Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Epidemiol Psychiatr Sci ; 31: e18, 2022 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-35352674

RESUMO

AIMS: To examine the association between benzodiazepine receptor agonist (BZRA) use and mortality in patients hospitalised for coronavirus disease 2019 (COVID-19). METHODS: A multicentre observational study was performed at Greater Paris University hospitals. The sample involved 14 381 patients hospitalised for COVID-19. A total of 686 (4.8%) inpatients received a BZRA at hospital admission at a mean daily diazepam-equivalent dose of 19.7 mg (standard deviation (s.d.) = 25.4). The study baseline was the date of admission, and the primary endpoint was death. We compared this endpoint between patients who received BZRAs and those who did not in time-to-event analyses adjusted for sociodemographic characteristics, medical comorbidities and other medications. The primary analysis was a Cox regression model with inverse probability weighting (IPW). RESULTS: Over a mean follow-up of 14.5 days (s.d. = 18.1), the primary endpoint occurred in 186 patients (27.1%) who received BZRAs and in 1134 patients (8.3%) who did not. There was a significant association between BZRA use and increased mortality both in the crude analysis (hazard ratio (HR) = 3.20; 95% confidence interval (CI) = 2.74-3.74; p < 0.01) and in the IPW analysis (HR = 1.61; 95% CI = 1.31-1.98, p < 0.01), with a significant dose-dependent relationship (HR = 1.55; 95% CI = 1.08-2.22; p = 0.02). This association remained significant in sensitivity analyses. Exploratory analyses indicate that most BZRAs may be associated with an increased mortality among patients hospitalised for COVID-19, except for diazepam, which may be associated with a reduced mortality compared with any other BZRA treatment. CONCLUSIONS: BZRA use may be associated with an increased mortality among patients hospitalised for COVID-19, suggesting the potential benefit of decreasing dose or tapering off gradually these medications when possible.


Assuntos
COVID-19 , Antagonistas de Receptores de GABA-A/efeitos adversos , COVID-19/mortalidade , Hospitalização , Humanos , Modelos de Riscos Proporcionais
2.
Cell Death Dis ; 6: e1930, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26492365

RESUMO

Salinomycin, isolated from Streptomyces albus, displays antimicrobial activity. Recently, a large-scale screening approach identified salinomycin and nigericin as selective apoptosis inducers of cancer stem cells. Growing evidence suggests that salinomycin is able to kill different types of non-stem tumor cells that usually display resistance to common therapeutic approaches, but the mechanism of action of this molecule is still poorly understood. Since salinomycin has been suggested to act as a K(+) ionophore, we explored its impact on mitochondrial bioenergetic performance at an early time point following drug application. In contrast to the K(+) ionophore valinomycin, salinomycin induced a rapid hyperpolarization. In addition, mitochondrial matrix acidification and a significant decrease of respiration were observed in intact mouse embryonic fibroblasts (MEFs) and in cancer stem cell-like HMLE cells within tens of minutes, while increased production of reactive oxygen species was not detected. By comparing the chemical structures and cellular effects of this drug with those of valinomycin (K(+) ionophore) and nigericin (K(+)/H(+) exchanger), we conclude that salinomycin mediates K(+)/H(+) exchange across the inner mitochondrial membrane. Compatible with its direct modulation of mitochondrial function, salinomycin was able to induce cell death also in Bax/Bak-less double-knockout MEF cells. Since at the concentration range used in most studies (around 10 µM) salinomycin exerts its effect at the level of mitochondria and alters bioenergetic performance, the specificity of its action on pathologic B cells isolated from patients with chronic lymphocytic leukemia (CLL) versus B cells from healthy subjects was investigated. Mesenchymal stromal cells (MSCs), proposed to mimic the tumor environment, attenuated the apoptotic effect of salinomycin on B-CLL cells. Apoptosis occurred to a significant extent in healthy B cells as well as in MSCs and human primary fibroblasts. The results indicate that salinomycin, when used above µM concentrations, exerts direct, mitochondrial effects, thus compromising cell survival.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Mitocôndrias/efeitos dos fármacos , Piranos/farmacologia , Equilíbrio Ácido-Base , Animais , Respiração Celular , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Humanos , Células Jurkat , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Nigericina/farmacologia , Consumo de Oxigênio , Valinomicina/farmacologia
3.
Oncogene ; 33(49): 5569-81, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24469031

RESUMO

Mitochondria, the key bioenergetic intracellular organelles, harbor a number of proteins with proven or hypothetical ion channel functions. Growing evidence points to the important contribution of these channels to the regulation of mitochondrial function, such as ion homeostasis imbalances profoundly affecting energy transducing processes, reactive oxygen species production and mitochondrial integrity. Given the central role of mitochondria in apoptosis, their ion channels with the potential to compromise mitochondrial function have become promising targets for the treatment of malignancies. Importantly, in vivo evidence demonstrates the involvement of the proton-transporting uncoupling protein, a mitochondrial potassium channel, the outer membrane located porin and the permeability transition pore in tumor progression/control. In this review, we focus on mitochondrial channels that have been assigned a definite role in cell death regulation and possess clear oncological relevance. Overall, based on in vivo and in vitro genetic and pharmacological evidence, mitochondrial ion channels are emerging as promising targets for cancer treatment.


Assuntos
Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Animais , Apoptose , Cálcio/metabolismo , Morte Celular , Homeostase , Humanos , Camundongos , Proteínas Mitocondriais/metabolismo , Permeabilidade , Porinas/metabolismo , Canais de Potássio/metabolismo , Espécies Reativas de Oxigênio , Proteína Desacopladora 1
4.
Clin Exp Allergy ; 44(1): 79-90, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24164338

RESUMO

BACKGROUND: Degranulation of mast cells is stimulated by store-operated Ca(2+) -entry (SOCE). In other cell types, Ca(2+) -entry is modified by ceramide. Exogenously added ceramide has been shown to trigger mast cell apoptosis. Effects of endogenously produced ceramide in mast cells remained, however, elusive. Ceramide may be produced from sphingomyelin by acid sphingomyelinase (Asm). OBJECTIVE: This study explored the impact of Asm on mast cell functions. METHODS: Mast cells were isolated from bone marrow (BMMCs) or peritoneal lavage of gene-targeted mice lacking Asm (asm(-/-)) and their wild-type littermates (asm(+/+)). BMMC maturation and apoptosis-associated annexin V binding were determined by flow cytometry. Asm activity was assessed enzymatically, cytosolic Ca(2+) activity ([Ca(2+)]i) utilizing Fura-2 fluorescence, current across the cell membrane by whole-cell patch clamp, degranulation from hexosaminidase-release and migration utilizing a transwell chamber. In vivo anaphylaxis was derived from decrease in body temperature. RESULTS: Peritoneal mast cell number, BMMC phenotype, spontaneous BMMC apoptosis as well as BMMC CD117, CD34 and FcεRI expression were similar in both genotypes. In asm(+/+) BMMCs, stimulation with antigen resulted in a fast ~2.5-fold increase in Asm activity. Release of Ca(2+) from internal stores and hence several Ca(2+) -dependent functions were strongly impaired in asm(-/-) BMMCs. Thus, antigen-induced increase in [Ca(2+)]i in IgE-sensitized cells, antigen- but not ionomycin-induced currents through Ca(2+) -activated K(+) -channels (KCa 3.1), IgE/antigen-triggered ß-hexosaminidase release, and antigen-induced migration were all lower in asm(-/-) BMMCs than in asm(+/+) BMMCs. Pharmacological inhibition of Asm by amitriptyline (500 nm, 3 h) in asm(+/+) BMMCs similarly decreased antigen-induced increase in [Ca(2+)]i , KCa 3.1 currents, ß-hexosaminidase release and migration. The decrease in body temperature upon the induction of systemic anaphylaxis was significantly less pronounced in asm(-/-) mice than in asm(+/+) mice, an observation pointing to in vivo significance of Asm. CONCLUSIONS AND CLINICAL RELEVANCE: Asm is a novel, powerful regulator of mast cell function and thus a potential target in the treatment of allergic reactions.


Assuntos
Mastócitos/imunologia , Mastócitos/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Anafilaxia/genética , Anafilaxia/imunologia , Anafilaxia/metabolismo , Animais , Antígenos/imunologia , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/genética , Degranulação Celular/imunologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Movimento Celular/imunologia , Ativação Enzimática/efeitos dos fármacos , Imunofenotipagem , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Mastócitos/ultraestrutura , Camundongos , Camundongos Knockout , Fenótipo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielina Fosfodiesterase/genética
5.
Handb Exp Pharmacol ; (216): 431-56, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23563670

RESUMO

Despite the high prevalence and devastating impact of psychiatric disorders, little is known about their etiopathology. In this review, we provide an overview on the participation of sphingolipids and enzymes responsible for their metabolism in mechanisms underlying psychiatric disorders. We focus on the pathway from sphingomyelin to proapoptotic ceramide and the subsequent metabolism of ceramide to sphingosine, which is in turn phosphorylated to yield anti-apoptotic sphingosine-1-phosphate (S1P).The sphingomyelinase/ceramide system has been linked to effects of reactive oxygen species and proinflammatory cytokines in the central nervous system as well as to synaptic transmission. Compared to ubiquitously expressed acid sphingomyelinase, acid and neutral ceramidase and neutral sphingomyelinase are highly active in brain regions. Depressed patients show elevated plasma ceramide levels and increased activities of acid sphingomyelinase which is functionally inhibited by many anti-depressive drugs. Exposure to alcohol is associated with an activation of acid and neutral sphingomyelinase observed in cell culture, mouse models and in alcohol-dependent patients and with increased concentrations of ceramide in various organs.Levels of sphingomyelin and ceramide are altered in erythrocytes and post-mortem brain tissues of schizophrenic patients in addition to changes in expression patterns for serine palmitoyltransferase and acid ceramidase leading to impaired myelination. After induction of anxiety-like behavior in animal models, higher serum levels of S1P were reported to lead to neurodegeneration. Correspondingly, S1P infusion appeared to increase anxiety-like behavior. Significantly upregulated levels of the endogenous ceramide catabolite N,N-dimethylsphingosine were observed in rat models of allodynia. Conversely, rats injected intrathecally with N,N-dimethylsphingosine developed mechanical allodynia. Moreover, S1P has been implicated in spinal nociceptive processing.The increasing interest in lipidomics and improved analytical methods led to growing insight into the connection between psychiatric and neurological disorders and sphingolipid metabolism and may once provide new targets and strategies for therapeutic intervention.


Assuntos
Encéfalo/metabolismo , Transtornos Mentais/metabolismo , Dor/metabolismo , Transdução de Sinais , Esfingolipídeos/metabolismo , Animais , Ceramidas/metabolismo , Humanos , Transtornos Mentais/terapia , Dor/prevenção & controle , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/metabolismo , Síndrome
6.
Horm Metab Res ; 45(6): 401-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23386414

RESUMO

Over the past decade a number of murine models of Graves' disease (GD) have been described. The full symptom complex, including typical orbital changes, however, could not yet be induced. In this report, we examined the influence of modified immunization protocols on orbital pathology. C57BL/6 and BALB/c mice were immunized against the human TSH receptor (TSHR), using either a TSHR encoding plasmid or a TSHR A-subunit adenovirus. Prior to immunization with the TSHR plasmid, regulatory T cells were depleted in one group of each strain. TSHR-stimulating antibodies (TSAbs) were evaluated and orbits were stained immunohistochemically for F4/80, uncoupling protein-1 (UCP-1) and the TSHR. We found that after depletion of regulatory T cells, incidence of TSAb was increased in TSHR plasmid immunized C57BL/6 mice. Examination of early immunized mice showed no antibody production. However, a TSHR epitope-specific cellular immune response could be detected by tetramer-analyses. Adenoviral immunization lead to TSAb production in all but one animal. Analysis of F4/80 positive cells in retrobulbar fat revealed no significant macrophage infiltration in the orbits of immunized mice. Immunohistochemical staining shows co-localization of F4/80 positive cells, UCP-1 and the TSHR in retrobulbar fat. Though targets for TSHR autoimmunity could clearly be shown, immunization methods were not efficient enough to cause clear signs of orbital inflammation.


Assuntos
Tecido Adiposo/metabolismo , Doença de Graves/genética , Canais Iônicos/genética , Proteínas Mitocondriais/genética , Órbita/metabolismo , Receptores da Tireotropina/genética , Animais , Modelos Animais de Doenças , Feminino , Doença de Graves/metabolismo , Humanos , Canais Iônicos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Transporte Proteico , Receptores da Tireotropina/metabolismo , Proteína Desacopladora 1
8.
Curr Med Chem ; 19(31): 5394-404, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22856664

RESUMO

We have previously shown that the mitochondrial potassium channel Kv1.3 (mtKv1.3) in T lymphocytes is a novel target of Bax. Mutation of Bax at lysine 128 (BaxK128E) abrogates its inhibitory effects on mtKv1.3 and prevents apoptosis. The importance of mtKv1.3 inhibition was underscored by the finding that membrane-permeant Kv1.3 inhibitors induced Bax/Bak-independent cell death and reduced the volume of an mtKv1.3-expressing tumor by 90% in a mouse model. However, the possible involvement of other Kv channels in apoptosis has not been clarified. Here we report that, like Kv1.3, Kv1.1 and Kv1.5 also interact with Bax. Transfection of Kvdeficient lymphocytes with Kv1.1 restores sensitivity to cell death in apoptosis-resistant CTLL-2 lymphocytes. SiRNA down-regulation of Kv1.3 and Kv1.5 expression in macrophages confers resistance to apoptosis. We further report that J774 macrophages express Kv1.3 and Kv1.5 in their mitochondria and that inhibition of both channels with specific membrane-permeant drugs can efficiently induce apoptosis in a macrophage cell line. Thus, our results indicate that the mechanism proposed for Kv1.3 can be extended to other Kv channels and suggest that membrane-permeant drugs may be a novel pharmacological tool for inducing apoptosis in macrophages, important players in the immune system. This result could be exploited for the depletion of tumor-associated macrophages, which have been shown to foster tumor growth.


Assuntos
Apoptose , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/genética , Macrófagos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estaurosporina/farmacologia , Transfecção , Proteína X Associada a bcl-2/metabolismo
9.
Cell Death Differ ; 18(3): 427-38, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20885444

RESUMO

Members of the Bcl-2 family play key roles as proapoptotic (e.g., Bax) and antiapoptotic (e.g., Bcl-x(L)) regulators of programmed cell death. We previously identified the mitochondrial potassium channel Kv1.3 as a novel target of Bax. Incubating Kv1.3-positive isolated mitochondria with Bax triggered apoptotic events, whereas Kv1.3-deficient mitochondria were resistant to this stimulus. Mutation of Bax at lysine 128 (BaxK128E) abrogated its effects on Kv1.3 and the induction of apoptotic changes in mitochondria. These data indicate a toxin-like action of Bax on Kv1.3 to trigger at least some of the mitochondrial changes typical for apoptosis. To gain insight into the mechanism of Bax-Kv1.3 interaction, we mutated Glu158 of Bcl-x(L) (corresponding to K128 in Bax) to lysine. This substitution turned Bcl-x(L) proapoptotic. Transfection of double knockout (Bax(-/-)/Bak(-/-)) mouse embryonic fibroblasts (DKO MEFs) with either wild-type Bax, BaxK128E, or Bcl-x(L)E158K showed that apoptosis induced by various stimuli was defective in DKO MEFs and BaxK128E-transfected cells, but was recovered upon transfection with Bcl-xLE158K or wild-type Bax. Both wild-type Bax and BaxK128E can form similar ion-conducting pores upon incorporation into planar lipid bilayers. Our results point to a physiologically relevant interaction of Bax with Kv1.3 and further indicate a crucial role of a distinct lysine in determining the proapoptotic character of Bcl2-family proteins.


Assuntos
Apoptose/genética , Fibroblastos/citologia , Lisina/genética , Mutação Puntual/genética , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Proteína X Associada a bcl-2/genética , Proteína bcl-X/genética , Sequência de Aminoácidos , Animais , Citocromos c/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Humanos , Ativação do Canal Iônico , Células Jurkat , Canal de Potássio Kv1.3/metabolismo , Bicamadas Lipídicas/metabolismo , Potencial da Membrana Mitocondrial , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estrutura Quaternária de Proteína , Homologia de Sequência de Aminoácidos , Proteína Killer-Antagonista Homóloga a bcl-2/química , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/química , Proteína X Associada a bcl-2/deficiência , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/química , Proteína bcl-X/metabolismo
10.
Urologe A ; 48(1): 6-11, 2009 Jan.
Artigo em Alemão | MEDLINE | ID: mdl-19156338

RESUMO

The risk of local and systemic lymphatic metastasis of a tumor increases with the size of the malignant neoplasia. Lymph vessels are generated in the tumor and seem to follow anatomically defined pathways. However, the precise molecular and biological mechanisms seem to be complex and require definition. At present, molecules belonging to the vascular endothelial growth factors family and podoplanin have been identified as key for the proliferation of the tumor's lacteals. Molecular mechanisms of the tumor origin and the hematogenic and lymphatic dissemination are increasingly better defined by the use of new diagnostic and therapeutic approaches for tumor patients. Simultaneously, we might be able to influence processes such as cell growth, apoptosis, angiogenesis, and lymphogenic dissemination by novel drugs and thereby develop novel approaches for tumor treatment. Chemokine receptors seem to control essential steps of lymphogenic dissemination such as migration, invasion, and proliferation of tumor cells.


Assuntos
Citocinas/imunologia , Linfonodos/imunologia , Metástase Linfática/imunologia , Modelos Imunológicos , Fator A de Crescimento do Endotélio Vascular/imunologia , Humanos
11.
Oncogene ; 27(51): 6590-5, 2008 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18679423

RESUMO

Cisplatin is one of the most effectively used chemotherapeutic agents for cancer treatment. However, in humans, important cytotoxic side effects are observed including dose-limiting renal damage and profound gastrointestinal symptomatology. The toxic responses to cisplatin in mice are similar to those in human patients. Here, we evaluated whether the acid sphingomyelinase (Asm) mediates at least some of the toxic in vivo effects of cisplatin. To this end, we determined the toxic effects of a single intraperitoneal dose of cisplatin (27 mg/kg) in wild type (Asm(+/+)) and Asm-deficient mice (Asm(-/-)). Tissue injury and apoptosis were determined histologically on hematoxylin-eosin and TUNEL (terminal deoxynucleotidyl transferase (TdT)-mediated nick end labeling) stainings 3, 12, 36 and 72 h after treatment. Our results revealed severe toxicity of cisplatin in Asm(+/+) mice with increased numbers of apoptotic cells in the thymus and small intestine. In marked contrast, Asm(-/-) mice were resistant to cisplatin and no apoptosis was observed in these organs after treatment. Moreover, cisplatin treatment primarily triggered apoptosis of endothelial cells in microvessels of intestine and thymus, an effect that was absent in mice lacking Asm. The data thus suggest that at least some toxic effects of cisplatin are mediated by the Asm in vivo resulting in early death of endothelial cells and consecutive organ damage.


Assuntos
Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Citoproteção/genética , Gastroenteropatias/induzido quimicamente , Trato Gastrointestinal/efeitos dos fármacos , Esfingomielina Fosfodiesterase/genética , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Cisplatino/farmacologia , Citoproteção/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Gastroenteropatias/patologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Genes p53 , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Esfingomielina Fosfodiesterase/fisiologia
12.
Oncogene ; 27(33): 4557-68, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18408751

RESUMO

The molecular causes for resistance of melanoma to apoptosis are currently only partly understood. In the present study, we examined gene transfer and expression of the proapoptotic BH3-only protein Noxa as an alternative approach to chemotherapy and investigated the molecular mechanisms regulating Noxa-induced apoptosis. Noxa gene transfer caused dysregulation of both mitochondria and, as shown for the first time, also the endoplasmic reticulum, resulting in the accumulation of reactive oxygen species. Interestingly, expression of Noxa not only triggered the classical mitochondrial caspase cascade, but also resulted in the activation of apoptosis signal-regulating kinase1 and its downstream effectors c-Jun N-terminal kinase and p38. The activation of these kinases was abolished by antioxidants. Moreover, inhibition of the kinases by RNA interference or pharmacological inhibitors significantly attenuated Noxa-induced apoptosis. Thus, our data provide evidence for the involvement of multiple pathways in Noxa-induced apoptosis that are triggered at mitochondria and the endoplasmic reticulum, and suggest Noxa gene transfer as a complementary approach to chemotherapy.


Assuntos
Apoptose , Retículo Endoplasmático/metabolismo , Sistema de Sinalização das MAP Quinases , Melanoma/metabolismo , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspases/genética , Caspases/metabolismo , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Técnicas de Transferência de Genes , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Melanoma/genética , Melanoma/terapia , Mitocôndrias/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Interferência de RNA , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Urologe A ; 46(9): 1262-5, 2007 Sep.
Artigo em Alemão | MEDLINE | ID: mdl-17598083

RESUMO

To convert the concept already successful in mice into clinical practice and commercialize it, a human anti-CD95-antibody must be produced. In a second step experiments must be performed on various normal healthy cells and tissues to determine whether these human anti-CD95-antibodies administered in very low doses have any effect on human cells (particularly hepatocytes) or at least cause only minimal side effects. If these studies yield positive results, then clinical trials can be conducted in which increasing doses are given to exclude an acute hepatotoxic effect and then the effect exerted by the antibody in combination with irradiation on tumor growth can be investigated.The advantage of this concept lies in the fact that systemic stimulus (low doses of anti-CD95-antibodies) is highly intensified by local radiotherapy and only then initiates cell death. Since the anti-CD95-antibodies trigger apoptosis primarily in tumor endothelia, this approach could be employed not only for prostate cancer and melanomas, which have already been tested, but also for many other tumors.


Assuntos
Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Ceramidas/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos da radiação , Receptor fas/imunologia , Animais , Anticorpos/toxicidade , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta a Droga , Endotélio/efeitos dos fármacos , Endotélio/patologia , Endotélio/efeitos da radiação , Humanos , Masculino , Melanoma Experimental , Camundongos , Neoplasias da Próstata/patologia , Esfingomielina Fosfodiesterase/metabolismo
16.
Cell Mol Life Sci ; 64(2): 181-91, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17131060

RESUMO

Rhinoviruses, which cause common cold, belong to the Picornaviridae family, small non-enveloped viruses (diameter 15-30 nm) containing a single-stranded RNA genome (about 7 kb). Over 100 different rhinoviral serotypes have been identified thus far, establishing rhinoviruses as the most diverse group of Picornaviridae. Based on receptor binding properties, rhinoviruses are divided into two classes: the major group binding to intracellular adhesion molecule-1 and the minor group binding to the very low density lipoprotein receptors. Interactions between virus and the receptor molecules cause a conformational change in the capsid, which is a prerequisite for viral uptake. Rhinoviruses trigger a chemokine response upon infection that may lead to exacerbation of the symptoms of common cold, i.e. asthma and inflammation. The following review aims to summarize the knowledge about rhinoviral infections and discusses therapeutical approaches against this almost perfectly adapted pathogen.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/química , Resfriado Comum/tratamento farmacológico , Resfriado Comum/virologia , Rhinovirus/metabolismo , Transdução de Sinais/genética , Antivirais/uso terapêutico , Capsídeo/ultraestrutura , Resfriado Comum/imunologia , Citocinas/imunologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Receptores de LDL/metabolismo , Receptores Virais/metabolismo , Rhinovirus/classificação
17.
Oncogene ; 25(41): 5612-25, 2006 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-16636669

RESUMO

We have previously shown that activation of the acid sphingomyelinase (ASM), the release of ceramide and the formation of ceramide-enriched membrane domains are central for the induction of apoptosis by CD95. Here, we demonstrate that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and CD95 activate the ASM via a redox mechanism resulting in release of ceramide and formation of ceramide-enriched membrane platforms. Ceramide-enriched membrane platforms serve to cluster DR5 upon stimulation. Antioxidants prevent TRAIL-mediated stimulation of ASM, the release of ceramide, the formation of ceramide-enriched membrane platforms and the induction of apoptosis by TRAIL. Further, ASM-deficient splenocytes fail to cluster DR5 in ceramide-enriched membrane domains upon TRAIL stimulation and resist TRAIL-induced apoptosis, events that were restored by addition of natural C(16)-ceramide. A dose-response analysis indicates that ceramide-enriched membrane platforms greatly sensitized tumor cells to TRAIL-induced apoptosis. Our data indicate that ceramide-enriched membrane platforms are required for the signaling of TRAIL-DR5 complexes under physiological conditions.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose/fisiologia , Ceramidas/metabolismo , Glicoproteínas de Membrana/fisiologia , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Linhagem Celular , Ativação Enzimática , Humanos , Imuno-Histoquímica , Camundongos , Oxirredução , Ligante Indutor de Apoptose Relacionado a TNF
18.
J Membr Biol ; 205(3): 147-57, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16362503

RESUMO

Cell proliferation and apoptosis are paralleled by altered regulation of ion channels that play an active part in the signaling of those fundamental cellular mechanisms. Cell proliferation must--at some time point--increase cell volume and apoptosis is typically paralleled by cell shrinkage. Cell volume changes require the participation of ion transport across the cell membrane, including appropriate activity of Cl- and K+ channels. Besides regulating cytosolic Cl- activity, osmolyte flux and, thus, cell volume, most Cl- channels allow HCO3- exit and cytosolic acidification, which inhibits cell proliferation and favors apoptosis. K+ exit through K+ channels may decrease intracellular K+ concentration, which in turn favors apoptotic cell death. K+ channel activity further maintains the cell membrane potential, a critical determinant of Ca2+ entry through Ca2+ channels. Cytosolic Ca2+ may trigger mechanisms required for cell proliferation and stimulate enzymes executing apoptosis. The switch between cell proliferation and apoptosis apparently depends on the magnitude and temporal organization of Ca2+ entry and on the functional state of the cell. Due to complex interaction with other signaling pathways, a given ion channel may play a dual role in both cell proliferation and apoptosis. Thus, specific ion channel blockers may abrogate both fundamental cellular mechanisms, depending on cell type, regulatory environment and condition of the cell. Clearly, considerable further experimental effort is required to fully understand the complex interplay between ion channels, cell proliferation and apoptosis.


Assuntos
Apoptose/fisiologia , Proliferação de Células , Canais Iônicos/fisiologia , Canais de Potássio/fisiologia , Animais , Canais de Cálcio/fisiologia , Tamanho Celular , Genes ras/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Canais de Ânion Dependentes de Voltagem/fisiologia , Receptor fas/fisiologia
19.
J Neural Transm (Vienna) ; 112(11): 1583-90, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16245071

RESUMO

Acid sphingomyelinase (A-SMase) and its reaction product ceramide may play a role in the pathophysiology of depressive disorders and in the therapeutic action of antidepressive drugs. In a prospective case-control study, A-SMase activity was measured in peripheral blood mononuclear cells of 17 patients with a major depressive episode who were free of antidepressant drug therapy for at least 10 days and 8 healthy volunteers. In the patient group, A-SMase activity was correlated to the score (n=17, r=0.64, P=0.005). The patient group exhibited higher A-SMase activity compared to healthy volunteers (T=2.09, df=21.33, P<0.05). In addition, we demonstrate that the antidepressants imipramine and amitriptyline induce a long-term reduction of the activity of A-SMase in cultured cells.


Assuntos
Química Encefálica/fisiologia , Encéfalo/enzimologia , Transtorno Depressivo Maior/enzimologia , Esfingomielina Fosfodiesterase/sangue , Regulação para Cima/fisiologia , Adulto , Antidepressivos Tricíclicos/farmacologia , Encéfalo/fisiopatologia , Química Encefálica/efeitos dos fármacos , Estudos de Casos e Controles , Células Cultivadas , Ceramidas/metabolismo , Transtorno Depressivo Maior/sangue , Transtorno Depressivo Maior/diagnóstico , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Feminino , Humanos , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Pessoa de Meia-Idade , Bainha de Mielina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estudos Prospectivos , Agregação de Receptores/efeitos dos fármacos , Agregação de Receptores/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingomielina Fosfodiesterase/análise , Esfingomielinas/metabolismo
20.
Urologe A ; 44(6): 608-13, 2005 Jun.
Artigo em Alemão | MEDLINE | ID: mdl-15912324

RESUMO

The spread of tumor cells of solid tumors to the regional lymph nodes is an important step in the progression of the disease and also an important prognostic factor. While the significance of the prognostic value of the lymphatic progression had been detected long ago, only increased knowledge of the molecular anatomy and mechanisms involved in the lymphatic spread of tumor cells provided a beginning insight into the understanding of lymphatic metastasis. One group of important molecular factors consists of proteins produced by the tumor cells inducing a proliferation of lymphatic vessels into the primary tumor. The vascular endothelial growth factors have been identified as key factors in this process. In addition there are hints for the fact that chemokines, which are cytokine-like proteins taking part in the regulation of processes of inflammation, and their chemokine receptors control cellular key steps of lymphatic metastasis of tumor cells such as migration, proliferation, and invasion. In conclusion new data point to the possible inhibition of lymphatic spread by selective blockade of growth factor receptors or chemokine receptors. The growing insight into cellular understanding of the mechanisms involved in the metastasis of tumor cells into the lymphatics and lymph nodes will hopefully facilitate the development of new diagnostic and therapeutic tools in the treatment of cancer patients.


Assuntos
Quimiocinas/imunologia , Linfangiogênese/imunologia , Metástase Linfática/imunologia , Biologia Molecular/métodos , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Humanos , Neoplasias/irrigação sanguínea
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...