Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Med Chem ; 67(5): 3307-3320, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38105611

ABSTRACT

Polo-like kinase 1 (PLK1), which is crucial in cell cycle regulation, is considered a promising anticancer drug target. Herein, we present the N-degron pathway-based proteolysis targeting chimera (PROTAC) for PLK1 degradation, targeting the Polo-box domain (PBD). We identified DD-2 as the most potent PROTAC that selectively induces PLK1 degradation in cancer cells, including HeLa and nonsmall cell lung cancer (NSCLC), through the N-degron pathway. DD-2 exhibited significant in vitro anticancer effects, inducing G2/M arrest and apoptosis in HeLa and NSCLC cell lines. DD-2 showed significant tumor growth inhibition in a xenograft mouse model using HeLa and NSCLC cell lines, highlighting its potential in cancer treatment. Furthermore, the combination of DD-2 with tyrosine kinase inhibitor (TKI), osimertinib, effectively suppressed tumor growth in double-mutated H1975 cell lines, emphasizing DD-2's potential in combination cancer therapies. Collectively, this study demonstrates the potential of the N-degron pathway, especially using DD-2, for targeted cancer therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins , Proteolysis Targeting Chimera , Protein Serine-Threonine Kinases , Polo-Like Kinase 1 , Apoptosis , Degrons , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , G2 Phase Cell Cycle Checkpoints , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Pharmaceutics ; 13(12)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34959352

ABSTRACT

Cancer has been identified as a leading cause of death worldwide, and the increasing number of cancer cases threatens to shorten the average life expectancy of people. Recently, we reported a 3-azido-3-deoxythymidine (AZT)-based amphipathic small molecule, ADG-2e that revealed a notable potency against tumor metastasis. To evaluate the anticancer potential of ADG-2e, we assessed its anticancer potency in vitro and in vivo. Anticancer screening of ADG-2e against cervical cancer cells, HeLa CCL2, and BT549 mammary gland ductal carcinoma showed significant inhibition of cancer cell proliferation. Furthermore, mechanistic investigations revealed that cancer cell death presumably proceeded through an oncosis mechanistic pathway because ADG-2e treated cells showed severe damage on the plasma membrane, a loss of membrane integrity, and leakage of α-tubulin and ß-actin. Finally, evaluation of the antitumorigenic potential of ADG-2e in mouse xenograft models revealed that this compound potentially inhibits cancer cell proliferation. Collectively, these findings suggest that ADG-2e can evolve as an anticancer agent, which may represent a model for nucleoside-based small molecule anticancer drug discovery.

3.
Diagnostics (Basel) ; 11(11)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34829485

ABSTRACT

Motion estimation and compensation are necessary for improvement of tumor quantification analysis in positron emission tomography (PET) images. The aim of this study was to propose adaptive PET imaging with internal motion estimation and correction using regional artificial evaluation of tumors injected with low-dose and high-dose radiopharmaceuticals. In order to assess internal motion, molecular sieves imitating tumors were loaded with 18F and inserted into the lung and liver regions in rats. All models were classified into two groups, based on the injected radiopharmaceutical activity, to compare the effect of tumor intensity. The PET study was performed with injection of F-18 fluorodeoxyglucose (18F-FDG). Respiratory gating was carried out by external trigger device. Count, signal to noise ratio (SNR), contrast and full width at half maximum (FWHM) were measured in artificial tumors in gated images. Motion correction was executed by affine transformation with estimated internal motion data. Monitoring data were different from estimated motion. Contrast in the low-activity group was 3.57, 4.08 and 6.19, while in the high-activity group it was 10.01, 8.36 and 6.97 for static, 4 bin and 8 bin images, respectively. The results of the lung target in 4 bin and the liver target in 8 bin showed improvement in FWHM and contrast with sufficient SNR. After motion correction, FWHM was improved in both regions (lung: 24.56%, liver: 10.77%). Moreover, with the low dose of radiopharmaceuticals the PET image visualized specific accumulated radiopharmaceutical areas in the liver. Therefore, low activity in PET images should undergo motion correction before quantification analysis using PET data. We could improve quantitative tumor evaluation by considering organ region and tumor intensity.

4.
Nanomedicine ; 34: 102394, 2021 06.
Article in English | MEDLINE | ID: mdl-33857687

ABSTRACT

Gold nanoparticles (AuNPs) have been widely used as nanocarriers in drug delivery to improve the efficiency of chemotherapy treatment and enhance early disease detection. The advantages of AuNPs include their excellent biocompatibility, easy modification and functionalization, facile synthesis, low toxicity, and controllable particle size. This study aimed to synthesize a conjugated citraconic anhydride link between morphologically homogeneous AuNPs and doxorubicin (DOX) (DOX-AuNP). The carrier was radiolabeled for tumor diagnosis using positron emission tomography (PET). The systemically designed DOX-AuNP was cleaved at the citraconic anhydride linker site under the mild acidic conditions of a cancer cell, thereby releasing DOX. Subsequently, the AuNPs aggregated via electrostatic attraction. HeLa cancer cells exhibited a high uptake of the radiolabeled DOX-AuNP. Moreover, PET tumor images were obtained using radiolabeled DOX-AuNP in cancer xenograft mouse models. Therefore, DOX-AuNP is expected to provide a valuable insight into the use of radioligands to detect tumors using PET.


Subject(s)
Gold/chemistry , Hydrogen-Ion Concentration , Metal Nanoparticles/chemistry , Uterine Cervical Neoplasms/diagnostic imaging , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/therapeutic use , Female , Gold/metabolism , HeLa Cells , Humans , Mice , Positron-Emission Tomography , Radioligand Assay , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Xenograft Model Antitumor Assays
5.
Cell Cycle ; 19(24): 3419-3436, 2020 12.
Article in English | MEDLINE | ID: mdl-33323015

ABSTRACT

Polo-like kinase 4 (Plk4) is a key regulator of centriole biogenesis. Studies have shown that Plk4 undergoes dynamic relocalization from a ring-like pattern around a centriole to a dot-like morphology at the procentriole assembly site and this event is central for inducing centriole biogenesis. However, the detailed mechanisms underlying Plk4's capacity to drive its symmetry-breaking ring-to-dot relocalization remain largely unknown. Here, we showed that Plk4 self-initiates this process in an autophosphorylation-dependent manner and that STIL, its downstream target, is not required for this event. Time-dependent analyses with mEOS-fused photoconvertible Plk4 revealed that a portion of ring-state Plk4 acquires a capacity, presumably through autophosphorylation, to linger around a centriole, ultimately generating a dot-state morphology. Interestingly, Plk4 WT, but not its catalytically inactive mutant, showed the ability to form a nanoscale spherical assembly in the cytosol of human cells or heterologous E. coli, demonstrating its autophosphorylation-dependent self-organizing capacity. At the biochemical level, Plk4 - unlike its N-terminal ßTrCP degron motif - robustly autophosphorylated the PC3 SSTT motif within its C-terminal cryptic polo-box, an event critical for inducing its physical clustering. Additional in vivo experiments showed that although STIL was not required for Plk4's initial ring-to-dot conversion, coexpressed STIL greatly enhanced Plk4's ability to generate a spherical condensate and recruit Sas6, a major component of the centriolar cartwheel structure. We propose that Plk4's autophosphorylation-induced clustering is sufficient to induce its ring-to-dot localization conversion and that subsequently recruited STIL potentiates this process to generate a procentriole assembly body critical for Plk4-dependent centriole biogenesis.


Subject(s)
Centrioles/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Biocatalysis , Cell Cycle/genetics , Cell Line, Tumor , Cytosol/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Osteosarcoma/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Protein Serine-Threonine Kinases/genetics , Proteolysis , RNA Interference , Transfection
6.
J Med Chem ; 63(23): 14905-14920, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33142063

ABSTRACT

Polo-like kinase-1 (Plk1) plays a key role in mitosis and has been identified as an attractive anticancer drug target. Plk1 consists of two drug-targeting sites, namely, N-terminal kinase domain (KD) and C-terminal polo-box domain (PBD). As KD-targeting inhibitors are associated with severe side effects, here we report on the pyrazole-based Plk1 PBD inhibitor, KBJK557, which showed a remarkable in vitro anticancer effect by inducing Plk1 delocalization, mitotic arrest, and apoptosis in HeLa cells. Further, in vivo optical imaging analysis and antitumorigenic activities in mouse xenograft models demonstrate that KBJK557 preferentially accumulates in cancer cells and selectively inhibits cancer cell proliferation. Pharmacokinetic profiles and partition coefficients suggest that KBJK557 was exposed in the blood and circulated through the organs with an intermediate level of clearance (t1/2, 7.73 h). The present investigation offers a strategy for specifically targeting cancer using a newly identified small-molecule inhibitor that targets the Plk1 PBD.


Subject(s)
Antineoplastic Agents/therapeutic use , Barbiturates/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Barbiturates/chemical synthesis , Barbiturates/metabolism , Barbiturates/pharmacokinetics , Carbocyanines/chemistry , Cell Cycle Proteins/metabolism , Drug Design , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemistry , G2 Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , Humans , Male , Mice, Inbred BALB C , Mice, Inbred ICR , Molecular Structure , Neoplasms/diagnosis , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
7.
Int J Mol Sci ; 21(12)2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32560337

ABSTRACT

89Zr is an emerging radionuclide that plays an essential role in immuno-positron emission tomography (PET) imaging. The long half-life of 89Zr (t1/2 = 3.3 days) is favorable for evaluating the in vivo distribution of monoclonal antibodies. Thus, the use of 89Zr is promising for monitoring antibody-based cancer therapies. Immuno-PET combines the sensitivity of PET with the specificity of antibodies. A number of studies have been conducted to investigate the feasibility of 89Zr immuno-PET imaging for predicting the efficacy of radioimmunotherapy and antibody therapies, imaging target expression, detecting target-expressing tumors, and the monitoring of anti-cancer chemotherapies. In this review, we summarize the current status of PET imaging using 89Zr in both preclinical and clinical studies by highlighting the use of immuno-PET for the targets of high clinical relevance. We also present 89Zr-PET applications other than immuno-PET, such as nanoparticle imaging and cell tracking. Finally, we discuss the limitations and the ongoing research being performed to overcome the remaining hurdles.


Subject(s)
Positron-Emission Tomography , Radioisotopes , Radiopharmaceuticals , Zirconium , Animals , Antigens, CD20 , Biomarkers , Biomarkers, Tumor , Disease Management , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunoconjugates , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/therapy , Positron-Emission Tomography/methods
8.
Int J Mol Sci ; 21(10)2020 May 20.
Article in English | MEDLINE | ID: mdl-32443730

ABSTRACT

To combat the escalating rise of antibacterial resistance, the development of antimicrobial peptides (AMPs) with a unique mode of action is considered an attractive strategy. However, proteolytic degradation of AMPs remains the greatest challenge in their transformation into therapeutics. Herein, we synthesized Fmoc-triazine amino acids that differ from each other by anchoring either cationic or hydrophobic residues. These unnatural amino acids were adopted for solid-phase peptide synthesis (SPPS) to synthesize a series of amphipathic antimicrobial peptidomimetics. From the antimicrobial screening, we found that the trimer, BJK-4 is the most potent short antimicrobial peptidomimetic without showing hemolytic activity and it displayed enhanced proteolytic stability. Moreover, the mechanism of action to kill bacteria was found to be an intracellular targeting.


Subject(s)
Biomimetic Materials/chemical synthesis , Pore Forming Cytotoxic Proteins/chemical synthesis , Triazines/chemistry , Amino Acids/chemistry , Animals , Biomimetic Materials/pharmacology , Cells, Cultured , Erythrocytes/drug effects , Escherichia coli/drug effects , Hemolysis , Hydrophobic and Hydrophilic Interactions , Mice , Pore Forming Cytotoxic Proteins/pharmacology , Protein Stability , Pseudomonas aeruginosa/drug effects , RAW 264.7 Cells , Sheep , Staphylococcus aureus/drug effects , Structure-Activity Relationship
9.
Eur J Med Chem ; 193: 112233, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32199136

ABSTRACT

Antimicrobial peptides (AMP) with anticancer activity have drawn remarkable attention in modern treatments. However, long peptide length and protease instability are the most addressing factors, which hampers their further development as therapeutic agents. In view of this, herein, we designed and synthesized a series of AZT-based cationic small molecule incorporating a variety of hydrophobic groups and cationic charges, including amine and guanidine groups to mimic the amphipathic structure of AMPs. These compounds were evaluated for their antibacterial activity against Gram-positive and Gram-negative bacteria. Through an extensive structure activity relationship study (SAR), we identified ADG-2e as the most potent antibacterial agent, which exhibited remarkable potency against drug resistant bacterial strains such as MRSA and MDRPA. Further, ADG-2e was examined for their anti-metastatic ability by investigating the cancer cell migration and invasiveness through scratch wound-healing assay and transwell invasive assay, respectively. In addition, time-lapse cell tracking analysis also performed for analyzing the cell movement pattern. Treatment of ADG-2e against metastatic breast cancer cells (MDA-MB-231) suppressed tumor cell migration by multi-directional lamellipodium formation, indicating their anti-metastatic potential. Thus, our cationic AZT based small molecules may evolve as an appealing class of antibacterial agents with anti-metastasis potential.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Zidovudine/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Female , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/growth & development , Humans , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured , Wound Healing/drug effects , Zidovudine/chemical synthesis , Zidovudine/chemistry
10.
Mol Cell Biol ; 40(10)2020 04 28.
Article in English | MEDLINE | ID: mdl-32152252

ABSTRACT

Cep57 has been characterized as a component of a pericentriolar complex containing Cep63 and Cep152. Interestingly, Cep63 and Cep152 self-assemble into a pericentriolar cylindrical architecture, and this event is critical for the orderly recruitment of Plk4, a key regulator of centriole duplication. However, the way in which Cep57 interacts with the Cep63-Cep152 complex and contributes to the structure and function of Cep63-Cep152 self-assembly remains unknown. We demonstrate that Cep57 interacts with Cep63 through N-terminal motifs and associates with Cep152 via Cep63. Three-dimensional structured illumination microscopy (3D-SIM) analyses suggested that the Cep57-Cep63-Cep152 complex is concentrically arranged around a centriole in a Cep57-in and Cep152-out manner. Cep57 mutant cells defective in Cep63 binding exhibited improper Cep63 and Cep152 localization and impaired Sas6 recruitment for procentriole assembly, proving the significance of the Cep57-Cep63 interaction. Intriguingly, Cep63 fused to a microtubule (MT)-binding domain of Cep57 functioned in concert with Cep152 to assemble around stabilized MTs in vitro Thus, Cep57 plays a key role in architecting the Cep63-Cep152 assembly around centriolar MTs and promoting centriole biogenesis. This study may offer a platform to investigate how the organization and function of the pericentriolar architecture are altered by disease-associated mutations found in the Cep57-Cep63-Cep152 complex.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Cell Cycle Proteins/analysis , Cell Line, Tumor , HEK293 Cells , Humans , Microtubule-Associated Proteins/analysis , Nuclear Proteins/analysis , Protein Interaction Domains and Motifs , Protein Interaction Maps
11.
Sci Rep ; 9(1): 15161, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641232

ABSTRACT

Considering the emergence of bacterial resistance and low proteolytic stability of antimicrobial peptides (AMPs), herein we developed a series of ultra-short triazine based amphipathic polymers (TZP) that are connected with ethylene diamine linkers instead of protease sensitive amide bond. The most potent oligomers, TZP3 and TZP5 not only displayed potent antibacterial action on various drug-resistant pathogens but also exhibited a strong synergic antibacterial activity in combination with chloramphenicol against multidrug-resistant Pseudomonas aeruginosa (MDRPA). Since most of atopic dermatitis (AD) infections are caused by bacterial colonization, we evaluated the potency of TZP3 and TZP5 on AD in vitro and in vivo. In vitro AD analysis of these two polymers showed significant inhibition against the release of ß-hexosaminidase and tumor necrosis factor (TNF-α) from RBL-2H3 cells. In AD-like skin lesions in BALB/c mice model, these two polymers displayed significant potency in suppressing dermal and epidermal thickness, mast cell infiltration and pro-inflammatory cytokines expression. Moreover, these polymers exhibited remarkable efficacy over the allergies caused by the imbalance of Th1/Th2 by regulating total IgE and IgG2a. Finally, the impact of treatment effects of these polymers was examined through analyzing the weights and sizes of spleen and lymph node of AD-induced mice.


Subject(s)
Anti-Bacterial Agents/pharmacology , Polymers/pharmacology , Surface-Active Agents/pharmacology , Triazines/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Cytokines/metabolism , Dermatitis, Atopic/blood , Dermatitis, Atopic/pathology , Disease Models, Animal , Drug Resistance, Microbial/drug effects , Enzyme Stability/drug effects , Erythrocytes/drug effects , Hemolysis , Hydrophobic and Hydrophilic Interactions , Immunoglobulin E/blood , Immunoglobulin G/blood , Inflammation Mediators/metabolism , Lymph Nodes/drug effects , Lymph Nodes/pathology , Mast Cells/drug effects , Mice, Inbred BALB C , Microbial Sensitivity Tests , Peptide Hydrolases/metabolism , Polymers/chemistry , Sheep , Skin/drug effects , Skin/pathology , Spleen/drug effects , Spleen/pathology , Triazines/chemistry
12.
Mol Pharm ; 16(12): 4867-4877, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31663746

ABSTRACT

Polo-like kinase 1 (Plk1) regulates cell cycle and cell proliferation, and is currently considered a potential biomarker in clinical trials for many cancers. A characteristic feature of Plks is their C-terminal polo-box domain (PBD). Pro-Leu-His-Ser-pThr (PLHS[pT])-the phosphopeptide inhibitor of the PBD of Plk1-induces apoptosis in cancer cells. However, because of the low cell membrane-penetration ability of PLHS[pT], new approaches are required to overcome these drawbacks. We therefore developed a vitamin E (VE) conjugate that is biodegradable by intracellular redox enzymes as an anticancer drug-delivery system. To ensure high efficiency of membrane penetration, we synthesized VE-S-S-PLHS[pT]KY (1) by conjugating PLHS[pT] to VE via a disulfide bond. We found that 1 penetrated cancer cell membranes, blocked cancer cell proliferation, and induced apoptosis in cancer cells through cell cycle arrest in the G2/M phase. We synthesized a radiolabeled peptide (124I-1), and the radioligand was evaluated in in vivo tumor uptake using positron emission tomography. This study shows that combination conjugates are an excellent strategy for specifically targeting Plk PBD. These conjugates have a dual function, with possible uses in anticancer therapy and tumor diagnosis.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Phosphopeptides/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Vitamin E/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Flow Cytometry , HeLa Cells , Humans , Mitosis/drug effects , Polo-Like Kinase 1
13.
Int J Mol Med ; 40(6): 1971-1982, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039468

ABSTRACT

Amyloid-ß (Aß), a main pathogenic factor of Alzheimer's disease (AD), induces apoptosis accompanied by caspase activation. However, limited caspase activation and the suppression of the intrinsic apoptotic pathway (IAPW) are frequently observed upon Aß treatment. In this study, we investigated whether these suppressive effects of Aß can be overcome; we also examined the death-related pathways. Single treatments of cells with Aß42 for up to 48 h barely induced caspase activation. In cells treated with Aß42 twice for 2 h followed by 22 h (2+22 h) or for longer durations, the apoptotic protease activating factor-1 (Apaf-1) apoptosome was formed and caspases-3 and -9 were activated to a certain extent, suggesting the activation of the IAPW. However, the Aß42-induced activation of the IAPW differed from that induced by treatment with other agents, such as staurosporine (STS) in that lower amounts of cytochrome c were released from the mitochondria, the majority of procaspase-9 in the Apaf-1 complex was not processed and caspase-3 was activated to a lesser extent in the peptide-treated cells. Thus, it seemed that the IAPW was not fully activated by Aß42. The 30- and 41/43-kDa fragments derived from procaspase-8 were detected, which appear to be produced through the IAPW without death-inducing signaling-complex (DISC) formation, a key feature of the extrinsic apoptotic pathway (EAPW). Bid cleavage was observed only after caspase-3 activity reached its maximal levels, suggesting that the cleavage may contribute in a limited capacity to the amplification process of the IAPW in the Aß-treated cells. Taken together, our data suggest that the IAPW, albeit functional only to a limited extent, plays a major role in Aß42-induced apoptosis without the EAPW.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/genetics , Apoptosis/genetics , Staurosporine/administration & dosage , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Apoptotic Protease-Activating Factor 1/genetics , Caspase 3/genetics , Caspase 9/genetics , Humans , Mitochondria/drug effects , Mitochondria/genetics , Signal Transduction/drug effects
14.
Eur J Med Chem ; 125: 551-564, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27718471

ABSTRACT

In this study, we report on the first chemical synthesis of ultra-short pyrazole-arginine based antimicrobial peptidomimetics derived from the newly synthesized N-alkyl/aryl pyrazole amino acids. Through the systematic tuning of hydrophobicity, charge, and peptide length, we identified the shortest peptide Py11 with the most potent antimicrobial activity. Py11 displayed greater antimicrobial activity against antibiotic-resistant bacteria, including MRSA, MDRPA, and VREF, which was approximately 2-4 times higher than that of melittin. Besides its higher selectivity (therapeutic index) toward bacterial cells than LL-37, Py11 showed highly increased proteolytic stability against trypsin digestion and maintained its antimicrobial activity in the presence of physiological salts. Interestingly, Py11 exhibited higher anti-biofilm activity against MDRPA compared to LL-37. The results from fluorescence spectroscopy and transmission electron microscopy (TEM) suggested that Py11 kills bacterial cells possibly by integrity disruption damaging the cell membrane, leading to the cytosol leakage and eventual cell lysis. Furthermore, Py11 displayed significant anti-inflammatory (endotoxin-neutralizing) activity by inhibiting LPS-induced production of nitric oxide (NO) and TNF-α. Collectively, our results suggest that Py11 may serve as a model compound for the design of antimicrobial and antisepsis agents.


Subject(s)
Antimicrobial Cationic Peptides/chemical synthesis , Biofilms/drug effects , Peptidomimetics/chemistry , Pyrazoles/chemical synthesis , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Bacteria/drug effects , Humans , Macrophages/drug effects , Microbial Sensitivity Tests , Microscopy, Electron, Transmission , Molecular Structure , Peptidomimetics/chemical synthesis , Peptidomimetics/pharmacology , Polymerase Chain Reaction , Pyrazoles/chemistry , Pyrazoles/pharmacology
15.
Mol Cells ; 39(12): 855-861, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27927007

ABSTRACT

Ginsenosides, which are the active materials of ginseng, have biological functions that include anti-osteoporotic effects. Aqueous ginseng extract inhibits osteoclast differentiation induced by receptor activator of NF-κB ligand (RANKL). Aqueous ginseng extract produces chromatography peaks characteristic of ginsenosides. Among these peaks, ginsenoside Re is a major component. However, the preventive effects of ginsenoside Re against osteoclast differentiation are not known. We studied the effect of ginsenoside Re on osteoclast differentiation, RANKL-induced tartrate-resistant acid phosphatase (TRAP) activity, and formation of multinucleated osteoclasts in vitro. Ginsenoside Re hampered osteoclast differentiation in a dose-dependent manner. In an in vivo zebrafish model, aqueous ginseng extract and ginsenoside Re had anti-osteoclastogenesis effects. These findings suggest that both aqueous ginseng extract and ginsenoside Re prevent bone resorption by inhibiting osteoclast differentiation. Ginsenoside Re could be important for promoting bone health.


Subject(s)
Bone Marrow Cells/drug effects , Ginsenosides/pharmacology , Macrophages/drug effects , Osteoclasts/drug effects , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Macrophages/cytology , Mice , Osteoclasts/cytology , Zebrafish
16.
Amino Acids ; 46(11): 2595-603, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25151148

ABSTRACT

In the last decade, drug delivery systems using biologically active molecules for cellular uptake of therapeutic targets have been studied for application and testing in clinical trials. For instance, the transactivator of transcription (TAT) peptide, or cell-penetrating peptide, was shown to deliver a variety of cargoes, including proteins, peptides, and nucleic acids. Polo-like kinase 1 (Plk1) plays key roles in the regulation of cell cycle events (e.g., mitotic progression). Plk1 was also shown to be activated and highly expressed in proliferating cells such as tumor cells. Amongst these phosphopeptides, Pro-Leu-His-Ser-p-Thr (PLHSpT), which is the minimal sequence for polo-box domain (PBD) binding, was shown to have an inhibitory effect and to induce apoptotic cell death. However, the phosphopeptide showed low cell membrane penetration. Thus, in our study, we synthesized Plk1 inhibitor TAT-PLHSpT to improve agent internalization into cells. TAT-PLHSpT was shown to internalize into the nucleus. The conjugation of TAT with PLHSpT inhibited cancer cell growth and survival. Moreover, it showed an increase in cellular uptake and inhibition of Plk1 kinase activity. Further studies are needed for biological evaluation of the new peptide in tumor-bearing animal models (in vivo). Our results prove that TAT-PLHSpT is a good candidate for specific PBD binding of Plk1 as a therapeutic agent for humans.


Subject(s)
Antineoplastic Agents/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Drug Carriers , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Acridine Orange/chemistry , Apoptosis , Binding Sites , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Dose-Response Relationship, Drug , Gene Products, tat/chemistry , HeLa Cells , Humans , Microscopy, Fluorescence , Mitosis , Neoplasms/chemistry , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Polo-Like Kinase 1
17.
Amino Acids ; 46(9): 2259-69, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24961649

ABSTRACT

Diversity of sequence and structure in naturally occurring antimicrobial peptides (AMPs) limits their intensive structure-activity relationship (SAR) study. In contrast, peptidomimetics have several advantages compared to naturally occurring peptide in terms of simple structure, convenient to analog synthesis, rapid elucidation of optimal physiochemical properties and low-cost synthesis. In search of short antimicrobial peptides using peptidomimetics, which provide facile access to identify the key factors involving in the destruction of pathogens through SAR study, a series of simple and short peptidomimetics consisting of multi-Lys residues and lipophilic moiety have been prepared and found to be active against several Gram-negative and Gram-positive bacteria containing methicillin-resistant Staphylococcus aureus (MRSA) without hemolytic activity. Based on the SAR studies, we found that hydrophobicity, +5 charges of multiple Lys residues, hydrocarbon tail lengths and cyclohexyl group were crucial for antimicrobial activity. Furthermore, membrane depolarization, dye leakage, inner membrane permeability and time-killing kinetics revealed that bacterial-killing mechanism of our peptidomimetics is different from the membrane-targeting AMPs (e. g. melittin and SMAP-29) and implied our peptidomimetics might kill bacteria via the intracellular-targeting mechanism as done by buforin-2.


Subject(s)
Anti-Infective Agents/pharmacology , Erythrocytes/metabolism , Hemolysis/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Peptidomimetics/pharmacology , Polylysine/pharmacology , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Erythrocytes/cytology , Humans , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Polylysine/chemical synthesis , Polylysine/chemistry
18.
J Drug Target ; 22(3): 191-199, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-24219340

ABSTRACT

Oleanolic acid (OA) is a pentacyclic triterpenoid found in various plant species. Triterpenoid compounds have been shown to inhibit tumor proliferation and to induce apoptosis in cancer cells. We synthesized an OA derivative and evaluated its inhibitory effects on cell proliferation in human colon cancer. Radioisotope-labeled OA was prepared for noninvasive monitoring of tumor progression in vitro and in vivo. The OA derivative decreased cell survival in a concentration-dependent manner and increased apoptosis in HT-29 cells. Furthermore, it induced downregulation of cyclin D1, Cox-2, Bcl-2 and Bcl-xL mRNA expression and upregulation of the mRNA expression of the anti-apoptotic Bax protein in HT29 cells. NF-κB p65 and IκB expression also decreased, whereas expression of the apoptosis marker, the cleaved form of PARP-1, significantly increased in OA derivative-treated HT-29 cells. Radioisotope-labeled OA (68Ga-NOTA-OA) showed significantly high tumor uptake, as assessed by biodistribution and positron emission tomography imaging analyses, at 1 h post-injection in the human colon cancer xenograft model. Our results demonstrate that the OA derivative has promising properties as an anticancer drug and as an imaging tool for tumor targeting.

19.
Amino Acids ; 45(5): 1149-56, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23907439

ABSTRACT

Peptides are ideal candidates for developing therapeutics. Polo-like kinase 1 is an important regulatory protein in the cell cycle and contains a C-terminal polo-box domain, which is the hallmark of this protein family. We developed a peptide inhibitor of polo-like kinase 1 that targets its polo-box domain. This new phosphopeptide, cRGDyK-S-S-CPLHSpT, preferentially penetrates the cancer cell membrane mediated by the integrin receptor, which is expressed at high levels by cancer cells. In the present study, using high performance liquid chromatography and mass spectroscopy, we determined the stability of cRGDyK-S-S-CPLHSpT and its cleavage by glutathione under typical conditions for cell culture. We further assessed the ability of the peptide to inhibit the proliferation of the U87MG glioma cell line. The phosphorylated peptide was stable, and the disulfide bond of cRGDyK-S-S-CPLHSpT was cleaved in 50 mM glutathione. This peptide inhibited the growth of cancer cells and changed their morphology. Therefore, we conclude that the phosphopeptide shows promise as a prodrug and has a high potential to act as an anticancer agent by inhibiting polo-like kinase 1 by binding its polo-box domain. These findings indicate the therapeutic potential of PLHSpT and peptides similarly targeted to surface receptors of cancer cells and to the functional domains of regulatory proteins.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Glioma/physiopathology , Phosphopeptides/pharmacology , Prodrugs/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glioma/drug therapy , Glioma/enzymology , Glioma/metabolism , Humans , Molecular Structure , Phosphopeptides/chemistry , Prodrugs/chemistry , Polo-Like Kinase 1
20.
Biomaterials ; 34(33): 8114-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23932293

ABSTRACT

Research into multifunctional nanoparticles is focused on creating an agent for use in an all-in-one multimodal imaging system that includes diagnostic imaging, drug delivery, and therapeutic monitoring. We designed a new dual-modality tumor-targeting agent with a new tumor-targeting molecule, oleanolic acid (OA), which is derived from a natural compound and coupled with a macrocyclic chelating agent such as 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), iron oxide nanoparticles (IONP), and radiolabeling components such as (68)Ga for dual-modality positron emission tomography (PET)/magnetic resonance imaging (MRI). We attempted to obtain fusion PET/MR images with the (68)Ga-NOTA-OA-IONP hybrid tumor-targeting imaging agent using colon cancer (HT-29) xenograft mice models. The HT-29 cancer cells showed high uptake of (68)Ga-NOTA-OA-IONP, which also had an inhibitory effect on the cells. Moreover, we obtained PET and MRI tumor images as well as fusion PET/MRI images of the tumors using (68)Ga-NOTA-OA-IONP. Therefore, the dual-modality cancer-targeting radiolabeled nanoparticle reported here is a potent imaging agent that is suitable for PET, MRI, and PET/MRI-based diagnosis of tumors; it also has the advantage of not only detecting tumor functionality, but also simultaneously aiding in tumor resolution.


Subject(s)
Ferric Compounds/chemistry , Magnetic Resonance Imaging/methods , Nanoparticles/chemistry , Oleanolic Acid/chemistry , Positron-Emission Tomography/methods , Animals , Cell Proliferation/drug effects , Colonic Neoplasms/diagnosis , HT29 Cells , Humans , Male , Mice , Mice, Nude , Nanoparticles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...