Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Metabolomics ; 16(9): 91, 2020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32851548

RESUMEN

INTRODUCTION: Repurposing of cationic amphiphilic drugs (CADs) emerges as an attractive therapeutic solution against various cancers, including leukemia. CADs target lysosomal lipid metabolism and preferentially kill cancer cells via induction of lysosomal membrane permeabilization, but the exact effects of CADs on the lysosomal lipid metabolism remain poorly illuminated. OBJECTIVES: We aimed to systematically monitor CAD-induced alterations in the quantitative lipid profiles of leukemia cell lines in order to chart effects of CADs on the metabolism of various lipid classes present in these cells. METHODS: We conducted this study on eight cultured cell lines representing two leukemia types, acute lymphoblastic leukemia and acute myeloid leukemia. Mass spectrometry-based quantitative shotgun lipidomics was employed to quantify the levels of around 400 lipid species of 26 lipid classes in the leukemia cell lines treated or untreated with a CAD, siramesine. RESULTS: The two leukemia types displayed high, but variable sensitivities to CADs and distinct profiles of cellular lipids. Treatment with siramesine rapidly altered the levels of diverse lipid classes in both leukemia types. These included sphingolipid classes previously reported to play key roles in CAD-induced cell death, but also lipids of other categories. We demonstrated that the treatment with siramesine additionally elevated the levels of numerous cytolytic lysoglycerophospholipids in positive correlation with the sensitivity of individual leukemia cell lines to siramesine. CONCLUSIONS: Our study shows that CAD treatment alters balance in the metabolism of glycerophospholipids, and proposes elevation in the levels of lysoglycerophospholipids as part of the mechanism leading to CAD-induced cell death of leukemia cells.


Asunto(s)
Muerte Celular/efectos de los fármacos , Leucemia/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos , Preparaciones Farmacéuticas , Línea Celular Tumoral , Supervivencia Celular , Humanos , Lipidómica , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Esfingolípidos/metabolismo
2.
J Infect Dis ; 220(8): 1312-1324, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31253993

RESUMEN

BACKGROUND: Viruses and other infectious agents cause more than 15% of human cancer cases. High-throughput sequencing-based studies of virus-cancer associations have mainly focused on cancer transcriptome data. METHODS: In this study, we applied a diverse selection of presequencing enrichment methods targeting all major viral groups, to characterize the viruses present in 197 samples from 18 sample types of cancerous origin. Using high-throughput sequencing, we generated 710 datasets constituting 57 billion sequencing reads. RESULTS: Detailed in silico investigation of the viral content, including exclusion of viral artefacts, from de novo assembled contigs and individual sequencing reads yielded a map of the viruses detected. Our data reveal a virome dominated by papillomaviruses, anelloviruses, herpesviruses, and parvoviruses. More than half of the included samples contained 1 or more viruses; however, no link between specific viruses and cancer types were found. CONCLUSIONS: Our study sheds light on viral presence in cancers and provides highly relevant virome data for future reference.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Metagenoma/genética , Neoplasias/virología , Anelloviridae/genética , Anelloviridae/aislamiento & purificación , Biopsia , Conjuntos de Datos como Asunto , Femenino , Herpesviridae/genética , Herpesviridae/aislamiento & purificación , Humanos , Masculino , Neoplasias/patología , Papillomaviridae/genética , Papillomaviridae/aislamiento & purificación , Parvovirus/genética , Parvovirus/aislamiento & purificación
3.
Pediatr Blood Cancer ; 63(11): 1935-42, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27447373

RESUMEN

BACKGROUND: Central nervous system (CNS) involvement is associated with relapse in childhood acute lymphoblastic leukemia (ALL) and is a diagnostic challenge. PROCEDURE: In a Nordic/Baltic prospective study, we assessed centralized flow cytometry (FCM) of locally fixed cerebrospinal fluid (CSF) samples versus local conventional cytospin-based cytology (CC) for detecting leukemic cells and evaluating kinetics of elimination of leukemic cells in CSF. RESULTS: Among 300 patients with newly diagnosed ALL, 87 (29%) had CSF involvement by FCM, while CC was positive in 30 (10%) of 299 patients with available CC data (P < 0.001). Patients with FCM+/CC+ had higher CSF leukemic blast counts compared to patients positive by FCM only (medians: 0.10 vs. 0.017 leukemic blasts/µl, P = 0.006). Patients positive by FCM had higher white blood cell counts in peripheral blood than patients negative by FCM (medians: 45 × 10(9) /l vs. 10 × 10(9) /l, P < 0.001), were younger (medians: 3 years vs. 4 years, P = 0.03), and more frequently had T-cell ALL (18/87 vs. 16/213, P = 0.001). At treatment day 15, five of 52 patients (10%) who had CSF positive by FCM at diagnosis remained so despite at least two doses of weekly intrathecal chemotherapy. CONCLUSIONS: Longer follow-up is needed to clarify whether FCM positivity has prognostic significance and is an indicator for intensified CNS-directed therapy.


Asunto(s)
Crisis Blástica/líquido cefalorraquídeo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adolescente , Niño , Preescolar , Femenino , Citometría de Flujo , Humanos , Lactante , Recién Nacido , Recuento de Leucocitos , Masculino , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras/líquido cefalorraquídeo
4.
Cold Spring Harb Protoc ; 2015(10): 904-7, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26430254

RESUMEN

Programmed cell death involving lysosomal membrane permeabilization (LMP) is a common phenomenon--more the rule than the exception under various cytotoxic stimuli and stressful cellular conditions. The protocol presented here is based on immunocytochemical staining of cathepsin B or L to visualize translocation from the lysosomal lumen to the cytosol. In healthy cells, cathepsins appear in localized punctate structures representing intact lysosomes, whereas LMP results in a diffuse staining pattern throughout the cytoplasm. LMP can be triggered upstream, downstream, or independently of the classical apoptotic death pathway involving mitochondrial outer membrane permeabilization (MOMP). Co-staining with antibodies recognizing the active form of Bax allows investigation of the order of events between LMP and MOMP in death signaling.


Asunto(s)
Catepsina B/análisis , Catepsina L/análisis , Citosol/enzimología , Inmunohistoquímica/métodos , Membranas Intracelulares/fisiología , Lisosomas/enzimología , Permeabilidad , Animales , Línea Celular , Humanos
6.
Cancer Cell ; 24(3): 379-93, 2013 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-24029234

RESUMEN

Lysosomal membrane permeabilization and subsequent cell death may prove useful in cancer treatment, provided that cancer cell lysosomes can be specifically targeted. Here, we identify acid sphingomyelinase (ASM) inhibition as a selective means to destabilize cancer cell lysosomes. Lysosome-destabilizing experimental anticancer agent siramesine inhibits ASM by interfering with the binding of ASM to its essential lysosomal cofactor, bis(monoacylglycero)phosphate. Like siramesine, several clinically relevant ASM inhibitors trigger cancer-specific lysosomal cell death, reduce tumor growth in vivo, and revert multidrug resistance. Their cancer selectivity is associated with transformation-associated reduction in ASM expression and subsequent failure to maintain sphingomyelin hydrolysis during drug exposure. Taken together, these data identify ASM as an attractive target for cancer therapy.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Inhibidores Enzimáticos/farmacología , Lisosomas/metabolismo , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Femenino , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Indoles/farmacología , Indoles/toxicidad , Ratones , Ratones Transgénicos , Fenotipo , Compuestos de Espiro/farmacología , Compuestos de Espiro/toxicidad , Tocoferoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Ther ; 12(10): 2018-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23920274

RESUMEN

Defective apoptosis signaling and multidrug resistance are major barriers for successful cancer treatment. To identify drugs capable of targeting treatment-resistant cancer cells, we screened small-molecule kinase inhibitor libraries for compounds that decrease the viability of apoptosis-resistant human MCF7-Bcl-2 breast cancer cells. SU11652, a multitargeting receptor tyrosine kinase inhibitor, emerged as the most potent compound in the screen. In addition to MCF7-Bcl-2 cells, it effectively killed HeLa cervix carcinoma, U-2-OS osteosarcoma, Du145 prostate carcinoma, and WEHI-S fibrosarcoma cells at low micromolar concentration. SU11652 accumulated rapidly in lysosomes and disturbed their pH regulation and ultrastructure, eventually leading to the leakage of lysosomal proteases into the cytosol. Lysosomal destabilization was preceded by an early inhibition of acid sphingomyelinase, a lysosomal lipase that promotes lysosomal membrane stability. Accordingly, Hsp70, which supports cancer cell survival by increasing lysosomal acid sphingomyelinase activity, conferred partial protection against SU11652-induced cytotoxicity. Remarkably, SU11652 killed multidrug-resistant Du145 prostate cancer cells as effectively as the drug-sensitive parental cells, and subtoxic concentrations of SU11652 effectively inhibited multidrug-resistant phenotype in Du145 prostate cancer cells. Notably, sunitinib, a structurally almost identical and widely used antiangiogenic cancer drug, exhibited similar lysosome-dependent cytotoxic activity, albeit with significantly lower efficacy. The significantly stronger lysosome-targeting activity of SU11652 suggests that it may display better efficacy in cancer treatment than sunitinib, encouraging further evaluation of its anticancer activity in vivo. Furthermore, our data provide a rationale for novel approaches to target drug-resistant cancers by combining classic chemotherapy with sunitinib or SU11652.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Indoles/administración & dosificación , Neoplasias/tratamiento farmacológico , Pirroles/administración & dosificación , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Lisosomas/efectos de los fármacos , Células MCF-7 , Neoplasias/genética , Neoplasias/patología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Sunitinib
8.
Cancer Lett ; 332(2): 265-74, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20598437

RESUMEN

Acquired therapy resistance is one of the prime obstacles for successful cancer treatment. Partial resistance is often acquired already during an early face of tumor development when genetic changes causing defects in classical caspase-dependent apoptosis pathway provide transformed cells with a growth advantage by protecting them against various apoptosis inducing stimuli including transforming oncogenes themselves and host immune system. Apoptosis defective cells are further selected during tumor progression and finally by apoptosis inducing treatments. Another form of resistance, multidrug resistance, arises during cancer treatment when cancer cells with effective efflux of cytotoxic agents escape the therapy. Remarkably, induction of lysosomal membrane permeabilization has recently emerged as an effective way to kill apoptosis resistant cancer cells and some lysosome targeting drugs can also re-sensitize multidrug resistant cells to classical chemotherapy. In this review, we highlight recent data on lysosomal cell death pathways and their implications for the future treatment of apoptosis defective and multidrug resistant aggressive tumors.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Resistencia a Antineoplásicos , Lisosomas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Catepsinas/metabolismo , Muerte Celular , Núcleo Celular/metabolismo , Citosol/metabolismo , Resistencia a Múltiples Medicamentos , Humanos , Membranas Intracelulares/metabolismo , Ratones , Esfingomielina Fosfodiesterasa/metabolismo
9.
PLoS One ; 7(10): e45381, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071517

RESUMEN

Microtubule-disturbing drugs inhibit lysosomal trafficking and induce lysosomal membrane permeabilization followed by cathepsin-dependent cell death. To identify specific trafficking-related proteins that control cell survival and lysosomal stability, we screened a molecular motor siRNA library in human MCF7 breast cancer cells. SiRNAs targeting four kinesins (KIF11/Eg5, KIF20A, KIF21A, KIF25), myosin 1G (MYO1G), myosin heavy chain 1 (MYH1) and tropomyosin 2 (TPM2) were identified as effective inducers of non-apoptotic cell death. The cell death induced by KIF11, KIF21A, KIF25, MYH1 or TPM2 siRNAs was preceded by lysosomal membrane permeabilization, and all identified siRNAs induced several changes in the endo-lysosomal compartment, i.e. increased lysosomal volume (KIF11, KIF20A, KIF25, MYO1G, MYH1), increased cysteine cathepsin activity (KIF20A, KIF25), altered lysosomal localization (KIF25, MYH1, TPM2), increased dextran accumulation (KIF20A), or reduced autophagic flux (MYO1G, MYH1). Importantly, all seven siRNAs also killed human cervix cancer (HeLa) and osteosarcoma (U-2-OS) cells and sensitized cancer cells to other lysosome-destabilizing treatments, i.e. photo-oxidation, siramesine, etoposide or cisplatin. Similarly to KIF11 siRNA, the KIF11 inhibitor monastrol induced lysosomal membrane permeabilization and sensitized several cancer cell lines to siramesine. While KIF11 inhibitors are under clinical development as mitotic blockers, our data reveal a new function for KIF11 in controlling lysosomal stability and introduce six other molecular motors as putative cancer drug targets.


Asunto(s)
Neoplasias de la Mama/patología , Muerte Celular/efectos de los fármacos , Proteínas del Citoesqueleto/fisiología , Lisosomas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Femenino , Células HeLa , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/fisiología , Antígenos de Histocompatibilidad Menor/fisiología , Miosinas/fisiología , Pirimidinas/farmacología , ARN Interferente Pequeño/farmacología , Tionas/farmacología , Tropomiosina/fisiología
10.
Mol Cancer Ther ; 9(1): 24-32, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20053771

RESUMEN

A complex of human alpha-lactalbumin and oleic acid (HAMLET) was originally isolated from human milk as a potent anticancer agent. It kills a wide range of transformed cells of various origins while leaving nontransformed healthy cells largely unaffected both in vitro and in vivo. Importantly, purified alpha-lactalbumins from other mammals form complexes with oleic acid that show biological activities similar to that of HAMLET. The mechanism by which these protein-lipid complexes kill tumor cells is, however, largely unknown. Here, we show that complex of bovine alpha-lactalbumin and oleic acid (BAMLET), the bovine counterpart of HAMLET, kills tumor cells via a mechanism involving lysosomal membrane permeabilization. BAMLET shows potent cytotoxic activity against eight cancer cell lines tested, whereas nontransformed NIH-3T3 murine embryonic fibroblasts are relatively resistant. BAMLET accumulates rapidly and specifically in the endolysosomal compartment of tumor cells and induces an early leakage of lysosomal cathepsins into the cytosol followed by the activation of the proapoptotic protein Bax. Ectopic expression of three proteins known to stabilize the lysosomal compartment, i.e. heat shock protein 70 (Hsp70), Hsp70-2, and lens epithelium-derived growth factor, confer significant protection against BAMLET-induced cell death, whereas the antiapoptotic protein Bcl-2, caspase inhibition, and autophagy inhibition fail to do so. These data indicate that BAMLET triggers lysosomal cell death pathway in cancer cells, thereby clarifying the ability of alpha-lactalbumin:oleate complexes to kill highly apoptosis-resistant tumor cells.


Asunto(s)
Lactalbúmina/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Neoplasias/patología , Ácido Oléico/farmacología , Animales , Autofagia/efectos de los fármacos , Inhibidores de Caspasas , Bovinos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citoprotección/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lactalbúmina/química , Ratones , Neoplasias/metabolismo , Ácido Oléico/química , Ácidos Oléicos/farmacología , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...