Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Metabolomics ; 16(9): 91, 2020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32851548

RESUMO

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.


Assuntos
Morte Celular/efeitos dos fármacos , Leucemia/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos , Preparações Farmacêuticas , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Lipidômica , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Esfingolipídeos/metabolismo
2.
J Infect Dis ; 220(8): 1312-1324, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31253993

RESUMO

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.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Metagenoma/genética , Neoplasias/virologia , Anelloviridae/genética , Anelloviridae/isolamento & purificação , Biópsia , Conjuntos de Dados como Assunto , Feminino , Herpesviridae/genética , Herpesviridae/isolamento & purificação , Humanos , Masculino , Neoplasias/patologia , Papillomaviridae/genética , Papillomaviridae/isolamento & purificação , Parvovirus/genética , Parvovirus/isolamento & purificação
3.
Pediatr Blood Cancer ; 63(11): 1935-42, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27447373

RESUMO

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.


Assuntos
Crise Blástica/líquido cefalorraquidiano , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Adolescente , Criança , Pré-Escolar , Feminino , Citometria de Fluxo , Humanos , Lactente , Recém-Nascido , Contagem de Leucócitos , Masculino , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras/líquido cefalorraquidiano
5.
Cancer Cell ; 24(3): 379-93, 2013 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-24029234

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/metabolismo , Inibidores Enzimáticos/farmacologia , Lisossomos/metabolismo , Esfingolipídeos/metabolismo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Feminino , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Indóis/farmacologia , Indóis/toxicidade , Camundongos , Camundongos Transgênicos , Fenótipo , Compostos de Espiro/farmacologia , Compostos de Espiro/toxicidade , Tocoferóis/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Cancer Ther ; 12(10): 2018-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23920274

RESUMO

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.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Indóis/administração & dosagem , Neoplasias/tratamento farmacológico , Pirróis/administração & dosagem , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células HeLa , Humanos , Lisossomos/efeitos dos fármacos , Células MCF-7 , Neoplasias/genética , Neoplasias/patologia , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Sunitinibe
7.
Cancer Lett ; 332(2): 265-74, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20598437

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Resistencia a Medicamentos Antineoplásicos , Lisossomos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Animais , Catepsinas/metabolismo , Morte Celular , Núcleo Celular/metabolismo , Citosol/metabolismo , Resistência a Múltiplos Medicamentos , Humanos , Membranas Intracelulares/metabolismo , Camundongos , Esfingomielina Fosfodiesterase/metabolismo
8.
PLoS One ; 7(10): e45381, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071517

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Morte Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/fisiologia , Lisossomos/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/fisiologia , Antígenos de Histocompatibilidade Menor/fisiologia , Miosinas/fisiologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/farmacologia , Tionas/farmacologia , Tropomiosina/fisiologia
9.
Mol Cancer Ther ; 9(1): 24-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20053771

RESUMO

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.


Assuntos
Lactalbumina/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Neoplasias/patologia , Ácido Oleico/farmacologia , Animais , Autofagia/efeitos dos fármacos , Inibidores de Caspase , Bovinos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lactalbumina/química , Camundongos , Neoplasias/metabolismo , Ácido Oleico/química , Ácidos Oleicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
10.
PLoS One ; 4(2): e4424, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19242560

RESUMO

BACKGROUND: Enhanced lysosomal trafficking is associated with metastatic cancer. In an attempt to discover cancer relevant lysosomal motor proteins, we compared the lysosomal proteomes from parental MCF-7 breast cancer cells with those from highly invasive MCF-7 cells that express an active form of the ErbB2 (DeltaN-ErbB2). METHODOLOGY/PRINCIPAL FINDINGS: Mass spectrometry analysis identified kinesin heavy chain protein KIF5B as the only microtubule motor associated with the lysosomes in MCF-7 cells, and ectopic DeltaN-ErbB2 enhanced its lysosomal association. KIF5B associated with lysosomes also in HeLa cervix carcinoma cells as analyzed by subcellular fractionation. The depletion of KIF5B triggered peripheral aggregations of lysosomes followed by lysosomal destabilization, and cell death in HeLa cells. Lysosomal exocytosis in response to plasma membrane damage as well as fluid phase endocytosis functioned, however, normally in these cells. Both HeLa and MCF-7 cells appeared to express similar levels of the KIF5B isoform but the death phenotype was weaker in KIF5B-depleted MCF-7 cells. Surprisingly, KIF5B depletion inhibited the rapamycin-induced accumulation of autophagosomes in MCF-7 cells. In KIF5B-depleted cells the autophagosomes formed and accumulated in the close proximity to the Golgi apparatus, whereas in the control cells they appeared uniformly distributed in the cytoplasm. CONCLUSIONS/SIGNIFICANCE: Our data identify KIF5B as a cancer relevant lysosomal motor protein with additional functions in autophagosome formation.


Assuntos
Autofagia , Cinesinas/deficiência , Lisossomos/metabolismo , Neoplasias/patologia , Organelas/metabolismo , Fagossomos/metabolismo , Morte Celular , Linhagem Celular Tumoral , Humanos , Cinesinas/fisiologia , Espectrometria de Massas , Transporte Proteico , Proteômica , Receptor ErbB-2 , Sirolimo
11.
Autophagy ; 4(4): 487-99, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18305408

RESUMO

A sigma-2 receptor ligand siramesine induces lysosomal leakage and cathepsin-dependent death of cancer cells in vitro and displays potent anti-cancer activity in vivo. The mechanism by which siramesine destabilizes lysosomes is, however, unknown. Here, we show that siramesine induces a rapid rise in the lysosomal pH that is followed by lysosomal leakage and dysfunction. The rapid accumulation of siramesine into cancer cell lysosomes, its ability to destabilize isolated lysosomes, and its chemical structure as an amphiphilic amine indicate that it is a lysosomotropic detergent. Notably, siramesine triggers also a substantial Atg6- and Atg7-dependent accumulation of autophagosomes that is associated with a rapid and sustained inhibition of mammalian target of rapamycin complex 1 (mTORC1; an inhibitor of autophagy). Siramesine fails, however, to increase the degradation rate of long-lived proteins. Thus, the massive accumulation of autophagosomes is likely to be due to a combined effect of activation of autophagy signaling and decreased autophagosome turnover. Importantly, pharmacological and RNA interference-based inhibition of autophagosome formation further sensitizes cancer cells to siramesine-induced cytotoxicity. These data identify siramesine as a lysosomotropic detergent that triggers cell death via a direct destabilization of lysosomes and cytoprotection by inducing the accumulation of autophagosomes. Threrefore, the combination of siramesine with inhibitors of autophagosome formation appears as a promising approach for future cancer therapy.


Assuntos
Antineoplásicos/metabolismo , Autofagia/fisiologia , Citoproteção , Detergentes/metabolismo , Indóis/metabolismo , Lisossomos/metabolismo , Fagossomos/metabolismo , Compostos de Espiro/metabolismo , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Detergentes/química , Feminino , Humanos , Concentração de Íons de Hidrogênio , Indóis/química , Membranas Intracelulares/metabolismo , Lisossomos/ultraestrutura , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Estrutura Molecular , Complexos Multiproteicos , Fosfolipídeos/metabolismo , Proteínas , Receptores sigma/metabolismo , Transdução de Sinais/fisiologia , Compostos de Espiro/química , Serina-Treonina Quinases TOR , Fatores de Transcrição/metabolismo , Transplante Heterólogo
12.
Cancer Res ; 67(5): 2217-25, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17332352

RESUMO

Vincristine is a microtubule-destabilizing antimitotic drug that has been used in cancer therapy for over 40 years. However, the knowledge on vincristine-induced cell death pathways is still sparse. Here, we show that vincristine induces dramatic changes in the lysosomal compartment and sensitizes cells to lysosomal membrane permeabilization. In HeLa cervix carcinoma cells, vincristine induced mitotic arrest and massive cell death associated with an early increase in the lysosomal volume and lysosomal leakage followed by the activation of the intrinsic apoptosis program. In contrast, the majority of vincristine-treated MCF-7 breast carcinoma cells resisted apoptosis. Instead, they adapted to the spindle assembly checkpoint and escaped the mitotic arrest as micronucleated and senescent cells with an increase in the volume and the activity of their lysosomal compartment. Consistent with its substantial effects on the lysosomes, vincristine greatly sensitized cultured cancer cells as well as orthotopic breast cancer xenografts in mice to the cytotoxicity induced by siramesine, a sigma-2 receptor ligand that kills cancer cells by destabilizing their lysosomes. Importantly, the combination of nontoxic concentrations of vincristine and siramesine resulted in massive cell death even in MCF-7 cells that were capable of escaping vincristine-induced spindle assembly checkpoint and cell death. Similar synergism was observed when siramesine was combined with a semisynthetic vincristine analogue, vinorelbine, or with microtubule-stabilizing paclitaxel. These data strongly suggest that combination therapies consisting of microtubule-disturbing and lysosome-destabilizing drugs may prove useful in the treatment of otherwise therapy-resistant human cancers.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Indóis/farmacologia , Lisossomos/efeitos dos fármacos , Compostos de Espiro/farmacologia , Vincristina/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Células HeLa , Humanos , Indóis/administração & dosagem , Camundongos , Camundongos SCID , Compostos de Espiro/administração & dosagem , Células Tumorais Cultivadas , Vincristina/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA