Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
J Endocrinol Invest ; 46(5): 991-999, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36469293

RESUMEN

PURPOSE: SGLT2 inhibitors (SGLT2i) and GLP1 receptor agonists (GLP1-RA) protect the kidney in type 2 diabetes (T2DM) subjects. The role of patient's phenotype years before starting the treatment in determining the kidney response to these drugs has never been evaluated. SUBJECTS AND METHODS: Clinical and biochemical parameters were collected in 92 T2DM patients with preserved kidney function from year -4 (T-4) to year +3 (T+3) from the introduction of semaglutide or empagliflozin (T0). Glomerular filtration rate (eGFR) slopes were evaluated to identify eGFR changes (ΔGFR) and predictors of treatment response. Urinary markers of kidney impairment were measured at T0, including KIM-1, TNFR1 and L-FABP. RESULTS: Characteristics of patients on semaglutide (n = 46) or empagliflozin (n = 37) were similar at T-4 and T0. ΔGFR from T0 to T+3 was -5.5 [-10.0; -0.7] vs -2.6 [-102.4] ml/min/1.73 m2 for GLP1-RA and SGLT2i, respectively (p = ns). Compared with patients with a slower eGFR decline, those with ΔGFR > 5 ml/min/1.73 m2 from T0 to T+3 (49%) or ΔGFR > 10 ml/min/1.73 m2 from T-4 to T+3 (25%) had similar characteristics and urinary markers at T-4 and T0. The latter group showed greater eGFR decline from T-3 to T0, which tended to be delayed more by SGLT2i than GLP1-RA (p = 0.09). CONCLUSION: In our cohort, subjects with T2DM and preserved renal function show similar eGFR response to treatment with GLP1-RA or SGLT2i. Baseline urinary biomarkers or prior phenotyping do not predict treatment response. An early eGFR decline identifies patients prone to lose more eGFR over time, who may benefit more from SGLT2i treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Estudios Prospectivos , Riñón
2.
Oncogene ; 34(12): 1499-509, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24704829

RESUMEN

The cyclic nucleotide phosphodiesterase 10A (PDE10) has been mostly studied as a therapeutic target for certain psychiatric and neurological conditions, although a potential role in tumorigenesis has not been reported. Here we show that PDE10 is elevated in human colon tumor cell lines compared with normal colonocytes, as well as in colon tumors from human clinical specimens and intestinal tumors from Apc(Min/+) mice compared with normal intestinal mucosa, respectively. An isozyme and tumor-selective role of PDE10 were evident by the ability of small-molecule inhibitors and small interfering RNA knockdown to suppress colon tumor cell growth with reduced sensitivity of normal colonocytes. Stable knockdown of PDE10 by short hairpin RNA also inhibits colony formation and increases doubling time of colon tumor cells. PDE10 inhibition selectively activates cGMP/cGMP-dependent protein kinase signaling to suppress ß-catenin levels and T-cell factor (TCF) transcriptional activity in colon tumor cells. Conversely, ectopic expression of PDE10 in normal and precancerous colonocytes increases proliferation and activates TCF transcriptional activity. These observations suggest a novel role of PDE10 in colon tumorigenesis and that inhibitors may be useful for the treatment or prevention of colorectal cancer.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Factores de Transcripción TCF/genética , beta Catenina/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HCT116 , Células HEK293 , Células HT29 , Humanos , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción TCF/metabolismo , Transcripción Genética , beta Catenina/metabolismo
3.
J Thromb Haemost ; 12(6): 860-70, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24674135

RESUMEN

BACKGROUND: A growing health problem, venous thromboembolism (VTE), including pulmonary embolism (PE) and deep vein thrombosis (DVT), requires refined diagnostic and therapeutic approaches. Neutrophils contribute to thrombus initiation and development in experimental DVT. Recent animal studies recognized neutrophil extracellular traps (NETs) as an important scaffold supporting thrombus stability. However, the hypothesis that human venous thrombi involve NETs has not undergone rigorous testing. OBJECTIVE: To explore the cellular composition and the presence of NETs within human venous thrombi at different stages of development. PATIENTS AND METHODS: We examined 16 thrombi obtained from 11 patients during surgery or at autopsy using histomorphological, immunohistochemical and immunofluorescence analyses. RESULTS: We classified thrombus regions as unorganized, organizing and organized according to their morphological characteristics. We then evaluated them, focusing on neutrophil and platelet deposition as well as micro-vascularization of the thrombus body. We observed evidence of NET accumulation, including the presence of citrullinated histone H3 (H3Cit)-positive cells. NETs, defined as extracellular diffuse H3Cit areas associated with myeloperoxidase and DNA, localized predominantly during the phase of organization in human venous thrombi. CONCLUSIONS: NETs are present in organizing thrombi in patients with VTE. They are associated with thrombus maturation in humans. Dissolution of NETs might thus facilitate thrombolysis. This finding provides new insights into the clinical development and pathology of thrombosis and provides new perspectives for therapeutic advances.


Asunto(s)
Trampas Extracelulares , Neutrófilos/patología , Tromboembolia Venosa/patología , Adolescente , Adulto , Anciano , Biomarcadores/análisis , Plaquetas/patología , Citrulina/análisis , ADN/análisis , Progresión de la Enfermedad , Trampas Extracelulares/química , Femenino , Histonas/análisis , Humanos , Inmunohistoquímica , Masculino , Microscopía Fluorescente , Microvasos/patología , Persona de Mediana Edad , Neutrófilos/metabolismo , Peroxidasa/análisis , Tromboembolia Venosa/sangre , Tromboembolia Venosa/metabolismo
4.
J Neurosci Res ; 91(3): 393-406, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23280445

RESUMEN

Malignant gliomas have low survival expectations regardless of current treatments. Nonsteroidal anti-inflammatory drugs (NSAIDs) prevent cell transformation and slow cancer cell growth by mechanisms independent of cyclooxygenase (COX) inhibition. Certain NSAIDs trigger the endoplasmic reticulum stress response (ERSR), as revealed by upregulation of molecular chaperones such as GRP78 and C/EBP homologous protein (CHOP). Although celecoxib (CELE) inhibits the sarcoendoplasmic reticulum Ca(2+) ATPase (SERCA), an effect known to induce ERSR, sulindac sulfide (SS) has not been reported to affect SERCA. Here, we investigated these two drugs for their effects on Ca(2+) homeostasis, ERSR, and glioma cell survival. Our findings indicate that SS is a reversible inhibitor of SERCA and that both SS and CELE bind SERCA at its cyclopiazonic acid binding site. Furthermore, CELE releases additional Ca(2+) from the mitochondria. In glioma cells, both NSAIDS upregulate GRP78 and activate ER-associated caspase-4 and caspase-3. Although only CELE upregulates the expression of CHOP, it appears that CHOP induction could be associated with mitochondrial poisoning. In addition, CHOP induction appears to be uncorrelated with the gliotoxicity of these NSAIDS in our experiments. Our data suggest that activation of ERSR is primarily responsible for the gliotoxic effect of these NSAIDS. Because SS has good brain bioavailability, has lower COX-2 inhibition, and has no mitochondrial effects, it represents a more appealing molecular candidate than CELE to achieve gliotoxicity via activation of ERSR.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glioma/metabolismo , Pirazoles/toxicidad , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Sulfonamidas/toxicidad , Sulindac/análogos & derivados , Celecoxib , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/fisiología , Inhibidores Enzimáticos/toxicidad , Glioma/enzimología , Humanos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/química , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Sulindac/toxicidad
5.
Curr Mol Med ; 12(10): 1244-52, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22834827

RESUMEN

Honokiol (3',5-di-(2-propenyl)-1,1'-biphenyl-2,4'-diol) is a bioactive natural product derived from Magnolia spp. Recent studies have demonstrated anti-inflammatory, anti-angiogenic, anti-oxidative and anticancer properties of honokiol in vitro and in preclinical models. Honokiol targets multiple signaling pathways including nuclear factor kappa B (NF-κB), signal transducers and activator of transcription 3 (STAT3), epidermal growth factor receptor (EGFR) and mammalian target of rapamycin (m-TOR), which have great relevance during cancer initiation and progression. Furthermore, pharmacokinetic profile of honokiol has revealed a desirable spectrum of bioavailability after intravenous administration in animal models, thus making it a suitable agent for clinical trials. In this review, we discuss recent data describing the molecular targets of honokiol and its anti-cancer activities against various malignancies in pre-clinical models. Evaluation of honokiol in clinical trials will be the next step towards its possible human applications.


Asunto(s)
Antineoplásicos Fitogénicos , Compuestos de Bifenilo , Lignanos , Neoplasias/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacocinética , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Lignanos/farmacocinética , Lignanos/farmacología , Lignanos/uso terapéutico , Magnolia , Ratones , FN-kappa B/metabolismo , Neoplasias/prevención & control , Preparaciones de Plantas/farmacología , Preparaciones de Plantas/uso terapéutico , Ratas , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Oncogene ; 31(48): 4979-86, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286762

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) have been widely reported to display strong efficacy for cancer chemoprevention, although their mechanism of action is poorly understood. The most well-documented effects of NSAIDs include inhibition of tumor cell proliferation and induction of apoptosis, but their effect on tumor cell invasion has not been well studied. Here, we show that the NSAID, sulindac sulfide (SS) can potently inhibit the invasion of human MDA-MB-231 breast and HCT116 colon tumor cells in vitro at concentrations less than those required to inhibit tumor cell growth. To study the molecular basis for this activity, we investigated the involvement of microRNA (miRNA). A total of 132 miRNAs were found to be altered in response to SS treatment, including miR-10b, miR-17, miR-21 and miR-9, which have been previously implicated in tumor invasion and metastasis. We confirmed that these miRNA can stimulate tumor cell invasion and show that SS can attenuate their invasive effects by downregulating their expression. Employing luciferase and chromatin immunoprecipitation assays, NF-κB was found to bind the promoters of all four miRNAs to suppress their expression at the transcriptional level. We show that SS can inhibit the translocation of NF-κB to the nucleus by decreasing the phosphorylation of IKKß and IκB. Analysis of the promoter sequences of the miRNAs suppressed by SS revealed that 81 of 115 sequences contained NF-κB-binding sites. These results show that SS can inhibit tumor cell invasion by suppressing NF-κB-mediated transcription of miRNAs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , MicroARNs/genética , FN-kappa B/antagonistas & inhibidores , Invasividad Neoplásica/prevención & control , Neoplasias/patología , Sulindac/farmacología , Transcripción Genética/efectos de los fármacos , Línea Celular Tumoral , Humanos
7.
Cell Death Differ ; 19(3): 378-86, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21979467

RESUMEN

MicroRNAs (miRNAs) have attracted attention because of their key regulatory functions in many biological events, including differentiation and tumorigenesis. Recent studies have reported the existence of a reciprocal regulatory loop between the family of let-7 miRNAs and an RNA-binding protein, Lin28, both of which have been documented for their important roles during cell differentiation. Hence, using bipotent K562 human leukemia cells and human CD34+ hematopoietic progenitor cells as research models, we demonstrate that let-7 and Lin28 have contrary roles in megakaryocytic (MK) differentiation with a dynamic balance; expression of miR-181 is capable of effectively repressing Lin28 expression, disrupting the Lin28-let-7 reciprocal regulatory loop, upregulating let-7, and eventually promoting MK differentiation. However, miR-181 lacks a significant effect on hemin-induced erythrocyte differentiation. These results demonstrate that miR-181 can function as a 'molecular switch' during hematopoietic lineage progression specific to MK differentiation, thus providing insight into future development of miRNA-oriented therapeutics.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/citología , Humanos , Células K562 , Megacariocitos/citología , MicroARNs/genética , Proteínas de Unión al ARN/genética
8.
Bioresour Technol ; 101(15): 5759-66, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20236820

RESUMEN

Partially hydrolyzed extracts from blood meal, feather meal, and meat and bone meal, as well as a variety of common surplus agricultural proteins were tested for their ability to promote the flocculation of clay. Partial alkaline or enzymatic hydrolyses of blood meal, feather meal, and meat and bone meal were performed to liberate proteins and peptides from their water-insoluble forms. Some of these extracts promoted flocculation. However, if hydrolysis was extensive, low molecular weight peptides were mainly produced, and these extracts did not promote flocculation. Beef skin gelatins and hydrolyzed fish collagen were found to promote flocculation when pH 5.5 buffer was added. Commercial preparations of peptone enzymatic digest and a mixture of keratin and hydrolyzed keratin did not promote flocculation.


Asunto(s)
Silicatos de Aluminio/química , Minerales/química , Péptidos/química , Proteínas/química , Productos Biológicos/química , Arcilla , Floculación
9.
J Intern Med ; 256(1): 22-9, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15189362

RESUMEN

OBJECTIVES: To evaluate, in a group of nondiabetic essential hypertensive patients with normal renal function, the relationship between albumin excretion rate (AER) and carotid-femoral pulse wave velocity (PWV), as an index of aortic stiffness. DESIGN: Cross-sectional study. SETTING: Outpatient hypertension clinic. SUBJECTS: Seventy patients with mild-to-moderate essential hypertension, aged 42 +/- 8 years, never pharmacologically treated. All subjects underwent routine laboratory tests, 24-h ambulatory blood pressure (BP) monitoring, measurement of carotid-femoral PWV, by means of a computerized method, and AER. RESULTS: Microalbuminuric patients (AER > or = 20 microg min(-1); n = 19), when compared with normoalbuminuric subjects, showed more elevated 24-h BP (136/88 +/- 10/10 vs. 128/83 +/- 7/6 mmHg; P < 0.001 and P = 0.013, for systolic and diastolic BP respectively) and higher values of carotid-femoral PWV (10.4 +/- 2 m s(-1) vs. 9.2 +/- 1.3; P = 0.006). This latter difference remained statistically significant, even after correction by ancova for 24-h systolic and diastolic BP, and body mass index (BMI, P = 0.016). Univariate regression analysis disclosed a tight correlation between AER and carotid-femoral PWV (r = 0.42; P = 0.0003). This association was confirmed in a multiple regression model (beta = 0.35; P = 0.009) in which, as independent variables, besides PWV, 24-h BP, age, serum glucose values, smoking status, gender and BMI, were added. CONCLUSIONS: Our results seem to confirm that microalbuminuria may represent the early renal manifestation of a widespread vascular dysfunction, and therefore it is an integrated marker of cardiovascular risk.


Asunto(s)
Albuminuria/fisiopatología , Aorta/fisiopatología , Hipertensión/fisiopatología , Hipertensión/orina , Adulto , Velocidad del Flujo Sanguíneo , Monitoreo Ambulatorio de la Presión Arterial , Arterias Carótidas/fisiopatología , Estudios Transversales , Elasticidad , Femenino , Arteria Femoral/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Pulso Arterial , Análisis de Regresión , Factores de Riesgo
10.
Perfusion ; 17(3): 187-9, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12017386

RESUMEN

Various methods of cerebral protection have been used during aortic arch surgery. We reviewed our experience with a modified technique for selective cerebral perfusion (SCP) administration during surgery on the thoracic aorta from October 1999. Conventionally, this technique requires an additional roller pump on the cardiopulmonary bypass (CPB) console. In order to simplify the extracorporeal circuit (ECC), the paediatric double-roller pump used for the administration of cardioplegia was utilized by adding a 'Y-connection' on the blood line of the cardioplegia circuit, upstream of the cardioplegia reservoir, to provide SCP blood flow. SCP administration with a Y-connection is both easy and fast to set up on the ECC circuit and does not create additional difficulties to the surgeon. It simplifies SCP delivery by allowing the perfusionist to use a standard ECC system set-up.


Asunto(s)
Aorta Torácica/cirugía , Circulación Cerebrovascular , Perfusión/métodos , Adulto , Anciano , Anciano de 80 o más Años , Diseño de Equipo , Circulación Extracorporea/instrumentación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Perfusión/instrumentación
11.
Eur J Emerg Med ; 8(3): 233-6, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11587471

RESUMEN

Pituitary apoplexy is a severe and potentially life-threatening condition that may be highly variable in its clinical presentation. We report a 37-year-old man presenting to the emergency department with diplopia that abruptly developed while he was eating canned and bottled food prepared at home. A computed tomography scanning revealed an isodense mass within the sellar region and, subsequently, a magnetic resonance imaging showed a pituitary apoplexy causing a compression of the right III and VI oculomotor nerves. There was no improvement with hydrocortisone therapy and the patient underwent a transsphenoidal excision of the mass with an uneventful course. Pituitary apoplexy may raise in the appropriate setting the suspicion of botulism. The abrupt-onset paralysis of oculomotor nerves has been described as the chief presenting sign of pituitary apoplexy in only few cases including this. A pathophysiology, differential diagnosis with botulism and other causes of multiple cranial nerve paralysis, and treatment are described.


Asunto(s)
Diplopía/etiología , Oftalmoplejía/etiología , Apoplejia Hipofisaria/complicaciones , Apoplejia Hipofisaria/diagnóstico , Enfermedad Aguda , Adulto , Diplopía/terapia , Urgencias Médicas , Humanos , Imagen por Resonancia Magnética , Masculino , Oftalmoplejía/terapia , Apoplejia Hipofisaria/fisiopatología , Apoplejia Hipofisaria/terapia , Neoplasias Hipofisarias/diagnóstico , Prolactinoma/diagnóstico , Resultado del Tratamiento
12.
Cancer Res ; 61(10): 3961-8, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11358813

RESUMEN

Exisulind (Aptosyn) is a novel antineoplastic drug being developed for the prevention and treatment of precancerous and malignant diseases. In colon tumor cells, the drug induces apoptosis by a mechanism involving cyclic GMP (cGMP) phosphodiesterase inhibition, sustained elevation of cGMP, and protein kinase G activation. We studied the effect of exisulind on bladder tumorigenesis induced in rats by the carcinogen, N-butyl-N-(4-hydroxybutyl) nitrosamine. Exisulind at doses of 800, 1000, and 1200 mg/kg (diet) inhibited tumor multiplicity by 36, 47, and 64% and tumor incidence by 31, 38, and 61%, respectively. Experiments on the human bladder tumor cell line, HT1376, showed that exisulind inhibited growth with a GI(50) of 118 microM, suggesting that the antineoplastic activity of the drug in vivo involved a direct effect on neoplastic urothelium. Exisulind also induced apoptosis as determined by DNA fragmentation, caspase activation, and morphology. Analysis of phosphodiesterase (PDE) isozymes in HT1376 cells showed PDE5 and PDE4 isozymes that were inhibited by exisulind with IC(50)s of 112 and 116 microM, respectively. Inhibition of PDE5 appears to be pharmacologically relevant, because treatment of HT1376 cells increased cGMP and activated protein kinase G at doses that induce apoptosis, whereas cyclic AMP levels were not changed. Immunocytochemistry showed that PDE5 was localized in discrete perinuclear foci in HT1376 cells. Immunohistochemistry showed that PDE5 was overexpressed in human squamous and transitional cell carcinomas compared with normal urothelium. The data lead us to conclude that future clinical trials of exisulind for human bladder cancer treatment and/or prevention should be considered and suggest a mechanism of action involving cGMP-mediated apoptosis induction.


Asunto(s)
Anticarcinógenos/farmacología , Sulindac/farmacología , Neoplasias de la Vejiga Urinaria/prevención & control , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , 3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Concentración 50 Inhibidora , Microscopía Fluorescente , Ratas , Ratas Endogámicas F344 , Sulindac/análogos & derivados , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/metabolismo
13.
Cancer Epidemiol Biomarkers Prev ; 9(11): 1155-62, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11097222

RESUMEN

The nonsteroidal anti-inflammatory drug sulindac and the ornithine decarboxylase inhibitor difluoromethylornithine (DFMO) are both potent inhibitors of colon carcinogenesis in experimental models of this disease. The combination of these two agents is undergoing evaluation as a strategy for colon cancer chemoprevention in humans with resected colon polyps. We evaluated the effects of the major sulfide and sulfone metabolites of sulindac and DFMO alone, or in combinations, on the growth and survival of Caco-2 colon cancer-derived cells and in clones of these cells transfected with an activated K-ras oncogene. Both the sulfide and sulfone metabolites of sulindac reduced cell viability, measured by colony-forming assays, primarily by inducing apoptosis. Expression of an activated K-ras oncogene caused cells treated with either sulindac sulfide or sulfone to undergo apoptosis earlier than nontransfected controls. However, clonogenic survival, measured 2 weeks after drug treatment, was the same in both Caco-2 and ras-transfected Caco-2 cells treated with sulindac metabolites. A 24-h treatment with DFMO caused a dose-dependent decrease in the colony-forming ability of cells expressing an activated K-ras but had no effect on the viability of the parental Caco-2 cells. The DFMO-dependent decrease in colony formation in K-ras-activated cells occurred in the absence of apoptosis. Assessment of cell survival by colony-forming assays indicated that these two agents acted in an additive manner when combined. These data indicate that K-ras can influence the kinetics of apoptosis induction by sulindac metabolites and cell survival in response to DFMO. However, cytotoxicity induced by these agents occurs via unique mechanisms. These studies suggest that the combination of DFMO and sulindac may be useful in human cancer prevention strategies.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Quimioprevención , Eflornitina/farmacología , Inhibidores Enzimáticos/farmacología , Sulindac/farmacología , Apoptosis/efectos de los fármacos , Células CACO-2 , Supervivencia Celular , Transformación Celular Neoplásica , Eflornitina/análogos & derivados , Genes ras/genética , Humanos , Sulindac/análogos & derivados
14.
Clin Cancer Res ; 6(10): 4136-41, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11051267

RESUMEN

Sulindac sulfone (Exisulind) induces apoptosis and exhibits cancer chemopreventive activity, but in contrast to sulindac, it does not inhibit cyclooxygenases 1 or 2. We found that sulindac sulfone and two potent derivatives, CP248 and CP461, inhibited the cyclic GMP (cGMP) phosphodiesterases (PDE) 2 and 5 in human colon cells, and these compounds caused rapid and sustained activation of the c-Jun NH2-terminal kinase 1 (JNK1). Rapid activation of stress-activated protein/ERK kinase 1 (SEK1) and mitogen-activated protein kinase kinase kinase (MEKK1), which are upstream of JNK1, was also observed. Other compounds that increase cellular levels of cGMP also activated JNK1, and an inhibitor of protein kinase G (PKG), Rp-8-pCPT-cGMPS, inhibited JNK1 activation by the sulindac sulfone derivatives. Expression of a dominant-negative JNK1 protein inhibited CP248-induced cleavage of poly(ADP-ribose) polymerase, a marker of apoptosis. Thus, it appears that sulindac sulfone and related compounds induce apoptosis, at least in part, through activation of PKG, which then activates the MEKK1-SEK1-JNK1 cascade. These studies also indicate a role for cGMP and PKG in the JNK pathway.


Asunto(s)
Apoptosis , GMP Cíclico/metabolismo , MAP Quinasa Quinasa 4 , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Sulindac/farmacología , Neoplasias del Colon/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Activación Enzimática , Genes Dominantes , Humanos , Proteína Quinasa 8 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Sulindac/análogos & derivados , Factores de Tiempo , Células Tumorales Cultivadas
15.
Cancer Res ; 60(13): 3338-42, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10910034

RESUMEN

Sulindac sulfone (exisulind), although a nonsteroidal anti-inflammatory drug derivative, induces apoptosis in tumor cells by a mechanism that does not involve cyclooxygenase inhibition. SW480 colon tumor cells contain guanosine 3',5'-monophosphate (cGMP) phosphodiesterase (PDE) isoforms of the PDE5 and PDE2 gene families that are inhibited by exisulind and new synthetic analogues. The analogues maintain rank order of potency for PDE inhibition, apoptosis induction, and growth inhibition. A novel mechanism for exisulind to induce apoptosis is studied involving sustained increases in cGMP levels and cGMP-dependent protein kinase (PKG) induction not found with selective PDE5 or most other PDE inhibitors. Accumulated beta-catenin, shown to be a substrate for PKG, is decreased by exisulind, suggesting a mechanism to explain apoptosis induction in neoplastic cells harboring adenomatous polyposis coli gene mutations.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Proteínas Quinasas/metabolismo , Sulindac/análogos & derivados , Transactivadores , 3',5'-GMP Cíclico Fosfodiesterasas/genética , Cadherinas/metabolismo , Neoplasias del Colon , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activación Enzimática , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Cinética , Piperidinas/farmacología , Quinazolinas/farmacología , Sulindac/farmacología , Células Tumorales Cultivadas , beta Catenina
16.
Biochem Pharmacol ; 58(7): 1097-107, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10484067

RESUMEN

We examined the activity of two metabolites of sulindac (a nonsteroidal anti-inflammatory drug), sulindac sulfide and sulindac sulfone (exisulind, Prevatec), and a novel highly potent analog of exisulind (CP248) on a series of human prostate epithelial cell lines. Marked growth inhibition was seen with the BPH-1, LNCaP, and PC3 cell lines with IC50 values of about 66 microM, 137 microM, and 64 nM for sulindac sulfide, exisulind, and CP248, respectively. DNA flow cytometry and 4',6'-diamido-2-phenylindole (DAPI) staining indicated that these three compounds also induced apoptosis in all of these cell lines. Similar growth inhibition also was seen with the PrEC normal human prostate epithelial cell line, but these cells were resistant to induction of apoptosis at concentrations up to 300 microM, 1 mM, and 750 nM of sulindac sulfide, exisulind, and CP248, respectively. Derivatives of LNCaP cells that stably overexpress bcl-2 remained sensitive to growth inhibition and induction of apoptosis by these compounds. In vitro enzyme assays indicated that despite its high potency in inhibiting growth and inducing apoptosis, CP248, like exisulind, lacked cyclooxygenase (COX-1 and COX-2) inhibitory activity even at concentrations up to 10 mM. Moreover, despite variations of COX-1 and COX-2 expression, the three benign and malignant prostate cell lines showed similar sensitivity to growth inhibition and induction of apoptosis by these three compounds. Therefore, sulindac derivatives can cause growth inhibition and induce apoptosis in human prostate cancer cells by a COX-1 and -2 independent mechanism, and this occurs irrespective of androgen sensitivity or increased expression of bcl-2. These compounds may be useful in the prevention and treatment of human prostate cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Neoplasias de la Próstata/tratamiento farmacológico , Sulindac/farmacología , Andrógenos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , División Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulindac/análogos & derivados , Células Tumorales Cultivadas
17.
Urology ; 53(2): 440-5, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9933075

RESUMEN

OBJECTIVES: Recent studies have shown that Exisulind, a sulfone metabolite of the nonsteroidal anti-inflammatory drug (NSAID) sulindac, has inhibitory activity in vitro with cultured human prostate cancer cells. To determine whether this effect might be pharmacologically relevant in vivo, we tested whether Exisulind therapy could suppress the growth of human prostate cancer cells in a nude mouse xenograft model. METHODS: Thirty athymic nude mice were injected subcutaneously in the flank with 1 x 10(7) LNCaP human prostate tumor cells. All mice received a control diet for 21 days. One group of mice was continued on this control diet for an additional 4 weeks, a second group was switched to a diet supplemented with 0.05% Exisulind (40% of maximal tolerated dose [MTD]), and a third group was switched to a diet supplemented with 0.1% Exisulind (80% MTD) for the additional 4 weeks. Tumor growth was measured through the 4-week test period, and subsequently tissue sections from the various groups were tested for apoptotic and dividing cells by quantified use of the TUNEL assay and a bromodeoxyuridine (BrdU) incorporation immunoassay. RESULTS: Tumors grew by 158%, 24%, and 18% for the control and 0.05% and 0.1% Exisulind groups, respectively (P = 0.02) during the 4-week test period. Immunohistochemical studies on excised tumors showed an increased number of apoptotic bodies in the treated groups versus the control group (P<0.0001) but no change in the number of BrdU positive cells. CONCLUSIONS: This is the first study to show a direct in vivo effect of an NSAID-derived drug, lacking cyclooxygenase inhibitory activity, in a xenograft model of prostate cancer. Clinical studies to evaluate the effects of Exisulind against prostate cancer in humans are warranted.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Sulindac/análogos & derivados , Animales , División Celular , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Sulindac/uso terapéutico
18.
Adv Exp Med Biol ; 470: 45-53, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10709673

RESUMEN

Sulindac sulfone (Exisulind), a metabolite of the non-steroidal anti-inflammatory drug, sulindac, was evalauted for its effects on the development of rectal polyps in patients with familial adenomatous polyposis. Three cohorts of 6 patients each were given doses of 200, 300, or 400 mg Exisulind twice daily. Hepatotoxicity, shown by elevation in blood transaminase levels, was the dose-limiting toxicity and occurred at the 400 mg bid dose. Due to this toxicity, all patients treated with the 400 mg dose were subsequently reduced to the 200 mg dose level. Subsequently, 2 of the 6 patients were dose-escalated to 400 mg bid dose. The patients were treated with Exisulind for a period of six months. Sixteen of 18 patients had regression of small polyps (> or = 6 mm in diameter) characterized by a flattening of the polyps and a macular "halo" appearance. Histopathologic examination of the polyp biopsy specimens showed a marked increase in the proportion of mucin producing cells in the glands after treatment with Exisulind at all dose levels. Ki-67 staining, a measure of cell proliferation, was higher in the polyps than in normal mucosa. There was no significant change in the proliferation index between baseline and six month values in any of the groups treated with Exisulind or in normal tissues. The median apoptotic labeling index, as determined by the TUNEL technique, was higher in the polyps than in normal-appearing mucosa. Overall, there was no significant change in the apoptotic labeling index between base-line and 6 months in normal-appearing mucosa however, the index in polyps was increased. These results suggest that treatment of FAP patients with Exisulind for a period of six months may lead to regression of small polyps, and that the mechanisms of Exisulind--induced regression appear to be through stimulation of mucus differentiation and apoptosis in glandular epithelium.


Asunto(s)
Poliposis Adenomatosa del Colon/tratamiento farmacológico , Poliposis Adenomatosa del Colon/patología , Antineoplásicos/uso terapéutico , Sulindac/análogos & derivados , Poliposis Adenomatosa del Colon/metabolismo , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/biosíntesis , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Antígeno Ki-67/biosíntesis , Sulindac/efectos adversos , Sulindac/uso terapéutico
19.
Acta Neurochir (Wien) ; 140(11): 1183-9, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9870066

RESUMEN

The site of leakage in a patients with rhinorrhea of various origin may be difficult to identify. The aim of our paper is to evaluate the contribution of cisternography in combination with single photon emission tomography (SPECT) to identify the fistulous track. From 1/1/1992 to 30/11/1997 we studied 20 patients with rhinorrhea posing a challenging diagnostic problem as to identification of the leakage site. Two mls of Indium DTPA (In 111) were injected into the subarachnoid space by the lumbar route. The tracer was followed by planar scintigraphy until it reached the cranial base and subsequently the SPECT acquisition started. A fistula was demonstrated in all of our cases including patients with no active leakage at the time of examination, patients with no bone defects on thin sliced CT scanning or patients with a normal MRI. At surgery the fistulous track was confirmed in all but two cases when a bilateral fistula was operatively identified only on one side. In conclusion whenever a CT scanning fails to demonstrate significant bone defects and MRI does not localize a fistulous track, SPECT cisternography via the lumbar route proved in our experience to be a reliable examination for a precise diagnosis.


Asunto(s)
Rinorrea de Líquido Cefalorraquídeo/diagnóstico , Neumoencefalografía , Tomografía Computarizada de Emisión de Fotón Único , Adolescente , Adulto , Anciano , Rinorrea de Líquido Cefalorraquídeo/etiología , Rinorrea de Líquido Cefalorraquídeo/cirugía , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Ácido Pentético , Estudios Prospectivos , Sensibilidad y Especificidad , Base del Cráneo/patología , Base del Cráneo/cirugía , Tomografía Computarizada por Rayos X
20.
Carcinogenesis ; 19(8): 1353-6, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9744528

RESUMEN

The sulfone derivative of the non-steroidal anti-inflammatory drug (NSAID), sulindac, has been reported to inhibit mammary and colon tumor formation in rodent models of chemically-induced carcinogenesis. Unlike its parent compound, this metabolite lacks cyclo-oxygenase inhibitory activity. A tumor induction protocol, consisting of NNK administration in the drinking water over several weeks to model chronic human exposure, was used to test whether the sulfone (called FGN-1) could inhibit the formation of primary lung tumors in mice. A total of 150 female, AIN76A-fed, A/J mice received 9 mg of NNK each. Concentrations of FGN-1 that had been previously determined not to affect body weight gain were added to the food at levels of 0, 250, 500 and 750 mg/kg of diet (30 mice/group) starting 2 weeks before NNK administration and continuing for 22 weeks. At that time pleural surface tumors were counted. Tumor incidence decreased significantly from 96 % in the control diet and 93% in the 250 FGN-1 mg/kg diet to 63 and 67% in the 500 and 750 mg FGN-1/kg diet groups, respectively (P < 0.001 by chi-square analysis). Lung tumor multiplicity decreased from 18.1+/-3 tumors/ mouse (mean+/-SEM, control diet) to 12.3+/-3 (250), 5.3+/-1 (500) and 2.1+/-1 (750) (P < 0.0005 by post hoc ANOVA). In previous studies using this carcinogenesis protocol, the maximum tolerated dose of sulindac inhibited lung tumor multiplicity by no more than 50% with no effect on incidence. This dose-dependent reduction in tumorigenesis by a non-toxic dose of FGN-1 indicates a strong chemopreventive activity against experimental induction of lung carcinogenesis. The greater potency of the sulfone over sulindac and its lack of toxic side effects because of its inability to affect cyclo-oxygenase activity suggests that clinical testing in individuals at high risk for lung cancer should be considered.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/prevención & control , Sulindac/análogos & derivados , Animales , Carcinógenos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Neoplasias Pulmonares/inducido químicamente , Ratones , Ratones Endogámicos A , Nitrosaminas , Sulindac/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA