Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
Artigo em Inglês | MEDLINE | ID: mdl-23295485

RESUMO

Human MTH1 (hMTH1) is an enzyme that hydrolyses several oxidized purine nucleoside triphosphates to their corresponding nucleoside monophosphates. Crystallographic studies have shown that the accurate mode of interaction between 8-oxoguanine and hMTH1 cannot be understood without determining the positions of the H atoms, as can be observed in neutron and/or ultrahigh-resolution X-ray diffraction studies. The hMTH1 protein prepared in the original expression system from Escherichia coli did not appear to be suitable for obtaining high-quality crystals because the hMTH1 protein had heterogeneous N-termini of Met1 and Gly2 that resulted from N-terminal Met excision by methionine aminopeptidase from the E. coli host. To obtain homogeneous hMTH1, the Gly at the second position was replaced by Lys. As a result, mutant hMTH1 protein [hMTH1(G2K)] with a homogeneous N-terminus could be prepared and high-quality crystals which diffracted to near 1.1 Šresolution using synchrotron radiation were produced. The new crystals belonged to space group P2(1)2(1)2(1), with unit-cell parameters a = 46.36, b = 47.58, c = 123.89 Å.


Assuntos
Enzimas Reparadoras do DNA/química , Monoéster Fosfórico Hidrolases/química , Substituição de Aminoácidos , Cristalização/métodos , Cristalografia por Raios X , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/isolamento & purificação , Glicina/química , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Humanos , Lisina/química , Mutação , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/isolamento & purificação , Conformação Proteica
6.
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.

7.
Biochim Biophys Acta ; 1811(3): 153-62, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21146630

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is secreted from adipose tissue and is considered to be a risk factor for both atherosclerosis and insulin resistance. Here we report for the first time that PAI-1 expression is enhanced by oxidized low-density lipoprotein (OxLDL) and its lipid component lysophosphatidylcholine (LPC) in mouse 3T3-L1 adipocytes. In fully differentiated 3T3-L1 cells, OxLDL treatment increased the mRNA expression and protein secretion of PAI-1 in a dose- and time-dependent manner, whereas native LDL had no effect. The addition of an anti-CD36 antibody suppressed OxLDL-stimulated PAI-1 expression by 50%, suggesting that adipose-derived CD36 contributes to roughly half of the PAI-1 expression stimulated by OxLDL. In addition, pharmacological experiments showed that the OxLDL-stimulated enhancement in PAI-1 expression was mediated through the generation of reactive oxygen species (ROS) and phosphorylation of extracellular signal-regulated kinase 1/2. Furthermore, LPC, a major lipid component of OxLDL, was responsible for the enhanced expression of PAI-1 as phospholipase A(2)-treated acetyl LDL, which generates LPC, strongly stimulated PAI-1 expression, whereas acetyl LDL itself had no such activity. These data demonstrate that the uptake of OxLDL and, in particular, its lipid component LPC into adipocytes triggers aberrant ROS-mediated PAI-1 expression, which may be involved in the pathogenesis of metabolic syndrome.


Assuntos
Adipócitos/metabolismo , Lipoproteínas LDL/farmacologia , Lisofosfatidilcolinas/farmacologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Células 3T3-L1 , Adipócitos/patologia , Animais , Anticorpos/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Camundongos , Fatores de Tempo
8.
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
9.
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
10.
J Electron Microsc (Tokyo) ; 59(3): 215-26, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19995890

RESUMO

The dihydropyridine receptor (DHPR) is a protein complex that consists of five distinct subunits of alpha(1), alpha(2), beta, gamma and delta and functions as a voltage-dependent L-type Ca(2+) channel. Here we purified the alpha(1)-beta complex (approximately 250 kDa) from the rabbit skeletal muscle DHPR and reconstructed its three-dimensional (3D) structure to 38 A resolution by single particle analysis of negative staining electron microscopy. The alpha(1)-beta structure exhibited two unique regions: a pseudo-4-fold petaloid region and an elongated region. X-ray crystallographic models of a homologous voltage-dependent K(+) channel and the beta subunit fit well into the individual regions of the alpha(1)-beta structure, revealing that the two regions correspond to the transmembrane alpha(1) and the cytoplasmic beta subunits, respectively. In addition, 3D reconstruction and immuno-electron microscopic analysis performed on the independently purified DHPR demonstrated that the alpha(1)-beta complex was located in the large globular portion of the DHPR, and the N-terminal region of the beta subunit was extended to the leg-shaped protrusion of the DHPR, which includes the alpha(2)delta subunits. Our results propose a model in which the beta subunit may regulate ion channel function by acting as a hinge between alpha(1) and alpha(2)delta subunits of the DHPR.


Assuntos
Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/ultraestrutura , Microscopia Eletrônica/métodos , Animais , Canais de Cálcio Tipo L/isolamento & purificação , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Músculo Esquelético/química , Coelhos
11.
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
12.
Biochim Biophys Acta ; 1725(3): 385-93, 2005 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-15978731

RESUMO

The binding of flunitrazepam (FNZP) by human alpha1-acid glycoprotein (hAGP) and the relationships between the extent of drug binding and desialylation and the genetic variants of hAGP were examined. The photolabeling specificity of [3H]FNZP was confirmed by findings in which other hAGP-binding ligands inhibited the formation of covalent bonds between [3H]FNZP and hAGP. The photolabeling of asialo-hAGP suggested that sialic acid does not involve in the binding of [3H]FNZP. No difference in the labeling could be found between the F1*S variants and A variant. Similarly, FNZP did not show a difference in binding affinity to the two genetic variants of hAGP. Sequence analysis of the photolabeled peptide indicated a sequence corresponding to Tyr91-Arg105 of hAGP.


Assuntos
Sítios de Ligação , Flunitrazepam/metabolismo , Orosomucoide/química , Marcadores de Fotoafinidade/metabolismo , Sequência de Aminoácidos , Assialoglicoproteínas/metabolismo , Brometo de Cianogênio , Variação Genética , Humanos , Dados de Sequência Molecular , Orosomucoide/análogos & derivados , Orosomucoide/genética , Orosomucoide/metabolismo , Fragmentos de Peptídeos/química , Tripsina
13.
Ann N Y Acad Sci ; 1043: 696-701, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16037295

RESUMO

Previous observations by us have clarified that proteins modified by advanced glycation end products (AGEs) are recognized as effective ligands by CD36-overexpressed CHO cells and undergo receptor-mediated endocytosis. CD36, a member of the class B scavenger receptor family, also acts as a fatty acid transporter in adipocytes. Oxidized low-density lipoprotein (Ox-LDL), a ligand for CD36, is known to upregulate CD36 by activating peroxisome proliferator-activated receptor gamma (PPAR-gamma) in macrophages, whereas PPAR-gamma ligands such as troglitazone and 15-deoxy-delta12,14-prostaglandin J2 decrease leptin secretion from adipocytes. The purpose of this study was to examine effects of AGE ligands on leptin expression in adipocytes. Glycolaldehyde-modified bovine serum albumin (GA-BSA) decreased leptin expression at both the protein and mRNA levels in 3T3-L1 adipocytes and mouse epididymal adipocytes. The binding to and subsequent endocytic degradation of GA-BSA by 3T3-L1 adipocytes were effectively inhibited by a neutralizing anti-CD36 antibody. These results indicate that the ligand interaction of GA-BSA with CD36 leads to downregulation of leptin expression in 3T3-L1 adipocytes, suggesting that AGE-induced leptin downregulation is linked to reduction of the insulin sensitivity in metabolic syndrome.


Assuntos
Acetaldeído/análogos & derivados , Adipócitos/fisiologia , Antígenos CD36/fisiologia , Leptina/genética , Soroalbumina Bovina/farmacologia , Células 3T3 , Acetaldeído/sangue , Adipócitos/efeitos dos fármacos , Adipócitos/imunologia , Animais , Técnicas de Cultura de Células , Regulação da Expressão Gênica/efeitos dos fármacos , Síndrome Metabólica/fisiopatologia , Camundongos , PPAR gama/fisiologia , RNA Mensageiro/genética , Receptores para Leptina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
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
15.
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
16.
FEBS Lett ; 537(1-3): 85-90, 2003 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-12606036

RESUMO

Interaction of advanced glycation end products (AGE) with AGE receptors induces several cellular phenomena potentially relating to diabetic complications. We here show that AGE-modified bovine serum albumin (BSA) is endocytosed by adipocytes via CD36. Upon differentiation, 3T3-L1 and human subcutaneous adipose cells showed marked increases in endocytic uptake and subsequent degradation of [(125)I]AGE-BSA, which were inhibited effectively by the anti-CD36 antibody. Ligand specificity of CD36 for modified BSAs was compared with that of LOX-1 and scavenger receptor class A. Effect of fucoidan on [(125)I]AGE-BSA binding showed a sharp contrast to that on [(125)I]-oxidized low density lipoprotein. These results implicate that CD36-mediated interaction of AGE-modified proteins with adipocytes might play a pathological role in obesity or insulin-resistance.


Assuntos
Adipócitos/fisiologia , Antígenos CD/fisiologia , Antígenos CD36/fisiologia , Endocitose/fisiologia , Produtos Finais de Glicação Avançada/farmacocinética , 1-Metil-3-Isobutilxantina/farmacologia , Células 3T3 , Animais , Anticorpos/farmacologia , Antígenos CD/imunologia , Transporte Biológico , Antígenos CD36/imunologia , Dexametasona/farmacologia , Humanos , Camundongos , Soroalbumina Bovina/farmacocinética , Pele
17.
J Atheroscler Thromb ; 10(1): 1-6, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12621157

RESUMO

Advanced glycation end products (AGE) -modified proteins behave as active ligands for several receptors belonging to the scavenger receptor family. Scavenger receptor class B type I (SR-BI) was identified as the first high density lipoprotein (HDL) receptor that mediates selective uptake of HDL-cholesteryl esters (HDL-CE). This study investigated whether AGE proteins serve as ligands for SR-BI and affect SR-BI-mediated cholesterol transport using Chinese hamster ovary (CHO) cells overexpressing hamster SR-BI (CHO-SR-BI cells). [125I] AGE-bovine serum albumin (AGE-BSA) underwent active endocytosis and subsequent lysosomal degradation by CHO-SR-BI cells, indicating that SR-BI serves as an AGE receptor. SR-BI-mediated selective uptake of HDL-CE by CHO-SR-BI cells was efficiently inhibited by AGE-BSA although AGE-BSA had no effect on HDL binding to CHO-SR-BI cells. In addition, AGE-BSA significantly inhibited the efflux of [3H] cholesterol from CHO-SR-BI cells to HDL. These findings suggest the possibility that AGE proteins in the circulation interfere with the functions of SR-BI in reverse cholesterol transport by inhibiting the selective uptake of HDL-CE, as well as cholesterol efflux from peripheral cells to HDL, thereby accelerating diabetes-induced atherosclerosis.


Assuntos
Antígenos CD36/efeitos dos fármacos , Colesterol/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , Proteínas de Membrana , Receptores de Lipoproteínas/antagonistas & inibidores , Animais , Transporte Biológico/efeitos dos fármacos , Antígenos CD36/metabolismo , Células CHO/efeitos dos fármacos , Bovinos , Ésteres do Colesterol/metabolismo , Cricetinae , Humanos , Lipoproteínas HDL/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Lipoproteínas/metabolismo , Receptores Depuradores , Receptores Depuradores Classe B
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
19.
Chem Biol Interact ; 143-144: 353-61, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12604222

RESUMO

In this study, we isolated a cDNA for tetrameric carbonyl reductase (CR) from pig heart. The pig CR showed high amino acid sequence identity (81%) with rabbit NADP(+)-dependent retinol dehydrogenase (NDRD). The purified recombinant pig CR and NDRD were about 100-kDa homotetramers and exhibited high reductase activity towards alkyl phenyl ketones, alpha-dicarbonyl compounds and all-trans-retinal. The identity of NDRD with the tetrameric CR was verified by protein sequencing of CR purified from rabbit heart. Both tetrameric CR and its mRNA were ubiquitously expressed in pig and rabbit tissues. The pig and rabbit enzymes belonged to the short-chain dehydrogenase/reductase family, and their sequences comprise a C-terminal SRL tripeptide, which is a variant of the type 1 peroxisomal targeting signal, SKL. Transfection of HeLa cells with vectors expressing pig CR demonstrated that the enzyme is localized in the peroxisomes. Thus, the tetrameric form of CR represents the first mammalian peroxisomal enzyme that reduces all-trans-retinal as the endogenous substrate.


Assuntos
Oxirredutases do Álcool/metabolismo , Oxirredutases do Álcool/química , Oxirredutases do Álcool/genética , Aldeído Redutase , Aldo-Ceto Redutases , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Suínos , Distribuição Tecidual
20.
Yakugaku Zasshi ; 123(8): 673-9, 2003 Aug.
Artigo em Japonês | MEDLINE | ID: mdl-12931663

RESUMO

Photoaffinity labeling is a useful and reliable method for 1) the identification of the ligand-target receptor and 2) the structural investigation of its binding site. Using photoaffinity labeling techniques, the binding sites of four typical calcium antagonists, 1,4-dihydropyridines, benzothiazepines, phenylalkylamines, and benzothiazines, were successfully identified within the primary structure of the skeletal muscle calcium channels. The results confirm pharmacological observations of the four antagonists, which had been proposed to interact allosterically with each other. Secondarily we demonstrated that human glutathione S-transferase class pi (GST pi) is specifically photolabeled by the antidiabetic agent sulfonylurea glibenclamide (GB) and it also inhibits the enzyme activities of glutathione conjugation by GB in a competitive manner for glutathione. These results indicate that GST pi is another target molecule of sulfonylurea since a subunit of ATP-sensitive potassium channels is well known to be a sulfonylurea receptor. This review focuses on photoaffinity labeling techniques as a useful tool for drug discovery and development.


Assuntos
Marcadores de Fotoafinidade , Sítios de Ligação , Bloqueadores dos Canais de Cálcio , Canais de Cálcio , Desenho de Fármacos , Glutationa Transferase , Hipoglicemiantes , Ligantes , Compostos de Sulfonilureia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA