Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Radiat Res ; 201(2): 174-187, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38329819

RESUMEN

Resistance to radiation remains a significant clinical challenge in non-small cell lung carcinoma (NSCLC). It is therefore important to identify the underlying molecular and cellular features that drive acquired resistance. We generated genetically matched NSCLC cell lines to investigate characteristics of acquired resistance. Murine Lewis lung carcinoma (LLC) and human A549 cells acquired an approximate 1.5-2.5-fold increase in radiation resistance as compared to their parental match, which each had unique intrinsic radio-sensitivities. The radiation resistance (RR) was reflected in higher levels of DNA damage and repair marker γH2AX and reduced apoptosis induction after radiation. Morphologically, we found that radiation resistance A549 (A549-RR) cells exhibited a greater nucleus-to-cytosol (N/C) ratio as compared to its parental counterpart. Since the N/C ratio is linked to the differentiation state, we next investigated the epithelial-to-mesenchymal transition (EMT) phenotype and cellular plasticity. We found that A549 cells had a greater radiation-induced plasticity, as measured by E-cadherin, vimentin and double-positive (DP) modulation, as compared to LLC. Additionally, migration was suppressed in A549-RR cells, as compared to A549 cells. Subsequently, we confirmed in vivo that the LLC-RR and A549-RR cells are also more resistance to radiation than their isogenic-matched counterpart. Moreover, we found that the acquired radiation resistance also induced resistance to cisplatin, but not carboplatin or oxaliplatin. This cross-resistance was attributed to induced elevation of thiol levels. Gamma-glutamylcysteine synthetase inhibitor buthionine sulfoximine (BSO) sensitized the resistant cells to cisplatin by decreasing the amount of thiols to levels prior to obtaining acquired radiation resistance. By generating radiation-resistance genetically matched NSCLC we were able to identify and overcome cisplatin cross-resistance. This is an important finding arguing for combinatorial treatment regimens including glutathione pathway disruptors in patients with the potential of improving clinical outcomes in the future.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Animales , Ratones , Cisplatino/farmacología , Cisplatino/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Carboplatino , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral
2.
J Appl Genet ; 65(1): 95-101, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37917375

RESUMEN

Burkitt lymphoma (BL) is a highly aggressive lymphoma that mainly affects children and young adults. Chemotherapy is effective in young BL patients but the outcome in adults is less satisfactory. Therefore, there is a need to enhance the cytotoxic effect of drugs used in BL treatment. Glutathione (GSH) is an important antioxidant involved in processes such as regulation of oxidative stress and drug detoxification. Elevated GSH levels have been observed in many cancers and were associated with chemoresistance. We previously identified GCLC, encoding an enzyme involved in GSH biosynthesis, as an essential gene in BL. We now confirm that knockout of GCLC decreases viability of BL cells and that the GCLC protein is overexpressed in BL tissues. Moreover, we demonstrate that buthionine sulfoximine (BSO), a known inhibitor of GCLC, decreases growth of BL cells but does not affect control B cells. Furthermore, we show for the first time that BSO enhances the cytotoxicity of compounds commonly used in BL treatment, doxorubicin, and cyclophosphamide. Given the fact that BSO itself was not toxic to control cells and well-tolerated in clinical trials, combination of chemotherapy with BSO may allow reduction of the doses of cytotoxic drugs required to obtain effective responses in BL patients.


Asunto(s)
Linfoma de Burkitt , Glutamato-Cisteína Ligasa , Niño , Humanos , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/genética , Dominio Catalítico , Ciclofosfamida/farmacología , Doxorrubicina/farmacología , Glutatión/metabolismo
3.
J Toxicol Environ Health B Crit Rev ; 26(8): 417-441, 2023 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-37606035

RESUMEN

Buthionine sulfoximine (BSO) is a synthetic amino acid that blocks the biosynthesis of reduced glutathione (GSH), an endogenous antioxidant cellular component present in tumor cells. GSH levels have been associated with tumor cell resistance to chemotherapeutic drugs and platinum compounds. Consequently, by depleting GSH, BSO enhances the cytotoxicity of chemotherapeutic agents in drug-resistant tumors. Therefore, the aim of this study was to conduct a systematic review with meta-analysis of preclinical studies utilizing BSO in cancer treatments. The systematic search was carried out using the following databases: PubMed, Web of Science, Scopus, and EMBASE up until March 20, 2023, in order to collect preclinical studies that evaluated BSO, alone or in association, as a strategy for antineoplastic therapy. One hundred nine investigations were found to assess the cytotoxic potential of BSO alone or in combination with other compounds. Twenty-one of these met the criteria for performing the meta-analysis. The evidence gathered indicated that BSO alone exhibits cytotoxic activity. However, this compound is generally used in combination with other antineoplastic strategies, mainly chemotherapy ones, to improve cytotoxicity to carcinogenic cells and treatment efficacy. Finally, this review provides important considerations regarding BSO use in cancer treatment conditions, which might optimize future studies as a potential adjuvant antineoplastic therapeutic tool.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Metionina Sulfoximina/uso terapéutico , Metionina Sulfoximina/toxicidad , Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
4.
Theranostics ; 10(1): 371-383, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31903126

RESUMEN

Rationale: Despite considerable advances, the reactive oxygen species (ROS)-mediated cancer treatment suffers from the problems of up-regulation of adaptive antioxidants in cancer cells as well as side effects to normal cells. Therefore, development of a new generation of cancer-specific nanomedicine capable of amplifying oxidative stress would be of great interest for accurate and effective cancer treatment. Methods: Herein, transferrin (Tf)-decorated, dihydroartemisinin (DHA), L-buthionine-sulfoximine (BSO), and CellROX-loaded liposomal nanoparticles (Tf-DBC NPs) were developed for precise cancer theranositcs. Tf-DBC NPs could specifically recognize cancer cells via Tf-Tf receptor binding and be uptaken into the lysosomes of cancer cells, where Tf-DBC NPs were activated to release Fe(II), DHA, and BSO. ROS was generated by DHA in the presence of Fe(II), and GSH was depleted by BSO to disrupt the redox balance in cancer cells. Furthermore, CellROX, as a fluorescent probe for imaging of intracellular oxidative stress, was used to monitor the therapeutic efficacy. Results: The integration of Tf, DHA, and BSO into the acidic pH-responsive liposomes selectively and effectively killed cancer cells and prevented the oxidative injury to normal cells. The high oxidative state was visualized at the tumor site and the amplification of oxidative stress enabled tumor eradication by Tf-DBC NPs, demonstrating the successful implementation of this novel strategy in vivo. Conclusion: Our study provides a new paradigm for the design of ROS-mediated therapeutics and offers a promising perspective for precise cancer treatment.


Asunto(s)
Artemisininas , Butionina Sulfoximina , Glutatión/metabolismo , Liposomas/química , Neoplasias/terapia , Especies Reactivas de Oxígeno/metabolismo , Animales , Artemisininas/farmacología , Artemisininas/uso terapéutico , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Portadores de Fármacos/química , Femenino , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Medicina de Precisión , Transferrina/química
5.
Arch Pharm Res ; 42(11): 990-999, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31482490

RESUMEN

In this study, we investigated the synergistic effect of L-buthionine sulfoximine (BSO) on the chlorin e6 (Ce6)-based photodynamic therapy (PDT) of cancer cells. Among various cancer cells, HCT116 cells have highest intracellular L-glutathione (GSH) level and SNU478 cells showed the lowest GSH level. BSO alone showed negligible intrinsic cytotoxicity against CCD986sk cells. Since HCT116 and SNU478 cells showed the highest and the lowest intracellular GSH levels, respectively, those were used to test synergistic effect on the Ce6-based PDT. In the absence of light, BSO and Ce6 combination did not practically increase reactive oxygen species (ROS) in either of HCT116 or SNU478 cells, while light irradiation increased ROS level dose-dependently. 10 µM BSO treatment significantly depleted total GSH level in cancer cells, i.e. total GSH level decreased to one-fourth of the control in HCT116 cells while it decreased to two-fifth of the control treatment at SNU478 cell. BSO showed synergistic effect on the ROS production in HCT116 cells while it has practically no benefits in ROS production of SNU478 cells. No synergistic effect was observed in viability of SNU478 cells because BSO itself was cytotoxic to SNU478 cells. However, BSO had negligible cytotoxicity against HCT116 cells and showed synergistic anticancer effect in combination with Ce6-based PDT. Furthermore, the addition of glutathione reduced ethyl ester (GSH-OEt), recovered intracellular GSH level, and cell viability with reduced the intracellular ROS level. We suggest that synergistic effect of BSO in the Ce6-based PDT should be considered with intrinsic intracellular GSH level of cancer cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Butionina Sulfoximina/farmacología , Fotoquimioterapia/métodos , Porfirinas/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Butionina Sulfoximina/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Clorofilidas , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Glutatión/metabolismo , Células HCT116 , Humanos , Ratones , Porfirinas/uso terapéutico , Especies Reactivas de Oxígeno
6.
Nano Lett ; 19(2): 805-815, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30592897

RESUMEN

Amplification of intracellular oxidative stress has been found to be an effective strategy to induce cancer cell death. To this end, we prepare a unique type of ultrasmall gallic acid-ferrous (GA-Fe(II)) nanocomplexes as the catalyst of Fenton reaction to enable persistent conversion of H2O2 to highly cytotoxic hydroxyl radicals (•OH). Then, both GA-Fe(II) and l-buthionine sulfoximine (BSO), an inhibitor of glutathione (GSH) synthesis, are coencapsulated within a stealth liposomal nanocarrier. Interestingly, the obtained BSO/GA-Fe(II)@liposome is able to efficiently amplify intracellular oxidative stress via increasing •OH generation and reducing GSH biosynthesis. After chelating with 99mTc4+ radioisotope, such BSO/GA-Fe(II)@liposome could be tracked under in vivo single-photon-emission-computed-tomography (SPECT) imaging, which illustrates the time-dependent tumor homing of such liposomal nanoparticles after intravenous injection. With GA-Fe(II)-mediated •OH production and BSO-mediated GSH depletion, treatment with such BSO/GA-Fe(II)@liposome would lead to dramatically enhanced intratumoral oxidative stresses, which then result in remarkably improved therapeutic efficacies of concurrently applied chemotherapy or radiotherapy. This work thus presents the concise fabrication of biocompatible BSO/GA-Fe(II)@liposome as an effective adjuvant nanomedicine to promote clinically used conventional cancer chemotherapy and radiotherapy, by greatly amplifying the intratumoral oxidative stress.


Asunto(s)
Butionina Sulfoximina/uso terapéutico , Compuestos Ferrosos/uso terapéutico , Ácido Gálico/uso terapéutico , Glutatión/antagonistas & inhibidores , Neoplasias Mamarias Animales/terapia , Estrés Oxidativo/efectos de los fármacos , Animales , Butionina Sulfoximina/administración & dosificación , Línea Celular Tumoral , Femenino , Compuestos Ferrosos/administración & dosificación , Ácido Gálico/administración & dosificación , Glutatión/metabolismo , Radical Hidroxilo/metabolismo , Liposomas/química , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/radioterapia , Ratones , Ratones Endogámicos BALB C , Tomografía Computarizada de Emisión de Fotón Único
7.
Biomed Pharmacother ; 103: 1223-1230, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29864902

RESUMEN

Diabetic nephropathy (DN) is a common complication of diabetes and the major cause of chronic kidney disease. Cyanidin 3-glucoside (C3G) is the most widespread anthocyanin in nature. In the present study, we aimed to investigate the possible effects of C3G on DN in db/db mice. We found that body weights and high levels of fasting blood glucose, serum insulin, C-peptide, glycosylated hemoglobin A1c, and systolic blood pressure in diabetic mice were significantly reduced by C3G. C3G also reduced the ratio of kidney to body weight and the levels of blood urea nitrogen (BUN), serum creatinine, urinary albumin content and albumin/creatinine ratio (ACR), ameliorated the pathological changes of kidneys, reduced the surface area of Bowman's capsule, glomerular tuft, Bowman's space, and decreased renal expression of collagen IV, fibronectin, transforming growth factor ß 1 (TGFß1), matrix metalloprotein 9 (MMP9) and α-smooth muscle actin (α-SMA) in db/db mice. The Lee's index, perirenal white adipose tissue weight, and high levels of blood and renal triglyceride and cholesterol were decreased by C3G. Moreover, C3G reduced systemic levels and renal expression of tumor necrosis factor ɑ (TNFɑ), IL-1ɑ, and monocyte chemotactic protein-1 (MCP-1), indicating the inhibition of inflammation. Furthermore, C3G increased glutathione (GSH) level and decreased GSSG level in kidneys of diabetic mice. The renal mRNA expression of glutamate-cysteine ligase catalytic subunit (GCLC) and glutamate-cysteine ligase modifier subunit (GCLM) was increased by C3G in diabetic mice. Buthionine sulphoximine (BSO), an inhibitor of GSH synthesis, inhibited the effects of C3G on glucose metabolic dysfunction and DN. The data demonstrates that enhancement of GSH pool is involved in the renal-protective effects of C3G. Overall, C3G could be a promising therapeutic option for attenuation of diabetes and DN.


Asunto(s)
Antocianinas/uso terapéutico , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Glucósidos/uso terapéutico , Glutatión/metabolismo , Animales , Antocianinas/farmacología , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/patología , Fibrosis , Glucosa/metabolismo , Glucósidos/farmacología , Inflamación/complicaciones , Inflamación/patología , Riñón/lesiones , Riñón/patología , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Obesidad/complicaciones , Obesidad/patología , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico
8.
PLoS One ; 12(3): e0174271, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28362858

RESUMEN

BACKGROUND & AIMS: Acquisition of anoikis resistance is a prerequisite for metastasis in hepatocellular carcinoma (HCC). However, little is known about how energy metabolism and antioxidant systems are altered in anoikis-resistant (AR) HCC cells. We evaluated anti-tumor effects of a combination treatment of 3-bromopyruvate (3-BP) and buthionine sulfoximine (BSO) in AR HCC cells. METHODS: We compared glycolysis, reactive oxygen species (ROS) production, and chemoresistance among Huh-BAT, HepG2 HCC cells, and the corresponding AR cells. Expression of hexokinase II, gamma-glutamylcysteine synthetase (rGCS), and epithelial-mesenchymal transition (EMT) markers in AR cells was assessed. Anti-tumor effects of a combination treatment of 3-BP and BSO were evaluated in AR cells and an HCC xenograft mouse model. RESULTS: AR HCC cells showed significantly higher chemoresistance, glycolysis and lower ROS production than attached cells. Expression of hexokinase II, rGCS, and EMT markers was higher in AR HCC cells than attached cells. A combination treatment of 3-BP/BSO effectively suppressed proliferation of AR HCC cells through apoptosis by blocking glycolysis and enhancing ROS levels. In xenograft mouse models, tumor growth derived from AR HCC cells was significantly suppressed in the group treated with 3-BP/BSO compared to the group treated with 3-BP or sorafenib. CONCLUSIONS: These results demonstrated that a combination treatment of 3-BP/BSO had a synergistic anti-tumor effect in an AR HCC model. This strategy may be an effective adjuvant therapy for patients with sorafenib-resistant HCC.


Asunto(s)
Anoicis/efectos de los fármacos , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Piruvatos/farmacología , Piruvatos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Hep G2 , Humanos , Niacinamida/análogos & derivados , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Sorafenib
9.
Pediatr Blood Cancer ; 63(8): 1349-56, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27092812

RESUMEN

BACKGROUND: Myeloablative therapy for high-risk neuroblastoma commonly includes melphalan. Increased cellular glutathione (GSH) can mediate melphalan resistance. Buthionine sulfoximine (BSO), a GSH synthesis inhibitor, enhances melphalan activity against neuroblastoma cell lines, providing the rationale for a Phase 1 trial of BSO-melphalan. PROCEDURES: Patients with recurrent/resistant high-risk neuroblastoma received BSO (3 gram/m(2) bolus, then 24 grams/m(2) /day infusion days -4 to -2), with escalating doses of intravenous melphalan (20-125 mg/m(2) ) days -3 and -2, and autologous stem cells day 0 using 3 + 3 dose escalation. RESULTS: Among 28 patients evaluable for dose escalation, one dose-limiting toxicity occurred at 20 mg/m(2) melphalan (grade 3 aspartate aminotransferase/alanine aminotransferase) and one at 80 mg/m(2) (streptococcal bacteremia, grade 4 hypotension/pulmonary/hypocalcemia) without sequelae. Among 25 patients evaluable for response, there was one partial response (PR) and two mixed responses (MRs) among eight patients with prior melphalan exposure; one PR and three MRs among 16 patients without prior melphalan; one stable disease with unknown melphalan history. Melphalan pharmacokinetics with BSO were similar to reports for melphalan alone. Melphalan Cmax for most patients was below the 10 µM concentration that showed neuroblastoma preclinical activity with BSO. CONCLUSIONS: BSO (75 gram/m(2) ) with melphalan (125 mg/m(2) ) is tolerable with stem cell support and active in recurrent/refractory neuroblastoma. Further dose escalation is feasible and may increase responses.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Butionina Sulfoximina/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Melfalán/uso terapéutico , Agonistas Mieloablativos/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Adolescente , Butionina Sulfoximina/efectos adversos , Niño , Preescolar , Sinergismo Farmacológico , Femenino , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutatión/uso terapéutico , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Melfalán/efectos adversos , Melfalán/farmacocinética , Recurrencia Local de Neoplasia/tratamiento farmacológico
10.
Artículo en Inglés | MEDLINE | ID: mdl-25613502

RESUMEN

The induction of heme oxygenase (HO), the rate-limiting enzyme in heme degradation, occurs as an adaptative response to oxidative stress and is consequent to decrease in cellular glutathione levels. Our previous studies demonstrated significant increase in survival rates of rats treated with glutathione depletors and submitted to transient cerebral ischemia. The aim of the present research was to test the effects of L-Buthionine sulfoximine (BSO), a glutathione depletor, during cerebral post-ischemic reperfusion. Cerebral ischemia was induced by bilateral clamping of common carotid arteries for 20 min. Each sample was used for glutathione ad lipid peroxidation level dosage and for evaluating the expression of heme oxygenase both after a single subcutaneous administration of BSO and without treatment. In the same experimental conditions, endothelial, inducible and neuronal Nitric Oxide Synthase (eNOS, iNOS and nNOS) and Dimethylarginine Dimethyl amine Hydrolases (DDAH-1 and DDAH-2) were also evaluated. Results obtained in the present study suggested that HO-1 over-expression may be implicated in the protective effect of BSO in post-ischemic reperfusion brain damage, although the involvement of other important stress mediators cannot be ruled out.


Asunto(s)
Isquemia Encefálica/complicaciones , Isquemia Encefálica/prevención & control , Butionina Sulfoximina/uso terapéutico , Glutatión/deficiencia , Fármacos Neuroprotectores/uso terapéutico , Daño por Reperfusión/complicaciones , Análisis de Varianza , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Encéfalo/metabolismo , Isquemia Encefálica/patología , Inyecciones Subcutáneas , Peroxidación de Lípido/efectos de los fármacos , Masculino , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
11.
Curr Drug Targets ; 16(1): 31-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25395102

RESUMEN

Aerobic metabolism of mammalian cells leads to the generation of reactive oxygen species (ROS). To cope with this toxicity, evolution provided cells with effective antioxidant systems like glutathione. Current anticancer therapies focus on the cancer dependence on oncogenes and non-oncogenes. Tumors trigger mechanisms to circumvent the oncogenic stress and to escape cell death. In this context we have studied 2-phenylethinesulfoxamine (PES), which disables the cell protective mechanisms to confront the proteotoxicity of damaged and unfolded proteins. Proteotoxic stress is increased in tumor cells, thus providing an explanation for the anticancer selectivity of PES. In addition, we have found that PES induces a severe oxidative stress and the activation of p53. The reduction of the cell content in glutathione by means of L-buthionine-sulfoximine (BSO) synergizes with PES. In conclusion, we have found that ROS constitutes a central element in a series of positive feed-back loops in the cell. ROS, p53, proteotoxicity, autophagy and mitochondrial dynamics are interconnected with the mechanisms leading to cell death, either apoptotic or necrotic. This network of interactions provides multiple targets for drug discovery and development in cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Descubrimiento de Drogas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Dinámicas Mitocondriales/efectos de los fármacos , Neoplasias/metabolismo , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo
12.
World J Gastroenterol ; 17(34): 3899-911, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22025878

RESUMEN

AIM: To evaluate the efficacy and the safety of azathioprine (AZA) and buthionine sulfoximine (BSO) by localized application into HepG2 tumor in vivo. METHODS: Different hepatoma and colon carcinoma cell lines (HepG2, HuH7, Chang liver, LoVo, RKO, SW-48, SW-480) were grown in minimal essencial medium supplemented with 10% fetal bovine serum and 1% antibiotic/antimycotic solution and maintained in a humidified 37 °C incubator with 5% CO2. These cells were pretreated with BSO for 24 h and then with AZA for different times. We examined the effects of this combination on some proteins and on cellular death. We also studied the efficacy and the safety of AZA (6 mg/kg per day) and BSO (90 mg/kg per day) in HepG2 tumor growth in vivo using athymic mice. We measured safety by serological markers such as aminotransferases and creatine kinase. RESULTS: The in vitro studies revealed a new mechanism of action for the AZA plus BSO combination in the cancer cells compared with other thiopurines (6-mercaptopurine, 6-methylmercaptopurine, 6-thioguanine and 6-methylthioguanine) in combination with BSO. The cytotoxic effect of AZA plus BSO in HepG2 cells resulted from necroptosis induction in a mitochondrial-dependent manner. From kinetic studies we suggest that glutathione (GSH) depletion stimulates c-Jun amino-terminal kinase and Bax translocation in HepG2 cells with subsequent deregulation of mitochondria (cytochrome c release, loss of membrane potential), and proteolysis activation leading to loss of membrane integrity, release of lactate dehydrogenase and DNA degradation. Some of this biochemical and cellular changes could be reversed by N-acetylcysteine (a GSH replenisher). In vivo studies showed that HepG2 tumor growth was inhibited when AZA was combined with BSO. CONCLUSION: Our studies suggest that a combination of AZA plus BSO could be useful for localized treatment of hepatocellular carcinoma as in the currently used transarterial chemoembolization method.


Asunto(s)
Antimetabolitos Antineoplásicos , Azatioprina , Butionina Sulfoximina , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Azatioprina/farmacología , Azatioprina/uso terapéutico , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Bovinos , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias
13.
J Steroid Biochem Mol Biol ; 114(1-2): 33-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19167492

RESUMEN

L-Buthionine sulfoximine (BSO) is a potent inhibitor of glutathione biosynthesis and studies have shown that it is capable of enhancing the apoptotic effects of several chemotherapeutic agents. Previous studies have shown that long-term antihormonal therapy leads to acquired drug resistance and that estrogen, which is normally a survival signal, is a potent apoptotic agent in these resistant cells. Interestingly, we have developed an antihormone-resistant breast cancer cell line, MCF-7:2A, which is resistant to estrogen-induced apoptosis but has elevated levels of glutathione. In the present study, we examined whether BSO is capable of sensitizing antihormone-resistant MCF-7:2A cells to estrogen-induced apoptosis. Our results showed that treatment of MCF-7:2A cells with 1nM E2 plus 100muM BSO combination for 1 week reduced the growth of these cells by almost 80-90% whereas the individual treatments had no significant effect on growth. TUNEL and 4',6-diamidino-2-phenylindole (DAPI) staining showed that the inhibitory effect of the combination treatment was due to apoptosis. Our data indicates that glutathione participates in retarding apoptosis in antihormone-resistant human breast cancer cells and that depletion of this molecule by BSO may be critical in predisposing resistant cells to estrogen-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Butionina Sulfoximina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Estradiol/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Butionina Sulfoximina/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Femenino , Glutatión/metabolismo , Humanos , Estructura Molecular , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico
14.
Salvador; s.n; 2009. 57 p. ilus.
Tesis en Portugués | LILACS | ID: lil-571294

RESUMEN

Segundo a Organização Mundial de Saúde (2002), a esquistossomose é a segunda maior doença tropical, causadora de 200 a 300 mil mortes por ano. Mesmo apresentando alguns quimioterápicos eficazes para o tratamento, como o praziquantel (PZQ) e a oxamniquina (OXQ), ocorrem muitos casos refratários e efeitos colaterais. Diante deste contexto, é necessária a busca racional de novos medicamentos e combinações para o tratamento desta doença. Uma possível solução é o estudo de drogas relacionadas com o estresse oxidativo do patógeno. Dentre estas podem ser analisadas: a artemisinina (ART) , que induz uma maior produção de radicais livres por inibição da formação da hemozoína em Plasmodium falciparum; a butionina sulfoximina (850), que impede a produção da glutationa; além do dietilditiocarbamato de sódio (DDC), que age como inibidor das superóxido dismutases. Portanto, o objetivo deste trabalho foi testar in vitro essas drogas, isoladas e combinadas, analisando a atividade das superóxido dismutases, alterações morfológicas e produção de hemozoína em vermes adultos, além de avaliar a toxicidade em esplenócitos. A ART apresentou efeito esquistossomicida em concentrações elevadas. O DDC mostrou um efeito esquistossomicida satifatório e inibiu as superóxido dismutases. Quanto à associação ART - DDC, esta inibiu a formação da hemozoína, apresentou danos nos tegumentos dos vermes e não apresentou citotoxicidade significativa. Estes resultados indicam que estas drogas são viáveis para estudos in vivo, podendo ser uma nova alternativa quimioterápica para esta patologia.


Asunto(s)
Humanos , Artemisininas/metabolismo , Butionina Sulfoximina/uso terapéutico , Ditiocarba/efectos adversos , Esquistosomiasis mansoni/parasitología , Esquistosomiasis/parasitología
15.
Antimicrob Agents Chemother ; 52(5): 1837-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18332173

RESUMEN

L-buthionine (S,R)-sulfoximine (BSO) at a dose of 220 mg/kg of body weight/day showed an anti-Trypanosoma cruzi effect in infected mice, increasing their survival rate and decreasing the parasitemias and parasite burden in the hearts. Treatment with BSO plus nifurtimox caused an increase in the survival rate in comparison to the rates with treatment with each drug alone.


Asunto(s)
Butionina Sulfoximina/farmacología , Enfermedad de Chagas/tratamiento farmacológico , Nifurtimox/farmacología , Trypanosoma cruzi/efectos de los fármacos , Enfermedad Aguda , Animales , Butionina Sulfoximina/uso terapéutico , Enfermedad de Chagas/mortalidad , Enfermedad de Chagas/parasitología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Ratones , Ratones Endogámicos BALB C , Nifurtimox/uso terapéutico , Tasa de Supervivencia , Tripanocidas/farmacología
16.
Int J Oncol ; 32(1): 121-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18097550

RESUMEN

Depletion of glutathione (GSH) by buthionine sulfoximine (BSO) has been reported to be toxic against some cancer cells and to sensitize many tumours including neuroblastoma (NB) to anticancer drugs. The balance between the production rate of reactive oxygen species (ROS) and the function of GSH affects the intracellular reduction-oxidation status, which is crucial for the regulation of several cellular physiological functions. To assess the role of glutathione in neuroblastoma therapy, the effect of sublethal concentrations of BSO was studied in a panel of neuroblastoma cell lines characterized by different MYCN status. We found that GSH depletion per se not accompanied by ROS overproduction, does not affect cell survival, and is not genotoxic but induces HO-1 expression in GI-ME-N cell line, a representative example of MYCN non-amplified NB cells, having the highest basal levels of GSH among the tested NB lines. These observations might open a novel therapeutic window based on the possibility of modulating the cellular 'activity' of GSH.


Asunto(s)
Butionina Sulfoximina/farmacología , Glutatión/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/mortalidad , Butionina Sulfoximina/uso terapéutico , Línea Celular Tumoral , Disulfuro de Glutatión/metabolismo , Hemo-Oxigenasa 1/biosíntesis , Humanos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/tratamiento farmacológico , Proteínas Nucleares/análisis , Proteínas Oncogénicas/análisis , Especies Reactivas de Oxígeno
17.
Leuk Res ; 31(9): 1293-301, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17306873

RESUMEN

Raised intracellular glutathione is one characteristics of high-risk childhood acute lymphoblastic leukemia (ALL). Depletion of glutathione by buthionine sulfoximine (BSO) has been reported to be toxic against some cancer cells. To assess the role of glutathione in ALL, the toxicity of BSO was studied in B-precursor ALL cell lines. BSO increased oxidative stress equally in all cell lines; however mitochondrial depolarization was observed only in BSO-sensitive cells. BSO up-regulated Bcl-2 protein, and antagonized the anti-ALL effect of prednisolone in BSO-resistant cells. A lack of mitochondrial death-signal activation by oxidative stress seemed to be associated with BSO-resistance in ALL.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Butionina Sulfoximina/uso terapéutico , Resistencia a Antineoplásicos , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Antiinflamatorios/farmacología , Proliferación Celular/efectos de los fármacos , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Oxidantes/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Prednisolona/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
18.
Parasitology ; 131(Pt 6): 747-57, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16336728

RESUMEN

Co-treatment of mice infected with different strains of Leishmania donovani with a non-ionic surfactant vesicle formulation of buthionine sulfoximine (BSO-NIV), and sodium stibogluconate (SSG), did not alter indicators of Th1 or Th2 responses but did result in a significant strain-independent up-regulation of IL6 and nitrite levels by stimulated splenocytes from treated mice compared to controls. The efficacy of BSO-NIV/SSG treatment was dependent on the host being able to mount a respiratory burst indicating that macrophages are important in controlling the outcome of treatment. In vitro studies showed that SSG resistance was associated with a greater resistance to killing by activated macrophages, treatment with hydrogen peroxide or potassium antimony tartrate. Longitudinal studies showed that a SSG resistant (SSG-R) strain was more virulent than a SSG susceptible (SSG-S) strain, resulting in significantly higher parasite burdens by 4 months post-infection. These results indicate that SSG exposure may favour the emergence of more virulent strains.


Asunto(s)
Gluconato de Sodio Antimonio/farmacología , Antiprotozoarios/farmacología , Leishmania donovani/efectos de los fármacos , Leishmania donovani/inmunología , Macrófagos/inmunología , Animales , Tartrato de Antimonio y Potasio/farmacología , Gluconato de Sodio Antimonio/uso terapéutico , Antiprotozoarios/uso terapéutico , Butionina Sulfoximina/farmacología , Butionina Sulfoximina/uso terapéutico , Cricetinae , Resistencia a Medicamentos , Femenino , Humanos , Peróxido de Hidrógeno/farmacología , Interleucina-6/análisis , Interleucina-6/biosíntesis , Leishmania donovani/patogenicidad , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/parasitología , Masculino , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Nitritos/análisis , Nitritos/metabolismo , Bazo/química , Bazo/parasitología , Virulencia
19.
Electrophoresis ; 26(6): 1155-62, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15706575

RESUMEN

A microchip electrophoresis method coupled with laser-induced fluorescence (LIF) detection was established for simultaneous determination of two kinds of intracellular signaling molecules (reactive oxygen species, ROS, and reduced glutathione, GSH) related to apoptosis and oxidative stress. As the probe dihydrorhodamine-123 (DHR-123) can be converted intracellularly by ROS to the fluorescent rhodamine-123 (Rh-123), and the probe naphthalene-2,3-dicarboxaldehyde (NDA) can react quickly with GSH to produce a fluorescent adduct, rapid determination of Rh-123 and GSH was achieved on a glass microchip within 27 s using a 20 mM borate buffer (pH 9.2). The established method was tested to measure the intracellular ROS and GSH levels in acute promyelocytic leukemia (APL)-derived NB4 cells. An elevation of intracellular ROS and depletion of GSH were observed in apoptotic NB4 cells induced by arsenic trioxide (As(2)O(3)) at low concentration (1-2 microM). Buthionine sulfoximine (BSO), in combination with As(2)O(3) enhanced the decrease of reduced GSH to a great extent. The combined treatment of As(2)O(3) and hydrogen peroxide (H(2)O(2)) led to an inverse relationship between the concentrations of ROS and GSH obtained, showing the proposed method can readily evaluate the generation of ROS, which occurs simultaneously with the consumption of the inherent antioxidant.


Asunto(s)
Apoptosis/fisiología , Electroforesis por Microchip/métodos , Glutatión/análisis , Leucemia Promielocítica Aguda/metabolismo , Especies Reactivas de Oxígeno/análisis , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/uso terapéutico , Butionina Sulfoximina/uso terapéutico , Colorantes Fluorescentes , Humanos , Rayos Láser , Leucemia Promielocítica Aguda/tratamiento farmacológico , Miniaturización , Óxidos/uso terapéutico , Reproducibilidad de los Resultados , Rodaminas , Transducción de Señal/fisiología , Espectrometría de Fluorescencia , Células Tumorales Cultivadas
20.
Eur J Cancer ; 40(13): 1934-40, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15315800

RESUMEN

The generation of reactive oxygen species (ROS) can be exploited therapeutically in the treatment of cancer. One of the first drugs to be developed that generates ROS was procarbazine. It is oxidised readily in an oxic environment to its azo derivative, generating ROS. Forty years ago, Berneis reported a synergistic effect in DNA degradation when procarbazine was combined with radiation; this was confirmed in preclinical in vivo modes. Early uncontrolled clinical trials suggested an enhancement of the radiation effect with procarbazine, but two randomised trials failed to confirm this. The role of ROS in cancer treatments and in the development of resistance to chemotherapy is now better understood. The possibility of exploiting ROS as a cancer treatment is re-emerging as a promising therapeutic option with the development of agents such as buthionine sulfoximine and motexafin gadolinium.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Procarbazina/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Butionina Sulfoximina/uso terapéutico , Terapia Combinada , ADN/metabolismo , Humanos , Metaloporfirinas/uso terapéutico , Neoplasias/radioterapia , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...