Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Int J Mol Sci ; 25(12)2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38928424

RESUMO

The SLC35 (Solute Carrier 35) family members acting as nucleotide sugar transporters are typically localized in the endoplasmic reticulum or Golgi apparatus. It is, therefore, intriguing that some reports document the presence of orphan transporters SLC35F1 and SLC35F6 within the endosomal and lysosomal system. Here, we compared the subcellular distribution of these proteins and found that they are concentrated in separate compartments; i.e., recycling endosomes for SLC35F1 and lysosomes for SLC35F6. Swapping the C-terminal tail of these proteins resulted in a switch of localization, with SLC35F1 being trafficked to lysosomes while SLC35F6 remained in endosomes. This suggested the presence of specific sorting signals in these C-terminal regions. Using site-directed mutagenesis, fluorescence microscopy, and cell surface biotinylation assays, we found that the EQERLL360 signal located in the cytoplasmic tail of human SLC35F6 is involved in its lysosomal sorting (as previously shown for this conserved sequence in mouse SLC35F6), and that SLC35F1 localization in the recycling pathway depends on two YXXΦ-type signals: a Y367KQF sequence facilitates its internalization from the plasma membrane, while a Y392TSL motif prevents its transport to lysosomes, likely by promoting SLC35F1 recycling to the cell surface. Taken together, these results support that some SLC35 members may function at different levels of the endosomal and lysosomal system.


Assuntos
Endossomos , Lisossomos , Humanos , Sequência de Aminoácidos , Membrana Celular/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Células HEK293 , Células HeLa , Lisossomos/metabolismo , Proteínas de Transporte de Nucleotídeos/metabolismo , Proteínas de Transporte de Nucleotídeos/genética , Sinais Direcionadores de Proteínas , Transporte Proteico
2.
Int J Mol Sci ; 24(21)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37958830

RESUMO

ABCB5ß is a member of the ABC transporter superfamily cloned from melanocytes. It has been reported as a marker of skin progenitor cells and melanoma stem cells. ABCB5ß has also been shown to exert an oncogenic activity and promote cancer metastasis. However, this protein remains poorly characterized. To elucidate its subcellular localization, we tested several anti-ABCB5 antibodies and prepared several tagged ABCB5ß cDNA constructs. We then used a combination of immunofluorescence and biochemical analyses to investigate the presence of ABCB5ß in different subcellular compartments of HeLa and MelJuSo cell lines. Treatment of the cells with the proteasome inhibitor MG132 showed that part of the population of newly synthesized ABCB5ß is degraded by the proteasome system. Interestingly, treatment with SAHA, a molecule that promotes chaperone-assisted folding, largely increased the expression of ABCB5ß. Nevertheless, the overall protein distribution in the cells remained similar to that of control conditions; the protein extensively colocalized with the endoplasmic reticulum marker calnexin. Taken together with cell surface biotinylation studies demonstrating that the protein does not reach the plasma membrane (even after SAHA treatment), the data indicate that ABCB5ß is a microsomal protein predominantly localized to the ER.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Retículo Endoplasmático , Humanos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Retículo Endoplasmático/metabolismo , Chaperonas Moleculares/metabolismo , Células HeLa , Isoformas de Proteínas/metabolismo , Trifosfato de Adenosina/metabolismo
3.
J Cell Mol Med ; 13(9B): 3960-72, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19243476

RESUMO

Melanomas remain associated with dismal prognosis because they are naturally resistant to apoptosis and they markedly metastasize. Up-regulated expression of sodium pump alpha sub-units has previously been demonstrated when comparing metastatic to non-metastatic melanomas. Our previous data revealed that impairing sodium pump alpha1 activity by means of selective ligands, that are cardiotonic steroids, markedly impairs cell migration and kills apoptosis-resistant cancer cells. The objective of this study was to determine the expression levels of sodium pump alpha sub-units in melanoma clinical samples and cell lines and also to characterize the role of alpha1 sub-units in melanoma cell biology. Quantitative RT-PCR, Western blotting and immunohistochemistry were used to determine the expression levels of sodium pump alpha sub-units. In vitro cytotoxicity of various cardenolides and of an anti-alpha1 siRNA was evaluated by means of MTT assay, quantitative videomicroscopy and through apoptosis assays. The in vivo activity of a novel cardenolide UNBS1450 was evaluated in a melanoma brain metastasis model. Our data show that all investigated human melanoma cell lines expressed high levels of the alpha1 sub-unit, and 33% of human melanomas displayed significant alpha1 sub-unit expression in correlation with the Breslow index. Furthermore, cardenolides (notably UNBS1450; currently in Phase I clinical trials) displayed marked anti-tumour effects against melanomas in vitro. This activity was closely paralleled by decreases in cMyc expression and by increases in apoptotic features. UNBS1450 also displayed marked anti-tumour activity in the aggressive human metastatic brain melanoma model in vivo. The alpha1 sodium pump sub-unit could represent a potential novel target for combating melanoma.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Melanoma/terapia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Antineoplásicos Alquilantes/farmacologia , Apoptose , Cardenolídeos/farmacologia , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos Nus , Microscopia de Vídeo/métodos , Metástase Neoplásica , Transplante de Neoplasias , Temozolomida
4.
J Med Chem ; 50(17): 4122-34, 2007 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-17658777

RESUMO

Amonafide (1), a naphthalimide which binds to DNA by intercalation and poisons topoisomerase IIalpha, has demonstrated activity in phase II breast cancer trials, but has failed thus far to enter clinical phase III because of dose-limiting bone marrow toxicity. Compound 17 (one of 41 new compounds synthesized) is a novel anticancer naphthalimide with a distinct mechanism of action, notably inducing autophagy and senescence in cancer cells. Compound 17 (2,2,2-trichloro-N-({2-[2-(dimethylamino)ethyl]-1,3-dioxo-2,3-dihydro-1H-benzo[de]isoquinolin-5-yl}carbamoyl)acetamide (UNBS3157)) was found to have a 3-4-fold higher maximum tolerated dose compared to amonafide and not to provoke hematotoxicity in mice at doses that display significant antitumor effects. Furthermore, 17 has shown itself to be superior to amonafide in vivo in models of (i) L1210 murine leukemia, (ii) MXT-HI murine mammary adenocarcinoma, and (iii) orthotopic models of human A549 NSCLC and BxPC3 pancreatic cancer. Compound 17, therefore, merits further investigation as a potential anticancer agent.


Assuntos
Acetamidas/síntese química , Antineoplásicos/síntese química , Isoquinolinas/síntese química , Naftalimidas/síntese química , Ureia/análogos & derivados , Acetamidas/farmacologia , Acetamidas/toxicidade , Adenina , Animais , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Apoptose , Autofagia , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Linhagem Celular Tumoral , Senescência Celular , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Contagem de Eritrócitos , Feminino , Humanos , Imidas/farmacologia , Imidas/toxicidade , Irinotecano , Isoquinolinas/farmacologia , Isoquinolinas/toxicidade , Contagem de Leucócitos , Dose Máxima Tolerável , Camundongos , Naftalimidas/farmacologia , Naftalimidas/toxicidade , Transplante de Neoplasias , Organofosfonatos , Contagem de Plaquetas , Relação Estrutura-Atividade , Inibidores da Topoisomerase I , Ureia/síntese química , Ureia/farmacologia , Ureia/toxicidade , Gencitabina
5.
Int J Oncol ; 27(3): 607-16, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16077908

RESUMO

To establish the pharmacological profile of a molecule with anti-cancer potential, it seems essential to add an in vivo approach to the first pharmacological experiments carried out in vitro. The present study aims to characterize the degree of sensitivity of seven syngeneic models (two leukemias and five solid tumors) to eleven molecules which have proven to be clinically reliable. We also used some of these models to investigate whether the molecular effects on the extent of growth in a subcutaneously grafted experimental model correlate with the effects of the same drug on the survival of the animals so grafted. Our data show that all the molecules demonstrated significant anti-tumor activities in two mouse leukemia models (with some discrepancies between the two). Two lymphoma models displayed weaker chemosensitivity profiles than the two leukemia models from which they were developed. Two other models, namely the MXT-HS mammary carcinoma and the B16 melanoma, appeared to be rather chemoresistant. However, a direct relationship was evident between the drug-induced decrease in the tumor growth rate and the increase observed in the survival periods of the MXT tumor-bearing mice. This relationship was also observed in the L1210_LYM lymphoma, though to a lesser extent, and was completely absent from the B16 melanoma model. Finally, our data indicated that we had developed a pair of metastasizing, as opposed to non-metastasizing, lymphoma and mammary carcinoma models. In conclusion, the present study shows that syngeneic mouse tumor models can be used as valuable in vivo experimental models for the screening of potential anti-cancer agents.


Assuntos
Antineoplásicos/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Neoplasias Experimentais/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais/estatística & dados numéricos , Feminino , Leucemia L1210/tratamento farmacológico , Leucemia L1210/patologia , Leucemia P388/tratamento farmacológico , Leucemia P388/patologia , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias Experimentais/patologia , Reprodutibilidade dos Testes
6.
J Neurosurg ; 102(4): 706-14, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15871514

RESUMO

OBJECT: Malignant gliomas consist of both heterogeneous proliferating and migrating cell subpopulations, with migrating glioma cells exhibiting less sensitivity to antiproliferative or proapoptotic drugs than proliferative cells. Therefore, the authors combined cimetidine, an antiinflammatory agent already proven to act against migrating epithelial cancer cells, with temozolomide to determine whether the combination induces antitumor activities in experimental orthotopic human gliomas compared with the effects of temozolomide alone. METHODS: Cimetidine added to temozolomide compared with temozolomide alone induced survival benefits in nude mice with U373 human glioblastoma multiforme (GBM) cells orthotopically xenografted in the brain. Computer-assisted phase-contrast microscopy analyses of 9L rat and U373 human GBM cells showed that cimetidine significantly decreased the migration levels of these tumor cells in vitro at concentrations at which tumor growth levels were not modified (as revealed on monotetrazolium colorimetric assay). Computer-assisted microscope analyses of neoglycoconjugate-based glycohistochemical staining profiles of 9L gliosarcomas grown in vivo revealed that cimetidine significantly decreased expression levels of endogenous receptors for fucose and, to a lesser extent, for N-acetyl-lactosamine moieties. Endogenous receptors of this specificity are known to play important roles in adhesion and migration processes of brain tumor cells. CONCLUSIONS: Cimetidine, acting as an antiadhesive and therefore an antimigratory agent for glioma cells, could be added in complement to the cytotoxic temozolomide compound to combat both migrating and proliferating cells in GBM.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Cimetidina/farmacologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Glioblastoma/tratamento farmacológico , Adjuvantes Imunológicos/administração & dosagem , Animais , Antineoplásicos Alquilantes/administração & dosagem , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/veterinária , Cimetidina/administração & dosagem , Dacarbazina/administração & dosagem , Feminino , Glioblastoma/patologia , Glioblastoma/veterinária , Camundongos , Camundongos Nus , Temozolomida , Transplante Heterólogo
7.
Neoplasia ; 10(12): 1383-92, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19048117

RESUMO

PURPOSE: The aims of the present work were to investigate the in vitro and in vivo antiangiogenic effects of chronic temozolomide treatment on various glioma models and to demonstrate whether bevacizumab (Avastin) increased the therapeutic benefits contributed by temozolomide in glioma. EXPERIMENTAL DESIGN: The expression levels of various antiangiogenic factors in four glioma cell lines were evaluated after chronic in vitro treatment with temozolomide by Western blot. Proliferation and migration assays were performed on human endothelial cells incubated with supernatants of glioma cells treated with and without temozolomide. Orthotopic glioma models were used to evaluate the antiangiogenic effects of temozolomide in vivo and the therapeutic benefits of different temozolomide treatment schedules used alone or in combination with bevacizumab. RESULTS: Temozolomide, a proautophagic and proapoptotic drug, decreased the expression levels of HIF-1alpha, ID-1, ID-2, and cMyc in the glioma models investigated, all of which playing major roles in angiogenesis and the switch to hypoxic metabolism. These changes could be, at least partly, responsible for the impairment of angiogenesis observed in vitro and in vivo. Moreover, combining bevacizumab with temozolomide increased the survival of glioma-bearing mice in comparison to each compound administered alone. CONCLUSIONS: In addition to the numerous mechanisms of action already identified for temozolomide, we report here that it also exerts antitumor effects by impairing angiogenic processes. We further emphasize that bevacizumab, which is an antiangiogenic drug with a different mechanism of action, could be useful in combination with temozolomide to increase the latter's therapeutic benefit in glioma patients.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos Monoclonais Humanizados , Bevacizumab , Dacarbazina/administração & dosagem , Endotélio Vascular/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica , Temozolomida
8.
Neoplasia ; 8(5): 402-12, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16790089

RESUMO

Non-small cell lung cancers (NSCLCs) are the leading cause of cancer deaths in most developed countries. Targeting heat shock protein 70 (Hsp70) expression and function, together with the induction of lysosomal membrane permeabilization (LMP), could overcome the multiple anti-cell death mechanisms evidenced in NSCLCs that are responsible for the failure of currently used chemotherapeutic drugs. Because cardenolides bind to the sodium pump, they affect multiple signaling pathways and thus have a number of marked effects on tumor cell behavior. The aim of the present study was to characterize in vitro and in vivo the antitumor effects of a new cardenolide (UNBS1450) on experimental human NSCLCs. UNBS1450 is a potent source of in vivo antitumor activity in the case of paclitaxel-and oxaliplatin-resistant subcutaneous human NCI-H727 and orthotopic A549 xenografts in nude mice. In vitro UNBS1450-mediated antitumor activity results from the induction of nonapoptotic cell death. UNBS1450 mediates the decrease of Hsp70 at both mRNA and protein levels, and this is at least partly due to UNBS1450-induced downregulation of NFAT5/TonEBP (a factor responsible for the transcriptional control of Hsp70). These effects were paralleled by the induction of LMP, as evidenced by acridine orange staining and immunofluorescence analysis for cathepsin B accumulation.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cardenolídeos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Lisossomos/química , Laranja de Acridina/farmacologia , Animais , Catepsina B/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Camundongos , Camundongos Nus
9.
Cancer ; 101(8): 1908-18, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15386340

RESUMO

BACKGROUND: Nonsmall cell lung carcinomas (NSCLCs) are associated with very dismal prognoses, and adjuvant chemotherapy, including irinotecan, taxanes, platin, and vinca alkaloid derivatives, offer patients only slight clinical benefits. Part of the chemoresistance of NSCLC results from the expression in NSCLC cells of a very large set of endogenous proteins, which antagonize chemotherapy-mediated attacks on these tumor cells. METHODS: The authors set up an orthotopic model of a human NSCLC by grafting A549 cells into the lungs of nude mice. They tried treating these A549 NSCLC orthotopic xenograft-bearing nude mice on the basis of various chemotherapeutic protocols, including chronic administrations of taxol, oxaliplatin, and irinotecan. A cyclooxygenase-2 (COX-2) inhibitor (NS-398) also was assayed in combination with taxol. The immunohistochemical expression levels of COX-2, prostaglandin E synthetase (PGES), ornithine decarboxylase (ODC), the lung-related resistance protein (LRP), and glutathione-S-transferase-alpha (GST-alpha), GST-mu, and GST-pi were quantitatively determined by means of computer-assisted microscopy in control and drug-treated NSCLC orthotopic xenografts. RESULTS: The orthotopic A549 xenograft model developed in 100% of the grafted mice, leading to brain metastases in approximately 61% mice and to liver metastases in approximately 40% of mice. The model was resistant to taxol and oxaliplatin and was only weakly sensitive to irinotecan. High levels of chemoresistant markers (i.e., COX-2, PGES, ODC, LRP, GST-alpha, GST-mu, and GST-pi) were observed in the nontreated A549 xenografts, although with dramatic variations in individual expression. Taxol and oxaliplatin significantly increased the levels of expression of COX-2, PGES, GST-mu, and GST-pi in a number of different experimental protocols. CONCLUSIONS: The A549 orthotopic xenograft model could be used to evaluate investigational chemotherapeutic agents to identify drugs rapidly that are more active than the drugs currently in use in hospitals.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Camptotecina/análogos & derivados , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Camptotecina/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Ciclo-Oxigenase 2 , Feminino , Glutationa S-Transferase pi , Glutationa Transferase/metabolismo , Humanos , Oxirredutases Intramoleculares/metabolismo , Irinotecano , Isoenzimas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteínas de Membrana , Camundongos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Compostos Organoplatínicos/administração & dosagem , Ornitina Descarboxilase/metabolismo , Oxaliplatina , Paclitaxel/administração & dosagem , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/metabolismo , Taxa de Sobrevida , Transplante Heterólogo , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA