Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 16(5)2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38473370

RESUMO

Multiple Myeloma (MM) is an incurable haematological malignancy caused by uncontrolled growth of plasma cells. MM pathogenesis is attributed to crosstalk between plasma cells and the bone marrow microenvironment, where extracellular vesicles (EVs) play a role. In this study, EVs secreted from a panel of MM cell lines were isolated from conditioned media by ultracentrifugation and fluorescently stained EVs were co-cultured with THP-1 monocyte cells. MM EVs from three cell lines displayed a differential yet dose-dependent uptake by THP-1 cells, with H929 EVs displaying the greatest EV uptake compared to MM.1s and U266 EVs suggesting that uptake efficiency is dependent on the cell line of origin. Furthermore, MM EVs increased the secretion of MMP-9 and IL-6 from monocytes, with H929 EVs inducing the greatest effect, consistent with the greatest uptake efficiency. Moreover, monocyte-conditioned media collected following H929 EV uptake significantly increased the migration and proliferation of MM cells. Finally, EV proteome analysis revealed differential cargo enrichment that correlates with disease progression including a significant enrichment of spliceosome-related proteins in H929 EVs compared to the U266 and MM.1s EVs. Overall, this study demonstrates that MM-derived EVs modulate monocyte function to promote tumour growth and metastasis and reveals possible molecular mechanisms involved.

2.
Invest Ophthalmol Vis Sci ; 64(12): 38, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37747402

RESUMO

Purpose: The purpose of this study was to isolate and culture human conjunctival mesenchymal stromal cells (Conj-MSCs) from cadaveric donor tissue, and to obtain and characterize their extracellular vesicles (EVs) and their effect on conjunctival epithelium. Methods: Stromal cells isolated from cadaveric donor conjunctival tissues were cultured and analyzed to determine whether they could be defined as MSCs. Expression of MSC markers was analyzed by flow cytometry. Cells were cultured in adipogenic, osteogenic, and chondrocyte differentiation media, and stained with Oil Red, Von Kossa, and Toluidine Blue, respectively, to determine multipotent capacity. EVs were isolated from cultured Conj-MSCs by differential ultracentrifugation. EV morphology was evaluated by atomic force microscopy, size distribution analyzed by dynamic light scattering, and EVs were individually characterized by nanoflow cytometry. The effect of EVs on oxidative stress and viability was analyzed in in vitro models using the conjunctival epithelial cell line IM-HConEpiC. Results: Cultured stromal cells fulfilled the criteria of MSCs: adherence to plastic; expression of CD90 (99.95 ± 0.03% positive cells), CD105 (99.04 ± 1.43%), CD73 (99.99 ± 0.19%), CD44 (99.93 ± 0.05%), and absence of CD34, CD11b, CD19, CD45 and HLA-DR (0.82 ± 0.91%); and in vitro differentiation into different lineages. Main Conj-MSC EV subpopulations were round, small EVs that expressed CD9, CD63, CD81, and CD147. Conj-MSC EVs significantly decreased the production of reactive oxygen species in IM-HConEpiCs exposed to H2O2 in similar levels than adipose tissue-MSC-derived EVs and ascorbic acid, used as controls. Conclusions: It is possible to isolate human Conj-MSCs from cadaveric tissue, and to use these cells as a source of small EVs with antioxidant activity on conjunctival epithelial cells.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Humanos , Peróxido de Hidrogênio/metabolismo , Células Cultivadas , Diferenciação Celular , Cadáver
3.
Int J Mol Sci ; 24(13)2023 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-37446263

RESUMO

The isomerase activity of Cyclophilin A is important for midbody abscission during cell division, however, to date, midbody substrates remain unknown. In this study, we report that the GTP-binding protein Septin 2 interacts with Cyclophilin A. We highlight a dynamic series of Septin 2 phenotypes at the midbody, previously undescribed in human cells. Furthermore, Cyclophilin A depletion or loss of isomerase activity is sufficient to induce phenotypic Septin 2 defects at the midbody. Structural and molecular analysis reveals that Septin 2 proline 259 is important for interaction with Cyclophilin A. Moreover, an isomerisation-deficient EGFP-Septin 2 proline 259 mutant displays defective midbody localisation and undergoes impaired abscission, which is consistent with data from cells with loss of Cyclophilin A expression or activity. Collectively, these data reveal Septin 2 as a novel interacting partner and isomerase substrate of Cyclophilin A at the midbody that is required for abscission during cytokinesis in cancer cells.


Assuntos
Citocinese , Septinas , Humanos , Citocinese/genética , Septinas/genética , Septinas/metabolismo , Ciclofilina A/genética , Ciclofilina A/metabolismo , Divisão Celular , Células HeLa
4.
Cell Cycle ; 22(8): 951-966, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36691345

RESUMO

The centrosome acts as a protein platform from which proteins are deployed to function throughout the cell cycle. Previously, we have shown that the prolyl isomerase Cyclophilin A (CypA) localizes to the centrosome in interphase and re-localizes to the midbody during mitosis where it functions in cytokinesis. In this study, investigation of CypA by SDS-PAGE during the cell cycle reveals that it undergoes a mobility shift during mitosis, indicative of a post-translational modification, which may correlate with its subcellular re-localization. Due to the lack of a phospho-specific antibody, we used site-directed mutagenesis to demonstrate that the previously identified serine 77 phosphorylation site within CypA is important for control of CypA centrosome localization. Furthermore, CypA is shown to interact with the mitotic NIMA-related kinase 2 (Nek2) during interphase and mitosis, while also interacting with the Nek2-antagonist PP1 during interphase but not during mitosis, suggesting a potential role for the Nek2-PP1 complex in CypA phospho-regulation. In support of this, Nek2 is capable of phosphorylating CypA in vitro. Overall, this work reveals that phosphorylation of CypA at serine 77 is important for its release from the centrosome during mitosis and may be regulated by the activity of Nek2 and PP1 during the cell cycle.


Assuntos
Ciclofilina A , Proteínas Serina-Treonina Quinases , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Peptidilprolil Isomerase/metabolismo , Quinases Relacionadas a NIMA/metabolismo , Mitose , Centrossomo/metabolismo , Serina/metabolismo , Proteínas de Ciclo Celular/metabolismo
5.
Cells ; 11(21)2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36359760

RESUMO

Daratumumab (DARA) has improved the outcome of treatment of multiple myeloma (MM). DARA acts via complement-dependent and -independent mechanisms. Resistance to DARA may result from upregulation of the complement inhibitory proteins CD55 and CD59, downregulation of the DARA target CD38 on myeloma cells or altered expression of the checkpoint inhibitor ligand programmed death ligand-1 (PD-L1) or other mechanisms. In this study, EVs were isolated from peripheral blood (PB) and bone marrow (BM) from multiple myeloma (MM) patients treated with DARA and PB of healthy controls. EV size and number and the expression of CD38, CD55, CD59 and PD-L1 as well as the EV markers CD9, CD63, CD81, CD147 were determined by flow cytometry. Results reveal that all patient EV samples express CD38, PD-L1, CD55 and CD59. The level of CD55 and CD59 are elevated on MM PB EVs compared with healthy controls, and the level of PD-L1 on MM PB EVs is higher in patients responding to treatment with DARA. CD147, a marker of various aspects of malignant behaviour of cancer cells and a potential target for therapy, was significantly elevated on MM EVs compared with healthy controls. Furthermore, mass spectrometry data suggests that MM PB EVs bind DARA. This study reveals a MM PB and BM EV protein signature that may have diagnostic and prognostic value.


Assuntos
Vesículas Extracelulares , Mieloma Múltiplo , Humanos , Antígeno B7-H1 , Antígenos CD55 , Antígenos CD59 , Proteínas do Sistema Complemento , Vesículas Extracelulares/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico
6.
PLoS One ; 17(9): e0274261, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36125986

RESUMO

Diffuse large B-cell lymphomas (DLBCLs) are the most common lymphoproliferative diseases in dogs. DLBCL diagnosis to date has relied on histopathological analysis; however liquid biopsies have gained attention in recent years as a source of diagnostic and prognostic information. Liquid biopsies can be a source of circulating DNA, miRNA, circulating tumour cells or extracellular vesicles (EVs). In this study EVs were isolated from the plasma of healthy dogs, and dogs with lymphoma, and adenocarcinoma by iodixanol density gradient centrifugation. These EVs were positive for the EV markers CD63 and TSG101 and the pan-B cell markers CD79a, CD21, CD45, CD20. NTA analysis revealed that the DLBCL and adenocarcinoma dogs had elevated plasma EVs relative to the healthy dogs. Furthermore, the modal size of lymphoma EVs had decreased relative to healthy dogs while adenocarcinoma EVs were unchanged. This study demonstrates that the plasma EV population is altered in canine lymphoma patients in a manner similar to previous studies on human lymphomas. The similar changes to the EV population in dogs, together with the similar pathological features and treatment protocols in canine and human non-Hodgkin lymphomas would make dogs a good comparative model for studying the role of EVs in DLBCL development and progression.


Assuntos
Adenocarcinoma , Ácidos Nucleicos Livres , Vesículas Extracelulares , Linfoma Difuso de Grandes Células B , MicroRNAs , Animais , Cães , Vesículas Extracelulares/genética , Humanos , Linfoma Difuso de Grandes Células B/veterinária , MicroRNAs/genética
7.
Transl Oncol ; 14(8): 101112, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33984826

RESUMO

Extracellular Vesicles (EVs) are a heterogenous population of particles that play an important role in cell-cell communication in physiological and pathophysiological situations. In this study we reveal that the peptidyl prolyl isomerase Cyclophilin A (CypA) is enriched in cancer-derived EVs from a range of haematopoietic malignancies. CypA-enriched blood cancer EVs were taken up by normal monocytes independent of EV surface trypsin-sensitive proteins and potently stimulated pro-inflammatory MMP9 and IL-6 secretion. Further characterisation revealed that CypA is intravesicular, however, it is not present in all EVs derived from the haematopoietic cells, instead, it is predominantly located in high density EVs with a range of 1.15-1.18 g/ml. Furthermore, loss of CypA expression in haematological cancer cells attenuates high density EV-induced pro-inflammatory MMP9 and IL-6 secretion from monocytes. Mechanistically, we reveal that homozygous loss or siRNA knockdown of CypA expression significantly reduced the secretion of EVs in the range of 100-200 nm from blood cancer cells under normal and hypoxic conditions. Overall, this work reveals a novel role for CypA in cancer cell EV biogenesis.

8.
J Infect Dis ; 219(1): 145-153, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29733369

RESUMO

Background: Listeria generate actin-rich tubular protrusions at the plasma membrane that propel the bacteria into neighboring cells. The precise molecular mechanisms governing the formation of these protrusions remain poorly defined. Methods: In this study, we demonstrate that the prolyl cis-trans isomerase (PPIase) cyclophilin A (CypA) is hijacked by Listeria at membrane protrusions used for cell-to-cell spreading. Results: Cyclophilin A localizes within the F-actin of these structures and is crucial for their proper formation, as cells depleted of CypA have extended actin-rich structures that are misshaped and are collapsed due to changes within the F-actin network. The lack of structural integrity within the Listeria membrane protrusions hampers the microbes from spreading from CypA null cells. Conclusions: Our results demonstrate a crucial role for CypA during Listeria infections.


Assuntos
Extensões da Superfície Celular/metabolismo , Extensões da Superfície Celular/microbiologia , Ciclofilina A/metabolismo , Listeria/metabolismo , Listeriose/metabolismo , Células A549 , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actinas/ultraestrutura , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Extensões da Superfície Celular/ultraestrutura , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Listeria/patogenicidade , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Peptidilprolil Isomerase/metabolismo
9.
Hum Mol Genet ; 25(9): 1824-35, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26908619

RESUMO

Skeletal dysplasias are a clinically and genetically heterogeneous group of bone and cartilage disorders. Whilst >450 skeletal dysplasias have been reported, 30% are genetically uncharacterized. We report two Irish Traveller families with a previously undescribed lethal skeletal dysplasia characterized by fetal akinesia, shortening of all long bones, multiple contractures, rib anomalies, thoracic dysplasia, pulmonary hypoplasia and protruding abdomen. Single nucleotide polymorphism homozygosity mapping and whole exome sequencing identified a novel homozygous stop-gain mutation in NEK9 (c.1489C>T; p.Arg497*) as the cause of this disorder. NEK9 encodes a never in mitosis gene A-related kinase involved in regulating spindle organization, chromosome alignment, cytokinesis and cell cycle progression. This is the first disorder to be associated with NEK9 in humans. Analysis of NEK9 protein expression and localization in patient fibroblasts showed complete loss of full-length NEK9 (107 kDa). Functional characterization of patient fibroblasts showed a significant reduction in cell proliferation and a delay in cell cycle progression. We also provide evidence to support possible ciliary associations for NEK9. Firstly, patient fibroblasts displayed a significant reduction in cilia number and length. Secondly, we show that the NEK9 orthologue in Caenorhabditis elegans, nekl-1, is almost exclusively expressed in a subset of ciliated cells, a strong indicator of cilia-related functions. In summary, we report the clinical and molecular characterization of a lethal skeletal dysplasia caused by NEK9 mutation and suggest that this disorder may represent a novel ciliopathy.


Assuntos
Ciclo Celular/fisiologia , Cílios/patologia , Genes Recessivos/genética , Mutação/genética , Quinases Relacionadas a NIMA/genética , Osteocondrodisplasias/etiologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Lactente , Masculino , Osteocondrodisplasias/patologia , Linhagem , Polimorfismo de Nucleotídeo Único/genética
10.
Mediators Inflamm ; 2015: 146282, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26491220

RESUMO

Mitotic catastrophe, as defined in 2012 by the International Nomenclature Committee on Cell Death, is a bona fide intrinsic oncosuppressive mechanism that senses mitotic failure and responds by driving a cell to an irreversible antiproliferative fate of death or senescence. Thus, failed mitotic catastrophe can promote the unrestrained growth of defective cells, thereby representing a major gateway to tumour development. Furthermore, the activation of mitotic catastrophe offers significant therapeutic advantage which has been exploited in the action of conventional and targeted anticancer agents. Yet, despite its importance in tumour prevention and treatment, the molecular mechanism of mitotic catastrophe is not well understood. A better understanding of the signals that determine cell fate following failed or defective mitosis will reveal new opportunities to selectively target and enhance the programme for therapeutic benefit and reveal biomarkers to predict patient response. This review is focused on the molecular mechanism of mitotic catastrophe induction and signalling and highlights current strategies to exploit the process in cancer therapy.


Assuntos
Mitose/efeitos dos fármacos , Neoplasias/genética , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Humanos , Mitose/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
11.
Curr Mol Pharmacol ; 9(2): 148-64, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25986562

RESUMO

Cyclophilins belong to a group of proteins that possess peptidyl prolyl isomerase activity and catalyse the cis-trans conversion of proline peptide bonds. Cyclophilin members play important roles in protein folding and as molecular chaperones, in addition to a well-established role as host factors required for completion of the virus life cycle. Members of the cyclophilin family are overexpressed in a range of human malignancies including hepatocellular cancer, pancreatic cancer, nonsmall cell lung cancer, gastric cancer, colorectal cancer and glioblastoma multiforme, however, their precise role in tumourigenesis remains unclear. In recent years, mounting evidence supports a role for prolyl isomerisation during mammalian cell division; a process with striking similarity to plasma membrane remodelling during virus replication. Here, we summarise our current understanding of the role of cyclophilins in cancer. We review the function of cyclophilins during mammalian cell division and during HIV-1 infection, and highlight common processes involving members of the ESCRT and Rab GTPase families.


Assuntos
Ciclofilinas/metabolismo , Infecções por HIV/complicações , HIV/fisiologia , Neoplasias/virologia , Replicação Viral , Sequência de Aminoácidos , Animais , Divisão Celular , Ciclofilinas/química , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Humanos , Dados de Sequência Molecular , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Estrutura Terciária de Proteína , Proteínas rab de Ligação ao GTP/metabolismo
12.
Nat Commun ; 5: 4632, 2014 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-25130259

RESUMO

Gliomas are the most common primary tumours affecting the adult central nervous system and respond poorly to standard therapy. Myc is causally implicated in most human tumours and the majority of glioblastomas have elevated Myc levels. Using the Myc dominant negative Omomyc, we previously showed that Myc inhibition is a promising strategy for cancer therapy. Here, we preclinically validate Myc inhibition as a therapeutic strategy in mouse and human glioma, using a mouse model of spontaneous multifocal invasive astrocytoma and its derived neuroprogenitors, human glioblastoma cell lines, and patient-derived tumours both in vitro and in orthotopic xenografts. Across all these experimental models we find that Myc inhibition reduces proliferation, increases apoptosis and remarkably, elicits the formation of multinucleated cells that then arrest or die by mitotic catastrophe, revealing a new role for Myc in the proficient division of glioma cells.


Assuntos
Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Glioma/patologia , Mitose/fisiologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Animais , Apoptose/fisiologia , Astrocitoma/fisiopatologia , Astrocitoma/terapia , Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Glioblastoma/fisiopatologia , Glioblastoma/terapia , Glioma/fisiopatologia , Glioma/terapia , Xenoenxertos , Humanos , Camundongos , Camundongos Transgênicos , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Enzimas Ativadoras de Ubiquitina/fisiologia
13.
Cell Cycle ; 11(7): 1340-53, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22421161

RESUMO

Failed cytokinesis leads to tetraploidy, which is an important intermediate preceding aneuploidy and the onset of tumorigenesis. The centrosome is required for the completion of cytokinesis through the transport of important components to the midbody; however, the identity of molecular components and the mechanism involved remains poorly understood. In this study, we report that the peptidyl prolyl isomerase cyclophilin A (cypA) is a centrosome protein that undergoes cell cycle-dependent relocation to the midzone and midbody during cytokinesis in Jurkat cells implicating a role during division. Depletion of cypA does not disrupt mitotic spindle formation or progression through anaphase; however, it leads to cytokinesis defects through an inability to resolve intercellular bridges, culminating in delayed or failed cytokinesis. Defective cytokinesis is also evident by an increased prevalence of midbody-arrested cells. Expression of wild-type cypA reverses the cytokinesis defect in knockout cells, whereas an isomerase mutant does not, indicating that the isomerisation activity of cypA is required for cytokinesis. In contrast, wild-type cypA and the isomerase mutant localize to the centrosome and midbody, suggesting that localization to these structures is independent of isomerase activity. Depletion of cypA also generates tetraploid cells and supernumerary centrosomes. Finally, colony formation in soft agar is impaired in cypA-knockout cells, suggesting that cypA confers clonogenic advantage on tumor cells. Collectively, this data reveals a novel role for cypA isomerase activity in the completion of cytokinesis and the maintenance of genome stability.


Assuntos
Centrossomo/metabolismo , Ciclofilina A/metabolismo , Citocinese , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citocinese/genética , Instabilidade Genômica , Humanos , Células Jurkat , Poliploidia , Fuso Acromático/fisiologia
14.
Int J Cancer ; 130(3): 705-15, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21413012

RESUMO

Photodynamic therapy (PDT) is an established treatment modality for cancer. ADPM06 is an emerging non-porphyrin PDT agent which has been specifically designed for therapeutic application. Recently, we have demonstrated that ADPM06-PDT is well tolerated in vivo and elicits impressive complete response rates in various models of cancer when a short drug-light interval is applied. Herein, the mechanism of action of ADPM06-PDT in vitro and in vivo is outlined. Using a drug and light combination that reduces the clonogenicity of MDA-MB-231 cells by >90%, we detected a well-orchestrated apoptotic response accompanied by the activation of various caspases in vitro. The generation of reactive oxygen species (ROS) upon photosensitizer irradiation was found to be the key instigator in the observed apoptotic response, with the endoplasmic reticulum (ER) found to be the intracellular site of initial PDT damage, as determined by induction of a rapid ER stress response post-PDT. PDT-induced apoptosis was also found to be independent of p53 tumor suppressor status. A robust therapeutic response in vivo was demonstrated, with a substantial reduction in tumor proliferation observed, as well as a rapid induction of apoptosis and initiation of ER stress, mirroring numerous aspects of the mechanism of action of ADPM06 in vitro. Finally, using a combination of (18) F-labeled 3'-deoxy-3'-fluorothymidine ((18) F-FLT) nuclear and optical imaging, a considerable decrease in tumor proliferation over 24-hr in two models of human cancer was observed. Taken together, this data clearly establishes ADPM06 as an exciting novel PDT agent with significant potential for further translational development.


Assuntos
Apoptose , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Pirróis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Células HCT116 , Humanos , Medições Luminescentes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tomografia por Emissão de Pósitrons , Desdobramento de Proteína/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cell Cycle ; 10(22): 3886-96, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22071694

RESUMO

Protracted mitotic arrest leads to cell death; however, the molecular signals that link these distinct processes remain poorly understood. Here we report that the pro-apoptotic BH3-only family member Bim undergoes phosphorylation in K562 cells following treatment with the microtubule targeting agents Taxol and Nocodazole. The phosphorylation of two Bim isoforms, BimEL and BimL, at the mitochondria correlates with mitotic arrest and precedes cell death induced by Taxol. It was also found that Bim undergoes transient phosphorylation during normal mitosis in K562 cells. In addition, siRNA silencing of Bim reduces sensitivity to Taxol-induced cell death. The transition of K562 cells from mitosis to G1 results in the loss of BimEL and BimL phosphorylation and correlates with the degradation of cyclin B1. The Cdk1 inhibitors, RO-3306 and Purvalanol A, block Bim phosphorylation in mitotically arrested cells. Importantly, it was found that cyclin B1 co-immunoprecipitates with endogenous Bim in mitotic extracts. Furthermore, active recombinant Cdk1/cyclin B1 phosphorylates BimEL and BimL in vitro and Serine 44 on BimL has been identified as a Cdk1 phosphorylation site. Collectively, these results suggest that Cdk1/cyclin B1-dependent hyper-phosphorylation of Bim during prolonged mitotic arrest is an important cell death signal.


Assuntos
Proteína Quinase CDC2/fisiologia , Ciclina B1/fisiologia , Mitose , Apoptose , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Proteína Quinase CDC2/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Ciclina B1/genética , Ciclina B1/metabolismo , Humanos , Células K562 , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Nocodazol/farmacologia , Paclitaxel/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Purinas/farmacologia , Quinolinas/farmacologia , Interferência de RNA , Tiazóis/farmacologia
16.
Biochem Soc Trans ; 37(Pt 4): 910-3, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19614617

RESUMO

The role of aneuploidy in tumorigenesis remains poorly understood, although the two have been known to be linked for more than 100 years. Recent studies indicate that aneuploidy can promote tumour cell growth and cell death and that the cellular outcome is dependent on the extent of aneuploidy induced. The mitotic checkpoint plays a pivotal role in the maintenance of genome stability and has been the focus of work investigating the distinct outcomes of aneuploidy. In the present article, we review the molecular mechanisms involved and discuss the potential of the mitotic checkpoint as a therapeutic target in cancer therapy.


Assuntos
Aneuploidia , Neoplasias/genética , Animais , Centrossomo/metabolismo , Instabilidade Genômica/genética , Instabilidade Genômica/fisiologia , Humanos
17.
J Pharmacol Exp Ther ; 329(1): 38-47, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19131583

RESUMO

Microtubule assembly and disassembly is required for the maintenance of cell structure, mobility, and division. However, the cellular and biochemical implications of microtubule disruption are not fully understood. Using a proteomic approach, we found that the peptidyl-prolyl isomerase, cyclophilin A, was increased in plasma membrane extracts from chronic myeloid leukemia cells after microtubule disruption. In addition, we found that two peptidyl-prolyl isomerases, cyclophilin A and pin1, are overexpressed up to 10-fold in hematological malignancies compared with normal peripheral blood mononuclear cells. Although previous reports suggest that cyclophilin A is localized to the cytosol of mammalian cells, we found that cyclophilin A and pin1 are both localized to the nucleus and nuclear domains in hematopoietic cells. Microtubule disruption of hematopoietic cells caused a dramatic subcellular redistribution of cyclophilin A and pin1 from the nucleus to the cytosol and plasma membrane. We suggest that this accounts for the increased cyclophilin A at the plasma membrane of chronic myeloid leukemia cells after microtubule disruption. The subcellular redistribution of cyclophilin A and pin1 occurred in a c-Jun NH(2)-terminal kinase- and serine protease-dependent manner. Moreover, the altered subcellular localization of the peptidyl-prolyl isomerases occurred in a dose- and time-dependent manner after microtubule disruption and was found to correlate with G(2)/M arrest and precede induced cell death. These results suggest that the function of peptidyl-prolyl isomerases may be influenced by microtubule dynamics throughout the cell cycle, and their altered localization may be an important part of the mechanism by which microtubule-disrupting agents exert their cytostatic effects.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Núcleo Celular/metabolismo , Ciclofilina A/metabolismo , Citoplasma/metabolismo , Neoplasias Hematológicas/metabolismo , Microtúbulos/efeitos dos fármacos , Oxazepinas/farmacologia , Paclitaxel/farmacologia , Peptidilprolil Isomerase/metabolismo , Pirróis/farmacologia , Western Blotting , Divisão Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Fase G2/efeitos dos fármacos , Neoplasias Hematológicas/patologia , Humanos , Hidrólise , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Peptidilprolil Isomerase de Interação com NIMA , Mapeamento de Peptídeos , Coloração pela Prata , Tripsina/química
18.
Mol Pharmacol ; 70(1): 60-70, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16571652

RESUMO

We have demonstrated previously that certain members of a series of novel pyrrolo-1,5-benzoxazepine (PBOX) compounds potently induce apoptosis in a variety of human chemotherapy-resistant cancer cell lines and in primary ex vivo material derived from cancer patients. A better understanding of the molecular mechanisms underlying the apoptotic effects of these PBOX compounds is essential to their development as antineoplastic therapeutic agents. This study sought to test the hypothesis that proapoptotic PBOX compounds target the microtubules. We show that a representative proapoptotic PBOX compound, PBOX-6, induces apoptosis in both the MCF-7 and K562 cell lines. An accumulation of cells in G2/M precedes apoptosis in response to PBOX-6. PBOX-6 induces prometaphase arrest and causes an accumulation of cyclin B1 levels and activation of cyclin B1/CDK1 kinase in a manner similar to that of two representative antimicrotubule agents, nocodazole and paclitaxel. Indirect immunofluorescence demonstrates that both PBOX-6 and another pro-apoptotic PBOX compound, PBOX-15, cause microtubule depolymerization in MCF-7 cells. They also inhibit the assembly of purified tubulin in vitro, whereas a nonapoptotic PBOX compound (PBOX-21) has no effect on either the cellular microtubule network or on the assembly of purified tubulin. This suggests that the molecular target of the pro-apoptotic PBOX compounds is tubulin. PBOX-6 does not bind to either the vinblastine or the colchicine binding site on tubulin, suggesting that it binds to an as-yet-uncharacterised novel site on tubulin. The ability of PBOX-6 to bind tubulin and cause microtubule depolymerization confirms it as a novel candidate for antineoplastic therapy.


Assuntos
Apoptose/efeitos dos fármacos , Oxazepinas/farmacologia , Pirróis/farmacologia , Tubulina (Proteína)/metabolismo , Antineoplásicos Fitogênicos/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Sítios de Ligação , Ligação Competitiva , Western Blotting , Proteína Quinase CDC2/metabolismo , Carbamatos/farmacologia , Divisão Celular , Linhagem Celular Tumoral , Colchicina/metabolismo , Colchicina/farmacologia , Ciclina B/metabolismo , Ciclina B1 , Relação Dose-Resposta a Droga , Fase G2 , Humanos , Células K562 , Metáfase/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Oxazepinas/metabolismo , Pirróis/metabolismo , Fatores de Tempo , Vimblastina/metabolismo , Vimblastina/farmacologia
19.
J Med Chem ; 48(13): 4367-77, 2005 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-15974589

RESUMO

We have recently developed five novel pyrrolo-1,5-benzoxazepines as proapoptotic agents. Their JNK-dependent induction of apoptosis in tumor cells suggested their potential as novel anticancer agents. The core structure of the apoptotic agent 6 was investigated, and the SARs were expanded with the design and synthesis of several analogues. To define the apoptotic mechanism of the new compounds and the localization of their drug target, two analogues of 6 were designed and synthesized to delineate events leading to JNK activation. The cell-penetrating compound 16 induced apoptosis in tumor cells, while its nonpenetrating analogue, 17, was incapable of inducing apoptosis or activating JNK. Plasma membrane permeabilization of tumor cells resulted in 17-induced JNK activation, suggesting that the pyrrolo-1,5-benzoxazepine molecular target is intracellular. Interestingly, compound 6 displayed cytotoxic activity against a panel of human tumor cell lines but demonstrated negligible toxicity in vivo with no effect on the animals' hematology parameters.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzoxazinas/síntese química , Benzoxazinas/farmacologia , Tiazepinas/síntese química , Tiazepinas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Benzoxazinas/química , Benzoxazinas/farmacocinética , Transporte Biológico , Linhagem Celular Tumoral , Desenho de Fármacos , Células HL-60 , Humanos , Células K562 , Modelos Moleculares , Estrutura Molecular , Tiazepinas/química , Tiazepinas/farmacocinética
20.
J Pharmacol Exp Ther ; 310(3): 1084-95, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15143129

RESUMO

Overexpression of the Bcl-2 proto-oncogene in tumor cells confers resistance against chemotherapeutic drugs. In this study, we describe how the novel pyrrolo-1,5-benzoxazepine compound 7-[[dimethylcarbamoyl]oxy]-6-(2-naphthyl)pyrrolo-[2,1-d] (1,5)-benzoxazepine (PBOX-6) selectively induces apoptosis in Bcl-2-overexpressing cancer cells, whereas it shows no cytotoxic effect on normal peripheral blood mononuclear cells. PBOX-6 overcomes Bcl-2-mediated resistance to apoptosis in chronic myelogenous leukemia (CML) K562 cells by the time- and dose-dependent phosphorylation and inactivation of antiapoptotic Bcl-2 family members Bcl-2 and Bcl-XL. PBOX-6 also induces Bcl-2 phosphorylation and apoptosis in wild-type T leukemia CEM cells and cells overexpressing Bcl-2. This is in contrast to chemotherapeutic agents such as etoposide, actinomycin D, and ultraviolet irradiation, whereby overexpression of Bcl-2 confers resistance against apoptosis. In addition, PBOX-6 induces Bcl-2 phosphorylation and apoptosis in wild-type Jurkat acute lymphoblastic leukemia cells and cells overexpressing Bcl-2. However, Jurkat cells containing a Bcl-2 triple mutant, whereby the principal Bcl-2 phosphorylation sites are mutated to alanine, demonstrate resistance against Bcl-2 phosphorylation and apoptosis. PBOX-6 also induces the early and transient activation of c-Jun NH2-terminal kinase (JNK) in CEM cells. Inhibition of JNK activity prevents Bcl-2 phosphorylation and apoptosis, implicating JNK in the upstream signaling pathway leading to Bcl-2 phosphorylation. Collectively, these findings identify Bcl-2 phosphorylation and inactivation as a critical step in the apoptotic pathway induced by PBOX-6 and highlight its potential as an effective antileukemic agent.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oxazepinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pirróis/farmacologia , Antibióticos Antineoplásicos/farmacologia , Carbazóis/farmacologia , Dactinomicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/farmacologia , Humanos , Indóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Células Jurkat , Células K562 , Leucemia/patologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Frações Subcelulares , Células Tumorais Cultivadas , Raios Ultravioleta , Proteína X Associada a bcl-2 , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...