Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Antimicrob Chemother ; 78(11): 2723-2730, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37757461

RESUMEN

BACKGROUND: The glycopeptide teicoplanin is considered first-line treatment for severe infections caused by Gram-positive bacteria. Individualized treatment of teicoplanin is gaining interest. As only protein-unbound drug is pharmacologically active, a sensitive assay measuring unbound and total teicoplanin is indispensable for pharmacological research and dose optimization. OBJECTIVES: To develop and validate a UPLC-MS/MS method to quantify unbound and total teicoplanin in human serum. METHODS: The developed assay was validated according to the ICH guideline M10 on Bioanalytical Method Validation and study sample analysis. Unbound teicoplanin was obtained by ultrafiltration. The assay was cross-validated with a quantitative microsphere (QMS) immunoassay in a side-by-side comparison using 40 patient samples. RESULTS: With the developed and validated method, all main teicoplanin components (A2-1, A2-2/A2-3, A2-4/A2-5 and A3-1) can be quantified. Total run time was 5.5 min. Concentration range was 2.5-150 mg/L for total and 0.1-25 mg/L for unbound teicoplanin. Precision (coefficient of variation) and accuracy (bias) of total teicoplanin were 5.97% and 107%, respectively, and 7.17% and 108%, respectively, for unbound teicoplanin.Bland-Altman analysis showed total concentrations measured with the UPLC-MS/MS method were equivalent to the results of the QMS immunoassay. A total of 188 samples from 30 patients admitted to the ICU and haematology department were measured; total concentrations ranged between 2.92 and 98.5 mg/L, and unbound concentrations ranged between 0.37 and 30.7 mg/L. CONCLUSIONS: The developed method provided rapid, precise and accurate measurement of unbound and total teicoplanin. The developed method is now routinely applied in pharmacological research and clinical practice.


Asunto(s)
Espectrometría de Masas en Tándem , Teicoplanina , Humanos , Cromatografía Liquida , Glicopéptidos
2.
Am J Transplant ; 11(3): 439-49, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21342445

RESUMEN

Mycophenolic acid is the active ingredient of the immunosuppressant mycophenolate mofetil that is widely used in transplantation medicine and autoimmunity. Mycophenolic acid inhibits inosine monophosphate dehydrogenase, an enzyme involved in biosynthesis of guanine nucleotides required for lymphocyte clonal expansion. Here, we present novel insights into the mechanisms underlying mycophenolic acid-mediated suppression of human CD4+ T cells. Upon CD3/CD28 stimulation, mycophenolic acid inhibited T cell IL-17, IFN-γ and TNF-α production but not IL-2 production. Phenotypic analysis showed that drug treatment enhanced the expression of negative co-stimulators PD-1, CTLA-4 and the transcription factor FoxP3 and decreased the expression of positive co-stimulators CD27 and CD28, whereas CD25 was unaffected. Mycophenolic acid-treated cells were anergic, but not suppressive, and at the same time proved hyperblastoid with high metabolic activity. Moreover, a reduced Akt/mTOR and STAT5 signaling was observed. Interestingly, the co-stimulatory molecule CD70 was uniquely and dose-dependently upregulated on mycophenolic acid-treated T cells and found to be directly linked to target enzyme inhibition. CD70 on mycophenolic acid-treated cells proved functional: an anti-CD70 agonist was found to restore both STAT5 and Akt/mTOR signaling and may thereby prevent apoptosis and promote survival. These novel insights may contribute to optimization of protocols for MPA-based immunosuppressive regimens.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Ligando CD27/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Nucleótidos de Guanina/metabolismo , Ácido Micofenólico/farmacología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citometría de Flujo , Humanos , IMP Deshidrogenasa/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
3.
Ann Rheum Dis ; 69(1): 276-83, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19188194

RESUMEN

OBJECTIVE: IL-18 is a pluripotent cytokine that has been implicated in the development of rheumatoid arthritis. A soluble form of the IL-18 receptor accessory protein (sIL-18Rbeta) with unknown function has recently been identified. This study examined the ability of sIL-18Rbeta to inhibit IL-18 biological activities and to modulate immune responses during collagen-induced arthritis (CIA). METHODS: Adenoviruses encoding sIL-18Rbeta were administered intravenously in type II collagen-immunised DBA/1 mice. Humoral responses were analysed by determining anti-bovine collagen type II (BCII) antibody levels by ELISA. Cytokine production by splenic T cells and cytokine levels in serum were measured by Luminex multi-analyte technology. CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) were measured by flow cytometry. RESULTS: Intravenous delivery of Ad5.sIL-18Rbeta in collagen-immunised mice led to enhanced transgene expression in splenic antigen-presenting cells (APC). A co-culture of these sIL-18Rbeta-transduced APC with purified splenic CD3(+) T cells led to a marked inhibition of IL-18-induced IFNgamma, IL-4 and IL-17 production by CD3(+) T cells. Remarkably, systemic treatment with Ad5.sIL-18Rbeta caused an exacerbation of arthritis, and histological evaluation of knee joints showed increased cartilage and bone erosion. No significant differences were observed in anti-BCII antibodies, but the aggravation was accompanied by decreased IFNgamma (-30%) and IL-4 (-44%) and increased IL-17 (+84%) production by splenic CD3(+) T cells. In addition, reduced circulating levels of CD4(+)CD25(+)Foxp3(+) Treg and anti-inflammatory IL-10 were shown. CONCLUSION: This study identifies sIL-18Rbeta as a novel IL-18 inhibitor, which promotes CIA after intravenous overexpression by affecting Treg levels and supporting a T helper type 17 response.


Asunto(s)
Artritis Experimental/inmunología , Receptores de Interleucina-18/inmunología , Subgrupos de Linfocitos T/inmunología , Adenoviridae/genética , Animales , Artritis Experimental/patología , Complejo CD3/análisis , Células Cultivadas , Citocinas/biosíntesis , Citometría de Flujo/métodos , Vectores Genéticos , Inmunomodulación/inmunología , Interferón gamma/biosíntesis , Interleucina-18/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Solubilidad , Bazo/inmunología , Linfocitos T Reguladores/inmunología , Transfección
4.
Br J Pharmacol ; 154(1): 153-64, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18297096

RESUMEN

BACKGROUND AND PURPOSE: The p38 kinase regulates the release of proinflammatory cytokines including tumour-necrosis factor-alpha (TNFalpha) and is regarded as a potential therapeutic target in rheumatoid arthritis (RA). Using the novel p38 inhibitor Org 48762-0, we investigated the therapeutic potential of p38 inhibition and compared this to anti-mouse (m)TNFalpha antibody treatment in murine collagen-induced arthritis (CIA). EXPERIMENTAL APPROACH: Pharmacological profiles of Org 48762-0 were characterized in kinase assays, cellular assays and in lipopolysaccharide (LPS)-induced inflammation in mice. The effects of Org 48762-0 and of mTNFalpha-neutralization on established arthritis were examined in murine CIA. KEY RESULTS: Org 48762-0 potently inhibited p38alpha kinase with a high degree of kinase selectivity. In cellular assays, Org 48762-0 reduced LPS-induced TNFalpha release. Oral administration of Org 48762-0 in mice showed drug-like pharmacokinetic properties and inhibited LPS-induced cytokine production. These pharmacological characteristics of Org 48762-0 prompted a comparison of therapeutic efficacy with mTNFalpha-neutralization in CIA. Org 48762-0 and anti-mTNFalpha antibody treatment equally inhibited development of arthritis when evaluated macroscopically. Radiological analyses revealed protection against bone damage for both treatments, although statistical difference was reached with Org 48762-0 treatment only. Further, micro-computed tomographical and histopathological analyses confirmed the protective effects of Org 48762-0 on joint damage. CONCLUSIONS AND IMPLICATIONS: Pharmacological targeting of p38 kinase provided good protection against joint tissue damage in CIA. In our experiments, neutralization of mTNFalpha produced less prominent suppression of bone damage. Our data suggest a therapeutic potential for selective and potent p38 inhibitors in RA.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Piridinas/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Antiinflamatorios/uso terapéutico , Artritis Experimental/patología , Western Blotting , Cartílago/patología , Endotoxemia/tratamiento farmacológico , Inhibidores Enzimáticos/farmacocinética , Femenino , Inflamación/inducido químicamente , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Prednisolona/uso terapéutico , Especificidad por Sustrato , Tomografía Computarizada por Rayos X
5.
Arthritis Rheum ; 54(5): 1518-28, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16646036

RESUMEN

OBJECTIVE: An important mechanism contributing to cartilage destruction in arthritis is chondrocyte desensitization toward its main anabolic factor, insulin-like growth factor 1 (IGF-1). In this study, we sought to determine the role of suppressor of cytokine signaling 3 (SOCS-3) in the induction of IGF-1 desensitization of murine chondrocytes. METHODS: Chondrocyte responsiveness to IGF-1 was assessed by 35S-sulfate incorporation into proteoglycans (PGs), via aggrecan messenger RNA expression, using quantitative real-time polymerase chain reaction or insulin receptor substrate 1 (IRS-1) tyrosine phosphorylation (Western blot analysis). IGF-1 desensitization of patellar chondrocytes was studied in zymosan-induced arthritis. IGF-1 desensitization was induced in patellar cartilage explants or the H4 chondrocyte cell line, exposed to interleukin-1alpha (IL-1alpha). SOCS-3 protein expression was assessed by immunohistochemistry or by Western blot analysis of protein extracts. The role of SOCS-3 in IGF-1 signaling was elucidated by adenoviral overexpression. RESULTS: Exposure of murine articular cartilage to IL-1 caused a significant decrease in IGF-1-induced PG synthesis. This effect also occurred in inducible nitric oxide synthase-knockout mice, revealing the involvement of a secondary IL-1-induced factor other than nitric oxide. We showed that IL-1 significantly up-regulated SOCS-3 transcription and protein synthesis in H4 chondrocytes. In contrast, IL-18 was unable to induce SOCS-3 expression and failed to induce chondrocyte IGF-1 desensitization. Histologic analysis of samples from arthritic knee joints revealed high expression of SOCS-3 in chondrocytes. Through adenoviral overexpression of SOCS-3, we obtained direct evidence that SOCS-3 inhibits IGF-1-mediated cell signaling, since IRS-1 phosphorylation was reduced. CONCLUSION: This study demonstrates that IL-1-induced SOCS-3 expression is a novel mechanism of IGF-1 desensitization in chondrocytes; in conjunction with nitric oxide it can contribute to cartilage damage during arthritis.


Asunto(s)
Cartílago/patología , Condrocitos/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Interleucina-1/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína 3 Supresora de la Señalización de Citocinas
6.
Arthritis Rheum ; 52(7): 2202-11, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15986350

RESUMEN

OBJECTIVE: To discern the mode of interleukin-1 (IL-1) inhibition of soluble IL-1 receptor accessory protein (sIL-1RAcP) by comparison with IL-1 receptor antagonist (IL-1Ra) in arthritis. METHODS: Adenoviral vectors encoding either sIL-1RAcP or IL-1Ra were administered systemically before onset of collagen-induced arthritis in DBA/1 mice. Anti-bovine type II collagen IgG and IL-6 were quantified in serum. Proliferative response of splenic T cells was determined in the presence of sIL-1RAcP or IL-1Ra. The effect on IL-1 inhibition of recombinant sIL-1RAcP and IL-1Ra was further examined in vitro, using NF-kappaB luciferase reporter cell lines. Quantitative polymerase chain reaction was used to determine the relative messenger RNA expression of the IL-1 receptors. RESULTS: Adenoviral overexpression of both sIL-1RAcP and IL-1Ra resulted in amelioration of the collagen-induced arthritis. Both IL-1 antagonists reduced the circulating levels of antigen-specific IgG2a antibodies, but only IL-1Ra was able to inhibit lymphocyte proliferation. By using purified lymphocyte populations derived from NF-kappaB reporter mice, we showed that sIL-1RAcP inhibits IL-1-induced NF-kappaB activity in B cells but not T cells, whereas IL-1Ra inhibited IL-1 on both cell types. A study in a panel of NF-kappaB luciferase reporter cells showed that the sIL-1RAcP inhibits IL-1 signaling on cells expressing either low levels of membrane IL-1RAcP or high levels of IL-1RII. CONCLUSION: We show that the sIL-1RAcP ameliorated experimental arthritis without affecting T cell immunity, in contrast to IL-1Ra. Our results provide data in support of receptor competition by sIL-1RAcP as an explanation for the different mode of IL-1 antagonism in comparison with IL-1Ra.


Asunto(s)
Artritis Experimental/terapia , Terapia Genética , Interleucina-1/antagonistas & inhibidores , Proteínas/genética , Receptores de Interleucina-1/genética , Adenoviridae/genética , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Bovinos , Proliferación Celular , Modelos Animales de Enfermedad , Expresión Génica , Interleucina-1/metabolismo , Proteína Accesoria del Receptor de Interleucina-1 , Masculino , Ratones , Ratones Endogámicos DBA , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
7.
Ann Rheum Dis ; 64(3): 408-14, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15256380

RESUMEN

BACKGROUND: Dendritic cells orchestrate pivotal immunological processes mediated by the production of cytokines and chemokines. OBJECTIVE: To assess whether neutralisation of tumour necrosis factor alpha (TNF alpha) during maturation of dendritic cells affects their phenotype and behaviour, which might explain the beneficial effects of TNF alpha neutralisation in rheumatoid arthritis. METHODS: Immature and fully matured dendritic cells were cultured from blood monocytes from patients with rheumatoid arthritis and healthy controls following standardised protocols. TNF alpha was neutralised by addition of the p55 soluble TNF alpha receptor, PEGsTNFRI. The effect of TNF alpha neutralisation on the phenotype (CD14, CD16, CD32, CD64, CD80, CD83, CD86, and MHC) of dendritic cells was investigated by flow cytometry. Expression of chemokines (CCL17, CCL18, CCL19, CCL22, CCL3, and CXCL8) and production of IL1 beta and IL6 during dendritic cell differentiation and maturation were examined. RESULTS: Neutralisation of TNF alpha during the differentiation and maturation of dendritic cells did not result in an altered dendritic cell phenotype in the rheumatoid patients or the healthy controls. In contrast, the expression of CCL17, CCL18, CCL19, CCL22, CCL3, and CXCL8 by dendritic cells was significantly reduced when TNF alpha activity was inhibited during lipopolysaccharide triggered dendritic cell maturation. The production of IL1 beta and IL6 by mature dendritic cells was inhibited by PEGsTNFRI. CONCLUSIONS: Inhibition of TNF alpha activity during dendritic cell maturation leads to the development of semi-mature cells. These data suggest a novel pathway by which the neutralisation of TNF alpha might exert its therapeutic effects.


Asunto(s)
Artritis Reumatoide/inmunología , Células Dendríticas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Quimiocinas CC/sangre , Células Dendríticas/inmunología , Células Dendríticas/patología , Relación Dosis-Respuesta Inmunológica , Humanos , Inmunofenotipificación , Interleucina-1/biosíntesis , Interleucina-6/biosíntesis , Polietilenglicoles/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/fisiología
8.
Gene Ther ; 11(7): 581-90, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14973543

RESUMEN

To achieve a disease-regulated transgene expression for physiologically responsive gene therapy of arthritis, a hybrid promoter was constructed. The human IL-1 beta enhancer region (-3690 to -2720) upstream of the human IL-6 promoter region (-163 to +12) was essential in mounting a robust response in HIG-82 synovial fibroblasts and in RAW 264,7 macrophages. A replication-deficient adenovirus was engineered with luciferase (Luc) controlled by the IL-1/IL-6 promoter (Ad5.IL-1/IL-6-Luc). LPS caused a 23- and 4.6-fold induction of Luc. activity in RAW cells infected with Ad5.IL-1/IL-6-Luc or the conventional Ad5.CMV-Luc construct, respectively. Next, adenoviruses (10(6) ffu) were injected into the knees of C57Bl/6 mice. An intra-articular injection of zymosan, 3 days after Ad5.IL-1/IL-6-Luc, increased Luc. activity by 39-fold but had no effect in the Ad5.CMV-Luc joints. The constitutive CMV promoter was rapidly silenced and could not be reactivated in vivo. In contrast, the IL-1/IL-6 promoter could be reactivated by Streptococcal cell wall (SCW)-induced arthritis up to 21 days after infection. Next the IL-1/IL-6 promoter was compared to the C3-Tat/HIV-LTR two-component system in wild-type, IL-6(-/-) and IL-1(-/-) gene knockout mice. Both systems responded well to LPS-, zymosan- and SCW-induced arthritis. However, the basal activity of the IL-1/IL-6 promoter was lower and IL-6 independent. This study showed that the IL-1/IL-6 promoter is feasible to achieve disease-regulated transgene expression for treatment of arthritis.


Asunto(s)
Adenoviridae/genética , Artritis Infecciosa/terapia , Citocinas/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Regiones Promotoras Genéticas , Animales , Antígenos Bacterianos , Artritis Infecciosa/inmunología , Regulación de la Expresión Génica , Inyecciones Intraarticulares , Interleucina-1/genética , Interleucina-6/genética , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Streptococcus/inmunología , Transducción Genética/métodos , Zimosan
9.
Arthritis Rheum ; 50(1): 103-11, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14730606

RESUMEN

OBJECTIVE: To investigate in vivo and in vitro whether macrophages have an intermediate role in transforming growth factor beta (TGFbeta)-induced osteophyte formation. METHODS: In vivo, synovial lining macrophages were selectively depleted by injection of clodronate-laden liposomes 7 days prior to injection of 20 ng or 200 ng of TGFbeta into murine knee joints 3 times, on alternate days. Total knee joint sections were obtained on day 7 after the last injection and stained with Safranin O. Production of bone morphogenetic protein 2 (BMP-2) and BMP-4 was determined by immunolocalization. The interaction between murine macrophages and mesenchymal cells (precursors with chondrogenic potential) was studied in vitro using a Transwell system in which RAW macrophages were cocultured with C3H10T1/2 mesenchymal cells. Spheroid neocartilage formation was quantified microscopically after staining with May-Grünwald-Giemsa. RESULTS: Triple injections of 20 ng or 200 ng of TGFbeta into normal murine knee joints induced significant osteophyte formation at the lateral and medial sites of the patella and femur on day 7 after the last injection. Strikingly, removal of synovial lining macrophages prior to TGFbeta injection resulted in a drastic reduction of osteophyte formation (by 70% and 64% after injection of 20 ng and 200 ng of TGFbeta, respectively). Synovial lining cells produced BMP-2 and BMP-4 after TGFbeta stimulation, whereas BMP-2 and BMP-4 were absent in the synovial tissue after macrophage depletion. In vitro, clustering and spheroid formation of C3H10T1/2 was induced by TGFbeta concentrations of >1 ng/ml. However, in the Transwell system, in the presence of murine macrophages, 0.5 ng/ml of TGFbeta was very effective in generating large spheroids, suggestive of macrophage-derived (co)factors. In coculture supernatants, TGFbeta concentrations were not elevated in the presence of macrophages, indicating generation of other growth factors involved in spheroid formation. CONCLUSION: These findings indicate that macrophages are crucial intermediate factors in osteophyte formation induced by TGFbeta, probably by inducing other chondrogenic signals.


Asunto(s)
Condrocitos/citología , Macrófagos/fisiología , Membrana Sinovial/citología , Factor de Crecimiento Transformador beta/farmacología , Animales , Antimetabolitos/farmacología , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Condrocitos/efectos de los fármacos , Ácido Clodrónico/farmacología , Liposomas , Macrófagos/efectos de los fármacos , Mesodermo/citología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Osteoartritis/metabolismo , Osteoartritis/patología , Periostio/metabolismo , Periostio/patología , Células Madre/citología , Células Madre/efectos de los fármacos , Membrana Sinovial/inmunología
10.
Arthritis Rheum ; 48(10): 2949-58, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14558102

RESUMEN

OBJECTIVE: To investigate whether the soluble form of interleukin-1 (IL-1) receptor accessory protein (sIL-1RAcP), whose physiologic function remains to be established, can serve as a specific inhibitor of IL-1 signaling in vitro, and to evaluate its applicability in collagen-induced arthritis (CIA). METHODS: Soluble IL-1RAcP was cloned from murine liver complementary DNA and expressed by the use of either an adenoviral vector (AdRGD) for sIL-1RAcP or a stable-transfected NIH3T3 fibroblast cell line. The ability of affinity-purified sIL-1RAcP to inhibit IL-1 signaling was tested on NF-kappaB luciferase reporter fibroblasts and quantified by luminometer. To investigate therapeutic efficacy, sIL-1RAcP was both locally (knee joint) and systemically overexpressed in collagen-immunized male DBA/1 mice. Severity of arthritis was monitored visually, and the pathologic process in the joint was examined histologically. Serum was obtained from mice to quantify IL-6 and anti-bovine type II collagen (BCII) antibody levels. RESULTS: Incubation of the NF-kappaB reporter fibroblast with purified sIL-1RAcP protein showed a marked reduction of IL-1-induced, but not tumor necrosis factor-induced, NF-kappaB activation. This showed a novel role for sIL-1RAcP as a specific inhibitor of IL-1 signaling. Local transplantation of sIL-1RAcP-producing NIH3T3 fibroblasts into the knee before onset of CIA had little or no effect on general disease severity in these mice. Histologic evaluation of the knee joints receiving sIL-1RAcP cell transplantation showed a marked reduction in both joint inflammation and bone and cartilage erosion. Local treatment with sIL-1RAcP had no profound effect on serum levels of IL-6 and anti-BCII antibodies, which is indicative of the ongoing presence of arthritis in distal joints. In contrast to local treatment, systemic treatment with the AdRGD for sIL-1RAcP markedly ameliorated CIA in all joints. CONCLUSION: In this study we demonstrated that sIL-1RAcP is a biologically active and innovative inhibitor of IL-1, and treatment of mice with sIL-1RAcP had a profound prophylactic effect on collagen-induced arthritis.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Experimental/terapia , Interleucina-1/antagonistas & inhibidores , Proteínas/genética , Adenoviridae/genética , Animales , Artritis Experimental/patología , Clonación Molecular , Expresión Génica , Terapia Genética , Interleucina-1/metabolismo , Proteína Accesoria del Receptor de Interleucina-1 , Articulación de la Rodilla/patología , Masculino , Ratones , Ratones Endogámicos DBA , FN-kappa B/metabolismo , Células 3T3 NIH/fisiología , Células 3T3 NIH/trasplante , Proteínas/metabolismo , Transducción de Señal , Solubilidad
11.
Gene Ther ; 10(12): 1004-11, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12776157

RESUMEN

Elevated concentrations of interleukin-18 (IL-18) are found in both serum and synovial fluid of patients suffering from rheumatoid arthritis (RA) and this cytokine has recently been implicated in the development of experimental arthritis. In this present study, we developed an IL-18 neutralizing intervention and examined its efficacy for local intra-articular treatment of experimental arthritis. To this end we constructed an adenoviral vector containing the murine IL-18 binding protein isoform c gene (AdCMVIL-18BPc). The constructed adenoviral vector was validated on replication deficiency, transfection efficacy and ability to express biological functional IL-18BPc. Intra-articular overexpression of IL-18BPc significantly reduced incidence of collagen-induced arthritis (CIA) in treated kneejoints. Affected kneejoints of IL-18BPc-treated mice showed less severe arthritis, characterized by reduction of inflammation and destruction of bone and cartilage. Local intra-articular IL-1BPc treatment in both knees provided additional protection against CIA incidence and severity in distal paws. Measurement of serum levels of specific collagen type (CII) Abs revealed a moderate reduction of circulating IgG2a anti-CII Abs, while IgG1 anti-CII Abs remained at similar level. The present study underlines the involvement of IL-18 as an important proinflammatory cytokine in onset of experimental arthritis. Furthermore, it shows that endogenous IL-18 can be blocked efficiently through local adenoviral overexpression of IL-18BPc, indicating that treatment with IL-18BPc might contribute to joint protection in RA.


Asunto(s)
Artritis Experimental/terapia , Terapia Genética/métodos , Glicoproteínas/genética , Adenoviridae/genética , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Autoanticuerpos/sangre , Colágeno Tipo II/inmunología , Citocinas/biosíntesis , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Interleucina-18/metabolismo , Interleucina-6/sangre , Articulación de la Rodilla/inmunología , Masculino , Ratones , Ratones Endogámicos DBA
12.
Br J Cancer ; 87(1): 113-8, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12085266

RESUMEN

During chemo- and radiation therapy, the balance between epithelial cell proliferation, differentiation, and cell death at the villus tip is disrupted by premature death of dividing epithelial cells. This will subsequently lead to the onset of mucosal barrier injury in the whole gastrointestinal tract. Up till now there is no validated method to treat side effects occurring due to therapy. An approach to manage this side effect might be to reversibly arrest growth of epithelial stem cells during therapy using Transforming Growth Factor-beta2. A Transforming Growth Factor-beta2 enriched fraction prepared from bovine milk was shown to protect small intestinal epithelial cells against cell cycle specific chemotherapeutic agents by arresting the cells in G1-phase. Secondly, in a rat model for induced small intestinal damage, oral supplementation of rats exposed to methotrexate with the Transforming Growth Factor-beta2 enriched fraction significantly reduced the chemotherapy-associated weight loss and ileal villus atrophy by reducing cell proliferation in the normal stem cell population. Thus oral supplementation with a bovine milk fraction enriched for Transforming Growth Factor-beta2 attenuated the side effects of chemotherapy in the small intestine in rats.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Ciclo Celular/efectos de los fármacos , Inmunosupresores/farmacología , Intestino Delgado/patología , Metotrexato/efectos adversos , Factor de Crecimiento Transformador beta/farmacología , Administración Oral , Animales , Atrofia , Muerte Celular , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Intestino Delgado/efectos de los fármacos , Ratas , Factor de Crecimiento Transformador beta2 , Pérdida de Peso
13.
Biochim Biophys Acta ; 1431(2): 421-32, 1999 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-10350617

RESUMEN

The receptors for insulin (IR) and epidermal growth factor (EGFR) are members of the tyrosine kinase receptor (TKR) family. Despite homology of their cytosolic TK domains, both receptors induce different cellular responses. Tyrosine phosphorylation of insulin receptor substrate (IRS) molecules is a specific IR post-receptor response. The EGFR specifically activates phospholipase C-gamma1 (PLC-gamma1). Recruitment of substrate molecules with Src homology 2 (SH2) domains or phosphotyrosine binding (PTB) domains to phosphotyrosines in the receptor is one of the factors creating substrate specificity. In addition, it has been shown that the TK domains of the IR and EGFR show preferences to phosphorylate distinct peptides in vitro, suggesting additional mechanisms of substrate recognition. We have examined to what extent the substrate preference of the TK domain contributes to the specificity of the receptor in vivo. For this purpose we determined whether the IR TK domain, in situ, is able to tyrosine-phosphorylate substrates normally used by the EGFR. A chimaeric receptor, consisting of an EGFR in which the juxtamembrane and tyrosine kinase domains were exchanged by their IR counterparts, was expressed in CHO-09 cells lacking endogenous EGFR. This receptor was found to activate PLC-gamma1, indicating that the IR TK domain, in situ, is able to tyrosine phosphorylate substrates normally used by the EGFR. These findings suggest that the IR TK domain, in situ, has a low specificity for selection and phosphorylation of non-cognate substrates.


Asunto(s)
Calcio/metabolismo , Factor de Crecimiento Epidérmico/química , Isoenzimas/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Fosfolipasas de Tipo C/metabolismo , Animales , Células CHO , Señalización del Calcio , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cricetinae , Citosol/metabolismo , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/biosíntesis , Receptores ErbB/química , Proteína Quinasa 3 Activada por Mitógenos , Fosfolipasa C gamma , Fosforilación , Fosfotirosina/metabolismo , Receptor de Insulina/biosíntesis , Especificidad por Sustrato , Transfección
14.
Biochem J ; 337 ( Pt 2): 263-8, 1999 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9882623

RESUMEN

Receptor-mediated activation of phosphatidylcholine phosphatidohydrolase or phospholipase D (PLD) was studied in Chinese hamster ovary (CHO) cells expressing the cholecystokinin-A (CCK-A) receptor. Cells were labelled with [3H]myristic acid for 24 h and PLD-catalysed [3H]phosphatidylethanol formation was measured in the presence of 1% (v/v) ethanol. Cholecystokinin-(26-33)-peptide amide (CCK8) increased PLD activity both time- and dose-dependently. Maximal activation of protein kinase C (PKC) with 1 microM PMA or sustained elevation of the cytosolic free Ca2+ concentration ([Ca2+]i) with 1 microM thapsigargin increased PLD activity to 50% and 70% of the maximal value obtained with CCK8 respectively. The stimulatory effects of CCK8, PMA and thapsigargin were abolished in cells in which PKC was downregulated or inhibited by chelerythrine. PMA/Ca2+-stimulated PLD activity was absent in a homogenate of PKC-downregulated cells but could be restored upon addition of purified rat brain PKC. CCK8-induced PLD activation was inhibited by 90% in the absence of external Ca2+, demonstrating that receptor-mediated activation of PKC in itself does not significantly add to PLD activation but requires a sustained increase in [Ca2+]i. Taken together, the results presented demonstrate that, in CHO-CCK-A cells, receptor-mediated PLD activation is completely dependent on PKC, but that the extent to which PLD becomes activated depends largely, if not entirely, on the magnitude and duration of the agonist-induced increase in [Ca2+]i.


Asunto(s)
Calcio/metabolismo , Citosol/metabolismo , Fosfolipasa D/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Colecistoquinina/metabolismo , Alcaloides , Animales , Benzofenantridinas , Encéfalo/enzimología , Células CHO , Cricetinae , Regulación hacia Abajo , Activación Enzimática , Glicerofosfolípidos/metabolismo , Fenantridinas/farmacología , Proteína Quinasa C/aislamiento & purificación , Inhibidores de Proteínas Quinasas , Ratas , Receptor de Colecistoquinina A , Receptores de Colecistoquinina/genética , Proteínas Recombinantes/metabolismo , Sincalida/farmacología , Acetato de Tetradecanoilforbol/farmacología , Tapsigargina/farmacología
15.
Br J Pharmacol ; 124(5): 935-45, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9692779

RESUMEN

1. Many G protein-coupled receptors contain potential phosphorylation sites for protein kinase C (PKC), the exact role of which is poorly understood. In the present study, a mutant cholecystokininA (CCK(A)) receptor was generated in which the four consensus sites for PKC action were changed in an alanine. Both the wild-type (CCK(A)WT) and mutant (CCK(A)MT) receptor were stably expressed in Chinese hamster ovary (CHO) cells. 2. Binding of [3H]-cholecystokinin-(26-33)-peptide amide (CCK-8) to membranes prepared from CHO-CCK(A)WT cells and CHO-CCK(A)MT cells revealed no difference in binding affinity (Kd values of 0.72 nM and 0.86 nM CCK-8, respectively). 3. The dose-response curves for CCK-8-induced cyclic AMP accumulation and inositol 1,4,5-trisphosphate (Ins(1,4,5)P3) formation were shifted to the left in CHO-CCK(A)MT cells. This leftward shift was mimicked by the potent inhibitor of protein kinase activity, staurosporine. However, the effect of staurosporine was restricted to CHO-CCK(A)WT cells. This demonstrates that attenuation of CCK-8-induced activation of adenylyl cyclase and phospholipase C-beta involves a staurosporine-sensitive kinase, which acts directly at the potential sites of PKC action on the CCK(A) receptor in CCK-8-stimulated CHO-CCK(A)WT cells. 4. The potent PKC activator, 12-O-tetradecanoylphorbol 13-acetate (TPA), evoked a rightward shift of the dose-response curve for CCK-8-induced cyclic AMP accumulation in CHO-CCK(A)WT cells but not CHO-CCK(A)MT cells. This is in agreement with the idea that PKC acts directly at the CCK(A) receptor to attenuate adenylyl cyclase activation. 5. In contrast, TPA evoked a rightward shift of the dose-response curve for CCK-8-induced Ins(1,4,5)P3 formation in both cell lines. This demonstrates that high-level PKC activation inhibits CCK-8-induced Ins(1,4,5)P3 formation also at a post-receptor site. 6. TPA inhibition of agonist-induced Ca2+ mobilization was only partly reversed in CHO-CCK(A)MT cells. TPA also inhibited Ca2+ mobilization in response to the G protein activator, Mas-7. These findings are in agreement with the idea that partial reversal of agonist-induced Ca2+ mobilization is due to the presence of an additional site of PKC inhibition downstream of the receptor and that the mutant receptor itself is not inhibited by the action of PKC. 7. The data presented demonstrate that the predicted sites for PKC action on the CCK(A) receptor are the only sites involved in TPA-induced uncoupling of the receptor from its G proteins. In addition, the present study unveils a post-receptor site of PKC action, the physiological relevance of which may be that it provides a means for the cell to inhibit phospholipase C-beta activation by receptors that are not phosphorylated by PKC.


Asunto(s)
Mutación , Proteína Quinasa C/metabolismo , Receptores de Colecistoquinina/genética , Secuencia de Aminoácidos , Animales , Células CHO , Calcio/antagonistas & inhibidores , Cricetinae , AMP Cíclico/antagonistas & inhibidores , Activación Enzimática , Proteínas de Unión al GTP/metabolismo , Inositol 1,4,5-Trifosfato/antagonistas & inhibidores , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Ratas , Receptor de Colecistoquinina A , Receptores de Colecistoquinina/biosíntesis , Receptores de Colecistoquinina/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sincalida/metabolismo , Sincalida/farmacología , Acetato de Tetradecanoilforbol/farmacología
16.
Eur J Pharmacol ; 346(2-3): 345-51, 1998 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-9652379

RESUMEN

The aminosteroid 1-(6-¿[17beta-3-methoxyestra- 1,3,5(10)-trien- 17-yl]-amino¿hexyl)- 1H-pyrrole-2,5-dione (U73122) and its inactive analogue 1-(6-¿[17beta-3-methoxyestra-1,3,5(10)-trien- 17-yl]-amino¿hexyl-2,5-pyrrolidine-dione (U73343) are widely used to study the involvement of G protein-coupled 1-phosphatidylinositol-phosphodiesterase, or phospholipase C, in receptor-mediated cell activation. The present work shows that both aminosteroids inhibit cholecystokinin-(26-33)-peptide amide (CCK-8)-induced phospholipase D activation equipotently in Chinese hamster ovary cells expressing the cholecystokinin-A receptor (CHO-CCK(A) cells). In addition, the two aminosteroids virtually completely inhibited thapsigargin- and 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced phospholipase D activation. Since the latter two drugs mimic inositol 1,4,5-trisphosphate-mediated Ca2+ mobilisation and 1,2-diacylglycerol-mediated protein kinase C activation. respectively, this suggests that both U73122 and U73343 act downstream of phospholipase C to inhibit receptor-mediated phospholipase D activation. U73122, but not U73343. effectively inhibited both TPA/Ca2+-stimulated phospholipase D activation and TPA/phosphatidylserine-stimulated protein kinase C activation in a homogenate of CHO-CCK(A) cells. The data presented suggest that U73122 may act at the level of protein kinase C to inhibit activation of phospholipase D. The exact site of action of U73343 is presently unknown.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Fosfolipasa D/antagonistas & inhibidores , Pirrolidinonas/farmacología , Fosfolipasas de Tipo C/metabolismo , Animales , Células CHO , Cricetinae , Activación Enzimática/efectos de los fármacos , Fosfolipasa D/metabolismo , Fosfolípidos/biosíntesis , Fosfolípidos/aislamiento & purificación , Receptores de Colecistoquinina/antagonistas & inhibidores , Receptores de Colecistoquinina/metabolismo , Espectrometría de Fluorescencia , Fosfolipasas de Tipo C/antagonistas & inhibidores
17.
Br J Pharmacol ; 123(6): 1189-97, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9559904

RESUMEN

1. The rat CCK(A) and CCK(B) receptors were stably expressed in Chinese hamster ovary (CHO-09) cells in order to compare modes of signal transduction and effects of protein kinase C (PKC) thereupon. 2. Spectrofluorophotometry of Fura-2-loaded cells revealed that both receptors retained their pharmacological characteristics following expression in CHO cells. Sulphated cholecystokinin-(26-33)-peptide amide (CCK-8-S) increased the cytosolic Ca2+ concentration ([Ca2+]i) in CCK(A) cells, measured as an increase in Fura-2 fluorescence emission ratio, 1000 fold more potently than its non-sulphated form (CCK-8-NS) (EC50 values of 0.19 nM and 0.18 microM, respectively). By contrast, CCK-8-S and CCK-8-NS were equally potent in CCK(B) cells (EC50 values of 0.86 nM and 1.18 nM, respectively). The CCK(A) receptor agonist JMV-180 increased [Ca2+]i only in CCK(A) cells. Likewise, pentagastrin increased [Ca2+]i only in CCK(B) cells. Finally, CCK-8-S-induced Ca2+ signalling through the CCK(A) receptor was most potently inhibited by the CCK(A) receptor antagonist L364,718, whereas the CCK(B) receptor antagonist L365,260 was more potent in CCK(B) cells. 3. Receptor-mediated activation of adenylyl cyclase was measured in the presence of the inhibitor of cyclic nucleotide phosphodiesterase activity, 3-isobutyl-1-methylxanthine. CCK-8-S and, to a lesser extent, CCK-8-NS, but not JMV-180 or pentagastrin, stimulated the accumulation of cyclicAMP in CCK(A) cells. By contrast, none of these agonists increased cyclicAMP in CCK(B) cells. 4. Short-term (3 min) pretreatment with the PKC activator 12-O-tetradecanoylphorbol 13-acetate (TPA) evoked a rightward shift of the dose-response curve for the Ca2+ mobilizing effect of CCK-8-S in both cell lines. In addition, short-term TPA pretreatment markedly reduced CCK-8-S-induced cyclicAMP accumulation in CCK(A) cells. In both cases, the inhibitory effect of TPA was abolished by the PKC inhibitors, GF-109203X and staurosporine, whereas no inhibition was observed with the inactive phorbol ester, 4-alpha-phorbol 12-myristate 13-acetate. 5. During prolonged TPA treatment, the cells gradually recovered from phorbol ester inhibition and in the case of CCK-8-S-induced Ca2+ mobilization complete recovery was achieved after 24 h of TPA treatment. Western blot analysis revealed that this recovery was paralleled by down-regulation of PKC-alpha, suggesting the involvement of this PKC isotype in the inhibitory action of TPA. 6. This study demonstrates that following expression in CHO cells (i) both CCK(A) and CCK(B) receptors are coupled to Ca2+ mobilization, (ii) only CCK(A) receptors are coupled to cyclicAMP formation and (iii) with both receptors signalling is inhibited by PKC.


Asunto(s)
Proteína Quinasa C/metabolismo , Receptores de Colecistoquinina/fisiología , Transducción de Señal , Animales , Células CHO , Calcio/metabolismo , Cricetinae , AMP Cíclico/biosíntesis , Regulación hacia Abajo , Ratas , Receptor de Colecistoquinina A , Receptor de Colecistoquinina B , Receptores de Colecistoquinina/metabolismo , Proteínas Recombinantes/metabolismo , Sincalida/metabolismo , Espectrometría de Fluorescencia , Acetato de Tetradecanoilforbol/farmacología , Tritio
18.
Pflugers Arch ; 435(3): 422-8, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9426300

RESUMEN

Receptor phosphorylation in response to agonist stimulation is a key regulatory principle in signal transduction. Previous work has suggested the concerted action of protein kinase C (PKC) and a staurosporine-insensitive receptor kinase in homologous phosphorylation of the cholecystokinin (CCK) receptor in freshly isolated rat pancreatic acinar cells [Gates, Ulrich, Miller (1993) Am J Physiol 264:G840-G847]. The present study shows that down-regulation of PKC by prolonged (2 h) treatment with 0.1 muM 12-O-tetradecanoylphorbol-13-acetate (TPA) markedly reduced basal CCK receptor phosphorylation as well as that induced by TPA (0.1 muM) and cholecystokinin-(26-33)-peptide amide (CCK8, 0.1 muM). The phosphorylation level reached was the same with both stimulants and equalled basal phosphorylation in untreated control cells. The absence of any CCK8-stimulated phosphorylation reflecting the activity of a putative staurosporine-insensitive receptor kinase raises the intriguing possibility that a basal level of PKC-mediated receptor phosphorylation is required for the action of such a receptor kinase. Immunoblot analysis revealed that the decrease in receptor phosphorylation coincided with a marked reduction of PKC-alpha and, to a lesser extent, PKC-epsilon. In addition, TPA-induced inhibition of the increase in cytosolic free Ca2+ concentration ([Ca2+]i) evoked by the high-affinity CCK receptor agonist JMV-180 was completely reversed. The time-course of recovery closely matched that of the reduction of PKC-alpha. Finally, digital imaging microscopy of individual PKC down-regulated cells revealed a marked increase in the duration of JMV-180-evoked oscillatory changes in [Ca2+]i. Taken together, the present findings are in agreement with the idea that PKC-alpha-mediated receptor phosphorylation leads to a shortening of the duration of the [Ca2+]i oscillations and eventually to inhibition of high-affinity Ca2+ signalling through the native CCK receptor in pancreatic acinar cells.


Asunto(s)
Isoenzimas/metabolismo , Páncreas/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Colecistoquinina/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes , Fura-2 , Immunoblotting , Cinética , Fosforilación , Ratas , Sincalida/análogos & derivados , Sincalida/farmacología , Estaurosporina/farmacología , Acetato de Tetradecanoilforbol/farmacología
19.
EMBO J ; 16(12): 3519-32, 1997 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-9218794

RESUMEN

Digital-imaging microscopy was performed to study the effect of Coxsackie B3 virus infection on the cytosolic free Ca2+ concentration and the Ca2+ content of the endoplasmic reticulum (ER). During the course of infection a gradual increase in the cytosolic free Ca2+ concentration was observed, due to the influx of extracellular Ca2+. The Ca2+ content of the ER decreased in time with kinetics inversely proportional to those of viral protein synthesis. Individual expression of protein 2B was sufficient to induce the influx of extracellular Ca2+ and to release Ca2+ from ER stores. Analysis of mutant 2B proteins showed that both a cationic amphipathic alpha-helix and a second hydrophobic domain in 2B were required for these activities. Consistent with a presumed ability of protein 2B to increase membrane permeability, viruses carrying a mutant 2B protein exhibited a defect in virus release. We propose that 2B gradually enhances membrane permeability, thereby disrupting the intracellular Ca2+ homeostasis and ultimately causing the membrane lesions that allow release of virus progeny.


Asunto(s)
Membrana Celular/virología , Retículo Endoplásmico/virología , Enterovirus Humano B/metabolismo , Proteínas Virales/metabolismo , Animales , Células COS , Calcio/metabolismo , Cationes Bivalentes , Permeabilidad de la Membrana Celular , Retículo Endoplásmico/metabolismo , Enterovirus Humano B/fisiología , Expresión Génica , Células HeLa , Humanos , Membranas Intracelulares/virología , Estructura Secundaria de Proteína , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral
20.
Eur J Pharmacol ; 325(1): 93-9, 1997 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-9151944

RESUMEN

Recently a molecular model was proposed for the binding site of the antagonist 3S(-)-N-(2,3-dihydro-1-methyl-2-oxo-5-phenyl-1H-1,4-benzodiazepine-3-yl) -1H-indole-2-carboxamide (devazepide) on the cholecystokinin-A (CCK(A)) receptor (Van der Bent et al., 1994. Drug Design Discov. 12, 129-148). Fifteen amino acids were identified, including hydrophilic ones such as Ser139, Asn349 and Ser379, that might interact with the carboxamide moiety in devazepide. To provide mutational evidence for this model, wild-type and mutant receptors (S139A, N349A and S379A) were transiently expressed and compared with respect to the ability of devazepide to inhibit binding of radiolabelled cholecystokinin-(26-33)-peptide amide (CCK-8) and CCK-8-evoked Ca2+ mobilization. The data presented suggest the involvement of the three residues in antagonist binding, although to a different extent. However, it does not seem likely that hydrogen bonds are the driving force in view of the relatively minor changes in receptor affinity and activity.


Asunto(s)
Benzodiazepinonas/metabolismo , Antagonistas de Hormonas/metabolismo , Receptores de Colecistoquinina/genética , Receptores de Colecistoquinina/metabolismo , Animales , Benzodiazepinonas/farmacología , Sitios de Unión , Células CHO/metabolismo , Células CHO/ultraestructura , Calcio/metabolismo , Cricetinae , Análisis Mutacional de ADN , ADN Complementario/genética , Devazepida , Antagonistas de Hormonas/farmacología , Enlace de Hidrógeno , Radioisótopos de Yodo , Cinética , Mutagénesis , Receptor de Colecistoquinina A , Receptores de Colecistoquinina/antagonistas & inhibidores , Sincalida/metabolismo , Sincalida/farmacología , Especificidad por Sustrato , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA