Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 88: 781-790, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32439472

RESUMEN

There is ample evidence for the role of the immune system in developing chronic pain following peripheral nerve injury. Especially Toll-like receptors (TLRs) and their associated signaling components and pro-inflammatory cytokines such as IL-1ß, induced after injury, are involved in nociceptive processes and believed to contribute to the manifestation of chronic neuropathic pain states. Whereas the inhibition of the kinase function of IRAK-4, a central kinase downstream of TLRs and IL-1 receptors (IL-1Rs), seems efficacious in various chronic inflammatory and autoimmune models, it's role in regulating chronic neuropathic pain remained elusive to date. Here, we examined whether pharmacological inhibition of IRAK-4 kinase activity using PF-06650833 and BMS-986147, two clinical-stage kinase inhibitors, is effective for controlling persistent pain following nerve injury. Both inhibitors potently inhibited TLR-triggered cytokine release in human peripheral blood mononuclear cell (PBMC) as well as human and rat whole blood cultures. BMS-986147 showing favorable pharmacokinetic (PK) properties, significantly inhibited R848-triggered plasma TNF levels in a rat in vivo cytokine release model after single oral dosing. However, BMS-986147 dose dependently reversed cold allodynia in a rat chronic constriction injury (CCI) model following intrathecal administration only, supporting the notion that central neuro-immune modulation is beneficial for treating chronic neuropathic pain. Although both inhibitors were efficacious in inhibiting IL-1ß- or TLR-triggered cytokine release in rat dorsal root ganglion cultures, only partial efficacy was reached in IL-1ß-stimulated human glial cultures indicating that inhibiting IRAK-4́'s kinase function might be partially dispensable for human IL-1ß driven neuroinflammation. Overall, our data demonstrate that IRAK-4 inhibitors could provide therapeutic benefit in chronic pain following nerve injury, and the central driver for efficacy in the neuropathic pain model as well as potential side effects of centrally available IRAK-4 inhibitors warrant further investigation to develop effective analgesia for patients in high unmet medical need.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Neuralgia , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Quinasas Asociadas a Receptores de Interleucina-1 , Leucocitos Mononucleares , Neuralgia/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
2.
ALTEX ; 37(1): 164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31960940

RESUMEN

In this manuscript, which appeared in ALTEX (2019), 36(4), 682- 699, doi:10.14573/altex.1909271 , the affiliation of Hennicke Kamp should be Experimental Toxicology and Ecology, BASF SE, Ludwigshafen, Germany. Further, the reference to an article by Bal-Price et al. (2015) should have the following doi:10.1007/s00204-015-1464-2 .

3.
ALTEX ; 36(4): 682-699, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31658359

RESUMEN

Only few cell-based test methods are described by Organisation for Economic Co-operation and Development (OECD) test guidelines or other regulatory references (e.g., the European Pharmacopoeia). The majority of toxicity tests still falls into the category of non-guideline methods. Data from these tests may nevertheless be used to support regulatory decisions or to guide strategies to assess compounds (e.g., drugs, agrochemicals) during research and development if they fulfill basic requirements concerning their relevance, reproducibility and predictivity. Only a method description of sufficient clarity and detail allows interpretation and use of the data. To guide regulators faced with increasing amounts of data from non-guideline studies, the OECD formulated Guidance Document 211 (GD211) on method documentation for the purpose of safety assessment. As GD211 is targeted mainly at regulators, it leaves scientists less familiar with regulation uncertain as to what level of detail is required and how individual questions should be answered. Moreover, little attention was given to the description of the test system (i.e., cell culture) and the steps leading to it being established in the guidance. To address these issues, an annotated toxicity test method template (ToxTemp) was developed (i) to fulfill all requirements of GD211, (ii) to guide the user concerning the types of answers and detail of information required, (iii) to include acceptance criteria for test elements, and (iv) to define the cells sufficiently and transparently. The fully annotated ToxTemp is provided here, together with reference to a database containing exemplary descriptions of more than 20 cell-based tests.


Asunto(s)
Pruebas de Toxicidad/métodos , Animales , Estudios de Evaluación como Asunto , Humanos , Organización para la Cooperación y el Desarrollo Económico , Reproducibilidad de los Resultados , Proyectos de Investigación , Pruebas de Toxicidad/normas
4.
J Med Chem ; 62(13): 6391-6397, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31244106

RESUMEN

Genome-wide-association studies in chronic low back pain patients identified sepiapterin reductase as a high interest target for developing new analgesics. Here we used 19F NMR fragment screening for the discovery of novel, ligand-efficient SPR inhibitors. We report the crystal structures of six chemically diverse inhibitors complexed with SPR, identifying relevant interactions and binding modes in the sepiapterin pocket. Exploration of our initial fragment screening hit led to double-digit nanomolar inhibitors of SPR with excellent ligand efficiency.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Amidas/química , Inhibidores Enzimáticos/química , Oxidorreductasas de Alcohol/metabolismo , Amidas/síntesis química , Amidas/metabolismo , Cristalografía por Rayos X , Descubrimiento de Drogas , Estabilidad de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Humanos , Ligandos , Espectroscopía de Resonancia Magnética , Microsomas Hepáticos/metabolismo , Estructura Molecular , Unión Proteica , Relación Estructura-Actividad
5.
Pharmacol Res Perspect ; 7(4): e00482, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31236277

RESUMEN

Small molecule inhibitors selectively targeting the immunoproteasome subunit ß5i are currently being developed for the treatment of autoimmune disorders. However, patients carrying loss-of-function mutations in the gene encoding ß5i (Psmb8) suffer from the proteasome-associated autoinflammatory syndromes (PRAAS) emphasizing the need to study pharmacological inhibition of immunoproteasome function in human cells. Here, we characterized the immunomodulatory potential of the selective ß5i inhibitor ONX 0914 and Bortezomib, a pan-proteasome inhibitor, in human peripheral blood mononuclear cells (PBMCs). Both compounds efficiently blocked pro-inflammatory cytokine secretion in human whole blood and PBMC cultures stimulated with toll-like receptor (TLR) agonists. Furthermore, the compounds inhibited T cell cytokine production induced by recall antigen CMVpp65 or by polyclonal stimulation. The viability of PBMCs, however, was rapidly decreased in the presence of ONX 0914 and Bortezomib demonstrated by decreased residual cytosolic ATP and increased Annexin V surface binding. Interestingly, HLA-DR + monocytes were rapidly depleted from the cultures in the presence of ONX 0914 as a ß5i-selective inhibitor and Bortezomib. In conclusion, the anti-inflammatory potential of ß5i-selective inhibitors is correlating with a cytotoxicity increase in human PBMC subsets ex vivo. Our results provide important insights into the anti-inflammatory mechanism of action of ß5i-inhibitors which currently hold the promise as a novel therapy for autoinflammatory diseases.


Asunto(s)
Leucocitos Mononucleares/citología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Proteínas de la Matriz Viral/inmunología , Bortezomib/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Regulación hacia Abajo , Antígenos HLA-DR/metabolismo , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Oligopéptidos/farmacología
6.
J Enzyme Inhib Med Chem ; 29(3): 408-19, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23663081

RESUMEN

This study is based on our attempts to further explore the structure-activity relationship (SAR) of VX-148 (3) in an attempt to identify inosine 5'-mono-phosphate dehydrogenase (IMPDH) inhibitors superior to mycophenolic acid. A five-point pharmacophore developed using structurally diverse, known IMPDH inhibitors guided further design of novel analogs of 3. Several conventional as well as novel medicinal chemistry strategies were tried. The combined structure- and ligand-based approaches culminated in a few analogs with either retained or slightly higher potency. The compounds which retained the potency were also checked for their ability to inhibit human peripheral blood mononuclear cells proliferation. This study illuminates the stringent structural requirements and strict SAR for IMPDH II inhibition.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , IMP Deshidrogenasa/antagonistas & inhibidores , IMP Deshidrogenasa/química , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Ácido Micofenólico/química , Ácido Micofenólico/farmacología , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Relación Estructura-Actividad
7.
J Comput Aided Mol Des ; 26(11): 1277-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23117549

RESUMEN

IMPDH (Inosine 5'-monophosphate dehydrogenase) catalyzes a rate-limiting step in the de novo biosynthesis of guanine nucleotides. IMPDH inhibition in sensitive cell types (e.g., lymphocytes) blocks proliferation (by blocking RNA and DNA synthesis as a result of decreased cellular levels of guanine nucleotides). This makes it an interesting target for cancer and autoimmune disorders. Currently available IMPDH inhibitors such as mycophenolic acid (MPA, uncompetitive inhibitor) and nucleoside analogs (e.g., ribavirin, competitive inhibitor after intracellular activation by phosphorylation) have unfavorable tolerability profiles which limit their use. Hence, the quest for novel IMPDH inhibitors continues. In the present study, a ligand-based virtual screening using IMPDH inhibitor pharmacophore models was performed on in-house compound collection. A total of 50,000 virtual hits were selected for primary screen using in vitro IMPDH II inhibition up to 10 µM. The list of 2,500 hits (with >70 % inhibition) was further subjected to hit confirmation for the determination of IC(50) values. The hits obtained were further clustered using maximum common substructure based formalism resulting in 90 classes and 7 singletons. A thorough inspection of these yielded 7 interesting classes in terms of mini-SAR with IC(50) values ranging from 0.163 µM to little over 25 µM. The average ligand efficiency was found to be 0.3 for the best class. The classes thus discovered represent structurally novel chemotypes which can be taken up for further development.


Asunto(s)
Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , IMP Deshidrogenasa/metabolismo , Preparaciones Farmacéuticas/química , Clonación Molecular , Inhibidores Enzimáticos/farmacología , Humanos , IMP Deshidrogenasa/genética , Modelos Químicos , Estructura Molecular , Conformación Proteica
8.
Handb Exp Pharmacol ; (204): 151-66, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695639

RESUMEN

Classic PDE5 inhibitors interact with and block the catalytic site of PDE5. They have been clinically validated for treatment of erectile dysfunction as well as reduction of pulmonary arterial pressure, improvement of exercise capacity, quality of life, and arterial oxygenation in patients with secondary pulmonary hypertension. Minor side effects are visual disturbances, headache, migraine, back pain, and interaction with nitrates (hypotension). Some of those side effects presumably can be ameliorated by improving selectivity and pharmacokinetics; other side effects probably are target related due to inhibition of basic physiological processes. Target related side effects may be bypassed by using PDE5 inhibitors with a different mode of action: PDE5, like PDE2, PDE6, PDE10, and PDE11, is a multidomain protein with an N-terminal tandem GAF domain, which in case of PDE5, is allosterically activated by cGMP. Potential inhibitors acting at the PDE5 GAF domain would be expected to inhibit only pathophysiologically upregulated PDE5 activity, whereas basal activity of PDE5 would remain unaffected.Here, we summarize a high-throughput screening campaign to identify inhibitors of the regulatory GAF domain of human PDE5. To target the regulatory domain independently from the catalytic site, we used a chimeric reporter enzyme: The hPDE5 GAF-tandem domain functionally replaced the GAF domain in the cyanobacterial adenylyl cyclase CyaB1. We identified inhibitors that target the GAF domain and also inhibitors that target the bacterial cyclase.Compounds binding to the PDE5 GAF domain were reanalysed with native human PDE5 to demonstrate inhibition using capillary electrophoresis. This identified 16 compounds that act on the GAF domain of PDE5. Two compounds fulfilled the initial requirement to inhibit, exclusively, activated PDE5, but not basal PDE5 activity.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/química , Inhibidores de Fosfodiesterasa 5/farmacología , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos , Estructura Terciaria de Proteína
9.
J Cell Physiol ; 226(8): 1970-80, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21520048

RESUMEN

Lung fibroblast proliferation and differentiation into myofibroblasts are pathological key events during development of lung fibrosis. Cyclic nucleotide signaling is described as a negative modulator of these cellular processes, and cyclic nucleotide degrading type 4 phosphodiesterases (PDE4) are important regulators of these pathways. In this study, we elucidated expression and the role of individual subtypes of PDE4 in primary normal human lung fibroblast (NHLF) in controlling cytokines-induced proliferation and conversion to myofibroblasts by short-interfering RNAs (siRNAs) induced knockdown. We verified the expression of PDE4A, B, and D, while PDE4C was only minor or even not expressed in NHLF. An efficient liposome-mediated transfection method for mRNA silencing and a knockdown of the expressed PDE4 subtypes was achieved in these cells. This knockdown was further validated by PDE4 protein expression analysis and PDE4 activity measurements. Functionally, the knockdown of PDE4A and PDE4B inhibited proliferation induced by the cytokine combination of bFGF and IL-1ß, whereas knockdown of PDE4D was ineffective. In contrast, TGF-ß induced differentiation into myofibroblasts was affected by knockdown of PDE4B and PDE4D, but not by PDE4A knockdown. In summary, our data allow to assign different PDE4 subtypes to distinct functions of human lung fibroblasts and highlight the predominant role of PDE4B in controlling pathophysiological processes of human lung fibroblasts. This provides a scientific rationale for focused therapeutic targeting of PDE4B to treat respiratory diseases with fibrotic lesions in the lung.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/fisiología , Citocinas/metabolismo , Fibroblastos/enzimología , Pulmón/enzimología , Miofibroblastos/enzimología , Aminopiridinas/farmacología , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Ciclopropanos/farmacología , Citocinas/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Interleucina-1beta/metabolismo , Isoenzimas/metabolismo , Pulmón/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Piridinas/farmacología , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Transfección , Factor de Crecimiento Transformador beta/metabolismo
10.
J Cell Physiol ; 223(2): 317-26, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20082309

RESUMEN

Interleukin-1beta (IL-1beta) and basic fibroblast growth factor (bFGF) are important regulators of proliferation, and their expression is increased in lungs of patients with asthma, idiopathic pulmonary fibrosis (IPF), or chronic obstructive pulmonary disease (COPD). We investigated the effect of IL-1beta and bFGF on proliferation of human lung fibroblasts and the role of COX-2, PGE(2), and cAMP in this process. Furthermore, the effect of phosphodiesterase (PDE) 3 and 4 inhibition was analyzed. In primary human lung fibroblasts low concentrations of IL-1beta (<10 pg/ml) potentiated the bFGF-induced DNA synthesis, whereas higher concentrations revealed antiproliferative effects. Higher concentrations of IL-1beta-induced COX-2 mRNA and protein associated with an increase in PGE(2) and cAMP, and all of these parameters were potentiated by bFGF. The PDE4 inhibitor piclamilast concentration-dependently reduced proliferation by a partial G1 arrest. The PDE3 inhibitor motapizone was inactive by itself but enhanced the effect of the PDE4 inhibitor. This study demonstrates that bFGF and IL-1beta act in concert to fine-tune lung fibroblast proliferation resulting in amplification or reduction. The antiproliferative effect of IL-1beta is likely attributed to the induction of COX-2, which is further potentiated by bFGF, and the subsequent generation of PGE(2) and cAMP. Inhibition of PDE4 inhibition (rather than PDE3) may diminish proliferation of human lung fibroblasts and therefore could be useful in the therapy of pathological remodeling in lung diseases.


Asunto(s)
AMP Cíclico/metabolismo , Citocinas/metabolismo , Fibroblastos/metabolismo , Sustancias de Crecimiento/agonistas , Pulmón/metabolismo , Inhibidores de Fosfodiesterasa 4 , Benzamidas/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Citocinas/farmacología , Replicación del ADN/efectos de los fármacos , Replicación del ADN/fisiología , Dinoprostona/agonistas , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Genes cdc/efectos de los fármacos , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Pulmón/inmunología , Pulmón/fisiopatología , Inhibidores de Fosfodiesterasa 3 , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/metabolismo , Piridazinas/farmacología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
BMC Gastroenterol ; 9: 69, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19765284

RESUMEN

BACKGROUND: The NO--cGMP system plays a key role in the regulation of sinusoidal tonus and liver blood flow with phosphodiesterase-5 (PDE-5) terminating the dilatory action of cGMP. We, therefore, investigated the effects of PDE-5 inhibitors on hepatic and systemic hemodynamics in rats. METHODS: Hemodynamic parameters were monitored for 60 min. after intravenous injection of sildenafil and vardenafil [1, 10 and 100 microg/kg (sil1, sil10, sil100, var1, var10, var100)] in anesthetized rats. RESULTS: Cardiac output and heart rate remained constant. After a short dip, mean arterial blood pressure again increased. Systemic vascular resistance transiently decreased slightly. Changes in hepatic hemodynamic parameters started after few minutes and continued for at least 60 min. Portal (var10 -31%, sil10 -34%) and hepatic arterial resistance (var10 -30%, sil10 -32%) decreased significantly (p < 0.05). At the same time portal venous (var10 +29%, sil10 +24%), hepatic arterial (var10 +34%, sil10 +48%), and hepatic parenchymal blood flow (var10 +15%, sil10 +15%) increased significantly (p < 0.05). The fractional liver blood flow (total liver flow/cardiac output) increased significantly (var10 26%, sil10 23%). Portal pressure remained constant or tended to decrease. 10 microg/kg was the most effective dose for both PDE-5 inhibitors. CONCLUSION: Low doses of phosphodiesterase-5 inhibitors have distinct effects on hepatic hemodynamic parameters. Their therapeutic use in portal hypertension should therefore be evaluated.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hemodinámica/efectos de los fármacos , Imidazoles/farmacología , Hígado/irrigación sanguínea , Inhibidores de Fosfodiesterasa 5 , Piperazinas/farmacología , Sulfonas/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Modelos Animales , Purinas/farmacología , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos , Citrato de Sildenafil , Triazinas/farmacología , Diclorhidrato de Vardenafil , Resistencia Vascular/efectos de los fármacos
12.
FEBS J ; 274(18): 4812-24, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17697115

RESUMEN

Different inhibitors of the Ca(2+)/calmodulin-stimulated phosphodiesterase 1 family have been described and used for the examination of phosphodiesterase 1 in cellular, organ or animal models. However, the inhibitors described differ in potency and selectivity for the different phosphodiesterase family enzymes, and in part exhibit additional pharmacodynamic actions. In this study, we demonstrate that phosphodiesterase 1C is expressed in the human glioblastoma cell line A172 with regard to mRNA, protein and activity level, and that lower activities of phosphodiesterase 2, phosphodiesterase 3, phosphodiesterase 4 and phosphodiesterase 5 are also present. The identity of the phosphodiesterase 1C activity detected was verified by downregulation of the mRNA and protein through human phosphodiesterase 1C specific small interfering RNA. In addition, the measured K(m) values (cAMP, 1.7 microm; cGMP, 1.3 microm) are characteristic of phosphodiesterase 1C. We demonstrate that treatment with the Ca(2+) ionophore ionomycin increases intracellular Ca(2+) in a concentration-dependent way without affecting cell viability. Under conditions of enhanced intracellular Ca(2+) concentration, a rapid increase in cAMP levels caused by the adenylyl cyclase activator forskolin was abolished, indicating the involvement of Ca(2+)-activated phosphodiesterase 1C. The reduction of forskolin-stimulated cAMP levels was reversed by phosphodiesterase 1 inhibitors in a concentration-dependent way. Using this cellular system, we compared the cellular potency of published phosphodiesterase 1 inhibitors, including 8-methoxymethyl-3-isobutyl-1-methylxanthine, vinpocetine, SCH51866, and two established phosphodiesterase 1 inhibitors developed by Schering-Plough (named compounds 31 and 30). We demonstrate that up to 10 microm 8-methoxymethyl-3-isobutyl-1-methylxanthine and vinpocetine had no effect on the reduction of forskolin-stimulated cAMP levels by ionomycin, whereas the more selective and up to 10 000 times more potent phosphodiesterase 1 inhibitors SCH51866, compound 31 and compound 30 inhibited the ionomycin-induced decline of forskolin-induced cAMP at nanomolar concentrations. Thus, our data indicate that SCH51866 and compounds 31 and 30 are effective phosphodiesterase 1 inhibitors in a cellular context, in contrast to the weakly selective and low-potency phosphodiesterase inhibitors 8-methoxymethyl-3-isobutyl-1-methylxanthine and vinpocetine. A172 cells therefore represent a suitable system in which to study the cellular effect of phosphodiesterase 1 inhibitors. 8-Methoxymethyl-3-isobutyl-1-methylxanthine and vinpocetine seem not to be suitable for the study of phosphodiesterase 1-mediated functions.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/farmacología , Calcio/metabolismo , Calmodulina/metabolismo , Línea Celular Tumoral , Colforsina/farmacología , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Activación Enzimática/efectos de los fármacos , Glioblastoma/enzimología , Humanos , Ionomicina/antagonistas & inhibidores , Ionomicina/farmacología , Inhibidores de Fosfodiesterasa/clasificación
13.
Eur J Pharmacol ; 572(1): 12-22, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17659276

RESUMEN

Pulmonary fibroblast to myofibroblast conversion is a pathophysiological feature of idiopathic pulmonary fibrosis and COPD. This conversion is induced by transforming growth factor (TGF)-beta derived from epithelial cells as well as activated macrophages that have infiltrated the lung. Preventing this conversion might be a favourable therapeutic approach. Within this study we examined the activity of different members of the phosphodiesterase (PDE) family in primary human lung fibroblasts and various lung fibroblast cell lines both before and after TGF-beta induced differentiation to myofibroblasts as reflected by the expression of alpha-smooth muscle actin. We showed that the predominant PDE activities in lung fibroblasts are attributed to PDE5, PDE1 and to a smaller extent to PDE4. cyclic GMP (cGMP)-hydrolyzing activity declines by about half after differentiation to myofibroblasts in all pulmonary fibroblasts investigated, which is accompanied by a down-regulation of PDE5 protein. Lung fibroblast to myofibroblast differentiation is blocked by treatment with the PDE4 inhibitor piclamilast alone, depending on the TGF-beta concentration applied, and in combination with prostaglandin E(2) (PGE(2)) in a synergistic manner. Despite the high PDE5 activity the PDE5 inhibitor sildenafil by itself as well as in combination with brain natriuretic peptide or the nitric oxide-donor DETA-NONOate shows no inhibiting effects. However, combining sildenafil with the guanylyl cyclase (GC) activator BAY58-2667 and ODQ (which sensitizes GC for activation by BAY58-2667) suppressed TGF-beta induced differentiation. In summary, our data indicate that drugs interfering with the cyclic AMP (cAMP)-as well as with the NO-cGMP-pathway offer the therapeutic opportunity to prevent the differentiation of pulmonary fibroblasts to myofibroblasts in lung fibrosis.


Asunto(s)
Activadores de Enzimas/farmacología , Fibroblastos/efectos de los fármacos , Guanilato Ciclasa/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Actinas/metabolismo , Benzamidas/farmacología , Benzoatos/farmacología , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/farmacología , Dinoprostona/farmacología , Sinergismo Farmacológico , Fibroblastos/citología , Fibroblastos/metabolismo , Fibrosis , Humanos , Inmunohistoquímica , Isoenzimas/metabolismo , Pulmón/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Péptido Natriurético Encefálico/farmacología , Donantes de Óxido Nítrico/farmacología , Compuestos Nitrosos/farmacología , Oxadiazoles/farmacología , Piperazinas/farmacología , Purinas/farmacología , Piridinas/farmacología , Quinoxalinas/farmacología , Citrato de Sildenafil , Guanilil Ciclasa Soluble , Sulfonas/farmacología , Factor de Crecimiento Transformador beta/farmacología
14.
Circulation ; 115(17): 2331-9, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17438150

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a life-threatening disease, characterized by vascular smooth muscle cell hyperproliferation. The calcium/calmodulin-dependent phosphodiesterase 1 (PDE1) may play a major role in vascular smooth muscle cell proliferation. METHODS AND RESULTS: We investigated the expression of PDE1 in explanted lungs from idiopathic PAH patients and animal models of PAH and undertook therapeutic intervention studies in the animal models. Strong upregulation of PDE1C in pulmonary arterial vessels in the idiopathic PAH lungs compared with healthy donor lungs was noted on the mRNA level by laser-assisted vessel microdissection and on the protein level by immunohistochemistry. In chronically hypoxic mouse lungs and lungs from monocrotaline-injected rats, PDE1A upregulation was detected in the structurally remodeled arterial muscular layer. Long-term infusion of the PDE1 inhibitor 8-methoxymethyl 3-isobutyl-1-methylxanthine in hypoxic mice and monocrotaline-injected rats with fully established pulmonary hypertension reversed the pulmonary artery pressure elevation, structural remodeling of the lung vasculature (nonmuscularized versus partially muscularized versus fully muscularized small pulmonary arteries), and right heart hypertrophy. CONCLUSIONS: Strong upregulation of the PDE1 family in pulmonary artery smooth muscle cells is noted in human idiopathic PAH lungs and lungs from animal models of PAH. Inhibition of PDE1 reverses structural lung vascular remodeling and right heart hypertrophy in 2 animal models. The PDE1 family may thus offer a new target for therapeutic intervention in pulmonary hypertension.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/terapia , Hidrolasas Diéster Fosfóricas/metabolismo , Arteria Pulmonar/enzimología , 1-Metil-3-Isobutilxantina/farmacología , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Animales , División Celular , Enfermedad Crónica , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , ADN/biosíntesis , Modelos Animales de Enfermedad , Humanos , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/terapia , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Inhibidores de Fosfodiesterasa/farmacología , Arteria Pulmonar/citología , Ratas , Regulación hacia Arriba/fisiología
15.
Cell Signal ; 17(3): 331-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15567064

RESUMEN

Human platelets contain the cyclic nucleotide-hydrolyzing phosphodiesterases (PDEs) 2, 3 and 5. The cGMP-PDE5 inhibitors Sildenafil and Zaprinast have been demonstrated to potentiate the anti-platelet aggregatory effect of NO donors like sodium nitroprusside (SNP) in vitro but the mechanisms of Sildenafil's action on the secretory function of human platelets have not been analysed in detail. In the present paper, we show (1) that both compounds potentiate the SNP-induced increase in cGMP in human platelets concentration-dependently. (2) However, whereas Sildenafil plus SNP treatment only partially inhibits thrombin-induced release of serotonin, the less selective Zaprinast plus SNP cause a complete inhibition. (3) The inhibition mediated by Sildenafil plus SNP is limited to low compound concentrations at which cAMP levels are increased, probably due to cGMP-mediated inhibition of PDE3. (4) High concentrations of Sildenafil (plus SNP) neither affect cAMP levels, likely due to the activation of PDE2, nor inhibits the release of serotonin. Thus, increases in both cyclic nucleotides seem to control platelet function. (5) Accordingly, treatment with increasing concentrations of Sildenafil plus SNP and a selective PDE2 inhibitor, which by its own has no effect, induced a concentration-dependent increase in cAMP and complete inhibition of platelet activation. In summary, our data indicate that Sildenafil inhibits secretory function of human platelets at least in part due to the cGMP-mediated effects on intracellular cAMP and that entire inhibition of serotonin release from thrombin-activated platelets is controlled by both cyclic nucleotides.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/fisiología , Plaquetas/efectos de los fármacos , Fosfodiesterasa I/fisiología , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/fisiología , Piperazinas/farmacología , 3',5'-GMP Cíclico Fosfodiesterasas , Plaquetas/metabolismo , AMP Cíclico/biosíntesis , GMP Cíclico/biosíntesis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Humanos , Técnicas In Vitro , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Activación Plaquetaria/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Purinas , Purinonas/farmacología , Piridazinas/farmacología , Serotonina/metabolismo , Citrato de Sildenafil , Sulfonas
16.
Mutat Res ; 544(2-3): 167-72, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14644318

RESUMEN

Agents inducing O(6)-methylguanine (O(6)MeG) in DNA, such as N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), are not only highly mutagenic and carcinogenic but also cytotoxic because of the induction of apoptosis. In CHO fibroblasts, apoptosis triggered by O(6)MeG requires cell proliferation and MutSalpha-dependent mismatch repair and is related to the induction of DNA double-strand breaks (DSBs). Furthermore, it is mediated by Bcl-2 degradation and does not require p53 for which the cells were mutated [Cancer Res. 60 (2000) 5815]. Here we studied cytotoxicity and apoptosis induced by MNNG in a pair of human lymphoblastoid cells expressing wild-type p53 (TK6) and mutant p53 (WTK1) and show that TK6 cells are more sensitive than WTK1 cells to cell killing (determined by a metabolic assay) and apoptosis. Apoptosis was a late response observed <24h after treatment and was related to accumulation of p53 and upregulation of Fas/CD95/Apo-1 receptor as well as Bax. The data indicate that MNNG induces apoptosis in lymphoblastoid cells by activating the p53-dependent Fas receptor-driven pathway. This is in contrast to CHO fibroblasts in which, in response to O(6)MeG, the mitochondrial damage pathway becomes activated.


Asunto(s)
Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Metilnitronitrosoguanidina/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2 , Proteína p53 Supresora de Tumor/fisiología , Receptor fas/fisiología , Animales , Células CHO , Línea Celular , Cricetinae , Daño del ADN/efectos de los fármacos , Humanos , Cinética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína X Asociada a bcl-2 , Receptor fas/efectos de los fármacos
17.
DNA Repair (Amst) ; 2(1): 49-60, 2003 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-12509267

RESUMEN

The anthracycline doxorubicin (adriamycin) is an important chemotherapeutic agent used in the treatment of solid epithelial and mesenchymal tumors as well as leukemias. A variety of mechanisms has been proposed to be involved in doxorubicin-induced cytotoxicity such as DNA intercalation, oxidative stress, DNA strand breakage by inhibition of topoisomerase II, activation of death receptors, and altered p53 expression. Concerning doxorubicin resistance and p53 status data reported are contradictory. Here, we show that mouse fibroblasts deficient in p53 (p53(-/-)) are more resistant to doxorubicin than p53 wild-type (p53 wt) cells. This is in contrast to other genotoxic agents (UV-light, alkylating drugs) for which p53(-/-) fibroblasts proved to be more sensitive. Resistance of p53(-/-) cells to doxorubicin is related to reduced induction of apoptosis. This is not likely to be due to altered apoptotic signaling since the expression of Bax and Bcl-2 was unchanged and the induction of Fas/CD95/APO-1 receptor and caspase-8 was the same in p53(-/-) and p53 wt cells on treatment with doxorubicin. However, we observed a clearly lower level of doxorubicin-induced DNA strand breaks in p53(-/-) cells compared to the wt. P170 glycoprotein was equally expressed and the accumulation and elimination of the drug occurred with identical kinetics in both cell types. p53 deficient cells were cross-resistant to another topoisomerase II inhibitor etoposide, which also provoked increased DNA strand breakage in p53 wt cells. Based on the data we conclude that the p53 status significantly impacts the generation of DNA strand breaks because of drug-induced topoisomerase inhibition rather than death receptor signaling. Since human tumors are frequently mutated in p53 the findings bear clinical implications.


Asunto(s)
Apoptosis/fisiología , Daño del ADN/fisiología , Doxorrubicina/metabolismo , Resistencia a Antineoplásicos/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Técnicas In Vitro , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Inhibidores de Topoisomerasa II
18.
Int J Mol Med ; 10(4): 473-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12239596

RESUMEN

Ras-homologous GTPases are involved in the regulation of genotoxic stress-induced gene expression and cell death. Since they need C-terminal isoprenylation for correct intracellular localization and function, we investigated whether depletion of cells from isopren precursor moieties using the HMG-CoA reductase inhibitor lovastatin affects cellular sensitivity to DNA damaging drugs. Here we show that lovastatin renders cells highly resistant to the tumor-therapeutic compound doxorubicin. Desensitization by lovastatin was reverted by co-treatment with GGPP indicating that inhibition of protein geranylgeranylation is involved in acquired doxorubicin resistance. Lovastatin does not influence cellular sensitivity to DNA damaging compounds such as cisplatin, methyl methanesulfonate and ionizing radiation. The frequency of apoptotic cell death induced by doxorubicin was not affected by lovastatin as shown by both annexin V and DNA fragmentation assay. However, lovastatin releases cells from doxorubicin induced G2 blockage. Furthermore, lovastatin protects cells from doxorubicin-induced DNA strand breakage without affecting drug uptake or the expression of multidrug resistance protein (mdr-1). Since lovastatin confers cross-resistance to the topoisomerase II specific inhibitor etoposide, we suggest desensitization by the statin to be related to topoisomerase II function. The finding that lovastatin renders cells resistant to doxorubicin and etoposide by reducing their genotoxic and cytotoxic effects might have clinical implications for cancer therapy.


Asunto(s)
Doxorrubicina/toxicidad , Resistencia a Antineoplásicos/fisiología , Etopósido/toxicidad , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/farmacología , Animales , Apoptosis/efectos de los fármacos , Células CHO , Ciclo Celular/efectos de los fármacos , Ensayo Cometa , Cricetinae , ADN/efectos de los fármacos , Doxorrubicina/farmacología , Etopósido/farmacología
19.
Int J Oncol ; 21(2): 289-96, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12118323

RESUMEN

The present investigation was performed to elucidate the role of purine nucleotides as potential indicators of chemosensitivity of malignant tumors. Drug-sensitive (s) and -resistant (r) tumor cell lines grown as monolayers (s: T47D, MCF-7 wild-type; r: NCI/ADR-RES, MCF-7/MDR) or as multicellular spheroids (T47D; NCI/ADR-RES) were exposed to 0.1, 1.0, and 10.0 microM Doxorubicin for up to 24 h. Purine nucleotides were assayed using HPLC and with some selected spheroids using imaging bioluminescence. The data show that in the time frame of the experiments reproducible and statistically significant changes in the nucleotides only occur at the highest drug concentration investigated. Under these conditions and using monolayer cultures, Doxorubicin caused a significant increase in ATP and GTP in sensitive but not in resistant cancer cells. Consequently, this differential change may be exploited for drug sensitivity testing in vitro. Doxorubicin exposure to spheroids was associated with significant increases in ATP and GTP in both sensitive and resistant variants. However, the kinetic of the changes in GTP was largely different between T47D and NCI/ADR-RES spheroids with a long-lasting, almost 3-fold elevation and a smaller, relatively short transient increase in GTP, respectively. Supplementing experiments with Doxorubicin treatment under inhibition of oxidative phosphorylation with Oligomycin abolished the drug-induced ATP and GTP peaks at persistent increases in ADP and AMP. Assuming that the spheroids may represent the in vivo situation to a better degree than monolayer cultures, experimental in vivo studies should clarify whether kinetic changes in GTP could be used as differential markers for the chemosensitivity of solid tumors. The experiments using Oligomycin support the hypothesis that purine nucleotides may be recycled from DNA fragments that result from the interaction of the drug with the DNA strands.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antineoplásicos/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Guanosina Trifosfato/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Cromatografía Líquida de Alta Presión , Resistencia a Múltiples Medicamentos , Humanos , Técnicas In Vitro , Mediciones Luminiscentes , Esferoides Celulares , Células Tumorales Cultivadas/metabolismo
20.
Mol Biol Cell ; 13(1): 348-61, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11809844

RESUMEN

UV light targets both membrane receptors and nuclear DNA, thus evoking signals triggering apoptosis. Although receptor-mediated apoptosis has been extensively investigated, the role of DNA damage in apoptosis is less clear. To analyze the importance of DNA damage induced by UV-C light in apoptosis, we compared nucleotide excision repair (NER)-deficient Chinese hamster ovary cells (lines 27-1 and 43-3B mutated for the repair genes ERCC3 and ERCC1, respectively) with the corresponding DNA repair-proficient fibroblasts (CHO-9 and ERCC1 complemented 43-3B cells). NER-deficient cells were hypersensitive as to the induction of apoptosis, indicating that apoptosis induced by UV-C light is due to unrepaired DNA base damage. Unrepaired lesions, however, do not activate the apoptotic pathway directly because apoptosis upon UV-C irradiation requires DNA replication and cell proliferation. It is also shown that in NER-deficient cells unrepaired lesions are converted into DNA double-strand breaks (DSBs) and chromosomal aberrations by a replication-dependent process that precedes apoptosis. We therefore propose that DSBs arising from replication of DNA containing nonrepaired lesions act as an ultimate trigger of UV-C-induced apoptosis. Induction of apoptosis by UV-C light was related to decline in the expression level of Bcl-2 and activation of caspases. Decline of Bcl-2 and subsequent apoptosis might also be caused, at least in part, by UV-C-induced blockage of transcription, which was more pronounced in NER-deficient than in wild-type cells. This is in line with experiments with actinomycin D, which provoked Bcl-2 decline and apoptosis. UV-C-induced apoptosis due to nonrepaired DNA lesions, replication-dependent formation of DSBs, and activation of the mitochondrial damage pathway is independent of functional p53 for which the cells are mutated.


Asunto(s)
Apoptosis , División Celular/efectos de la radiación , Daño del ADN , Rayos Ultravioleta/efectos adversos , Animales , Células CHO , Inhibidores de Caspasas , Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Cromatina/efectos de la radiación , Aberraciones Cromosómicas , Cricetinae , Inhibidores de Cisteína Proteinasa/farmacología , Reparación del ADN , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Cinética , Ratones , Mutación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tolerancia a Radiación/fisiología , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA