Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Immunopharmacol Immunotoxicol ; 36(2): 165-75, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24611750

RESUMEN

Myeloid-derived suppressor cells (MDSCs), one of the major orchestrators of immunosuppressive network are present in the tumor microenvironment suppress antitumor immunity by subverting Th1 response in tumor site and considered as a great obstacle for advancement of different cancer immunotherapeutic protocols. Till date, various pharmacological approaches have been explored to modulate the suppressive functions of MDSCs in vivo. The present study describes our endeavor to explore a possibility of eradicating MDSCs by the application of a copper chelate, namely copper N-(2-hydroxy acetophenone) glycinate (CuNG), previously found to be a potential immunomodulator that can elicit antitumorogenic Th1 response in doxorubicin-resistant EAC (EAC/Dox) bearing mice. Herein, we demonstrated that CuNG treatment could reduce Gr-1+CD11b+ MDSC accumulation in ascitic fluid and spleen of EAC/Dox tumor model. Furthermore, we found that CuNG mediated reduction in MDSCs is associated with induction of Th1 response and reduction in Treg cells. Moreover, we observed that CuNG could deplete MDSCs by inducing Fas-FasL mediated apoptotic cell death where death receptor Fas expression is enhanced in MDSCs and FasL is provided by activated T cells. However, MDSC expansion from bone marrow cells and their differentiation was not affected by CuNG. Altogether, these findings suggest that the immunomodulatory property of CuNG is attributed to, at least in part, by its selective cytotoxic action on MDSCs. So, this preclinical study unveils a new mechanism of regulating MDSC levels in drug-resistant cancer model and holds promise of translating the findings into clinical settings.


Asunto(s)
Antineoplásicos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Quelantes/farmacología , Cobre/inmunología , Cobre/farmacología , Células Mieloides/inmunología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Doxorrubicina/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Células Mieloides/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Receptor fas/inmunología
2.
Biometals ; 26(3): 517-34, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23733180

RESUMEN

Drug induced toxicity and drug resistance are the major impediments to successful application of cancer chemotherapy. Therefore, selective targeting of the key biochemical events of the malignant cells may have a great therapeutic potential in specifically kill the cancer cells. We have evaluated in vitro the cytotoxic efficacy of a previously reported copper complex viz. copper N-(2-hydroxy acetophenone) glycinate (CuNG) on different drug sensitive and resistant cancer cell lines by MTT, annexin V positivity and caspase 3 activation assays. We have also investigated the underlying signalling events in CuNG mediated apoptosis of cancer cells by Western blotting technique. We have found that CuNG preferentially induces apoptosis to malignant cells irrespective of drug sensitivity and spares the normal cells. Our studies disclose that CuNG causes cellular redox imbalance in cancer cells through depletion of intracellular GSH level. CuNG mediated depletion of intracellular GSH level induces mitochondrial superoxide generation, which detaches cyto C from mitochondrial membrane through lipid peroxidation. The detached cyto C then release into the extra mitochondrial milieu in Bax mediated pathway where CuNG facilitates the binding of Bax through dissociation of hexokinase II from mitochondrial membrane. The present study opens the possibility of developing effective chemotherapeutic drugs by synthesizing numerous chemical compounds capable of targeting cellular redox environment and thus specifically kills cancer cells of broad spectrum.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Quelantes/farmacología , Glutatión/metabolismo , Glicina/análogos & derivados , Compuestos Organometálicos/farmacología , Animales , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quelantes/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glicina/química , Glicina/farmacología , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Células K562 , Ratones , Células 3T3 NIH , Compuestos Organometálicos/química , Relación Estructura-Actividad
3.
Biomed Pharmacother ; 92: 509-518, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28575808

RESUMEN

Vanadium compounds are well known for their therapeutic interventions against several diseases. Various biochemical attributes of vanadium complexes inspired us to evaluate the cancer cell killing efficacy of the vanadium complex, viz., vanadyl N-(2-hydroxyacetophenone) glycinate [VO(NG)2]. Previously we showed that VO(NG)2 is an effective anticancer agent in in vitro and in vivo cancer models and imposed miniscule side effects. Herein we report that VO(NG)2 is significantly cytotoxic to various cancer cell lines. Furthermore, this redox active vanadyl complex altered the redox homeostatsis of many human cancer cell lines significantly. VO(NG)2 actuates programmed cell death in human colorectal carcinoma cells(HCT-116) through mitochondrial outer membrane permeabilization but in caspase independent manner, possibly by altering cellular redox status and by inflicting DNA damage. Thus, the present work is an attempt to provide many evidences regarding the potent and selective chemotherapeutic efficacy of the novel VO(NG)2.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Vanadio/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/fisiología , Células Cultivadas , Neoplasias Colorrectales/tratamiento farmacológico , Células HCT116 , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Bases de Schiff/química , Bases de Schiff/farmacología , Bases de Schiff/uso terapéutico , Oligoelementos/química , Oligoelementos/farmacología , Oligoelementos/uso terapéutico , Vanadio/química , Vanadio/uso terapéutico
4.
Environ Toxicol Pharmacol ; 56: 383-392, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29145169

RESUMEN

Cancer chemotherapy suffers from selectivity and undesired toxicity of the drugs. Since zinc is a biocompatible tracer element and cysteine derivatives are used in cancer chemoprevention, we intend to develop a complex of zinc and cysteine-derivatives as potent, non-toxic anticancer agents. Herein, we synthesized and characterized cysteine based ligand, 2-[(2-Hydroxy-3-methoxy-benzylidene)-amino]-3-mercapto-propionic acid and its Zn-complex, which are found to be non-toxic towards normal human PBMC. Data also revealed that only Zn-complex exhibited remarkable apoptosis in drug-sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 cancer cells as assessed by MTT, Cell cycle and AnnexinV binding assay. Moreover, Zn-complex altered ROS and GSH level of the respective cell lines. Finally, treatment of Zn-complex in Swiss albino mice did not show any systemic toxicity in preliminary trials in normal mice and remarkably increased the life-span of EAC bearing mice. In conclusion, the synthesized Zn-complex may be developed for efficacious, multidrug resistance reversal, non-toxic chemotherapeutic agents in future.


Asunto(s)
Carcinoma de Ehrlich/tratamiento farmacológico , Complejos de Coordinación/administración & dosificación , Cisteína/química , Zinc/química , Animales , Apoptosis , Carcinoma de Ehrlich/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Progresión de la Enfermedad , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Ratones , Especies Reactivas de Oxígeno/metabolismo , Bases de Schiff/química , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Free Radic Res ; 50(4): 426-46, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26733073

RESUMEN

Multidrug resistance (MDR) in cancer represents a variety of strategies employed by tumor cells to evade the beneficial cytotoxic effects of structurally different anticancer drugs and thus confers impediments to the successful treatment of cancers. Efflux of drugs by MDR protein-1, functional P-glycoprotein and elevated level of reduced glutathione confer resistance to cell death or apoptosis and thus provide a possible therapeutic target for overcoming MDR in cancer. Previously, we reported that a Schiff base ligand, potassium-N-(2-hydroxy 3-methoxy-benzaldehyde)-alaninate (PHMBA) overcomes MDR in both in vivo and in vitro by targeting intrinsic apoptotic/necrotic pathway through induction of reactive oxygen species (ROS). The present study describes the synthesis and spectroscopic characterization of a copper chelate of Schiff base, viz., copper (II)-N-(2-hydroxy-3-methoxy-benzaldehyde)-alaninate (CuPHMBA) and the underlying mechanism of cell death induced by CuPHMBA in vitro. CuPHMBA kills both the drug-resistant and sensitive cell types irrespective of their drug resistance phenotype. The cell death induced by CuPHMBA follows apoptotic pathway and moreover, the cell death is associated with intrinsic mitochondrial and extrinsic receptor-mediated pathways. Oxidative stress plays a pivotal role in the process as proved by the fact that antioxidant enzyme; polyethylene glycol conjugated-catalase completely blocked CuPHMBA-induced ROS generation and abrogated cell death. To summarize, the present work provides a compelling rationale for the future clinical use of CuPHMBA, a redox active copper chelate in the treatment of cancer patients, irrespective of their drug-resistance status.


Asunto(s)
Antineoplásicos/farmacología , Quelantes/farmacología , Complejos de Coordinación/farmacología , Cobre/química , Especies Reactivas de Oxígeno/agonistas , Bases de Schiff/farmacología , Alanina/análogos & derivados , Animales , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Benzaldehídos/química , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/patología , Catalasa/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quelantes/síntesis química , Complejos de Coordinación/antagonistas & inhibidores , Complejos de Coordinación/síntesis química , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Oxidación-Reducción , Polietilenglicoles/química , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Bases de Schiff/antagonistas & inhibidores , Bases de Schiff/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA