inhibition of cloning efficiency on plates; (b) inhibition of cell growth and induction of apoptosis in subconfluent/confluent cells; and (c) inhibition of transcription of two c-Myc-regulated genes (ODC and p53). Int-H1-S6A,F8A was active in the 1-10 microM range. Int-H1-S6A,F8A may represent a lead molecule for peptidomimetic compounds that have a similar three-dimensional structure but are more resistant to peptidases and, therefore, suitable for in vivo treatment of experimentally induced tumors.


Assuntos
Proteínas de Homeodomínio/farmacologia , Proteínas Nucleares , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Fatores de Transcrição , Sequência de Aminoácidos , Proteína do Homeodomínio de Antennapedia , Apoptose , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Adesão Celular/efeitos dos fármacos , Contagem de Células , Divisão Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Genes myc/efeitos dos fármacos , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas/metabolismo , Ensaio Tumoral de Célula-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA