Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
ACS Omega ; 8(27): 24467-24476, 2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37457445

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-34375687

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/therapeutic use , Bradykinin/therapeutic use , Cell Line, Tumor , Doxorubicin/therapeutic use , Drug Carriers/therapeutic use , Methacrylates , Mice , Nanomedicine , Neoplasms/drug therapy , Polymers/therapeutic use
3.
Histochem Cell Biol ; 156(5): 409-421, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34331103

ABSTRACT

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.


Subject(s)
Nuclear Pore Complex Proteins/genetics , Cell Movement , Cells, Cultured , HeLa Cells , Humans , Nuclear Pore Complex Proteins/metabolism
4.
BMC Cancer ; 18(1): 519, 2018 May 03.
Article in English | MEDLINE | ID: mdl-29724197

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Nucleus/pathology , Cell Transformation, Neoplastic/pathology , Nuclear Pore Complex Proteins/metabolism , Vimentin/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Phosphorylation , RNA, Small Interfering/metabolism , Vimentin/genetics
5.
J Control Release ; 277: 23-34, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29530390

ABSTRACT

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.


Subject(s)
Cell Membrane Permeability/drug effects , Cell-Penetrating Peptides/administration & dosage , Drug Delivery Systems/methods , Drug Design , Nanoparticles/administration & dosage , Serum Albumin, Human/administration & dosage , Cell Membrane Permeability/physiology , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/metabolism , Dose-Response Relationship, Drug , HeLa Cells , Humans , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Serum Albumin, Human/chemical synthesis , Serum Albumin, Human/metabolism , Structure-Activity Relationship
6.
Glia ; 64(11): 1938-61, 2016 11.
Article in English | MEDLINE | ID: mdl-27464357

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/metabolism , Brain/cytology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Microglia/enzymology , Microglia/immunology , Animals , Animals, Newborn , Antigens/metabolism , Antigens, CD/metabolism , COS Cells/drug effects , COS Cells/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Chlorocebus aethiops , Ectodysplasins/metabolism , Embryo, Mammalian , Eye/embryology , Eye/growth & development , Eye/metabolism , Female , Furin/genetics , Furin/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Interleukin-12/pharmacology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Microfilament Proteins/metabolism , Microglia/classification , Microglia/drug effects , Proteoglycans/metabolism , Rats , Rats, Wistar
7.
Clin Cancer Res ; 21(13): 3041-51, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25779950

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia/drug therapy , Lymphoma/drug therapy , Peptides/pharmacology , Receptors, Interleukin-11/antagonists & inhibitors , Amino Acid Sequence , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Ligands , Molecular Sequence Data
8.
J Alzheimers Dis ; 42(2): 587-605, 2014.
Article in English | MEDLINE | ID: mdl-24916544

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative process involving amyloid-ß (Aß) peptide deposition, neuroinflammation, and progressive memory loss. Here, we evaluated whether oral administration of retinoic acid receptor (RAR)α,ß agonist Am80 (tamibarotene) or specific retinoid X receptor (RXR) pan agonist HX630 or their combination could improve deficits in an AD model, 8.5-month-old amyloid-ß protein precursor 23 (AßPP23) mice. Co-administration of Am80 (0.5 mg/kg) and HX630 (5 mg/kg) for 17 days significantly improved memory deficits (Morris water maze) in AßPP23 mice, whereas administration of either agent alone produced no effect. Only co-administration significantly reduced the level of insoluble Aß peptide in the brain. These results thus indicate that effective memory improvement via reduction of insoluble Aß peptide in 8.5-month-old AßPP23 mice requires co-activation of RARα,ß and RXRs. RARα-positive microglia accumulated Aß plaques in the AßPP23 mice. Rat primary microglia co-treated with Am80/HX630 showed increased degradation activity towards 125I-labeled oligomeric Aß1-42 peptide in an insulin-degrading enzyme (IDE)-dependent manner. The co-administration increased mRNA for IDE and membrane-associated IDE protein in vivo, suggesting that IDE contributes to Aß clearance in Am80/HX630-treated AßPP23 mice. Am80/HX630 also increased IL-4Rα expression in microglial MG5 cells. The improvement in memory of Am80/HX630-treated AßPP23 mice was correlated with the levels and signaling of hippocampal interleukin-4 (IL-4). Therefore, Am80/HX630 may promote differentiation of IL-4-responsive M2-like microglia and increase their activity for clearance of oligomeric Aß peptides by restoring impaired IL-4 signaling in AßPP23 mice. Combination treatment with RAR and RXR agonists may be an effective approach for AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Antipsychotic Agents/therapeutic use , Benzazepines/therapeutic use , Benzoates/therapeutic use , Tetrahydronaphthalenes/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Benzazepines/chemistry , Benzazepines/pharmacology , Benzoates/chemistry , Benzoates/pharmacology , Brain/drug effects , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Drug Therapy, Combination , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Insulysin/genetics , Insulysin/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Mutation/genetics , Rats , Rats, Wistar , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/pharmacology , Time Factors
9.
Article in English | MEDLINE | ID: mdl-23295485

ABSTRACT

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.


Subject(s)
DNA Repair Enzymes/chemistry , Phosphoric Monoester Hydrolases/chemistry , Amino Acid Substitution , Crystallization/methods , Crystallography, X-Ray , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , Glycine/chemistry , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Humans , Lysine/chemistry , Mutation , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Protein Conformation
10.
Int Immunopharmacol ; 12(1): 158-68, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22155625

ABSTRACT

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.


Subject(s)
Adenylyl Cyclase Inhibitors , Anaphylatoxins/pharmacology , Chemotactic Factors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Immunologic Factors/pharmacology , Adenylyl Cyclases/metabolism , Anaphylatoxins/antagonists & inhibitors , Calcium/metabolism , Cell Line , Cells, Cultured , Chemotactic Factors/antagonists & inhibitors , Chemotaxis/drug effects , Cyclic AMP/metabolism , Histamine Release/drug effects , Humans , Imines/pharmacology , Immunologic Factors/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mast Cells/drug effects , Mast Cells/physiology , Neutrophils/drug effects , Neutrophils/physiology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology
11.
J Pharm Sci ; 100(9): 3968-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21480235

ABSTRACT

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.


Subject(s)
Antioxidants/pharmacology , Cysteine/analysis , Renal Dialysis , Serum Albumin/physiology , Aged , Aged, 80 and over , Cells, Cultured , Chromatography, High Pressure Liquid , Endothelium, Vascular/metabolism , Female , Humans , Male , Middle Aged , Reactive Oxygen Species/metabolism , Serum Albumin/chemistry
12.
Biochim Biophys Acta ; 1811(3): 153-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21146630

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Lipoproteins, LDL/pharmacology , Lysophosphatidylcholines/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Reactive Oxygen Species/metabolism , 3T3-L1 Cells , Adipocytes/pathology , Animals , Antibodies/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mice , Time Factors
13.
Blood ; 117(3): 920-7, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21063027

ABSTRACT

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.


Subject(s)
Leukemia/metabolism , Lymphoma/metabolism , Neuropilin-1/metabolism , Oligopeptides/pharmacology , Acute Disease , Amino Acid Sequence , Apoptosis/drug effects , Binding Sites/genetics , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , K562 Cells , Leukemia/genetics , Leukemia/pathology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Lymphoma/genetics , Lymphoma/pathology , Molecular Sequence Data , Neuropilin-1/genetics , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Library , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding , RNA Interference , U937 Cells
14.
J Leukoc Biol ; 87(6): 965-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20089669

ABSTRACT

Skp derived from Escherichia coli attracts leukocytes as a pure chemotactic ligand of the C5a receptor. We identified the submolecular region of Skp that binds and activates the C5a receptor to be -Gln103-Asp104-Arg105- using synthetic peptide fragments and site-directed mutants of Skp. As the C5a amino acid residue equivalent to Gln103 of Skp is Leu72, we prepared a Gln103Leu-Skp mutant as a recombinant protein. With this mutation, Skp gained secretagogue functions including induction of the respiratory burst and granule release reactions and leukotriene generation, in addition to the chemoattraction displayed by C5a. However, when we substituted Leu72 with Gln in C5a, the L72Q-C5a mutant largely lost its secretagogue function. These functional conversions were reproduced using synthetic peptides mimicking the receptor-binding/-activating regions of the recombinant proteins. Receptor-binding assays using the mimicking peptides demonstrated only a small difference between the Leu72-C5a and Gln72-C5a peptides. Consistently, L72Q-C5a apparently antagonized C5a secretagogue function. These results indicate that the difference between a chemotactic response and a combined chemotactic/secretory response can be attributed not to the nature of the receptor but to guidance by the ligand, at least in the case of C5a receptor-mediated leukocyte responses.


Subject(s)
Chemotactic Factors/metabolism , Chemotaxis, Leukocyte/physiology , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Molecular Chaperones/metabolism , Neutrophils/metabolism , Peptide Fragments/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Calcium/metabolism , Cytoplasm , Guinea Pigs , Histamine/metabolism , Male , Molecular Sequence Data , Mutation/genetics , Protein Binding , Receptor, Anaphylatoxin C5a/genetics , Recombinant Proteins/metabolism , Respiratory Burst , Sequence Homology, Amino Acid
15.
J Electron Microsc (Tokyo) ; 59(3): 215-26, 2010.
Article in English | MEDLINE | ID: mdl-19995890

ABSTRACT

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.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/ultrastructure , Microscopy, Electron/methods , Animals , Calcium Channels, L-Type/isolation & purification , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Muscle, Skeletal/chemistry , Rabbits
16.
Cancer Sci ; 101(1): 137-42, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19843070

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Benzamides , Drug Resistance, Neoplasm , Enzyme Activation , Humans , Imatinib Mesylate , K562 Cells , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
17.
Biol Pharm Bull ; 32(7): 1307-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19571405

ABSTRACT

The purpose of this study is to investigate whether a synthetic retinoid Am80 (tamibarotene) exhibits any improving effects on amyloid precursor protein (APP)23 mice, a model of Alzheimer's disease. Am80 was orally administered in feed to 20-week (5-month)-old APP23 mice at a dose of 0 (control) or 0.5 mg/kg/d for 14 weeks. The Am80 treatment reduced significantly the insoluble Abeta levels in brain, in particular Abeta(42), while it gave no apparent effects on the soluble Abeta levels. The results suggest that oral administration of Am80 may have potency to reduce the extracellular Abeta(42) of insoluble and possibly oligomeric or protofibril forms, which are related to the cause and/or progression of Alzheimer's disease. The Am80 treatment showed no significant effect on spatial learning and memory of APP23 mice by Morris water maze analysis. The main reason for the absence of significance seems based on the large deviation and some mice both in the treated and the non-treated groups would neither swim nor make efforts to reach the platform.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/antagonists & inhibitors , Benzoates/pharmacology , Brain/drug effects , Peptide Fragments/antagonists & inhibitors , Retinoids/pharmacology , Tetrahydronaphthalenes/pharmacology , Administration, Oral , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Benzoates/administration & dosage , Brain/metabolism , Disease Models, Animal , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Retinoids/administration & dosage , Tetrahydronaphthalenes/administration & dosage
18.
J Neuroimmunol ; 208(1-2): 70-9, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19211155

ABSTRACT

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.


Subject(s)
CD40 Antigens/biosynthesis , Gene Expression Regulation, Enzymologic/immunology , Microglia/immunology , Microglia/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , CD40 Antigens/deficiency , CD40 Antigens/metabolism , Cells, Cultured , Enzyme Induction/genetics , Enzyme Induction/immunology , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Inflammation Mediators/toxicity , Lipopolysaccharides/physiology , Microglia/enzymology , Nitric Oxide/biosynthesis , Nitric Oxide/toxicity , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/toxicity
19.
J Biol Chem ; 284(6): 3396-407, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19068484

ABSTRACT

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.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Cycle Proteins/metabolism , DNA Replication/physiology , DNA, Fungal/metabolism , Origin Recognition Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/genetics , Cell Cycle Proteins/genetics , Checkpoint Kinase 2 , Chromosomes, Fungal/physiology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA, Fungal/genetics , G1 Phase/physiology , Origin Recognition Complex/genetics , Phosphorylation/physiology , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , S Phase/physiology , Saccharomyces cerevisiae Proteins/genetics
20.
J Biol Chem ; 283(17): 11752-62, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18292083

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic , Leukemia/metabolism , Lymphoma/metabolism , Catalysis , Cell Line, Tumor , Cell Survival , Chemistry, Pharmaceutical/methods , Drug Design , Humans , K562 Cells , Ligands , Peptide Library , Peptides/chemistry , Pinocytosis
SELECTION OF CITATIONS
SEARCH DETAIL