Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Omega ; 8(27): 24467-24476, 2023 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-37457445

RESUMO

Triple-negative breast cancer is an aggressive subtype with a high recurrence rate, potential for metastasis, and a poor prognosis. The chemokine receptor, CXCR4, is a promising molecular target in breast cancer therapy. Here, we have developed a CXCR4-targeted antitumor peptidomimetic (named CTCE-KLAK), which is a fusion of the CXCR4 receptor antagonist CTCE-9908 and the D-form of proapoptotic peptide (KLAKLAK)2, for the treatment of breast cancer. First, we investigated the in vitro antitumor activity of CTCE-KLAK against various breast cancer cells and noncancerous mammary epithelial cells. CTCE-KLAK showed cell-selective cytotoxicity and induced rapid necrotic cell death in breast cancer cells but not in normal cells. In contrast, unconjugated peptides such as the carboxylate analogues of CTCE-9908 and D(KLAKLAK)2 were not cytotoxic to these cells. The tumor selectivity of CTCE-KLAK for cytotoxic activity depends on its internalization into tumor cells. There was no cleavage of caspase-3, caspase-7, or PARP1 in CTCE-KLAK-treated cells. In addition, cell death by CTCE-KLAK was not prevented by z-VAD-fmk, a pan-caspase inhibitor that inhibits cisplatin-induced cell death. These data indicate that the CTCE-KLAK conjugate is a cell-selective inducer of necrosis. Furthermore, we evaluated the in vivo antitumor activity of CTCE-KLAK in the 4T1 mouse metastatic breast cancer model. Intravenous administration of CTCE-KLAK significantly inhibited tumor growth and lung metastasis. Together, these findings suggest that the necrosis-inducing peptidomimetic CTCE-KLAK is a promising CXCR4-targeted agent for treating triple-negative breast cancer.

2.
J Control Release ; 337: 546-556, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34375687

RESUMO

Obstructed blood flow and erratic blood supply in the tumor region attenuate the distribution and accumulation of nanomedicines in the tumor. Therefore, improvement of these conditions is crucial for efficient drug delivery. In this study, we designed and synthesized a novel N-(2-hydroxypropyl)methacrylamide (HPMA)-based copolymer conjugate of BK, which possessed adequate systemic stability and tumor-selective action required to improve the accumulation of nanomedicines in the tumor. Levulinoyl-BK (Lev-BK) was conjugated to an HPMA-based polymer via an acid-cleavable hydrazone bond (P-BK). An acid-responsive release of Lev-BK from P-BK was observed, and P-BK alone after intradermal application showed below 10% of the BK activity, thus proving a reduction in the vascular permeability activity of BK when attached to the polymer carrier. P-BK pre-treatment improved blood flow in the tumor tissue by 1.4-1.7-fold, which was maintained for more than 4 h. In addition, P-BK pre-treatment increased the tumor accumulation of pegylated liposomal doxorubicin (PLD) by approximately 3-fold. Furthermore, P-BK pre-treatment led to superior antitumor activity of PLD and significantly improved the survival of tumor-bearing mice. The release of BK from P-BK in the acidic milieu of the tumor was a prerequisite for P-BK to exert its effect, as the vascular permeability enhancing activity of P-BK was negligible. Collectively, P-BK pre-treatment improved intratumoral blood flow and augmented tumor accumulation of nanomedicine, thereby resulting in a significant suppression of tumor growth. Therefore, these findings demonstrate that P-BK is a potential concomitant drug for improving the tumor delivery of nanomedicines.


Assuntos
Antineoplásicos , Neoplasias , Animais , Antineoplásicos/uso terapêutico , Bradicinina/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Metacrilatos , Camundongos , Nanomedicina , Neoplasias/tratamento farmacológico , Polímeros/uso terapêutico
3.
Histochem Cell Biol ; 156(5): 409-421, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34331103

RESUMO

Elevated expression of the nucleoporin Nup88, a constituent of the nuclear pore complex, is seen in various types of malignant tumors, but whether this overexpression contributes to the malignant phenotype has yet to be determined. Here, we investigated the effect of the overexpression of Nup88 on the migration and invasion of cervical cancer HeLa cells. The overexpression of Nup88 promoted a slight but significant increase in both migration and invasion, whereas knockdown of Nup88 by RNA interference suppressed these phenotypes. The observed phenotypes in Nup88-overexpressing HeLa cells were not due to the progression of the epithelial-to-mesenchymal transition or activation of NF-κB, which are known to be important for cell migration and invasion. Instead, we identified an upregulation of matrix metalloproteinase-12 (MMP-12) at both the gene and protein levels in Nup88-overexpressing HeLa cells. Upregulation of MMP-12 protein by the overexpression of Nup88 was also observed in one other cervical cancer cell line and two prostate cancer cell lines but not 293 cells. Treatment with a selective inhibitor against MMP-12 enzymatic activity significantly suppressed the invasive ability of HeLa cells induced by Nup88 overexpression. Taken together, our results suggest that overexpression of Nup88 can stimulate malignant phenotypes including invasive ability, which is promoted by MMP-12 expression.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares/genética , Movimento Celular , Células Cultivadas , Células HeLa , Humanos , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo
4.
BMC Cancer ; 18(1): 519, 2018 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-29724197

RESUMO

BACKGROUND: Nucleoporin Nup88, a component of nuclear pore complexes, is known to be overexpressed in several types of tumor tissue. The overexpression of Nup88 has been reported to promote the early step of tumorigenesis by inducing multinuclei in both HeLa cells and a mouse model. However, the molecular basis of how Nup88 leads to a multinucleated phenotype remains unclear because of a lack of information concerning its binding partners. In this study, we characterize a novel interaction between Nup88 and vimentin. We also examine the involvement of vimentin in the Nup88-dependent multinucleated phenotype. METHODS: Cells overexpressing tagged versions of Nup88, vimentin and their truncations were used in this study. Coprecipitation and GST-pulldown assays were carried out to analyze protein-protein interactions. Vimentin knockdown by siRNA was performed to examine the functional role of the Nup88-vimentin interaction in cells. The phosphorylation status of vimentin was analyzed by immunoblotting using an antibody specific for its phosphorylation site. RESULTS: Vimentin was identified as a Nup88 interacting partner, although it did not bind to other nucleoporins, such as Nup50, Nup214, and Nup358, in HeLa cell lysates. The N-terminal 541 amino acid residues of Nup88 was found to be responsible for its interaction with vimentin. Recombinant GST-tagged Nup88 bound to recombinant vimentin in a GST-pulldown assay. Although overexpression of Nup88 in HeLa cells was observed mainly at the nuclear rim and in the cytoplasm, colocalization with vimentin was only partially detected at or around the nuclear rim. Disruption of the Nup88-vimentin interaction by vimentin specific siRNA transfection suppressed the Nup88-dependent multinucleated phenotype. An excess amount of Nup88 in cell lysates inhibited the dephosphorylation of a serine residue (Ser83) within the vimentin N-terminal region even in the absence and presence of an exogenous phosphatase. The N-terminal 96 amino acid residues of vimentin interacted with both full-length and the N-terminal 541 residues of Nup88. CONCLUSIONS: Nup88 can affect the phosphorylation status of vimentin, which may contribute to the Nup88-dependent multinucleated phenotype through changing the organization of vimentin.


Assuntos
Biomarcadores Tumorais/metabolismo , Núcleo Celular/patologia , Transformação Celular Neoplásica/patologia , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Vimentina/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Fosforilação , RNA Interferente Pequeno/metabolismo , Vimentina/genética
5.
J Control Release ; 277: 23-34, 2018 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-29530390

RESUMO

Human serum albumin (HSA) is a superior carrier for delivering extracellular drugs. However, the development of a cell-penetrating HSA remains a great challenge due to its low membrane permeability. We report herein on the design of a series of palmitoyl-poly-arginine peptides (CPPs) and an evaluation of their cell-penetrating effects after forming a complex with HSA for use in intracellular drug delivery. The palmitoyl CPPs forms a stable complex with HSA by anchoring itself to the high affinity palmitate binding sites of HSA. Among the CPPs evaluated, a cyclic polypeptide composed of D-dodecaarginines, palmitoyl-cyclic-(D-Arg)12 was the most effective for facilitating the cellular uptake of HSA by HeLa cells. Such a superior cell-penetrating capability is primarily mediated by macropinocytosis. The effect of the CPP on pharmacological activity was examined using three drugs loaded in HSA via three different methods: a) an HSA-paclitaxel complex, b) an HSA-doxorubicin covalent conjugate and c) an HSA-thioredoxin fusion protein. The results showed that cell-penetrating efficiency was increased with a corresponding and significant enhancement in pharmacological activity. In conclusion, palmitoyl-cyclic-(D-Arg)12/HSA is a versatile cell-penetrating drug delivery system with great potential for use as a nano-carrier for a wide diversity of pharmaceutical applications.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Desenho de Fármacos , Nanopartículas/administração & dosagem , Albumina Sérica Humana/administração & dosagem , Permeabilidade da Membrana Celular/fisiologia , Peptídeos Penetradores de Células/síntese química , Peptídeos Penetradores de Células/metabolismo , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Albumina Sérica Humana/síntese química , Albumina Sérica Humana/metabolismo , Relação Estrutura-Atividade
6.
Glia ; 64(11): 1938-61, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27464357

RESUMO

To differentiate subtypes of microglia (MG), we developed a novel monoclonal antibody, 9F5, against one subtype (type 1) of rat primary MG. The 9F5 showed high selectivity for this cell type in Western blot and immunocytochemical analyses and no cross-reaction with rat peritoneal macrophages (Mφ). We identified the antigen molecule for 9F5: the 50- to 70-kDa fragments of rat glycoprotein nonmetastatic melanoma protein B (GPNMB)/osteoactivin, which started at Lys(170) . In addition, 9F5 immunoreactivity with GPNMB depended on the activity of furin-like protease(s). More important, rat type 1 MG expressed the GPNMB fragments, but type 2 MG and Mφ did not, although all these cells expressed mRNA and the full-length protein for GPNMB. These results suggest that 9F5 reactivity with MG depends greatly on cleavage of GPNMB and that type 1 MG, in contrast to type 2 MG and Mφ, may have furin-like protease(s) for GPNMB cleavage. In neonatal rat brain, amoeboid 9F5+ MG were observed in specific brain areas including forebrain subventricular zone, corpus callosum, and retina. Double-immunοstaining with 9F5 antibody and anti-Iba1 antibody, which reacts with MG throughout the CNS, revealed that 9F5+ MG were a portion of Iba1+ MG, suggesting that MG subtype(s) exist in vivo. We propose that 9F5 is a useful tool to discriminate between rat type 1 MG and other subtypes of MG/Mφ and to reveal the role of the GPNMB fragments during developing brain. GLIA 2016;64:1938-1961.


Assuntos
Anticorpos Monoclonais/metabolismo , Encéfalo/citologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Microglia/enzimologia , Microglia/imunologia , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Antígenos CD/metabolismo , Células COS/efeitos dos fármacos , Células COS/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Chlorocebus aethiops , Ectodisplasinas/metabolismo , Embrião de Mamíferos , Olho/embriologia , Olho/crescimento & desenvolvimento , Olho/metabolismo , Feminino , Furina/genética , Furina/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Interleucina-12/farmacologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/metabolismo , Microglia/classificação , Microglia/efeitos dos fármacos , Proteoglicanas/metabolismo , Ratos , Ratos Wistar
7.
Clin Cancer Res ; 21(13): 3041-51, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25779950

RESUMO

PURPOSE: The IL11 receptor (IL11R) is an established molecular target in primary tumors of bone, such as osteosarcoma, and in secondary bone metastases from solid tumors, such as prostate cancer. However, its potential role in management of hematopoietic malignancies has not yet been determined. Here, we evaluated the IL11R as a candidate therapeutic target in human leukemia and lymphoma. EXPERIMENTAL DESIGN AND RESULTS: First, we show that the IL11R protein is expressed in a variety of human leukemia- and lymphoma-derived cell lines and in a large panel of bone marrow samples from leukemia and lymphoma patients, whereas expression is absent from nonmalignant control bone marrow. Moreover, a targeted peptidomimetic prototype (termed BMTP-11), specifically bound to leukemia and lymphoma cell membranes, induced ligand-receptor internalization mediated by the IL11R, and resulted in a specific dose-dependent cell death induction in these cells. Finally, a pilot drug lead-optimization program yielded a new myristoylated BMTP-11 analogue with an apparent improved antileukemia cell profile. CONCLUSIONS: These results indicate (i) that the IL11R is a suitable cell surface target for ligand-directed applications in human leukemia and lymphoma and (ii) that BMTP-11 and its derivatives have translational potential against this group of malignant diseases.


Assuntos
Antineoplásicos/farmacologia , Leucemia/tratamento farmacológico , Linfoma/tratamento farmacológico , Peptídeos/farmacologia , Receptores de Interleucina-11/antagonistas & inibidores , Sequência de Aminoácidos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Concentração Inibidora 50 , Ligantes , Dados de Sequência Molecular
8.
Int Immunopharmacol ; 12(1): 158-68, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22155625

RESUMO

A recombinant complement anaphylatoxin, C4a, inhibited chemotaxis, respiratory burst and histamine release in mast cell-like HMC-1 cells that were treated with recombinant C5a anaphylatoxin. C4a also inhibited histamine release from HMC-1 cells that were induced by recombinant C3a. The inhibition of C5a- and C3a-induced leukocyte reactions by C4a was recapitulated in peripheral blood CD133(+) cell-derived differentiated mast cells. In HMC-1 cells, C4a inhibited cytoplasmic Ca(2+) influx, an event that precedes anaphylatoxin-induced chemotactic and secretary responses. A conditioned medium of HMC-1 cells after shortly treated with C4a also inhibited the anaphylatoxin-induced Ca(2+) influx even after removal of C4a, indicating that the effect of C4a is to liberate an autocrine inhibitor from the mast cells. The inhibitor secretion by C4a was prevented with pertussis toxin or with a phosphodiesterase inhibitor. Conversely, an adenylyl cyclase inhibitor reproduced the effect of C4a. C4a decreased the intracellular cyclic AMP concentration of HMC-1 cells, indicating that C4a elicited the Gi protein-adenylyl cyclase inhibition pathway. Neither C4a nor the conditioned medium, however, inhibited Ca(2+) influx and respiratory burst in C5a- or C3a-stimulated peripheral neutrophils, suggesting that these cells lack this inhibitory system. Additionally, in HMC-1 cells, C4a did not inhibit Ca(2+)-independent, Leu72Gln-C5a-stimulated chemotactic response. In agreement with this finding, C4a treatment inhibited ERK1/2 phosphorylation in HMC-1 cells stimulated with other anaphylatoxins but did not inhibit p38MAPK phosphorylation in cells stimulated with Leu72Gln-C5a. Taken together, these findings suggest that the autocrine inhibitory effect elicited by C4a is attributed to interruption of Ca(2+)-dependent intracellular signaling pathway.


Assuntos
Inibidores de Adenilil Ciclases , Anafilatoxinas/farmacologia , Fatores Quimiotáticos/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Fatores Imunológicos/farmacologia , Adenilil Ciclases/metabolismo , Anafilatoxinas/antagonistas & inibidores , Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , Fatores Quimiotáticos/antagonistas & inibidores , Quimiotaxia/efeitos dos fármacos , AMP Cíclico/metabolismo , Liberação de Histamina/efeitos dos fármacos , Humanos , Iminas/farmacologia , Fatores Imunológicos/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Mastócitos/fisiologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/farmacologia
9.
J Pharm Sci ; 100(9): 3968-76, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21480235

RESUMO

The purpose of this study was to quantitatively evaluate the extent of contribution of cysteine-34 ((34) Cys) on the antioxidant effect of human serum albumin (HSA) and to elucidate the physiological implication in hemodialysis (HD) patients. The ratio of oxidized albumin correlated directly with the thiol content in plasma of the HD patients who received intravenous iron. Moreover, the degree of oxidation of (34) Cys in HSA purified from the HD patients' plasma correlated with the thiol content in plasma. The radical scavenging activity of purified HSA was dependent on the degree of (34) Cys oxidation. A recombinant mutant, C34S, was produced to confirm the role of (34) Cys. The activity of C34S was about half of that of wild-type HSA (WT-HSA). Consistent with this observation, the protective effects of C34S were significantly lower than those of WT-HSA in suppressing cytotoxicity and vascular endothelial growth factor production induced by fenton reaction in a human vascular endothelial cell model. These results indicate that the (34) Cys residue of HSA may account for more than 40% of the antioxidant effect of HSA in vivo, and thus may exert a protective effect on vascular endothelium function via an antioxidative mechanism in chronic renal disease.


Assuntos
Antioxidantes/farmacologia , Cisteína/análise , Diálise Renal , Albumina Sérica/fisiologia , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Endotélio Vascular/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Espécies Reativas de Oxigênio/metabolismo , Albumina Sérica/química
10.
Blood ; 117(3): 920-7, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21063027

RESUMO

Targeted drug delivery offers an opportunity for the development of safer and more effective therapies for the treatment of cancer. In this study, we sought to identify short, cell-internalizing peptide ligands that could serve as directive agents for specific drug delivery in hematologic malignancies. By screening of human leukemia cells with a combinatorial phage display peptide library, we isolated a peptide motif, sequence Phe-Phe/Tyr-Any-Leu-Arg-Ser (F(F)/(Y)XLRS), which bound to different leukemia cell lines and to patient-derived bone marrow samples. The motif was internalized through a receptor-mediated pathway, and we next identified the corresponding receptor as the transmembrane glycoprotein neuropilin-1 (NRP-1). Moreover, we observed a potent anti-leukemia cell effect when the targeting motif was synthesized in tandem to the pro-apoptotic sequence (D)(KLAKLAK)2. Finally, our results confirmed increased expression of NRP-1 in representative human leukemia and lymphoma cell lines and in a panel of bone marrow specimens obtained from patients with acute lymphoblastic leukemia or acute myelogenous leukemia compared with normal bone marrow. These results indicate that NRP-1 could potentially be used as a target for ligand-directed therapy in human leukemias and lymphomas and that the prototype CGFYWLRSC-GG-(D)(KLAKLAK)2 is a promising drug candidate in this setting.


Assuntos
Leucemia/metabolismo , Linfoma/metabolismo , Neuropilina-1/metabolismo , Oligopeptídeos/farmacologia , Doença Aguda , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Sítios de Ligação/genética , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Imuno-Histoquímica , Células K562 , Leucemia/genética , Leucemia/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Linfoma/genética , Linfoma/patologia , Dados de Sequência Molecular , Neuropilina-1/genética , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Biblioteca de Peptídeos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Ligação Proteica , Interferência de RNA , Células U937
11.
Cancer Sci ; 101(1): 137-42, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19843070

RESUMO

BCR-ABL tyrosine kinase, generated from the reciprocal chromosomal translocation t(9;22), causes chronic myeloid leukemia (CML). BCR-ABL is inhibited by imatinib; however, several mechanisms of imatinib resistance have been proposed that account for loss of imatinib efficacy in patients with CML. Previously, we showed that overexpression of the efflux drug transporter P-glycoprotein partially contributed to imatinib resistance in imatinib-resistant K562 CML cells having no BCR-ABL mutations. To explain an additional mechanism of drug resistance, we established a subclone (K562/R) of the cells and examined the BCR-ABL signaling pathway in these and wild-type K562 (K562/W) cells. We found the K562/R cells were 15 times more resistant to imatinib than their wild-type counterparts. In both cell lines, BCR-ABL and its downstream signaling molecules, such as ERK1/2, ERK5, STAT5, and AKT, were phosphorylated in the absence of imatinib. In both cell lines, imatinib effectively reduced the phosphorylation of all the above, except ERK1/2, whose phosphorylation was, interestingly, only inhibited in the wild-type cells. We then observed that phospho-ERK1/2 levels decreased in the presence of siRNA targeting BCR-ABL, again, only in the K562/W cells. However, using an ERK1/2 inhibitor, U0126, we found that we could reduce phospho-ERK1/2 levels in K562/R cells and restore their sensitivity to imatinib. Taken together, we conclude that the BCR-ABL-independent activation of ERK1/2 contributes to imatinib resistance in K562/R cells, and that ERK1/2 could be a target for the treatment of CML patients whose imatinib resistance is due to this mechanism.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Fusão bcr-abl/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Benzamidas , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Humanos , Mesilato de Imatinib , Células K562 , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores
12.
J Neuroimmunol ; 208(1-2): 70-9, 2009 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-19211155

RESUMO

There may be two subtypes of microglia (MG) at least in the CNS. We separated the two types from rat mixed glial culture. mRNAs and proteins for inducible nitric oxide synthase (iNOS) and tumor necrosis factor-alpha (TNFalpha) were more induced in CD40(+) MG than CD40(-) MG after LPS stimulation. Although the expression level of LPS receptors showed a little difference between the subtypes, LPS-induced degradation of phosphorylated IkappaBalpha was marked in CD40(+) MG. These results strongly suggest that CD40(+) MG produce larger amount of NO and TNFalpha to exhibit neurotoxic action under certain pathological conditions in brains.


Assuntos
Antígenos CD40/biossíntese , Regulação Enzimológica da Expressão Gênica/imunologia , Microglia/imunologia , Microglia/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Animais , Antígenos CD40/deficiência , Antígenos CD40/metabolismo , Células Cultivadas , Indução Enzimática/genética , Indução Enzimática/imunologia , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Mediadores da Inflamação/toxicidade , Lipopolissacarídeos/fisiologia , Microglia/enzimologia , Óxido Nítrico/biossíntese , Óxido Nítrico/toxicidade , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/toxicidade
13.
J Biol Chem ; 284(6): 3396-407, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19068484

RESUMO

The initiation of chromosomal DNA replication is tightly regulated to achieve genome replication just once per cell cycle and cyclin-dependent kinase (CDK) plays an important role in this process. Adenine nucleotides that bind to the origin recognition complex (ORC) are also suggested to be involved in this process. Of the six subunits of the Saccharomyces cerevisiae ORC (Orc1-6p), both Orc1p and Orc5p have ATP binding activity, and both Orc2p and Orc6p are phosphorylated by CDK in cells. In this study we constructed a series of yeast strains expressing phospho-mimetic mutants of Orc2p or Orc6p and found that expression of a Ser-188 mutant of Orc2p (Orc2-5Dp) delays G1-S transition and S phase progression and causes the accumulation of cells with 2C DNA content. Using antibody that specifically recognizes Ser-188-phosphorylated Orc2p, we showed that Ser-188 is phosphorylated by CDK in a cell cycle-regulated manner. Expression of Orc2-5Dp caused phosphorylation of Rad53p and inefficient loading of the six minichromosome maintenance proteins. These results suggest that the accumulation of cells with 2C DNA content is due to inefficient origin firing and induction of the cell cycle checkpoint response and that dephosphorylation of Ser-188 of Orc2p in late M or G1 phase may be involved in pre-RC formation. In vitro, a purified mutant ORC containing Orc2-5Dp lost Orc5p ATP binding activity. This is the first demonstration of a link between phosphorylation of the ORC and its ability to bind ATP, which may be important for the cell cycle-regulated initiation of DNA replication.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Replicação do DNA/fisiologia , DNA Fúngico/metabolismo , Complexo de Reconhecimento de Origem/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Trifosfato de Adenosina/genética , Proteínas de Ciclo Celular/genética , Quinase do Ponto de Checagem 2 , Cromossomos Fúngicos/fisiologia , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , DNA Fúngico/genética , Fase G1/fisiologia , Complexo de Reconhecimento de Origem/genética , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Proteínas Serina-Treonina Quinases/genética , Fase S/fisiologia , Proteínas de Saccharomyces cerevisiae/genética
14.
J Biol Chem ; 283(17): 11752-62, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18292083

RESUMO

Ligand-directed delivery of agents to leukemia and lymphoma cells has the potential to yield new mechanistic disease insights and targeted therapies. Here we set out to target the macropinocytotic pathway with a combinatorial approach. From the screening of acute T-lymphoblastic leukemia Molt-4 cells with a random phage-display peptide library, we isolated a phage displaying the sequence CAYHRLRRC. This peptide contains a lymph node-homing motif (Cys-Ala-Tyr) and a cell-penetrating motif (Arg-Leu-Arg-Arg). Binding of this ligand-directed phage to a large panel of leukemia/lymphoma cells and to patient-derived samples was much higher than to non-leukemia control cells. CAYHRLRRC phage internalization into Molt-4 cells is both energy- and temperature-dependent. Flow cytometry with fluorescein-labeled peptide and endocytosis blocking with specific inhibitors revealed that CAYHRLRRC is indeed taken up through macropinocytosis in Molt-4 and K562 human leukemia cells. Unexpectedly, the cell surface receptor for the CAYHRLRRC peptide is not a heparan sulfate proteoglycan as it would be predicted for other cell-penetrating peptides. Confirming this interpretation, a CAYHRLRRC-directed peptidomimetic-induced cell death in all the leukemia and lymphoma cells was evaluated, whereas a control transactivator of transcription protein (tat)-directed proapoptotic peptidomimetic was non-selective. In summary, the targeting peptide CAYHRLRRC is selectively internalized through macropinocytosis in leukemia and lymphoma cells and has potential as a drug lead for ligand-directed anti-leukemia therapies.


Assuntos
Antineoplásicos/farmacologia , Regulação Leucêmica da Expressão Gênica , Leucemia/metabolismo , Linfoma/metabolismo , Catálise , Linhagem Celular Tumoral , Sobrevivência Celular , Química Farmacêutica/métodos , Desenho de Fármacos , Humanos , Células K562 , Ligantes , Biblioteca de Peptídeos , Peptídeos/química , Pinocitose
15.
Pharm Res ; 25(4): 827-35, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17934801

RESUMO

PURPOSE: The purpose of this study was to investigate the contribution of drug transporters in acquired imatinib-resistance. Specifically, we focused on the efflux transporters, P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), and an influx transporter, organic cation transporter 1 (OCT1). MATERIALS AND METHODS: We established imatinib-resistant K562 cells (K562/IM). Real-time PCR or Western blot analyses were performed to examine the mRNA or protein levels. Alamar blue method was used in the cytotoxicity assay. The transport activities and intracellular imatinib levels were measured by flow cytometry and HPLC, respectively. RESULTS: K562/IM displayed a 47-fold increase in resistance to imatinib over the parent K562 cells. Both P-gp and BCRP were overexpressed in K562/IM relative to K562. Furthermore, the intracellular imatinib level was markedly reduced in K562/IM. Interestingly, cyclosporin A, a P-gp inhibitor, but not fumitremorgin C, a BCRP inhibitor, restored both imatinib-sensitivity and the intracellular imatinib level. By contrast, no significant difference in the expression and function of OCT1 was observed between K562/IM and K562. CONCLUSIONS: P-gp, rather than BCRP or OCT1, is partially responsible for the development of imatinib-resistance due to constitutive and functional overexpression, leading to reduced intracellular accumulation of imatinib in K562/IM.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/metabolismo , Resistencia a Medicamentos Antineoplásicos , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Transportador 1 de Cátions Orgânicos/genética , Piperazinas/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Pirimidinas/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/genética , Antineoplásicos/farmacologia , Benzamidas , Western Blotting , Sobrevivência Celular , Cromatografia Líquida de Alta Pressão , Ciclosporina/farmacologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Citometria de Fluxo , Humanos , Mesilato de Imatinib , Indóis/farmacologia , Células K562 , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Transportador 1 de Cátions Orgânicos/metabolismo , Piperazinas/farmacologia , Reação em Cadeia da Polimerase , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Regulação para Cima
16.
J Immunol ; 178(12): 8183-94, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548657

RESUMO

TWEAK (TNF-like weak inducer of apoptosis) is a TNF superfamily member implicated in several mechanisms. Although fibroblast growth factor inducible 14 (Fn14)/TweakR has been reported as its receptor, an as yet unrecognized surface molecule(s) might modulate TWEAK function(s). Thus, we set out to identify TWEAK-binding proteins by screening a combinatorial peptide library. Cyclic peptides containing a consensus motif (WXDDG) bound to TWEAK specifically. These peptides were similar to CD163, a scavenger receptor cysteine-rich domain family member, restricted to the monocyte/macrophage lineage and responsible for the uptake of circulating haptoglobin-hemoglobin (Hp-Hb) complexes. Sequence profile analysis suggested that TWEAK mimicked the CD163 natural ligand (Hp-Hb). Consistently, we show dose-dependent TWEAK binding to CD163 and blockade by an anti-CD163 Ab. In a competition assay, both soluble CD163 and Fn14/TweakR were able to compete off TWEAK binding to coated Fn14/TweakR or CD163, respectively. Flow-cytometry and immunofluorescence assays showed that human monocytes (Fn14/TweakR negative and CD163 positive) bind TWEAK, thus blocking the recognition of CD163 and reducing the activation mediated by a specific mAb in these cells. We demonstrate that monocytes can sequester TWEAK from supernatants, thus preventing tumor cell apoptosis; this effect was reverted by preincubation with the peptide mimicking CD163 or with a mAb anti-CD163, indicating specificity. Finally, we show that recombinant human TWEAK binding to CD163-transfected Chinese hamster ovary cells is inhibited by the presence of either unlabeled TWEAK or the Hp-Hb complex. Together, these data are consistent with the hypothesis that CD163 either acts as a TWEAK scavenger in pathological conditions or serves as an alternate receptor for TWEAK in cells lacking Fn14/TweakR.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Receptores de Superfície Celular/metabolismo , Fatores de Necrose Tumoral/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD/química , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/química , Antígenos de Diferenciação Mielomonocítica/genética , Células CHO , Cricetinae , Cricetulus , Citocina TWEAK , Humanos , Radioisótopos do Iodo/análise , Ligantes , Dados de Sequência Molecular , Monócitos/efeitos dos fármacos , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Mapeamento de Interação de Proteínas , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Receptor de TWEAK , Fatores de Necrose Tumoral/química , Fatores de Necrose Tumoral/genética
17.
FEBS Lett ; 552(2-3): 259-63, 2003 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-14527696

RESUMO

Cardiac ATP-sensitive K(+) (K(ATP)) channels are proposed to contribute to cardio-protection and ischemic preconditioning. Although mRNAs for all subunits of K(ATP) channels (Kir6.0 and sulfonylurea receptors SURs) were detected in hearts, subcellular localization of their proteins and the subunit combination are not well elucidated. We address these questions in rat hearts, using anti-peptide antibodies raised against each subunit. By immunoblot analysis, all of the subunits were detected in microsomal fractions including sarcolemmal membranes, while they were not detected in mitochondrial fractions at all. Immunoprecipitation and sucrose gradient sedimentation of the digitonin-solubilized microsomes indicated that Kir6.2 exclusively assembled with SUR2A. The molecular mass of the Kir6.2-SUR2A complex estimated by sucrose sedimentation was 1150 kDa, significantly larger than the calculated value for (Kir6.2)(4)-(SUR2A)(4), suggesting a potential formation of micellar complex with digitonin but no indication of hybrid channel formation under the conditions. These findings provide additional information on the structural and functional relationships of cardiac K(ATP) channel proteins involving subcellular localization and roles for cardioprotection and ischemic preconditioning.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Trifosfato de Adenosina/metabolismo , Miocárdio/metabolismo , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Centrifugação com Gradiente de Concentração , Microssomos/metabolismo , Mitocôndrias Cardíacas/metabolismo , Canais de Potássio/química , Canais de Potássio/genética , Canais de Potássio/imunologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Testes de Precipitina , Subunidades Proteicas , Ratos , Receptores de Droga/metabolismo , Frações Subcelulares/metabolismo , Receptores de Sulfonilureias
18.
Artigo em Inglês | MEDLINE | ID: mdl-11863294

RESUMO

Four molecular forms of transferrins with different iron-binding states were separated by HPLC using a pyridinium polymer column. The elution order was monoferric transferrin bound to the C-site, holotransferrin, apotransferrin and monoferric transferrin bound to the N-site. Human sera were also analyzed with the column, and ICP-MS combined with HPLC was used to detect iron in each peak. Transferrin peaks separated by HPLC were also confirmed by an immunological method. The percentages of iron saturation in transferrins obtained by the HPLC method were compared with the values calculated from clinical data.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Ferro/metabolismo , Piridinas/química , Transferrina/isolamento & purificação , Eletroforese em Gel de Poliacrilamida , Humanos , Espectrometria de Massas/métodos , Polímeros , Ligação Proteica , Espectrofotometria Ultravioleta , Transferrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA