Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Pharmacol Res ; 91: 47-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25462173

RESUMO

The anticancer efficacy of anthracyclines is limited by cumulative dose-dependent early and delayed cardiotoxicity resulting in congestive heart failure. Mechanisms responsible for anthracycline-induced heart damage are controversially discussed and effective preventive measures are preferable. Here, we analyzed the influence of the lipid lowering drug lovastatin on anthracycline-induced late cardiotoxicity three month after treatment of C57BL/6 mice with five low doses of doxorubicin (5×3mg/kg BW; i.p.). Doxorubicin increased the cardiac mRNA levels of BNP, IL-6 and CTGF, while the expression of ANP remained unchanged. Lovastatin counteracted these persisting cardiac stress responses evoked by the anthracycline. Doxorubicin-induced fibrotic alterations were neither detected by histochemical collagen staining of heart sections nor by analysis of the mRNA expression of collagens. Extensive qRT-PCR-array based analyses revealed a large increase in the mRNA level of heat shock protein Hspa1b in doxorubicin-treated mice, which was mitigated by lovastatin co-treatment. Electron microscopy together with qPCR-based analysis of mitochondrial DNA content indicate that lovastatin attenuates doxorubicin-stimulated hyperproliferation of mitochondria. This was not paralleled by increased expression of oxidative stress responsive genes or senescence-associated proteins. Echocardiographic analyses disclosed that lovastatin protects from the doxorubicin-induced decrease in the left ventricular posterior wall diameter (LVPWD), while constrictions in fractional shortening (FS) and ejection fraction (EF) evoked by doxorubicin were not amended by the statin. Taken together, the data suggest beneficial effects of lovastatin against doxorubicin-induced delayed cardiotoxicity. Clinical studies are preferable to scrutinize the usefulness of statins for the prevention of anthracycline-induced late cardiotoxicity.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Cardiotônicos/uso terapêutico , Cardiotoxicidade/tratamento farmacológico , Doxorrubicina/efeitos adversos , Lovastatina/uso terapêutico , Animais , Cardiotônicos/farmacologia , Cardiotoxicidade/genética , Cardiotoxicidade/patologia , Fator de Crescimento do Tecido Conjuntivo/genética , Dano ao DNA , DNA Mitocondrial/metabolismo , Feminino , Fibrose , Perfilação da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Interleucina-6/genética , Lovastatina/farmacologia , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Peptídeo Natriurético Encefálico/genética
2.
J Biol Chem ; 287(46): 38590-9, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012366

RESUMO

To investigate the potency of the topoisomerase II (topo II) poisons doxorubicin and etoposide to stimulate the DNA damage response (DDR), S139 phosphorylation of histone H2AX (γH2AX) was analyzed using rat cardiomyoblast cells (H9c2). Etoposide caused a dose-dependent increase in the γH2AX level as shown by Western blotting. By contrast, the doxorubicin response was bell-shaped with high doses failing to increase H2AX phosphorylation. Identical results were obtained by immunohistochemical analysis of γH2AX focus formation, comet assay-based DNA strand break analysis, and measuring the formation of the topo II-DNA cleavable complex. At low dose, doxorubicin activated ataxia telangiectasia mutated (ATM) but not ATM and Rad3-related (ATR). Both the lipid-lowering drug lovastatin and the Rac1-specific inhibitor NSC23766 attenuated doxorubicin- and etoposide-stimulated H2AX phosphorylation, induction of DNA strand breaks, and topo II-DNA complex formation. Lovastatin and NSC23766 acted in an additive manner. They did not attenuate doxorubicin-induced increase in p-ATM and p-Chk2 levels. DDR stimulated by topo II poisons was partially blocked by inhibition of type I p21-associated kinases. DDR evoked by the topoisomerase I poison topotecan remained unaffected by lovastatin. The data show that the mechanisms involved in DDR stimulated by topo II poisons are agent-specific with anthracyclines lacking DDR-stimulating activity at high doses. Pharmacological inhibition of Rac1 signaling counteracts doxorubicin- and etoposide-stimulated DDR by disabling the formation of the topo II-DNA cleavable complex. Based on the data we suggest that Rac1-regulated mechanisms are required for DNA damage induction and subsequent activation of the DDR following treatment with topo II but not topo I poisons.


Assuntos
Dano ao DNA , DNA Topoisomerases Tipo II/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Antineoplásicos/farmacologia , Morte Celular , Linhagem Celular , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/efeitos dos fármacos , Histonas/química , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Imuno-Histoquímica/métodos , Neoplasias/tratamento farmacológico , Venenos/química , Ratos , Transdução de Sinais , Inibidores da Topoisomerase II/farmacologia , Proteínas rac1 de Ligação ao GTP/química , Proteínas rho de Ligação ao GTP/metabolismo
3.
J Cell Sci ; 123(Pt 10): 1652-62, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20406886

RESUMO

Functional recovery and regeneration of corticospinal tract (CST) fibers following spinal cord injury by compression or dorsal hemisection in mice was monitored after application of the enzyme-deficient Clostridium botulinum C3-protein-derived 29-amino-acid fragment C3bot(154-182). This peptide significantly improved locomotor restoration in both injury models as assessed by the open-field Basso Mouse Scale for locomotion test and Rotarod treadmill experiments. These data were supported by tracing studies showing an enhanced regenerative growth of CST fibers in treated animals as visualized by anterograde tracing. Additionally, C3bot(154-182) stimulated regenerative growth of raphespinal fibers and improved serotonergic input to lumbar alpha-motoneurons. These in vivo data were confirmed by in vitro data, showing an enhanced axon outgrowth of alpha-motoneurons and hippocampal neurons cultivated on normal or growth-inhibitory substrates after application of C3bot(154-182). The observed effects were probably caused by a non-enzymatic downregulation of active RhoA by the C3 peptide as indicated by pull-down experiments. By contrast, C3bot(154-182) did not induce neurite outgrowth in primary cultures of dorsal root ganglion cells. In conclusion, C3bot(154-182) represents a novel, promising tool to foster axonal protection and/or repair, as well as functional recovery after traumatic CNS injury.


Assuntos
ADP Ribose Transferases/farmacologia , Toxinas Botulínicas/farmacologia , Clostridium botulinum/metabolismo , Neurônios Motores/efeitos dos fármacos , Regeneração Nervosa , Fragmentos de Peptídeos/farmacologia , Traumatismos da Medula Espinal/fisiopatologia , Medula Espinal/efeitos dos fármacos , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Atividade Motora/efeitos dos fármacos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Tratos Piramidais/efeitos dos fármacos , Tratos Piramidais/fisiologia , Recuperação de Função Fisiológica , Serotonina/genética , Serotonina/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/cirurgia , Traumatismos da Medula Espinal/tratamento farmacológico , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
4.
Toxicol Appl Pharmacol ; 261(1): 66-73, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22712078

RESUMO

Liver is the main detoxifying organ and therefore the target of high concentrations of genotoxic compounds, such as environmental carcinogens and anticancer drugs. Here, we investigated the usefulness of lovastatin, which is nowadays widely used for lipid lowering purpose, as a hepatoprotective drug following the administration of the anthracycline derivative doxorubicin in vivo. To this end, BALB/c mice were exposed to either a single high dose or three consecutive low doses of doxorubicin. Acute and subacute hepatotoxicities were analyzed with or without lovastatin co-treatment. Lovastatin protected the liver against doxorubicin-induced acute pro-inflammatory and pro-fibrotic stress responses as indicated by an attenuated mRNA expression of tumor necrosis factor alpha (TNFα) and connective tissue growth factor (CTGF), respectively. Hepatoprotection by lovastatin was due to a reduced induction of DNA damage following doxorubicin treatment. The statin also mitigated subacute anthracycline-provoked hepatotoxicity as shown on the level of doxorubicin- and epirubicin-stimulated CTGF mRNA expression as well as histopathologically detectable fibrosis and serum concentration of marker enzymes of hepatotoxicity (GPT/GLDH). Kidney damage following doxorubicin exposure was not detectable under our experimental conditions.Moreover, lovastatin showed multiple inhibitory effects on doxorubicin-triggered hepatic expression of genes involved in oxidative stress response, drug transport, DNA repair, cell cycle progression and cell death. Doxorubicin also stimulated the formation of ceramides. Ceramide production, however, was not blocked by lovastatin, indicating that hepatoprotection by lovastatin is independent of the sphingolipid metabolism. Overall, the data show that lovastatin is hepatoprotective following genotoxic stress induced by anthracyclines. Based on the data, we hypothesize that statins might be suitable to lower hepatic injury following anthracycline-based anticancer therapy.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Doxorrubicina/toxicidade , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Animais , Antibióticos Antineoplásicos/administração & dosagem , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Fator de Crescimento do Tecido Conjuntivo/genética , Dano ao DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/administração & dosagem , Epirubicina/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo
5.
Cell Motil Cytoskeleton ; 66(11): 967-75, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19504561

RESUMO

Low molecular weight GTP-binding proteins of the Rho family control the organization of the actin cytoskeleton in eukaryotic cells. RhoA governs the formation of actin stress fibers and is responsible for the formation of the contractile ring in cytokinesis. Cytokinesis completion requires RhoA inactivation resulting in disassembly of the contractile ring. Cytokinesis thus requires switching of RhoA activity. This switch of RhoA activity is blocked by Rho-modifying bacterial protein toxins that either activate or inactivate RhoA by covalent modifications. Exoenzyme C3 from Clostridium limosum (C3-lim) and Clostridium difficile toxin B (TcdB) inactivate RhoA by mono-ADP-ribosylation and mono-glucosylation, respectively. Cytotoxic necrotizing factors (CNF), produced by either Yersinia pseudotuberculosis (CNFY) or uropathogenic strains of E. coli (CNF1), deamidate and thereby activate RhoA. This study provides evidence that RhoA-activating as well as RhoA-inactivating toxins cause inhibition of cytokinesis and cell division. The toxins' effects on cytokinesis were analyzed in Hela cells synchronized using the thymidine double block technique. Treatment of G2-phase cells with either the RhoA-activating CNFY or CNF1 or the RhoA-inactivating C3-lim or TcdB resulted in cytokinesis inhibition, as evidenced by the formation of a 4N population on flow cytometry, the inhibition of contractile ring formation, and the formation of bi-nucleated cells. While TcdB and CNF1 modify a broad-spectrum of Rho proteins, C3-lim and CNFY specifically target RhoA. Since C3-lim and CNFY both caused cytokinesis inhibition, our study re-inforces the critical role of RhoA (not Rac1 or Cdc42) in cytokinesis and cell division.


Assuntos
Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Citocinese/efeitos dos fármacos , Proteínas de Escherichia coli/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/fisiologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Células HeLa , Humanos
6.
Int J Biochem Cell Biol ; 45(8): 1767-75, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732113

RESUMO

RhoB is the only member of the Rho subfamily of small GTPases, which is classified as an immediate early gene product. RhoB is up-regulated in response to growth factors as well as cytotoxic and genotoxic agents. Clostridial glucosylating toxins have been reported to evoke pronounced RhoB expression, based on the inactivation of Rho/Ras proteins. In this study, we report on a long lasting expression of RhoB in cultured cells upon activation of Rho proteins by the cytotoxic necrotizing factor 1 (CNF1) from Escherichia coli. The observations of this study highlight a new pathway involving Rac1, which positively regulates the activity of the rhoB promoter and RhoB expression. Conversely, the isomeric cytotoxic necrotizing factor from Yersinia pseudotuberculosis (CNFy) drives GTP-loading of basal RhoB but fails to cause activation of the rhoB promoter and thus its expression. CNF1 inhibits cytokinesis and induces the formation of bi-nucleated (tetraploid) cells. Upon long term treatment with CNF1, RhoB(-/-) mouse embryonic fibroblasts (MEFs) exhibit DNA fragmentation, phosphatidylserine exposure, and loss of membrane integrity, while RhoB(+/-) MEFs persist as bi-nucleated (tetraploid) cells without any signs of cell death. In conclusion, the cytoprotective RhoB response is not only evoked by bacterial protein toxins inactivating Rho/Ras proteins but also by the Rac1-activating toxin CNF1.


Assuntos
Toxinas Bacterianas/farmacologia , Citoproteção/efeitos dos fármacos , Proteínas de Escherichia coli/farmacologia , Escherichia coli/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HT29 , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Poliploidia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ativação Transcricional/efeitos dos fármacos , Yersinia pseudotuberculosis/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoB de Ligação ao GTP/genética
7.
Naunyn Schmiedebergs Arch Pharmacol ; 385(9): 883-90, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22644106

RESUMO

C3-like exoenzymes are produced by various microorganism including Clostridium botulinum (C3bot), Bacillus cereus and Staphylococcus aureus. C3bot is the prototype of C3-like exoenzymes that specifically ADP-ribosylates and thereby inactivates Rho(A/B/C). C3-like exoenzymes are not yet regarded as virulence factors, as the lack of cell entry domains results in a poor accessibility of the C3-like exoenzymes to cells. In this study, the sensitivity of various cell lines to C3bot has been reinvestigated. Primary monocytes as well as cultured macrophage-like cells including J774A.1 cells and RAW macrophages exhibit a tenfold higher sensitivity to C3bot than fibroblasts and epithelial cells. RhoA ADP-ribosylation by C3bot resulted in the formation of pronounced bipolar protrusions based on defective tail retraction. The formation of bipolar protrusion resulted in inhibited macrophage migration. These findings suggested that macrophages appear to be target cells of C3bot. Migration of macrophage is a prerequiste for their recruitment to the site of pathogen invasion or tissue damage. Inhibition of macrophage migration likely preserves the survival of C3-producing microorganisms. The observations of this study reinforce the paradigm of a role of C3-like exoenzymes as virulence factors.


Assuntos
ADP Ribose Transferases/metabolismo , Toxinas Botulínicas/metabolismo , Movimento Celular , Macrófagos/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Humanos , Camundongos , Monócitos/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Neurotherapeutics ; 9(1): 185-98, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21866396

RESUMO

Peripheral nerve injuries are frequently seen in trauma patients and due to delayed nerve repair, lifelong disabilities often follow this type of injury. Innovative therapies are needed to facilitate and expedite peripheral nerve regeneration. The purpose of this study was to determine the effects of a 1-time topical application of a 26-amino-acid fragment (C3(156-181)), derived from the Clostridium botulinum C3-exoenzyme, on peripheral nerve regeneration in 2 models of nerve injury and repair in adult rats. After sciatic nerve crush, different dosages of C3(156-181) dissolved in buffer or reference solutions (nerve growth factor or C3(bot)-wild-type protein) or vehicle-only were injected through an epineurial opening into the lesion sites. After 10-mm nerve autotransplantation, either 8.0 nmol/kg C3(156-181) or vehicle were injected into the proximal and distal suture sites. For a period of 3 to 10 postoperative weeks, C3(156-181)-treated animals showed a faster motor recovery than control animals. After crush injury, axonal outgrowth and elongation were activated and consequently resulted in faster motor recovery. The nerve autotransplantation model further elucidated that C3(156-181) treatment accounts for better axonal elongation into motor targets and reduced axonal sprouting, which are followed by enhanced axonal maturation and better axonal functionality. The effects of C3(156-181) are likely caused by a nonenzymatic down-regulation of active RhoA. Our results indicate the potential of C3(156-181) as a therapeutic agent for the topical treatment of peripheral nerve repair sites.


Assuntos
Axônios/efeitos dos fármacos , Complemento C3/uso terapêutico , Atividade Motora/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Neuropatia Ciática/tratamento farmacológico , Potenciais de Ação/efeitos dos fármacos , Animais , Complemento C3/química , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Lateralidade Funcional , Imunoprecipitação , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Condução Nervosa/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Peptídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/patologia , Neuropatia Ciática/fisiopatologia , Neuropatia Ciática/cirurgia , Estatísticas não Paramétricas , Fatores de Tempo , Transplante de Tecidos/métodos , Proteína rhoA de Ligação ao GTP/metabolismo
9.
Naunyn Schmiedebergs Arch Pharmacol ; 383(3): 247-52, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21193903

RESUMO

C3 exoenzyme from Clostridium botulinum, specifically ADP-ribosylates small GTP-binding proteins RhoA, B, and C. ADP-ribosylation causes functional inactivation of Rho proteins resulting in cessation of the complete downstream signaling. Rho proteins are general regulators of a lot of essential cellular functions, among others, the neuronal growth cone. Rho activation, triggered by neuronal injury, inhibits neuronal repair mechanisms. To prevent the detrimental effect of active Rho in the recovery of injured neuronal systems, C3 has become a promising drug to inactivate RhoA in neurons. During the advancement of C3 to a drug candidate, it was found that ADP-ribosyltransferase activity of C3, in fact, is not essential for axonal and dendritic growth and branching. Rather, a peptide fragment of C3 covering the surface exposed ARTT loop from C3 (C3(154-182) peptide) is sufficient to induce growth and branching of neurons comparable to the effect of full-length C3. Whereas full-length C3 also acts on astrocytes and microglia to induce-at least in an in vitro model-inflammation and glial scar formation, C3(154-182) peptide is inert and seems only to act on neurons. In addition to its axono- and dendritotrophic effects on cultured primary hippocampal neurons, C3(154-182) peptide enhanced functional recovery and regeneration in a mouse model of spinal cord injury. Thus, in a proof-of-principle experiment, C3 peptide was shown to be efficacious in post-traumatic neuro-regeneration.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Botulínicas/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , ADP Ribose Transferases/metabolismo , ADP Ribose Transferases/farmacologia , Animais , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/farmacologia , Humanos , Regeneração Nervosa/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/metabolismo
10.
FEBS Lett ; 583(19): 3133-9, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19744486

RESUMO

Mono-glucosylation of (H/K/N)Ras by Clostridium sordellii lethal toxin (TcsL) blocks critical survival signaling pathways, resulting in apoptotic cell death. One yet unsolved problem in studies on TcsL is the lack of a method allowing the specific detection of (H/K/N)Ras glucosylation. In this study, we identify the Ras(Mab 27H5) antibody as a glucosylation-sensitive antibody capable for the immunoblot detection of (H/K/N)Ras glucosylation in TcsL-treated cells. Alternative Ras antibodies including the K-Ras(Mab F234) antibody or the v-H-Ras(Mab Y13-159) antibody recognize Ras proteins regardless of glucosylation. (H/K)Ras are further shown to be more efficaciously glucosylated by TcsL than Rac1 in rat basophilic leukemia cells as well as in a cell-free system.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo , Animais , Anticorpos , Apoptose , Toxinas Bacterianas/farmacologia , Catálise , Linhagem Celular Tumoral , Glicosilação , Ratos , Proteínas rac1 de Ligação ao GTP/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA