Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
ESMO Open ; 9(2): 102217, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38320431

RESUMEN

INTRODUCTION: We report results from a phase I, three-part, dose-escalation study of peposertib, a DNA-dependent protein kinase inhibitor, in combination with avelumab, an immune checkpoint inhibitor, with or without radiotherapy in patients with advanced solid tumors. MATERIALS AND METHODS: Peposertib 100-400 mg twice daily (b.i.d.) or 100-250 mg once daily (q.d.) was administered in combination with avelumab 800 mg every 2 weeks in Part A or avelumab plus radiotherapy (3 Gy/fraction × 10 days) in Part B. Part FE assessed the effect of food on the pharmacokinetics of peposertib plus avelumab. The primary endpoint in Parts A and B was dose-limiting toxicity (DLT). Secondary endpoints were safety, best overall response per RECIST version 1.1, and pharmacokinetics. The recommended phase II dose (RP2D) and maximum tolerated dose (MTD) were determined in Parts A and B. RESULTS: In Part A, peposertib doses administered were 100 mg (n = 4), 200 mg (n = 11), 250 mg (n = 4), 300 mg (n = 6), and 400 mg (n = 4) b.i.d. Of DLT-evaluable patients, one each had DLT at the 250-mg and 300-mg dose levels and three had DLT at the 400-mg b.i.d. dose level. In Part B, peposertib doses administered were 100 mg (n = 3), 150 mg (n = 3), 200 mg (n = 4), and 250 mg (n = 9) q.d.; no DLT was reported in evaluable patients. Peposertib 200 mg b.i.d. plus avelumab and peposertib 250 mg q.d. plus avelumab and radiotherapy were declared as the RP2D/MTD. No objective responses were observed in Part A or B; one patient had a partial response in Part FE. Peposertib exposure was generally dose proportional. CONCLUSIONS: Peposertib doses up to 200 mg b.i.d. in combination with avelumab and up to 250 mg q.d. in combination with avelumab and radiotherapy were tolerable in patients with advanced solid tumors; however, antitumor activity was limited. GOV IDENTIFIER: NCT03724890.


Asunto(s)
Neoplasias , Piridazinas , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Quinazolinas/uso terapéutico
2.
Curr Cancer Drug Targets ; 6(3): 197-205, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16712457

RESUMEN

Exposure to ionizing radiation (IR) results in the formation of DNA double strand breaks, resulting in the activation of phosphatidylinositol 3'-kinase-like kinases ATM, ATR and DNK-PKcs. A physiologically important downstream target is the minor histone H2A variant, H2AX, which is rapidly phosphorylated on Ser 139 of the carboxyl tail after IR. Recent work suggests that phosphorylated H2AX (gamma-H2AX) plays an important role in the recruitment and/or retention of DNA repair and checkpoint proteins such as BRCA1, MRE11/RAD50/NBS1 complex, MDC1 and 53BP1. H2AX-/- mouse embryonic fibroblasts are radiation sensitive and demonstrate deficits in repairing DNA damage compared to their wildtype counterparts. Cells treated with peptide inhibitors of gamma-H2AX demonstrate increased radiosensitivity following radiation compared with untreated irradiated cells. Analysis of the kinetics of gamma-H2AX clearance after IR or other DNA damaging agents reveals a correlation between increased gamma-H2AX persistence and unrepaired DNA damage and cell death. These data highlight the potential of post-translational modifications of chromatin as a therapeutic target for enhancing the efficacy of radiotherapy. Therapies that either block gamma-H2AX foci formation by inhibiting upstream kinase activity or that directly inhibit H2AX function may interfere with DNA damage repair processes and warrant further investigation as potential radiosensitizing agents. Agents that increase persistence of gamma-H2AX after IR are likely to increase unrepaired DNA damage.


Asunto(s)
Histonas/efectos de la radiación , Neoplasias/genética , Neoplasias/radioterapia , Radioterapia , Antineoplásicos/farmacología , Biomarcadores , Daño del ADN/genética , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/efectos de la radiación , Humanos
3.
Br J Radiol ; 79(942): 497-503, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16714752

RESUMEN

The purpose of this study was to assess the efficacy and toxicity of intensity-modulated radiation therapy (IMRT) in the treatment of gastric cancer. Seven patients with gastric cancer were treated with IMRT. Six patients (all Stage III) received post-operative chemoradiotherapy with concurrent 5-fluorouracil and leucovorin. One received planned pre-operative radiation, though did not proceed to surgery. All patients were planned to receive 50.4 Gy in 1.8 Gy fractions. IMRT planning was compared with opposed anterior-posterior: posterior-anterior (AP/PA) and 3-field conventional three-dimensional plans. When compared with either AP/PA or 3-field plans, IMRT significantly reduced the volume exceeding the threshold dose of the liver and at least one kidney. Target coverage with IMRT was excellent, with 98+/-1% of the target receiving >or=100% of the dose. Compared with AP/PA and 3-field plans, IMRT plans had a greater percentage of target receiving the prescribed dose, but also a greater volume receiving >110% of the dose. IMRT was well tolerated; no patients developed acute gastrointestinal toxicity greater than grade 2. All seven experienced grade 2 nausea, three had grade 2 diarrhoea and two had grade 2 oesophagitis. Weight loss ranged from 0-12% (mean 6.1% and median 5.8%). IMRT in the treatment of gastric malignancies reduces the mean and above threshold doses to critical normal tissues. In an initial cohort of seven patients, 50.4 Gy delivered by IMRT is well tolerated and safe.


Asunto(s)
Adenocarcinoma/radioterapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Radioterapia de Intensidad Modulada/métodos , Neoplasias Gástricas/radioterapia , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/cirugía , Anciano , Anciano de 80 o más Años , Terapia Combinada , Fluorouracilo/administración & dosificación , Humanos , Leucovorina/administración & dosificación , Persona de Mediana Edad , Cuidados Preoperatorios , Radiometría , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador , Radioterapia de Intensidad Modulada/efectos adversos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/cirugía , Resultado del Tratamiento
4.
Semin Radiat Oncol ; 11(4): 338-45, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11677658

RESUMEN

Ionizing radiation (IR) has been extensively used to treat a variety of solid tumors to improve local control and overall survival in patients. Gene therapy strategies represent one experimental direction to improve radiocurability. These gene therapy strategies include (1) replacement of mutated or deleted tumor-suppressor genes, (2) delivery of prodrugs, (3) transduction of genes under the control of radiation-inducible promoters, and (4) genetically engineered viruses that replicate preferentially in tumor cells after IR. Although any one of these viral-based gene therapy approaches is unlikely to succeed independently, experimental results suggest that clinically important antitumor can be achieved when these strategies are combined with IR. Several of these strategies are currently being or soon will be evaluated in clinical trials. This review focuses on molecular mechanisms and potential clinical application of these viral-based gene therapy strategies to improve the therapeutic index of IR.


Asunto(s)
Terapia Genética/métodos , Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Virus/genética , Animales , Antineoplásicos/uso terapéutico , Terapia Combinada , Técnicas de Transferencia de Gen , Genes p53/genética , Vectores Genéticos/genética , Humanos , Neoplasias/tratamiento farmacológico , Profármacos/farmacología , Radiación Ionizante
5.
Mol Pharmacol ; 57(4): 792-6, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10727527

RESUMEN

Previous work has demonstrated that down-regulation of ceramide production after selection of cells with N-oleoylethanolamine (OE), an inhibitor of ceramidase, results in resistance to DNA damage-induced apoptosis. We report here that acute exposure of WEHI-231 cells (murine B-cell lymphoma) to OE activates neutral sphingomyelinase, induces ceramide production and increases intracellular reactive oxygen species. OE exposure also induces mitochondrial permeability, cytochrome c release, and apoptosis. Cells selected for resistance to OE exhibit little if any change in reactive oxygen species and cytochrome c release when exposed either to OE or to toxic doses of ceramide. Importantly, the OE resistant cells are also resistant to ionizing radiation-induced cytochrome c release and apoptosis. These findings demonstrate that down-regulation of neutral sphingomyelinase activity is associated with decreased DNA-damage-induced apoptosis. In addition, the data suggests that agents that modify extranuclear targets responsible for ceramide production select for cells resistant to ionizing radiation-induced apoptosis through alterations in mitochondrial function.


Asunto(s)
Apoptosis , Ceramidas/metabolismo , Grupo Citocromo c/metabolismo , Animales , Regulación hacia Abajo , Endocannabinoides , Activación Enzimática/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Etanolaminas/farmacología , Ratones , Mitocondrias/metabolismo , Ácidos Oléicos , Permeabilidad , Radiación Ionizante , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielina Fosfodiesterasa/efectos de la radiación , Células Tumorales Cultivadas
6.
Clin Cancer Res ; 6(2): 737-42, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10690561

RESUMEN

Although clonogenic or divisional death is the main mechanism by which DNA-damaging agents demonstrate antitumor activity, recent data indicate that strategies specifically designed to trigger apoptosis may also prove to be useful antitumor agents. Protein kinase C (PKC) isoenzymes are involved in the regulation of cell proliferation, differentiation, and survival. Whereas pharmacological inhibition of PKC activity triggers apoptosis in most mammalian cells, cell line and tissue differences in sensitivities to these inhibitors remain. Whereas PKC inhibitors have potential as antitumor agents, issue of kinase specificity and solubility have remained obstacles to their clinical use. In this report, we investigated the antitumor activity of the PKC inhibitor chelerythrine chloride (chelerythrine), a selective inhibitor of group A and B PKC isoforms. Chelerythrine exhibited cytotoxic activity against nine human tumor cell lines tested in vitro. On the basis of the finding that radioresistant and chemoresistant squamous cell carcinoma lines (HNSCC) undergo apoptosis rapidly after treatment with chelerythrine in vitro, we assessed the effects of this agent on p53-deficient SQ-20B HNSCC cells in vivo. The results demonstrate that chelerythrine treatment of nude mice bearing SQ-20B is associated with significant tumor growth delay. Significantly, treatment with chelerythrine resulted in minimal toxicity. These findings demonstrate a potential for chelerythrine as an antitumor drug against squamous cell carcinoma.


Asunto(s)
Antineoplásicos/toxicidad , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , Fenantridinas/toxicidad , Alcaloides , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Benzofenantridinas , Peso Corporal/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Ratones , Ratones Desnudos , Fenantridinas/uso terapéutico , Proteína Quinasa C/antagonistas & inhibidores , Trasplante Heterólogo , Células Tumorales Cultivadas
7.
Radiat Oncol Investig ; 7(5): 261-9, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10580895

RESUMEN

Many gene-therapy strategies under investigation aim to increase the efficacy of current cancer-treatment regimens. Promising results have been obtained in the laboratory and early clinical trials using viral-based motifs specifically designed to enhance the efficacy of ionizing radiation or chemotherapy. These strategies fall into two general categories: replication-incompetent viral shuttle vectors for the delivery of specific genes encoding a chemo/radiation modulator and attenuated replication-competent viruses with proposed replicative advantages in tumor cells. In this review, we discuss the rational, molecular mechanisms, and clinical application of these strategies with particular focus on recent research applying these viral-based strategies to improve the therapeutic index of ionizing radiation.


Asunto(s)
Terapia Genética/métodos , Neoplasias/radioterapia , Antineoplásicos/uso terapéutico , Terapia Combinada , Sinergismo Farmacológico , Técnicas de Transferencia de Gen , Genes p53/genética , Vectores Genéticos/genética , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Profármacos/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Transcripción Genética/efectos de la radiación , Replicación Viral/genética , Virus/genética
8.
Urology ; 54(3): 573-7, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10475376

RESUMEN

OBJECTIVES: Calphostin C, a highly specific protein kinase C inhibitor, induces apoptosis in the presence of visible light. We report the photoactivatable cytotoxicity of calphostin C in a series of well-characterized human bladder cancer cell lines: RT4, UM-UC-3, and 5637. METHODS: The human bladder cancer cell lines RT4, UM-UC-3, and 5637 were chosen on the basis of their p53, pRb and 9p21 deletion status. Using standard tissue culture techniques, the cytotoxicity of 10 to 100 nM calphostin C in combination with increasing exposures of visible light was examined. Controls consisted of cells treated with calphostin C without visible light and cells exposed to visible light without calphostin C treatment. Cell viability was determined by MTT assay. The induction of apoptosis by activated calphostin C was determined by 4,6-diamidino-2-phenylindole (DAPI) staining/fluorescence microscopy of nuclei. RESULTS: In the absence of light, calphostin C did not demonstrate a cytotoxic effect on any of the cell lines tested. Increasing the duration of light exposure resulted in a concomitant decrease in cell viability. Significant cell death was seen with calphostin C concentrations as low as 10 nM. These studies also demonstrated that calphostin C induced apoptosis by a mechanism independent of p53 and pRb status and the presence or absence of 9p21 deletions. CONCLUSIONS: We demonstrated the ability of activated calphostin C to induce apoptosis in a light-dependent and concentration-dependent fashion in a bladder cancer model system. Activated calphostin C cytotoxicity is independent of tumor genetic background and the status of p53 and pRb. Further development of calphostin C as a photosensitizer for photodynamic therapy of superficial bladder cancer may be warranted.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Naftalenos/uso terapéutico , Fotoquimioterapia , Proteína Quinasa C/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células Tumorales Cultivadas
9.
Clin Cancer Res ; 4(10): 2391-8, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9796970

RESUMEN

The development of novel therapeutic agents to modulate programmed cell death independent of genetic background or malignant potential is a primary goal of modern cancer therapy. In this report, the light activation- and concentration-dependent cytotoxicity of calphostin C, a photoactivatable perylenequinone, is carefully evaluated using a series of nine well-characterized human and rodent prostate cancer cell lines representing the spectrum of disease progression (e.g., variations in metastatic ability, ploidy, and tumor suppressor gene status). Treatment of these cancer cell lines with nanomolar concentrations of calphostin C in combination with increasing amounts of light exposure established a relationship between light and dose dependence of calphostin C cytotoxicity. The induction of apoptosis is rapid, as evidenced by the fact that immediately after treatment, cells exposed to calphostin C with light activation exhibit both morphological and biochemical changes consistent with apoptosis (cellular and nuclear shrinkage and chromatin condensation). For example, 78% of cells treated with 100 nM calphostin C in combination with 2 h of light activation underwent apoptosis within 24 h of treatment. DNA ladder formation could be detected within 12 h of treatment. In the absence of light activation, treatment with calphostin C at all concentrations tested had no acute or durable cytotoxic effects in any of the cell lines. Our findings demonstrate that calphostin C cytotoxicity is strictly light dependent. Furthermore, its efficacy is independent of the genetic background, p53 status, or in vivo malignant potential of a cell, making it a suitable candidate for the treatment of heterogeneous tumor cell populations.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Naftalenos/farmacología , Proteína p53 Supresora de Tumor/análisis , Animales , Apoptosis , Biotransformación , Humanos , Luz , Masculino , Naftalenos/farmacocinética , Metástasis de la Neoplasia , Ratas , Células Tumorales Cultivadas
10.
Cancer Res ; 57(19): 4340-7, 1997 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9331096

RESUMEN

Approximately 30% of cancer deaths result from the failure to control local and regional tumors. The goal of radiotherapy is to maximize local and regional tumor cell killing while minimizing normal tissue destruction. Attempts to enhance radiation-mediated tumor cell killing using halogenated pyrimidines, antimetabolites, and other DNA-damaging agents or sensitizers of hypoxic tumor cells have met with only modest clinical success. In an unique strategy to modify tumor radiosensitivity, we used an inhibitor of the protein kinase C group A and B isoforms, chelerythrine chloride (chelerythrine), to enhance the killing effects of ionizing radiation (IR). Protein kinase C activity plays a central role in cellular proliferation, differentiation, and apoptosis. Chelerythrine increases sphingomyelinase activity and enhances IR-mediated cell killing through induction of apoptotic tumor cell death in a radioresistant tumor model both in vitro and in vivo. Although previous reports have suggested that IR-mediated apoptosis correlates with tumor volume reduction, we demonstrate for the first time that lowering the apoptotic threshold increases tumor cell killing in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/radioterapia , Traumatismos Craneocerebrales/radioterapia , Inhibidores Enzimáticos/farmacología , Isoenzimas/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Esfingomielina Fosfodiesterasa/metabolismo , Alcaloides , Animales , Benzofenantridinas , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/enzimología , Ceramidas/farmacología , Quimioterapia Adyuvante , Terapia Combinada , Traumatismos Craneocerebrales/tratamiento farmacológico , Traumatismos Craneocerebrales/enzimología , Endopeptidasas/metabolismo , Activación Enzimática/efectos de los fármacos , Isoenzimas/metabolismo , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Trasplante Heterólogo
11.
Cancer Res ; 57(7): 1270-5, 1997 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9102212

RESUMEN

Ionizing radiation mediates cell death, in part, through chromosomal damage following one or more cell divisions. X-rays also induce programmed cell death (apoptosis) in some cell types both in vitro and in vivo. Both neutral and acidic sphingomyelinases, which generate the lipid second messenger ceramide, are reported to induce apoptosis following ionizing radiation and other death signals such as tumor necrosis factor alpha and Fas ligand. Herein we report that a loss of ceramide production from a neutral sphingomyelinase generates a radioresistant phenotype as measured by a marked decrease in apoptosis. A WEHI-231 subline made deficient in ceramide production was found to be resistant to apoptosis compared with the parental subline following treatment with X-rays. The resistant subline underwent two to three subsequent cell divisions following X-irradiation, confirming that X-rays induce cell death through both mitotic and apoptotic mechanisms. These data suggest that loss of ceramide production following X-rays represents an extranuclear mechanism for the development of radioresistance. Modulation of extranuclear signals may increase tumor cell killing following radiation and represent new cellular targets for cancer therapy.


Asunto(s)
Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Ceramidas/metabolismo , Linfocitos/efectos de la radiación , Alcaloides , Amidohidrolasas/antagonistas & inhibidores , Animales , Benzofenantridinas , División Celular/efectos de la radiación , Núcleo Celular/efectos de la radiación , Células Cultivadas , Ceramidasas , Relación Dosis-Respuesta en la Radiación , Endocannabinoides , Inhibidores Enzimáticos/farmacología , Etanolaminas/farmacología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , Ácidos Oléicos , Fenantridinas/farmacología , Esfingosina/análogos & derivados , Esfingosina/farmacología , Factores de Tiempo
12.
Semin Oncol ; 24(6): 633-8, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9422260

RESUMEN

Improving local tumor control is a goal of modern radiotherapy that will directly impact a majority of cancer patients. Numerous attempts to develop radiosensitizing agents have, however, met with limited clinical success. Recent advances in gene therapeutic approaches have allowed researchers to combine gene therapy with ionizing radiation to enhance tumor cell killing. In the following review, we discuss the current advances in the use of radiation-inducible promoters, the use of pro-drug converting enzymes, and cytotoxic cytokines to enhance the therapeutic efficacy of ionizing radiation.


Asunto(s)
Terapia Genética/tendencias , Neoplasias/terapia , Oncología por Radiación/tendencias , Radioterapia/tendencias , Adenoviridae , Animales , Terapia Combinada , Regulación de la Expresión Génica , Humanos , Radiación Ionizante , Fármacos Sensibilizantes a Radiaciones
13.
Cancer Res ; 56(12): 2711-4, 1996 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8665499

RESUMEN

We report that WEHI-231 undergo apoptosis following exposure to the protein kinase C inhibitors chelerythrine chloride and calphostin C. Following the addition of chelerythrine or calphostin C to WEHI-231 cells, ceramide production increased over baseline levels with a concurrent decrease in sphingomyelin. More detailed examinations determined that the ceramide accumulation resulted from activation of neutral, but not acidic, sphingomyelinase. These results suggest an antagonistic relationship between protein kinase C activity and ceramide in the signaling events preceding apoptosis.


Asunto(s)
Apoptosis , Ceramidas/biosíntesis , Naftalenos/farmacología , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Esfingomielinas/metabolismo , Alcaloides , Benzofenantridinas , Células Cultivadas , Hidrólisis , Esfingomielina Fosfodiesterasa/metabolismo
14.
Adv Exp Med Biol ; 406: 39-55, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8910670

RESUMEN

WEHI-231, a murine B-cell lymphoma, readily undergoes programmed cell death following surface immunoglobulin (Ig) cross-linking [1]. Ceramide has been shown to induce apoptosis in WEHI-231 following its exposure to anti-lg antibodies, dexamethasone, and irradiation [2]. Recently, Haimovitz-Friedman et al. have demonstrated in endothelial cells that PMA not only prevented ceramide mediated apoptosis, but inhibited the generation of ceramide following irradiation [3]. In this paper we use highly specific PKC inhibitors to explore the connection between PKC activity, ceramide signaling and apoptosis. Both chelerythrine chloride and calphostin C triggered rapid apoptosis in WEHI-231 and acted in synergy with exogenous ceramide to induce apoptosis. Detailed studies of chelerythrine's mechanism of action revealed that 30 minutes following addition of 10 microM chelerythrine, sphingomyelin and phosphatidylcholine (PC) mass decreased confirming our previous findings of neutral, but not acidic, sphingomyelinase activation following treatment with PKC inhibitors [4]. The novel observation that inhibition of PKC isoforms present in WEHI-231 leads to a rapid rise in cellular ceramide as a results of sphingomyelin hydrolysis further suggests an antagonistic relationship between PKC activity and ceramide in the signaling events preceding apoptosis.


Asunto(s)
Apoptosis/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Alcaloides , Animales , Benzofenantridinas , Sinergismo Farmacológico , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Linfoma de Células B/patología , Ratones , Naftalenos/farmacología , Fenantridinas/farmacología , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo , Esfingosina/farmacología , Células Tumorales Cultivadas
15.
Hum Mol Genet ; 3(11): 2005-10, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7874118

RESUMEN

Mutations or loss of the APC tumor-suppressor gene is important for the development of colorectal polyps and cancers, but little is known about the function of this gene in normal tissue. To study the role of APC and other genes in colonocytes in vivo, a system was developed whereby transient expression of genes is established in normal rodent colonic epithelium, using liposomal gene delivery by rectal catheter infusion. Expression of a beta-galactosidase reporter gene and of the human APC gene under a constitutive promoter is demonstrated. A high efficiency of transfection is maintained, with close to 100% of epithelial cells expressing the introduced gene. Expression is transient and does not persist beyond 4 days, consistent with the normal turnover time of gut epithelium, but it can be maintained by repeated treatments. Human APC was expressed for three weeks under these conditions at approximately one-tenth the level of the endogenous APC gene, and no toxicity was observed beyond that attributed to repeated rectal enemas. These results reveal that in vivo expression of exogenous gene is feasible using a liposomal delivery system and suggest a method to further study the physiologic role of APC or other genes in the interrelated process of colonic epithelial proliferation and differentiation.


Asunto(s)
Colon/química , Expresión Génica/genética , Técnicas de Transferencia de Gen , Genes APC , Liposomas/química , Animales , Secuencia de Bases , Colon/enzimología , Humanos , Mucosa Intestinal/química , Mucosa Intestinal/enzimología , Masculino , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , beta-Galactosidasa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA