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












Base de datos
Intervalo de año de publicación
1.
Am J Emerg Med ; 47: 347.e1-347.e3, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33745773

RESUMEN

Blunt cerebrovascular injury is a very rare complication of blunt trauma and a diagnostic challenge. A 14 year old male fell 10 m sustaining multi system trauma. The atypical Glasgow Coma Score was six with a fully preserved eye component. Initial whole-body CT scanning demonstrated multiple injuries but no obvious brain injury. Trauma management involved non-operative resuscitation and was successful, however profound coma occurred and brain stem reflexes disappeared on day two. Repeat brain CT scan demonstrated multiple cerebral and cerebellar ischemic lesions and no opacification of the vertebral or basilar arteries. Secondary analysis of the first CT scan demonstrated a small focal basilar artery dissection not initially reported. Our case report highlights an unusual cause of coma after traumatic brain injury where the clinical scenario mimics locked in syndrome. In such circumstances cerebrovascular injury, and in particular traumatic basilar artery dissection, must be actively excluded.


Asunto(s)
Arteria Basilar/lesiones , Heridas no Penetrantes/complicaciones , Adolescente , Disección Aórtica/diagnóstico , Arteria Basilar/diagnóstico por imagen , Arteria Basilar/patología , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/etiología , Escala de Coma de Glasgow , Humanos , Masculino
2.
Br J Anaesth ; 120(6): 1158-1164, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29793582

RESUMEN

BACKGROUND: The significance of cardiac troponin I (cTnI) elevation after trauma is debated. We therefore explored the association between cTnI elevation at admission after trauma and ICU mortality. METHODS: We performed a retrospective analysis from a prospectively constituted database, of patients admitted to ICU after trauma at a single centre, over a 36 month period. According to cTnI plasma concentration at admission, patients were categorised into three groups: normal (<0.05 ng ml-1), intermediate (0.05-0.99 ng ml-1), or high concentration (≥1.0 ng ml-1). Associations of pre-hospital conditions or cTnI elevation and mortality were analysed by multivariate logistic regression. RESULTS: Among the 994 patients, 177 (18%) had cTnI elevation at ICU admission. Of this total, 114 (11%) patients died in the ICU. The cTnI release was an independent predictor of ICU mortality with a concentration-response relationship [odds ratio (OR) 4.90 (2.19-11.16) and 14.83 (4.68-49.90) for intermediate and high concentrations, respectively] and Day 2 mortality [OR 2.23 (1.18-5.80) and 7.49 (2.77-20.12) for intermediate and high concentrations, respectively]. The severity of thoracic trauma [OR 2.25 (1.07-4.55) and 3.23 (2.00-5.27) for Abbreviated Injury Scale scores 1-2 and ≥3, respectively], out-of-hospital maximal heart rate ≥120 beats min-1 [OR 2.22 (1.32-3.69)], and out-of-hospital shock [OR 2.02 (1.20-3.38)] were independently associated with cTnI elevation. CONCLUSIONS: Release of cTnI was an independent predictor of ICU mortality, suggesting that this biomarker can be used in daily practice for early stratification of the risk of ICU death. Thoracic trauma was strongly associated with cTnI elevation.


Asunto(s)
Troponina I/sangre , Heridas y Lesiones/diagnóstico , Adulto , Biomarcadores/sangre , Bases de Datos Factuales , Femenino , Francia/epidemiología , Mortalidad Hospitalaria , Humanos , Unidades de Cuidados Intensivos/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Heridas y Lesiones/sangre , Adulto Joven
3.
Br J Anaesth ; 116(6): 847-54, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27199316

RESUMEN

BACKGROUND: We reviewed our experience with tracheal extubation in the operating room (E-OR) among cystic fibrosis patients requiring bilateral lung transplantation to evaluate safety and determine predictive factors of E-OR. METHODS: The charts of 89 recipients (from May 2007 to June 2013) were analysed. Patients were divided into E-OR and E-ICU (intensive care unit extubation) groups. Data are expressed as numbers (percentages) or medians [25th-75th percentiles]. RESULTS: There were 41 patients in the E-OR group (46%). Donor and recipient characteristics were similar between groups. Intraoperative complications occurred less frequently in the E-OR group, and fluid and transfusion requirements were lower. Postoperative courses were different in the E-OR group, including a lower rate of grade 3 primary graft dysfunction (0 compared with 19 patients, P<0.0001) and shorter ICU (5.0 [3.7-7.2] compared with 11.5 [7.0-15.5] days) and hospital stays (22.0 [18.0-25.5] compared with 33.0 [25.0-56.5] days, respectively; P<0.0001 for both). The 1 yr survival rates were similar: 95% in the E-OR group and 98% in the E-ICU group. A statistical model built on a development cohort of 60 randomly selected patients predicted 95% of E-OR instances in this cohort and 82% of E-OR instances in the validation cohort (28 patients). Predictive factors were complications during single-lung ventilation (second graft implantation), complications during bipulmonary ventilation (end of surgery), and the ratio of arterial partial pressure of oxygen to fractional inspired oxygen (end of surgery). CONCLUSIONS: Our protocol allowed for extubation of 46% of bilateral lung transplant patients without increased postoperative risks.


Asunto(s)
Extubación Traqueal/métodos , Trasplante de Pulmón/métodos , Adolescente , Adulto , Anciano , Presión Arterial , Transfusión Sanguínea/estadística & datos numéricos , Estudios de Cohortes , Cuidados Críticos , Fibrosis Quística/cirugía , Femenino , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Ventilación Unipulmonar , Quirófanos , Oxígeno/sangre , Complicaciones Posoperatorias/epidemiología , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
5.
Cancer Res ; 61(24): 8683-8, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11751385

RESUMEN

The N-nitroso-N-methylurea-induced rat mammary tumor model was used to conduct two types of studies: a prevention study designed to test the ability of the novel selective estrogen receptor modulator lasofoxifene (LAS) to inhibit the development of mammary tumors, and a treatment study designed to test the inhibitory effect of LAS on the growth of established tumors. The prevention study indicated that LAS markedly delayed the emergence of N-nitroso-N-methylurea-induced tumors to an extent similar to that obtained by the established antiestrogen tamoxifen (TAM). At the highest dose administered, both TAM and LAS reduced tumor incidence by 75% and total tumor number by 90% relative to the controls. LAS also reduced the multiplicity of tumors, i.e., the mean number of tumors per rat, and resulted in substantially smaller total tumor burden. In the treatment study, LAS significantly inhibited tumor growth compared with the controls. In addition, whereas none of the untreated tumors regressed completely over the experimental period, 40% of LAS-treated tumors regressed by >50% at the highest dose (10 mg/kg daily). The results of this study in a rat mammary tumor model indicate that LAS has both chemopreventive and chemotherapeutic effects quantitatively comparable with those of TAM.


Asunto(s)
Anticarcinógenos/farmacología , Antineoplásicos/farmacología , Neoplasias Mamarias Experimentales/prevención & control , Pirrolidinas/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tetrahidronaftalenos/farmacología , Animales , Anticarcinógenos/sangre , Antineoplásicos/sangre , Carcinógenos , Antagonistas de Estrógenos/farmacología , Femenino , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Metilnitrosourea , Pirrolidinas/sangre , Ratas , Ratas Sprague-Dawley , Moduladores Selectivos de los Receptores de Estrógeno/sangre , Tamoxifeno/farmacología , Tetrahidronaftalenos/sangre , Aumento de Peso/efectos de los fármacos
6.
J Pharmacol Exp Ther ; 291(2): 739-48, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10525095

RESUMEN

Phosphorylation of tyrosine residues on the epidermal growth factor (EGF) receptor (EGFr) is an important early event in signal transduction, leading to cell replication for major human carcinomas. CP-358,774 is a potent and selective inhibitor of the EGFr tyrosine kinase and produces selective inhibition of EGF-mediated tumor cell mitogenesis. To assess the pharmacodynamic aspects of EGFr inhibition, we devised an ex vivo enzyme-linked immunosorbent assay for quantification of EGFr-specific tyrosine phosphorylation in human tumor tissue specimens obtained from xenografts growing s.c. in athymic mice. When coupled with pharmacokinetic analyses, this measurement can be used to describe the extent and duration of kinase inhibition in vivo. CP-358,774 is an effective, orally active inhibitor of EGFr-specific tyrosine phosphorylation (ED(50) = 10 mg/kg, single dose). It has a significant duration of action, producing, on average, a 70% reduction in EGFr-associated phosphotyrosine over a 24-h period after a single 100 mg/kg dose. Inhibition of EGFr phosphotyrosine in an ex vivo assay format effectively estimates the potency and degree of inhibition of EGFr-dependent human LICR-LON-HN5 head and neck carcinoma tumor growth. Substantial growth inhibition of human tumor xenografts was achieved with p.o. doses of the compound (ED(50) = 10 mg/kg q.d. for 20 days). Combination chemotherapy with cisplatin produced a significant response above that of cisplatin alone with no detectable effects on body weight or lethal toxicity. Taken together, these observations suggest that CP-358,774 may be useful for the treatment of EGFr-driven human carcinomas.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/fisiología , Quinazolinas/farmacología , Tirosina/metabolismo , Animales , Peso Corporal/efectos de los fármacos , División Celular/efectos de los fármacos , Cisplatino/toxicidad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Clorhidrato de Erlotinib , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Fosforilación , Fosfotirosina/metabolismo , Polifarmacia , Quinazolinas/sangre , Factores de Tiempo , Trasplante Heterólogo/fisiología , Células Tumorales Cultivadas
7.
EMBO J ; 17(20): 5948-63, 1998 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9774339

RESUMEN

Virulence of poxviruses, the causative agents of smallpox, depends on virus-encoded growth factors related to the mammalian epidermal growth factor (EGF). Here we report that the growth factors of Shope fibroma virus, Myxoma virus and vaccinia virus (SFGF, MGF and VGF) display unique patterns of specificity to ErbB receptor tyrosine kinases; whereas SFGF is a broad-specificity ligand, VGF binds primarily to ErbB-1 homodimers, and the exclusive receptor for MGF is a heterodimer comprised of ErbB-2 and ErbB-3. In spite of 10- to 1000-fold lower binding affinity to their respective receptors, the viral ligands are mitogenically equivalent or even more potent than their mammalian counterparts. This remarkable enhancement of cell growth is due to attenuation of receptor degradation and ubiquitination, which leads to sustained signal transduction. Our results imply that signal potentiation and precise targeting to specific receptor combinations contribute to cell transformation at sites of poxvirus infection, and they underscore the importance of the often ignored low-affinity ligand-receptor interactions.


Asunto(s)
Poxviridae/patogenicidad , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Dimerización , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-2/fisiología , Receptor ErbB-3 , Receptor ErbB-4
8.
Mol Cell Biol ; 18(10): 6090-101, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9742126

RESUMEN

The recently isolated second family of neuregulins, NRG2, shares its primary receptors, ErbB-3 and ErbB-4, and induction of mammary cell differentiation with NRG1 isoforms, suggesting functional redundancy of the two growth factor families. To address this possibility, we analyzed receptor specificity of NRGs by using an engineered cellular system. The activity of isoform-specific but partly overlapping patterns of specificities that collectively activate all eight ligand-stimulatable ErbB dimers was revealed. Specifically, NRG2-alpha [corrected], like NRG1-beta [corrected], emerges as a narrow-specificity ligand, whereas NRG2-beta [corrected] is a pan-ErbB ligand that binds with different affinities to all receptor combinations, including those containing ErbB-1, but excluding homodimers of ErbB-2. The latter protein, however, displayed cooperativity with the direct NRG receptors. Apparently, signaling by all NRGs is funneled through the mitogen-activated protein kinase (MAPK). However, the duration and potency of MAPK activation depend on the identity of the stimulatory ligand-receptor ternary complex. We conclude that the NRG-ErbB network represents a complex and nonredundant machinery developed for fine-tuning of signal transduction.


Asunto(s)
Receptores ErbB/metabolismo , Glicoproteínas/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Activación Enzimática , Receptores ErbB/biosíntesis , Glicoproteínas/farmacología , Isomerismo , Ligandos , Factores de Crecimiento Nervioso/farmacología , Neurregulinas , Fosforilación , Proteínas Proto-Oncogénicas/biosíntesis , Receptor ErbB-2/biosíntesis , Receptor ErbB-3 , Receptor ErbB-4
9.
Cancer Res ; 57(21): 4838-48, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9354447

RESUMEN

The epidermal growth factor receptor (EGFR) is overexpressed in a significant percentage of carcinomas and contributes to the malignant phenotype. CP-358,774 is a directly acting inhibitor of human EGFR tyrosine kinase with an IC50 of 2 nM and reduces EGFR autophosphorylation in intact tumor cells with an IC50 of 20 nM. This inhibition is selective for EGFR tyrosine kinase relative to other tyrosine kinases we have examined, both in assays of isolated kinases and whole cells. At doses of 100 mg/kg, CP-358,774 completely prevents EGF-induced autophosphorylation of EGFR in human HN5 tumors growing as xenografts in athymic mice and of the hepatic EGFR of the treated mice. CP-358,774 inhibits the proliferation of DiFi human colon tumor cells at submicromolar concentrations in cell culture and blocks cell cycle progression at the G1 phase. This inhibitor produces a marked accumulation of retinoblastoma protein in its underphosphorylated form and accumulation of p27KIP1 in DiFi cells, which may contribute to the cell cycle block. Inhibition of the EGFR also triggers apoptosis in these cells as determined by formation of DNA fragments and other criteria. These results indicate that CP-358,774 has potential for the treatment of tumors that are dependent on the EGFR pathway for proliferation or survival.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/genética , Ciclo Celular/genética , División Celular/efectos de los fármacos , Fragmentación del ADN , ADN de Neoplasias/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas
10.
EMBO J ; 16(16): 4938-50, 1997 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9305636

RESUMEN

Signaling by epidermal growth factor (EGF)-like ligands is mediated by an interactive network of four ErbB receptor tyrosine kinases, whose mechanism of ligand-induced dimerization is unknown. We contrasted two existing models: a conformation-driven activation of a receptor-intrinsic dimerization site and a ligand bivalence model. Analysis of a Neu differentiation factor (NDF)-induced heterodimer between ErbB-3 and ErbB-2 favors a bivalence model; the ligand simultaneously binds both ErbB-3 and ErbB-2, but, due to low-affinity of the second binding event, ligand bivalence drives dimerization only when the receptors are membrane anchored. Results obtained with a chimera and isoforms of NDF/neuregulin predict that each terminus of the ligand molecule contains a distinct binding site. The C-terminal low-affinity site has broad specificity, but it prefers interaction with ErbB-2, an oncogenic protein acting as a promiscuous low-affinity subunit of the three primary receptors. Thus, ligand bivalence enables signal diversification through selective recruitment of homo- and heterodimers of ErbB receptors, and it may explain oncogenicity of erbB-2/HER2.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Glicoproteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Animales , Sitios de Unión , Células CHO , Cricetinae , Dimerización , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/química , Receptores ErbB/genética , Glicoproteínas/química , Humanos , Cinética , Ligandos , Modelos Moleculares , Mutagénesis , Neurregulinas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Receptor ErbB-2/química , Receptor ErbB-3 , Proteínas Recombinantes/metabolismo
11.
J Med Chem ; 38(19): 3806-12, 1995 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-7562911

RESUMEN

The erbB-2 oncogene encodes a transmembrane protein tyrosine kinase which plays a pivotal role in signal transduction and has been implicated when overexpressed in breast, ovarian, and gastric cancers. Naturally occurring benzoquinoid ansamycin antibiotics herbimycin A, geldanamycin (GDM), and dihydrogeldanamycin were found to potently deplete p185, the erbB-2 oncoprotein, in human breast cancer SKBR-3 cells in culture. Chemistry efforts to modify selectively the quinoid moiety of GDM afforded derivatives with greater potency in vitro and in vivo. Analogs demonstrated inhibition of p185 phosphotyrosine in cell culture and in vivo after systemic drug administration to nu/nu nude mice bearing Fisher rat embryo cells transfected with human erbB-2 (FRE/erbB-2). Specifically, dosed intraperitoneally at 100 mg/kg, 17-(allylamino)-17-demethoxygeldanamycin and other 17-amino analogs were effective at reducing p185 phosphotyrosine in subcutaneous flank FRE/erbB-2 tumors. Modifications to the 17-19-positions of the quinone ring revealed a broad structure-activity relationship in vitro.


Asunto(s)
Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacología , Quinonas/química , Quinonas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antibióticos Antineoplásicos/metabolismo , Benzoquinonas , Neoplasias de la Mama/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Lactamas Macrocíclicas , Ratones , Ratones Desnudos , Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinonas/metabolismo , Ratas , Rifabutina/análogos & derivados , Relación Estructura-Actividad , Transfección , Células Tumorales Cultivadas
12.
J Biol Chem ; 270(16): 9585-9, 1995 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-7721889

RESUMEN

Heregulin is a ligand for the erbB3 and erbB4 receptors, with a region of high homology to epidermal growth factor (EGF). Despite this homology, these ligands bind to their corresponding receptors with great specificity. We report here the synthesis of heregulin beta 177-241 and show that a region consisting of amino acids 177-226 is sufficient both for binding and stimulation of receptor phosphorylation. Studies of chimeric EGF/heregulin peptides revealed that amino acids 177-181 of heregulin provide the specificity for binding to the heregulin receptor. The substitution of amino acids 177-181 of heregulin for the N terminus of EGF produced a unique bifunctional agonist that binds with high affinity to both the EGF receptor and the heregulin receptor.


Asunto(s)
Proteínas Portadoras/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Glicoproteínas/metabolismo , Neurregulina-1 , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/química , Factor de Crecimiento Epidérmico/química , Glicoproteínas/química , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Receptor ErbB-3 , Sensibilidad y Especificidad , Relación Estructura-Actividad
13.
Biochem Pharmacol ; 49(2): 203-7, 1995 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-7840797

RESUMEN

Cyclopentenyl uracil, a non-cytotoxic inhibitor of uridine kinase, was found to effectively block the salvage of circulating uridine by host and tumor tissues in the intact mouse. Dose-response characteristics of the inhibition were determined. Large doses (1 g/kg) of cyclopentenyl uracil were required, and the effect of a single dose fell rapidly over a 24-hr period. A sustained inhibition of uridine salvage of > 64-79% could be maintained by multiple doses of 1 g/kg given on an every 8-hr schedule. Mice given cyclopentenyl uracil (1 g/kg) every 8 hr for 5 days continued to gain weight and showed no signs of toxicity; however, the combination of cyclopentenyl uracil with a non-toxic dose of N-(phosphonacetyl)-L-aspartic acid (PALA; 200 mg/kg daily for 5 days) was lethal to mice, indicating that circulating uridine modifies the toxicity of agents that act on enzymes of the de novo pyrimidine pathway. Although the duration of action and potency of cyclopentenyl uracil are not ideal, this is the first demonstration of an effective inhibition of uridine salvage in the intact mouse with a non-cytotoxic agent. This makes possible the evaluation of concurrent inhibition of de novo and salvage routes to pyrimidine nucleotides as an approach to chemotherapy.


Asunto(s)
Uracilo/análogos & derivados , Uridina Quinasa/antagonistas & inhibidores , Uridina/metabolismo , Animales , Leucemia L1210/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Nucleótidos de Pirimidina/biosíntesis , Uracilo/farmacología , Nucleótidos de Uracilo/metabolismo
14.
Biochem Biophys Res Commun ; 201(3): 1313-9, 1994 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-7912926

RESUMEN

Several benzoquinoid ansamycins, e.g., herbimycin A and geldanamycin, have been widely used as inhibitors of tyrosine kinases. We recently reported that exposure to herbimycin A and several analogs depletes the erbB2 gene product p185 in human breast cancer cells. In order to explore the mechanism of this specific degradation of p185, a biologically active ansamycin incorporating a photoaffinity label was synthesized. This compound, CP202509, specifically bound to a 100 kD protein (p100) in intact SKBr3 cells and in fibroblasts transfected with the c-erbB2 or v-src oncogenes. Binding of other ansamycin analogs to p100, as measured indirectly by their ability to inhibit CP202509 binding, correlated with their ability to lower p185 protein and phosphotyrosine in SKBr3 cells. These results suggest that the ansamycins may deplete tyrosine kinases through binding to this protein.


Asunto(s)
Antibacterianos/farmacología , Receptores ErbB/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Quinonas/farmacología , Marcadores de Afinidad , Antibacterianos/metabolismo , Benzoquinonas , Humanos , Técnicas In Vitro , Lactamas Macrocíclicas , Sustancias Macromoleculares , Peso Molecular , Unión Proteica , Quinonas/metabolismo , Receptor ErbB-2 , Rifabutina/análogos & derivados , Células Tumorales Cultivadas
15.
Cancer Res ; 54(10): 2724-30, 1994 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-7909494

RESUMEN

Herbimycin A, a benzoquinoid ansamycin, is widely used as an inhibitor of tyrosine kinases. We have examined the effects of herbimycin A and several analogues on p185, the tyrosine kinase encoded by the erbB2 gene in human breast cancer cells. Exposure to 0.35 microM herbimycin A reduced tyrosine phosphorylation of p185 in SKBr3 cells by 80% after 2 h, and the p185 protein level was reduced by 90% after 6 h. The reduction of p185 resulted primarily from increased degradation of p185; cellular protein synthesis was reduced only 16% in SKBr3 cells treated with herbimycin A, RNA synthesis was inhibited only 10%, and erbB2 mRNA levels were unchanged. Examination of the major cellular glycoproteins indicated that most glycoproteins were unaffected under conditions that substantially depleted p185. Studies with cell lines transfected with erbB2 containing defined deletions indicated that susceptibility to the depletion of p185 by herbimycin and its analogues required the domain encoded by amino acids 751-971. The benzoquinoid ansamycins therefore initiate a process of specific degradation of tyrosine kinases by a mechanism that remains unknown.


Asunto(s)
Antibacterianos/farmacología , Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Quinonas/farmacología , Benzoquinonas , Neoplasias de la Mama/genética , Femenino , Humanos , Lactamas Macrocíclicas , Fosforilación , Receptor ErbB-2 , Rifabutina/análogos & derivados , Transfección , Células Tumorales Cultivadas
16.
Cancer Res ; 54(3): 724-9, 1994 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-8306334

RESUMEN

Aphidicolin, an inhibitor of DNA polymerases alpha and delta, is cytotoxic in vitro against tumor cells. The poor solubility of aphidicolin has led to the development of aphidicolin glycinate (AG; NSC 303812), a water soluble ester currently in early clinical trials. The antitumor activity of AG was investigated in a series of transplantable murine tumors in vivo. The drug demonstrated activity against the i.p. implanted B16 melanoma, producing maximum increased life spans of 75% following i.p. administration every 3 h for three doses on days 1-9. Treatment schedules involving both single injections per day on days 1-9 and multiple injections per day on days 1, 5, and 9 were less effective, indicating that this antitumor activity is schedule dependent. Similarly, greater activity was observed against the i.p. M5076 sarcoma when three daily injections were given on days 1-9 (57% increased life span) than with a single injection either on days 1-9 (36% increased life span) or on days 1, 5, 9, and 13 (inactive). Further scheduling studies in the s.c. M5076 sarcoma model showed that a 7-day infusion was superior to both a 24-h infusion and a 7-day course of three bolus treatments per day. On the assumption that DNA polymerase inhibition is the basis for this antitumor activity, inhibition of DNA synthesis in BALB/c x DBA/2 F1 mice was investigated by measuring incorporation of [3H]thymidine (20 microCi, i.v.) into DNA of spleen and jejunum. At 2 h after administration of AG, inhibition of DNA synthesis was dose dependent (median inhibitory dose, 60 mg/kg in both tissues) and was > 99% at 300 mg/kg. The inhibition was rapid in onset; AG (100 mg/kg i.p.) produced maximal (> 98%) inhibition in both tissues at 30 min. Recovery occurred in the intestine within 16 h; in spleen recovery was delayed to 24 h, and was followed by a rebound incorporation at 48 h (203%). A comparison of the inhibition of thymidine incorporation in tumor cells (B16 melanoma and P388 leukemia) and normal jejunum revealed no significant differences in the extent of inhibition or the rapidity of recovery in these tissues. The rapid recovery of DNA synthesis inhibition supports the use of prolonged infusion schedules in clinical trials, but the lack of evidence of selectivity for tumor cells suggests that AG may be of limited therapeutic value as a single agent. Thus, we evaluated AG in combination with cisplatin in an in vivo model of cisplatin refractory human ovarian cancer.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Afidicolina/análogos & derivados , Cisplatino/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Afidicolina/administración & dosificación , Afidicolina/farmacología , Cisplatino/administración & dosificación , Reparación del ADN/fisiología , ADN Polimerasa Dirigida por ADN/fisiología , Modelos Animales de Enfermedad , Esquema de Medicación , Resistencia a Medicamentos/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Inhibidores de la Síntesis del Ácido Nucleico , Neoplasias Ováricas/tratamiento farmacológico
18.
J Biol Chem ; 264(2): 713-8, 1989 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-2910861

RESUMEN

Cyclopentenylcytosine (CPEC) is phosphorylated in L1210 cells with CPEC triphosphate as the major metabolite. Partially purified uridine-cytidine kinase catalyzes the initial phosphorylation of cyclopentenylcytosine with an apparent Km of 196 +/- 9 microM, and cyclopentenylcytosine is a competitive inhibitor of cytidine phosphorylation by this enzyme with a Ki value of 144 +/- 14 microM. Examination of the CTP synthetase activity in extracts of L1210 cells revealed a dose-dependent decrease on exposure of cells to CPEC. Synthesis of CPEC triphosphate by an enzymatic method permitted direct examination of the inhibition of partially purified CTP synthetase. CPEC triphosphate inhibited bovine CTP synthetase with a median inhibitory concentration of 6 microM, whereas CPEC mono- and diphosphates were ineffective. CTP synthetase showed a classical Michaelis-Menten hyperbolic plot of velocity and UTP concentration in the presence of saturating concentrations of ATP and glutamine, but CPEC triphosphate induced sigmoidal kinetic plots. The Hill coefficient was calculated to be 3.2.


Asunto(s)
Ligasas de Carbono-Nitrógeno , Citidina Trifosfato/farmacología , Citidina/análogos & derivados , Nucleótidos de Citosina/farmacología , Ligasas/antagonistas & inhibidores , Animales , Citidina/metabolismo , Citidina Trifosfato/análogos & derivados , Citidina Trifosfato/biosíntesis , Cinética , Leucemia L1210/enzimología , Leucemia P388/enzimología , Ligasas/aislamiento & purificación , Espectrometría de Masas , Ratones , Fosforilación
19.
Biochem Biophys Res Commun ; 156(3): 1383-9, 1988 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-2847733

RESUMEN

The human small cell lung carcinoma (SCLC) cell line NCI-H345 constitutively produces gastrin-releasing peptide (GRP), a peptide homologous to the mitogen bombesin. In addition, NCI-H345 cells express bombesin receptors and respond to bombesin with rapid activation of phospholipase C and mobilization of intracellular Ca2+. Treatment of NCI-H345 cells with a novel potent bombesin receptor antagonist [Leu13-psi-CH2NH-Leu14]bombesin blocked the increase in phosphatidylinositol turnover and cytoplasmic free Ca2+ ([Ca2+]i) stimulated by bombesin. Furthermore [Leu13-psi-CH2NH-Leu14]bombesin inhibited NCI-H345 colony formation in defined semisolid medium in the absence of exogenous GRP. The rapid, hormone-induced accumulation of inositol(1,4,5)trisphosphate was markedly more sensitive to antagonist inhibition than the hormone-induced Ca2+ transient, the sustained accumulation of inositol monophosphates, or colony formation in soft agarose. These data demonstrated inhibition of transmembrane signals associated with autocrine growth control in SCLC by a novel peptide receptor antagonist.


Asunto(s)
Carcinoma de Células Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores de Neurotransmisores/antagonistas & inhibidores , Bombesina/metabolismo , Calcio/metabolismo , Carcinoma de Células Pequeñas/patología , Comunicación Celular/efectos de los fármacos , Línea Celular , Citoplasma/metabolismo , Péptido Liberador de Gastrina , Humanos , Neoplasias Pulmonares/patología , Biosíntesis de Péptidos , Receptores de Bombesina , Receptores de Neurotransmisores/fisiología , Fosfolipasas de Tipo C/metabolismo
20.
Biochem J ; 255(2): 403-10, 1988 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-2849413

RESUMEN

Bombesin is an amphibian tetradecapeptide whose mammalian homologue, gastrin-releasing peptide (GRP), is produced by many small-cell lung-cancer (SCLC) cells, and which can function in an autocrine growth-promoting manner in SCLC. Studies reported here show that [Tyr4]bombesin and its congeners increase inositol 1,4,5-trisphosphate within seconds in NCI-H345, a SCLC cell line that constitutively produces GRP. After 30 min in the presence of 0.01 M-Li+ and [Tyr4]bombesin, there is marked accumulation of inositol monophosphates and inositol tetrakisphosphate. Pretreatment with phorbol 12-myristate 13-acetate (PMA) for 20 min inhibited the ability of [Tyr4]bombesin to induce phosphatidylinositol (PtdIns) turnover and to increase intracellular free Ca2+ ([Ca2+]i). Pretreatment with PMA for 48 h attenuated the ability of subsequently added PMA to decrease the response to [Tyr4]bombesin. Pretreatment with pertussis toxin (PT; 1 microgram/ml for 18-24 h) decreased by less than 30% [Tyr4]bombesin-induced increases in [Ca2+]i and PtdIns metabolites. However, interpretation of this result is complicated by the inability of PT to ADP-ribosylate completely its substrates in intact NCI-H345 cells. In contrast, pretreatment with cholera toxin (1 microgram/ml for 18-24 h) lowered basal [Ca2+]i and basal inositol phosphate concentrations, attenuated the response of NCI-H345 to subsequently added [Tyr4]bombesin, and was not mimicked by treatments that increase cellular cyclic AMP. These data demonstrate the activation of phospholipase C in SCLC by bombesin congeners. In addition, the results suggest a regulatory role for protein kinase C, a cholera-toxin substrate, and perhaps a pertussis-toxin substrate in the response of SCLC to bombesin.


Asunto(s)
Bombesina/farmacología , Carcinoma de Células Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfatidilinositoles/metabolismo , Adenosina Difosfato/metabolismo , Animales , Línea Celular , Toxina del Cólera/farmacología , Relación Dosis-Respuesta a Droga , Fosfatos de Inositol/aislamiento & purificación , Toxina del Pertussis , Ésteres del Forbol/farmacología , Relación Estructura-Actividad , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Factores de Virulencia de Bordetella/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...