Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Anticancer Res ; 41(2): 601-608, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33517264

RESUMEN

BACKGROUND/AIM: Hepcidin is a cationic acute phase reactant synthesized by the liver. It has bactericidal properties and is a major regulator of iron homeostasis. Cationic antimicrobial peptides represent an innate antimicrobial defense system. We hypothesized that, like other cationic antimicrobial peptides, hepcidin is cytotoxic to cancer cells. MATERIALS AND METHODS: The cytotoxicity of human hepcidin against myeloma cells was assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) and DNA fragmentation assays. Plasma membrane damage was quantified by propidium iodide (PI) staining. Cell membrane changes were visualized by scanning electron microscopy. RESULTS: Hepcidin impaired myeloma cell survival and induced DNA fragmentation. PI staining and scanning electron microscopy revealed hepcidin-induced disruption of the plasma membrane. CONCLUSION: Human hepcidin is an anti-cancer peptide that induces myeloma cell lysis, and therefore may play a role in innate anticancer immunity. To our knowledge, this is the first biological function ascribed to human hepcidin that is not related to its antimicrobial and iron-regulatory properties.


Asunto(s)
Antineoplásicos/farmacología , Hepcidinas/farmacología , Mieloma Múltiple/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Animales , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN , Metabolismo Energético/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/metabolismo , Mieloma Múltiple/ultraestructura
2.
Biomolecules ; 10(9)2020 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-32842611

RESUMEN

Direct-acting anticancer (DAA) peptides are cytolytic peptides that show promise as novel anticancer agents. DAA peptides bind to anionic molecules that are abundant on cancer cells relative to normal healthy cells, which results in preferential killing of cancer cells. Due to the mechanism by which DAA peptides kill cancer cells, it was thought that resistance would be difficult to achieve. Here, we describe the generation and characterization of two MDA-MB-231 breast cancer cell-line variants with reduced susceptibility to pleurocidin-family and mastoparan DAA peptides. Peptide resistance correlated with deficiencies in peptide binding to cell-surface structures, suggesting that resistance was due to altered composition of the cell membrane. Peptide-resistant MDA-MB-231 cells were phenotypically distinct yet remained susceptible to chemotherapy. Surprisingly, neither of the peptide-resistant breast cancer cell lines was able to establish tumors in immune-deficient mice. Histological analysis and RNA sequencing suggested that tumorigenicity was impacted by alternations in angiogenesis and extracellular matrix composition in the peptide-resistant MDA-MB-231 variants. Collectively, these data further support the therapeutic potential of DAA peptides as adjunctive treatments for cancer.


Asunto(s)
Antineoplásicos/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Peces/metabolismo , Animales , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos NOD , Células Tumorales Cultivadas
3.
Biochim Biophys Acta Biomembr ; 1862(8): 183228, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32126228

RESUMEN

Antimicrobial peptides (AMPs) constitute a diverse family of peptides with the ability to protect their host against microbial infections. In addition to their ability to kill microorganisms, several AMPs also exhibit selective cytotoxicity towards cancer cells and are collectively referred to as anticancer peptides (ACPs). Here a large library of AMPs, mainly derived from the porcine cathelicidin peptide, tritrpticin (VRRFPWWWPFLRR), were assessed for their anticancer activity against the Jurkat T cell leukemia line. These anticancer potencies were compared to the cytotoxicity of the peptides towards normal cells isolated from healthy donors, namely peripheral blood mononuclear cells (PBMCs) and red blood cells (RBCs; where hemolytic activity was assessed). Among the active tritrpticin derivatives, substitution of Arg by Lys enhanced the selectivity of the peptides towards Jurkat cells when compared to PBMCs. Additionally, the side chain length of the Lys residues was also optimized to further enhance the tritrpticin ACP selectivity at low concentrations. The mechanism of action of the peptides with high selectivity involved the permeabilization of the cytoplasmic membrane of Jurkat cells, without formation of apoptotic bodies. The incorporation of non-natural Lys-based cationic amino acids could provide a new strategy to improve the selectivity of other synthetic ACPs to enhance their potential for therapeutic use against leukemia cells.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Antineoplásicos/farmacología , Oligopéptidos/genética , Péptidos/genética , Animales , Antibacterianos/química , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Antineoplásicos/química , Dicroismo Circular , Eritrocitos/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Oligopéptidos/química , Péptidos/química , Péptidos/farmacología , Porcinos , Catelicidinas
4.
Cell Chem Biol ; 24(8): 969-980.e4, 2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28807783

RESUMEN

There is increasing interest in developing cationic host defense peptides (HDPs) and their synthetic derivatives as antimicrobial, immunomodulatory, and anti-biofilm agents. These activities are often evaluated without considering biologically relevant concentrations of salts or serum; furthermore certain HDPs have been shown to aggregate in vitro. Here we examined the effect of aggregation on the immunomodulatory activity of a synthetic innate defense regulator peptide, 1018 (VRLIVAVRIWRR-NH2). A variety of salts and solutes were screened to determine their influence on 1018 aggregation, revealing that this peptide "salts out" of solution in an anion-specific and concentration-dependent manner. Furthermore, the immunomodulatory activity of 1018 was found to be inhibited under aggregation-promoting conditions. A series of 1018 derivatives were synthesized with the goal of disrupting this self-assembly process. Indeed, some derivatives exhibited reduced aggregation while maintaining certain immunomodulatory functions, demonstrating that it is possible to engineer optimized synthetic HDPs to avoid unwanted peptide aggregation.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Inmunomodulación/efectos de los fármacos , Agregado de Proteínas , Péptidos Catiónicos Antimicrobianos/síntesis química , Biopelículas/efectos de los fármacos , Bronquiolos/efectos de los fármacos , Bronquiolos/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Ingeniería de Proteínas
5.
Biochem Cell Biol ; 95(1): 91-98, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28165293

RESUMEN

Lactoferrin (LF) is a mammalian host defense glycoprotein with diverse biological activities. Peptides derived from the cationic region of LF possess cytotoxic activity against cancer cells in vitro and in vivo. Bovine lactoferricin (LFcinB), a peptide derived from bovine LF (bLF), exhibits broad-spectrum anticancer activity, while a similar peptide derived from human LF (hLF) is not as active. In this work, several peptides derived from the N-terminal regions of bLF and hLF were studied for their anticancer activities against leukemia and breast-cancer cells, as well as normal peripheral blood mononuclear cells. The cyclized LFcinB-CLICK peptide, which possesses a stable triazole linkage, showed improved anticancer activity, while short peptides hLF11 and bLF10 were not cytotoxic to cancer cells. Interestingly, hLF11 can act as a cell-penetrating peptide; when combined with the antimicrobial core sequence of LFcinB (RRWQWR) through either a Pro or Gly-Gly linker, toxicity to Jurkat cells increased. Together, our work extends the library of LF-derived peptides tested for anticancer activity, and identified new chimeric peptides with high cytotoxicity towards cancerous cells. Additionally, these results support the notion that short cell-penetrating peptides and antimicrobial peptides can be combined to create new adducts with increased potency.


Asunto(s)
Antiinfecciosos/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Lactoferrina/farmacología , Fragmentos de Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Bovinos , Femenino , Hemólisis/efectos de los fármacos , Humanos , Células Jurkat , Células Tumorales Cultivadas
6.
Biochim Biophys Acta ; 1858(12): 3195-3204, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27693190

RESUMEN

Anti-cancer peptides (ACPs) are small cationic and hydrophobic peptides that are more toxic to cancer cells than normal cells. ACPs kill cancer cells by causing irreparable membrane damage and cell lysis, or by inducing apoptosis. Direct-acting ACPs do not bind to a unique receptor, but are rather attracted to several different molecules on the surface of cancer cells. Here we report that an amidated wasp venom peptide, Mastoparan, exhibited potent anti-cancer activities toward leukemia (IC50~8-9.2µM), myeloma (IC50~11µM), and breast cancer cells (IC50~20-24µM), including multidrug resistant and slow growing cancer cells. Importantly, the potency and mechanism of cancer cell killing was related to the amidation of the C-terminal carboxyl group. Mastoparan was less toxic to normal cells than it was to cancer cells (e.g., IC50 to PBMC=48µM). Mastoparan killed cancer cells by a lytic mechanism. Moreover, Mastoparan enhanced etoposide-induced cell death in vitro. Our data also suggest that Mastoparan and gemcitabine work synergistically in a mouse model of mammary carcinoma. Collectively, these data demonstrate that Mastoparan is a broad-spectrum, direct-acting ACP that warrants additional study as a new therapeutic agent for the treatment of various cancers.


Asunto(s)
Antineoplásicos/farmacología , Membrana Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Péptidos/farmacología , Venenos de Avispas/farmacología , Animales , Línea Celular Tumoral , Dicroismo Circular , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Gemcitabina
7.
Exp Mol Pathol ; 99(3): 426-34, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26344617

RESUMEN

Cationic antimicrobial peptides (CAPs) defend against pathogens and, in some cases, exhibit potent anticancer activities. We previously reported that the pleurocidin NRC-03 causes lysis of breast cancer and multiple myeloma cells. NRC-03 also reduces the EC50 of other cytotoxic compounds and prevents tumor growth in vivo. However, the therapeutic utility of NRC-03 may be limited by its susceptibility to degradation by proteases. The goal of this study was to characterize the anticancer activities of a d-amino acid analog of NRC-03 ([D]-NRC-03) that was predicted to be resistant to proteolytic degradation. Unlike NRC-03, [D]-NRC-03 was not degraded by human serum or trypsin and, in comparison to NRC-03, showed increased killing of breast cancer cells, including multidrug-resistant cells; however, [D]-NRC-03 was somewhat more cytotoxic than NRC-03 for several types of normal cells. Importantly, [D]-NRC-03 was more effective than NRC-03 in vivo since 4-fold less peptide was required for an equivalent inhibitory effect on the growth of breast cancer cell xenografts in immune-deficient mice. These findings demonstrate that a d-amino acid analog of NRC-03 overcomes a major limitation to the therapeutic use of NRC-03, namely peptide stability. Further modification of [D]-NRC-03 is required to improve its selectivity for cancer cells.


Asunto(s)
Aminoácidos/farmacología , Neoplasias de la Mama/patología , Proteínas de Peces/farmacología , Aminoácidos/química , Animales , Muerte Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Proteínas de Peces/química , Lenguado , Humanos , Ratones SCID
8.
Peptides ; 71: 276-85, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25836992

RESUMEN

The recent observation that certain cationic peptides possess potent antibiofilm activity demonstrated that small peptides could be used to treat biofilm-associated infections. Other so-called innate defense regulator peptides possess potent immunomodulatory properties such as leukocyte recruitment and suppression of harmful inflammation. A peptide that directly targets biofilm cells while favorably modulating the immune response would be particularly advantageous for treating serious skin infections caused by Staphylococcus aureus. In the present work, using SPOT-synthesized peptide arrays on cellulose membranes, we outline a strategy for systematically assessing the antibiofilm activity of hundreds of IDR-1002 (VQRWLIVWRIRK-NH2) and IDR-HH2 (VQLRIRVAVIRA-NH2) peptide variants against MRSA biofilms. In addition, the ability of these peptides to stimulate production of a monocyte chemoattractant protein (MCP-1) and suppress LPS-induced interleukin (IL)-1ß production in human peripheral blood mononuclear cells (PBMCs) was evaluated. These results informed the synthesis of second-generation peptides resulting in a new peptide, IDR-2009 (KWRLLIRWRIQK-NH2), with enhanced MCP-1 stimulatory activity, favorable IL-1ß suppression characteristics and strong antibiofilm activity against MRSA and Pseudomonas aeruginosa biofilms. This work provides a proof-of-concept that multiple peptide activities can be optimized simultaneously to generate novel sequences that possess a variety of biological properties.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Biopelículas/efectos de los fármacos , Factores Inmunológicos , Leucocitos Mononucleares/inmunología , Staphylococcus aureus Resistente a Meticilina/fisiología , Pseudomonas aeruginosa/fisiología , Péptidos Catiónicos Antimicrobianos/síntesis química , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Quimiocina CCL2/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Factores Inmunológicos/síntesis química , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Interleucina-1beta/inmunología , Lipopolisacáridos/farmacología
9.
Methods Mol Biol ; 1088: 159-70, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24146403

RESUMEN

Cationic antimicrobial (host defense) peptides (CAPs) are able to kill microorganisms and cancer cells, leading to their consideration as novel candidate therapeutic agents in human medicine. CAPs can physically associate with anionic membrane structures, such as those found on cancer cells, causing pore formation, intracellular disturbances, and leakage of cell contents. In contrast, normal cells are less negatively-charged and are typically not susceptible to CAP-mediated cell death. Because the interaction of CAPs with cells is based on charge properties rather than cell proliferation, both rapidly dividing and quiescent cancer cells, as well as multidrug-resistant cancer cells, are targeted by CAPs, making CAPS potentially valuable as anti-cancer agents. CAPs often exist as families of peptides with slightly different amino acid sequences. In addition, libraries of synthetic peptide variants based on naturally occurring CAP templates can be generated in order to improve upon their action. High-throughput screens are needed to quickly and efficiently assess the suitability of each CAP variant. Here we present the methods for assessing CAP-mediated cytotoxicity against cancer cells (suspension and adherent) and untransformed cells (measured using the tritiated thymidine-release or MTT assay), and for discriminating between cell death caused by necrosis (measured using lactate dehydrogenase- or (51)Cr-release assays), or apoptosis and necrosis (single-stranded DNA content measured by flow cytometry). In addition the clonogenic assay, which assesses the ability of single transformed cells to multiply and produce colonies, is described.


Asunto(s)
Antiinfecciosos/farmacología , Antineoplásicos/farmacología , Péptidos/farmacología , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Células Clonales , ADN/metabolismo , Citometría de Flujo , Hemólisis/efectos de los fármacos , Humanos , L-Lactato Deshidrogenasa/metabolismo , Péptidos/metabolismo , Suspensiones , Timidina/metabolismo , Tritio/metabolismo
10.
Nat Chem Biol ; 9(12): 761-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24231617

RESUMEN

Cationic host defense (antimicrobial) peptides were originally studied for their direct antimicrobial activities. They have since been found to exhibit multifaceted immunomodulatory activities, including profound anti-infective and selective anti-inflammatory properties, as well as adjuvant and wound-healing activities in animal models. These biological properties suggest that host defense peptides, and synthetic derivatives thereof, possess clinical potential beyond the treatment of antibiotic-resistant infections. In this Review, we provide an overview of the biological activities of host defense and synthetic peptides, their mechanism(s) of action and new therapeutic applications and challenges that are associated with their clinical use.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/farmacología , Animales , Antiinfecciosos/inmunología , Antiinfecciosos/farmacología , Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Inmunomodulación , Macrófagos/clasificación , Macrófagos/metabolismo , Cicatrización de Heridas/inmunología
11.
Exp Mol Pathol ; 95(2): 192-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23892223

RESUMEN

Cationic antimicrobial peptides such as bovine lactoferricin (LfcinB) constitute an important innate defense mechanism against many microbial pathogens. LfcinB also binds to and selectively kills human cancer cells via a mechanism that involves reactive oxygen species (ROS) generation and caspase activation. The antimicrobial core of LfcinB consists of only six amino acids (RRWQWR), referred to in this study as LfcinB6. Although free LfcinB6 is devoid of cytotoxic activity against cancer cells, we show here that adding a cell-penetrating hepta-arginine sequence via a glycine-glycine linker to LfcinB6 generates a peptide (MPLfcinB6) that is selectively cytotoxic for human T-leukemia and B-lymphoma cells. Flow cytometric analysis of propidium iodide and fluorescein isothiocyanate-dextran uptake by MPLfcinB6-treated cancer cells revealed extensive damage to the cell membrane, which was confirmed by scanning electron microscopy. MPLfcinB6-induced cytotoxicity was also associated with sequential ROS production and mitochondrial membrane permeabilization; however, neither ROS nor caspase activation caused by the loss of mitochondrial membrane integrity was essential for peptide-mediated cell death. We conclude that MPLfcinB6 selectively kills human T-leukemia and B-lymphoma cells by causing extensive and irreparable damage to the cell membrane.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/síntesis química , Péptidos Catiónicos Antimicrobianos/farmacología , Lactoferrina/farmacología , Leucemia , Linfoma de Células B , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/química , Bovinos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Lactoferrina/química , Potencial de la Membrana Mitocondrial , Microscopía Electrónica de Rastreo , Especies Reactivas de Oxígeno
12.
Leuk Lymphoma ; 54(10): 2255-62, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23350892

RESUMEN

Abstract Multiple myeloma is a common hematological malignancy that urgently requires new approaches to treatment, since the disease is not curable using current chemotherapeutic regimens. The aim of this study was to determine whether human and mouse multiple myeloma cells are killed by the pleurocidin-like cationic antimicrobial peptides NRC-03 and NRC-07, previously shown to be active against breast cancer cells. We demonstrate here that NRC-03 and NRC-07 bound to and rapidly killed multiple myeloma cells by causing extensive membrane damage, as well as DNA cleavage. NRC-03 showed greater binding to multiple myeloma cells and a more potent cytotoxic effect than NRC-07. In addition, intratumoral injections of NRC-03 impaired the growth of multiple myeloma xenografts in immune-deficient mice. We conclude that NRC-03 warrants further investigation for its possible use in the treatment of multiple myeloma.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas de Peces/farmacología , Mieloma Múltiple/patología , Animales , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Proteínas de Peces/administración & dosificación , Proteínas de Peces/toxicidad , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Nutr Biochem ; 24(1): 231-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22902327

RESUMEN

Angiogenesis plays an important role in tumor progression. Piperine, a major alkaloid constituent of black pepper, has diverse physiological actions including killing of cancer cells; however, the effect of piperine on angiogenesis is not known. Here we show that piperine inhibited the proliferation and G(1)/S transition of human umbilical vein endothelial cells (HUVECs) without causing cell death. Piperine also inhibited HUVEC migration and tubule formation in vitro, as well as collagen-induced angiogenic activity by rat aorta explants and breast cancer cell-induced angiogenesis in chick embryos. Although piperine binds to and activates the cation channel transient receptor potential vanilloid 1 (TRPV1), its effects on endothelial cells did not involve TRPV1 since the antiproliferative effect of piperine was not affected by TRPV1-selective antagonists, nor did HUVECs express detectable TRPV1 mRNA. Importantly, piperine inhibited phosphorylation of Ser 473 and Thr 308 residues of Akt (protein kinase B), which is a key regulator of endothelial cell function and angiogenesis. Consistent with Akt inhibition as the basis of piperine's action on HUVECs, inhibition of the phosphoinositide-3 kinase/Akt signaling pathway with LY-294002 also inhibited HUVEC proliferation and collagen-induced angiogenesis. Taken together, these data support the further investigation of piperine as an angiogenesis inhibitor for use in cancer treatment.


Asunto(s)
Alcaloides/farmacología , Inhibidores de la Angiogénesis/farmacología , Benzodioxoles/farmacología , Neovascularización Patológica/tratamiento farmacológico , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Animales , Aorta/efectos de los fármacos , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Cromonas/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Técnicas In Vitro , Masculino , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Wistar , Fase S/efectos de los fármacos , Serina/metabolismo , Canales Catiónicos TRPV/genética , Treonina/metabolismo
14.
Breast Cancer Res ; 13(5): R102, 2011 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-22023734

RESUMEN

INTRODUCTION: Cationic antimicrobial peptides (CAPs) defend against microbial pathogens; however, certain CAPs also exhibit anticancer activity. The purpose of this investigation was to determine the effect of the pleurocidin-family CAPs, NRC-03 and NRC-07, on breast cancer cells. METHODS: MTT (3-(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide) and acid phosphatase cell-viability assays were used to assess NRC-03- and NRC-07-mediated killing of breast carcinoma cells. Erythrocyte lysis was determined with hemolysis assay. NRC-03 and NRC-07 binding to breast cancer cells and normal fibroblasts was assessed with fluorescence microscopy by using biotinylated-NRC-03 and -NRC-07. Lactate dehydrogenase-release assays and scanning electron microscopy were used to evaluate the effect of NRC-03 and NRC-07 on the cell membrane. Flow-cytometric analysis of 3,3'-dihexyloxacarbocyanine iodide- and dihydroethidium-stained breast cancer cells was used to evaluate the effects of NRC-03 and NRC-07 on mitochondrial membrane integrity and reactive oxygen species (ROS) production, respectively. Tumoricidal activity of NRC-03 and NRC-07 was evaluated in NOD SCID mice bearing breast cancer xenografts. RESULTS: NRC-03 and NRC-07 killed breast cancer cells, including drug-resistant variants, and human mammary epithelial cells but showed little or no lysis of human dermal fibroblasts, umbilical vein endothelial cells, or erythrocytes. Sublethal doses of NRC-03 and, to a lesser extent, NRC-07 significantly reduced the median effective concentration (EC50) of cisplatin for breast cancer cells. NRC-03 and NRC-07 bound to breast cancer cells but not fibroblasts, suggesting that killing required peptide binding to target cells. NRC-03- and NRC-07-mediated killing of breast cancer cells correlated with expression of several different anionic cell-surface molecules, suggesting that NRC-03 and NRC-07 bind to a variety of negatively-charged cell-surface molecules. NRC-03 and NRC-07 also caused significant and irreversible cell-membrane damage in breast cancer cells but not in fibroblasts. NRC-03- and NRC-07-mediated cell death involved, but did not require, mitochondrial membrane damage and ROS production. Importantly, intratumoral administration of NRC-03 and NRC-07 killed breast cancer cells grown as xenografts in NOD SCID mice. CONCLUSIONS: These findings warrant the development of stable and targeted forms of NRC-03 and/or NRC-07 that might be used alone or in combination with conventional chemotherapeutic drugs for the treatment of breast cancer.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Peces/farmacología , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/patología , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Membrana Celular/efectos de los fármacos , Células Cultivadas , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Ratones SCID , Membranas Mitocondriales/efectos de los fármacos , Datos de Secuencia Molecular , Especies Reactivas de Oxígeno/metabolismo , Venas Umbilicales/citología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nutr Cancer ; 62(3): 379-89, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20358476

RESUMEN

Curcumin, the principal curcuminoid of tumeric, has potent anticancer activity. To determine the mechanism of curcumin-induced cytotoxicity in prostate cancer cells, we exposed PC3 prostate carcinoma cells to 25 to 100 microM curcumin for 24 to 72 h. Curcumin treatment of PC3 cells caused time- and dose-dependent induction of apoptosis and depletion of cellular reduced glutathione (GSH). Exogenous GSH and its precursor N-acetyl-cysteine, but not ascorbic acid (AA) or ebselen, decreased curcumin accumulation in PC3 cells and also prevented curcumin-induced DNA fragmentation. The failure of AA and ebselen to protect PC3 cells from curcumin-induced apoptosis argued against the involvement of reactive oxygen species; rather, GSH-mediated inhibition of curcumin-induced cytotoxicity was due to reduced curcumin accumulation in PC3 cells. Curcumin-treated PC3 cells showed apoptosis-inducing cellular ceramide accumulation and activation of p38 mitogen-activated protein kinase (MAPK) and c-jun N-terminal kinase (JNK). Caspase-3, caspase-8, and caspase-9 were activated, and cytochrome c and apoptosis-inducing factor (AIF) were released from mitochondria following curcumin treatment. Interestingly, curcumin-induced apoptosis was not prevented by p38 MAPK, JNK, or caspase inhibition. We conclude that curcumin-induced cytotoxicity was due to cellular ceramide accumulation and damage to mitochondria that resulted in apoptosis mediated by AIF and other caspase-independent processes.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/fisiología , Ceramidas/metabolismo , Curcumina/farmacología , Mitocondrias/fisiología , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Glutatión/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Exp Mol Pathol ; 82(1): 1-11, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16624283

RESUMEN

Strategies to promote intracellular ceramide accumulation in cancer cells may have therapeutic utility because ceramide is an important second messenger during apoptosis. Exposure to cell-permeable C(6) ceramide or tricyclodecan-9-yl-xanthate (an inducer of de novo ceramide synthesis and an inhibitor of sphingomyelin synthase) caused MDA-MB-435 human breast carcinoma cells to die by apoptosis. Concomitant treatment with the ceramidase inhibitor D-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol (MAPP) or the glucosylceramide synthase inhibitor 1-phenyl-2-palmitoylamino-3-morpholino-1-propanol (PPMP) potentiated the cytotoxic effect of C(6) ceramide, indicating that C(6) ceramide-mediated cytotoxicity was antagonized by the action of ceramidases and glucosylceramide synthase. Interestingly, treatment with PPMP alone, but not MAPP alone, also induced apoptosis in MDA-MB-435 cells, suggesting that conversion to glucosylceramide rather than catabolism by ceramidases prevented endogenous ceramide from reaching cytotoxic levels. C(6) ceramide-induced apoptosis in MDA-MB-435 cells was associated with the generation of reactive oxygen species, and was inhibited by the antioxidants N-acetylcysteine and glutathione. Although mitochondrial membrane integrity was disrupted in C(6) ceramide-treated MDA-MB-435 cells, apoptosis was not mediated by caspases because there was no protective effect by the pan-caspase inhibitor z-VAD-fmk. Collectively, these findings indicate that strategies to enhance intracellular ceramide accumulation in malignant cells might offer a novel approach to the treatment of breast cancer.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ceramidas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ceramidas/antagonistas & inhibidores , Fragmentación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...