Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters










Publication year range
1.
Emerg Microbes Infect ; 13(1): 2332652, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38517705

ABSTRACT

A diverse population of avian influenza A viruses (AIVs) are maintained in wild birds and ducks yet the zoonotic potential of AIVs in these environmental reservoirs and the host-virus interactions involved in mammalian infection are not well understood. In studies of a group of subtype H1N1 AIVs isolated from migratory wild birds during surveillance in North America, we previously identified eight amino acids in the polymerase genes PB2 and PB1 that were important for the transmissibility of these AIVs in a ferret model of human influenza virus transmission. In this current study we found that PB2 containing amino acids associated with transmissibility at 67, 152, 199, 508, and 649 and PB1 at 298, 642, and 667 were associated with more rapid viral replication kinetics, greater infectivity, more active polymerase complexes and greater kinetics of viral genome replication and transcription. Pathogenicity in the mouse model was also impacted, evident as greater weight loss and lung pathology associated with greater inflammatory lung cytokine expression. Further, these AIVs all contained the avian-type amino acids of PB2-E627, D701, G590, Q591 and T271. Therefore, our study provides novel insights into the role of the AIV polymerase complex in the zoonotic transmission of AIVs in mammals.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza in Birds , Mice , Animals , Humans , Influenza A Virus, H1N1 Subtype/genetics , Amino Acids/genetics , Host Microbial Interactions , Viral Proteins/genetics , Viral Proteins/metabolism , Ferrets , Influenza A virus/metabolism , Birds , Nucleotidyltransferases , Virus Replication/genetics , Phylogeny
2.
Biomater Res ; 27(1): 124, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38031117

ABSTRACT

BACKGROUND: Recently, increased attention has been given on exosomes as ideal nanocarriers of drugs owing to their intrinsic properties that facilitate the transport of biomolecular cargos. However, large-scale exosome production remains a major challenge in the clinical application of exosome-based drug delivery systems. Considering its biocompatibility and stability, bovine milk is a suitable natural source for large-scale and stable exosome production. Because the active-targeting ability of drug carriers is essential to maximize therapeutic efficacy and minimize side effects, precise membrane functionalization strategies are required to enable tissue-specific delivery of milk exosomes with difficulty in post-isolation modification. METHODS: In this study, the membrane functionalization of a milk exosome platform modified using a simple post-insertion method was examined comprehensively. Exosomes were engineered from bovine milk (mExo) with surface-tunable modifications for the delivery of tumor-targeting doxorubicin (Dox). The surface modification of mExo was achieved through the hydrophobic insertion of folate (FA)-conjugated lipids. RESULTS: We have confirmed the stable integration of functionalized PE-lipid chains into the mExo membrane through an optimized post-insertion technique, thereby effectively enhancing the surface functionality of mExo. Indeed, the results revealed that FA-modified mExo (mExo-FA) improved cellular uptake in cancer cells via FA receptor (FR)-mediated endocytosis. The designed mExo-FA selectively delivered Dox to FR-positive tumor cells and triggered notable tumor cell death, as confirmed by in vitro and in vivo analyses. CONCLUSIONS: This simple and easy method for post-isolation modification of the exosomal surface may be used to develop milk-exosome-based drug delivery systems.

3.
Commun Biol ; 6(1): 970, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37740002

ABSTRACT

USP47 is widely involved in tumor development, metastasis, and other processes while performing a more regulatory role in inflammatory responses, myocardial infarction, and neuronal development. In this study, we investigate the functional and biochemical properties of USP47, whereby depleting USP47 inhibited cancer cell growth in a p53-dependent manner-a phenomenon that enhances during the simultaneous knockdown of USP7. Full-length USP47 shows higher deubiquitinase activity than the catalytic domain. The crystal structures of the catalytic domain, in its free and ubiquitin-bound states, reveal that the misaligned catalytic triads, ultimately, become aligned upon ubiquitin-binding, similar to USP7, thereby becoming ready for catalysis. Yet, the composition and lengths of BL1, BL2, and BL3 of USP47 differ from those for USP7, and they contribute to the observed selectivity. Our study provides molecular details of USP47 regulation, substrate recognition, and the hotspots for drug discovery by targeting USP47.


Subject(s)
Cell Transformation, Neoplastic , Ubiquitin , Humans , Ubiquitin-Specific Peptidase 7/genetics , Catalysis , Catalytic Domain
4.
Cell Mol Life Sci ; 80(4): 112, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37004621

ABSTRACT

Recently, a number of reports on the importance of USP35 in cancer have been published. However, very little is known about the exact mechanism by which USP35 activity is regulated. Here, we show the possible regulation of USP35 activity and the structural specificity affecting its function by analyzing various fragments of USP35. Interestingly, the catalytic domain of USP35 alone does not exhibit deubiquitinating activity; in contrast, the C-terminal domain and insertion region in the catalytic domain is required for full USP35 activity. Additionally, through its C-terminal domain, USP35 forms a homodimer that prevents USP35 degradation. CHIP bound to HSP90 interacts with and ubiquitinates USP35. However, when fully functional USP35 undergoes auto-deubiquitination, which attenuates CHIP-mediated ubiquitination. Finally, USP35 dimer is required for deubiquitination of the substrate Aurora B and regulation of faithful mitotic progression. The properties of USP35 identified in this study are a unique homodimer structure, regulation of deubiquitinating activity through this, and utilization of a novel E3 ligase involved in USP35 auto-deubiquitination, which adds another complexity to the regulation of deubiquitinating enzymes.


Subject(s)
Neoplasms , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Ubiquitination
5.
Bioeng Transl Med ; 8(1): e10362, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36684086

ABSTRACT

Adoptive cell therapy (ACT) with antigen-specific T cells is a promising treatment approach for solid cancers. Interleukin-2 (IL-2) has been utilized in boosting the efficacy of ACT. However, the clinical applications of IL-2 in combination with ACT is greatly limited by short exposure and high toxicities. Herein, a complex coacervate was designed to intratumorally deliver IL-2 in a sustained manner and protect against proteolysis. The complex coacervate consisted of fucoidan, a specific IL-2 binding glycosaminoglycan, and poly-l-lysine, a cationic counterpart (FPC2). IL-2-laden FPC2 exhibited a preferential bioactivity in ex vivo expansion of CD8+T cells over Treg cells. Additionally, FPC2 was embedded in pH modulating injectable gel (FPC2-IG) to endure the acidic tumor microenvironment. A single intratumoral administration of FPC2-IG-IL-2 increased expansion of tumor-infiltrating cytotoxic lymphocytes and reduced frequencies of myeloid populations. Notably, the activation and persistency of tumor-reactive T cells were observed only in the tumor site, not in the spleen, confirming a localized effect of FPC2-IG-IL-2. The immune-favorable tumor microenvironment induced by FPC2-IG-IL-2 enabled adoptively transferred TCR-engineered T cells to effectively eradicate tumors. FPC2-IG delivery system is a promising strategy for T-cell-based immunotherapies.

6.
J Mol Cell Biol ; 14(6)2022 11 17.
Article in English | MEDLINE | ID: mdl-35704671

ABSTRACT

Fas-associated factor 1 (FAF1) is a scaffolding protein that plays multiple functions, and dysregulation of FAF1 is associated with many types of diseases such as cancers. FAF1 contains multiple ubiquitin-related domains (UBA, UBL1, UBL2, UAS, and UBX), each domain interacting with a specific partner. In particular, the interaction of UBL1 with heat shock protein 70 (Hsp70) is associated with tumor formation, although the molecular understanding remains unknown. In this study, the structural analysis revealed that His160 of FAF1 is important for its interaction with Hsp70. The association of Hsp70 with FAF1 is required for the interaction with IQGAP1. FAF1 negatively regulates RhoA activation by FAF1-Hsp70 complex formation, which then interacts with IQGAP1. These steps play a key role in maintaining the stability of cell-to-cell junction. We conclude that FAF1 plays a critical role in the structure and function of adherens junction during tissue homeostasis and morphogenesis by suppressing RhoA activation, which induces the activation of Rho-associated protein kinase, phosphorylation of myosin light chain, formation of actin stress fiber, and disruption of adherens junction. In addition, depletion of FAF1 increased collective invasion in a 3D spheroid cell culture. These results provide insight into how the FAF1-Hsp70 complex acts as a novel regulator of the adherens junction integrity. The complex can be a potential therapeutic target to inhibit tumorigenesis and metastasis.


Subject(s)
HSP70 Heat-Shock Proteins , Neoplasms , Humans , HSP70 Heat-Shock Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Ubiquitin/metabolism , Neoplasms/metabolism , rhoA GTP-Binding Protein/metabolism
7.
Biochem Biophys Rep ; 25: 100871, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33367116

ABSTRACT

Human prostaglandin E2 receptor 4 (EP4) is one of the four subtypes of prostaglandin E2 (PGE2) receptors and belongs to the rhodopsin-type G protein-coupled receptor (GPCR) family. Particularly, EP4 is expressed in various cancer cells and is involved in cancer-cell proliferation by a G protein signaling cascade. To prepare an active form of EP4 for biochemical characterization and pharmaceutical application, this study designed a recombinant protein comprising human EP4 fused to the P9 protein (a major envelope protein of phi6 phage) and overexpressed the P9-EP4 fusion protein in the membrane fraction of E. coli. The solubilized P9-EP4 with sarkosyl (a strong anionic detergent) was purified by affinity chromatography. The purified protein was stabilized with amphiphilic polymers derived from poly-γ-glutamate. The polymer-stabilized P9-EP4 showed specific interaction with the alpha subunits of Gs or Gi proteins, and a high content of α-helical structure by a circular dichroism spectroscopy. Furthermore, the polymer-stabilized P9-EP4 showed strong heat resistance compared with P9-EP4 in detergents. The functional preparation of EP4 and its stabilization with amphiphilic polymers could facilitate both the biochemical characterization and pharmacological applications targeting EP4.

8.
Int J Mol Sci ; 21(24)2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33317068

ABSTRACT

Inhibition of the molecular chaperone heat shock protein 90 (Hsp90) represents a promising approach for cancer treatment. BIIB021 is a highly potent Hsp90 inhibitor with remarkable anticancer activity; however, its clinical application is limited by lack of potency and response. In this study, we aimed to investigate the impact of replacing the hydrophobic moiety of BIIB021, 4-methoxy-3,5-dimethylpyridine, with various five-membered ring structures on the binding to Hsp90. A focused array of N7/N9-substituted purines, featuring aromatic and non-aromatic rings, was designed, considering the size of hydrophobic pocket B in Hsp90 to obtain insights into their binding modes within the ATP binding site of Hsp90 in terms of π-π stacking interactions in pocket B as well as outer α-helix 4 configurations. The target molecules were synthesized and evaluated for their Hsp90α inhibitory activity in cell-free assays. Among the tested compounds, the isoxazole derivatives 6b and 6c, and the sole six-membered derivative 14 showed favorable Hsp90α inhibitory activity, with IC50 values of 1.76 µM, 0.203 µM, and 1.00 µM, respectively. Furthermore, compound 14 elicited promising anticancer activity against MCF-7, SK-BR-3, and HCT116 cell lines. The X-ray structures of compounds 4b, 6b, 6c, 8, and 14 bound to the N-terminal domain of Hsp90 were determined in order to understand the obtained results and to acquire additional structural insights, which might enable further optimization of BIIB021.


Subject(s)
Antineoplastic Agents/chemical synthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Binding Sites , HCT116 Cells , HSP90 Heat-Shock Proteins/chemistry , Humans , Isoxazoles/chemistry , MCF-7 Cells , Mice , Protein Binding , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship
9.
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
10.
Eur J Med Chem ; 208: 112756, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32942186

ABSTRACT

Prompted by the urgent demand for identification of new anticancer agents with improved potency and efficacy, a new series of arylamides incorporating the privileged 2-anilinoquinoline scaffold has been designed, synthesized, and biologically assessed. Aiming at extensive evaluation of the target compounds' potency and spectrum, a panel of 60 clinically important cancer cell lines representing nine cancer types has been used. Compounds 9a and 9c, with piperazine substituted phenyl ring, emerged as the most active members surpassing the anticancer potencies of the FDA-approved drug imatinib. They elicited sub-micromolar or one-digit micromolar GI50 values over the majority of tested cancer cells including multidrug resistant (MDR) cells like colon HCT-15, renal TK-10 and UO-31, and ovarian NCI/ADR-RES. In vitro mechanistic study showed that compounds 9a and 9c could trigger morphological changes, apoptosis and cell cycle arrest in HCT-116 colon cancer cells. Besides, compound 9c altered microtubule polymerization pattern in a similar fashion to paclitaxel. Kinase screening of 9c disclosed its inhibitory activity over B-RAFV600E and C-RAF kinases with IC50 values of 0.888 µM and 0.229 µM, respectively. Taken together, the current report presents compounds 9a and 9c as promising broad-spectrum potent anticancer candidates, which could be considered for further development of new anticancer drugs.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Quinolines/pharmacology , Acrylamides/chemical synthesis , Acrylamides/metabolism , Aniline Compounds/chemical synthesis , Aniline Compounds/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Drug Design , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Molecular Structure , Mutation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins B-raf/genetics , Quinolines/chemical synthesis , Quinolines/metabolism , Structure-Activity Relationship , Vero Cells
11.
Int J Mol Sci ; 21(11)2020 Jun 04.
Article in English | MEDLINE | ID: mdl-32512887

ABSTRACT

Ubiquitination and deubiquitination play a critical role in all aspects of cellular processes, and the enzymes involved are tightly regulated by multiple factors including posttranslational modifications like most other proteins. Dysfunction or misregulation of these enzymes could have dramatic physiological consequences, sometimes leading to diseases. Therefore, it is important to have a clear understanding of these regulatory processes. Here, we have reviewed the posttranslational modifications of deubiquitinating enzymes and their consequences on the catalytic activity, stability, abundance, localization, and interaction with the partner proteins.


Subject(s)
Deubiquitinating Enzymes/metabolism , Protein Processing, Post-Translational , Catalysis , Humans , Phosphorylation , Protein Transport , Ubiquitin/metabolism , Ubiquitination
12.
Cancers (Basel) ; 12(5)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370049

ABSTRACT

p53 is activated in response to cellular stresses such as DNA damage, oxidative stress, and especially ribosomal stress. Although the regulations of p53 by E3 ligase and deubiquitinating enzymes (DUBs) have been described, the cellular roles of DUB associated with ribosomal stress have not been well studied. In this study, we report that Ubiquitin Specific Protease 47 (USP47) functions as an important regulator of p53. We show that ubiquitinated ribosomal protein S2 (RPS2) by Mouse double minute 2 homolog (MDM2) is deubiquitinated by USP47. USP47 inhibits the interaction between RPS2 and MDM2 thereby alleviating RPS2-mediated suppression of MDM2 under normal conditions. However, dissociation of USP47 leads to RPS2 binding to MDM2, which is required for the suppression of MDM2, consequently inducing up-regulation of the p53 level under ribosomal stress. Finally, we show that depletion of USP47 induces p53 and therefore inhibits cell proliferation, colony formation, and tumor progression in cancer cell lines and a mouse xenograft model. These findings suggest that USP47 could be a potential therapeutic target for cancer.

13.
Biochem Biophys Res Commun ; 523(2): 542-547, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31928715

ABSTRACT

The MDM2-p53 pathway is crucial for maintenance of p53 homeostasis. Some ribosomal proteins (RPs) play critical roles in regulating p53 by interacting with MDM2. However, the role and functional mechanism of each RP in MDM2-p53 pathway still remain unknown. In this study, we found that Ribosomal Protein S2 (RPS2) is a new regulator of MDM2-P53 signaling pathway to regulate p53 protein level. Here, we characterized that RPS2 interacts with MDM2 through the RING finger domain of MDM2. RPS2 is ubiquitinated by MDM2 and the ubiquitinated status of RPS2 regulates the stability of p53, which is activated in response to cellular stresses such as DNA damage, oxidative stress, and especially ribosomal stress. In addition, p53 is not induced in RPS2 knockdown even in the ribosomal stressed condition, indicating that RPS2 is essential for the stabilization of p53. Collectively, our data suggest that RPS2 plays a critical role in the regulation of p53 signaling including the ribosomal stress response.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , HeLa Cells , Humans , Protein Domains , Protein Stability , Proto-Oncogene Proteins c-mdm2/genetics , Ribosomal Proteins/genetics , Signal Transduction , Stress, Physiological , Tumor Suppressor Protein p53/genetics , Ubiquitination
15.
ACS Appl Mater Interfaces ; 11(40): 36905-36916, 2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31523951

ABSTRACT

Herein, we report a detailed study on the optoelectronic properties, photovoltaic performance, structural conformation, morphology variation, charge carrier mobility, and recombination dynamics in bulk heterojunction solar cells comprising a series of donor-acceptor conjugated polymers as electron donors based on benzodithiophene (BDT) and 5,8-bis(5-bromothiophen-2-yl)-6,7-difluoro-2,3-bis(3-(octyloxy)phenyl)quinoxaline as a function of the BDT's thienyl substitution (alkyl (WF3), alkylthio (WF3S), and fluoro (WF3F)). The synergistic positive effects of the fluorine substituents on the minimization of the bimolecular recombination losses, the reduction of the series resistances (RS), the increment of the shunt resistances (RSh), the suppression of the trap-assisted recombination losses, the balanced charge transport, the finer nanoscale morphology, and the deeper highest occupied molecular orbital (EHOMO) are manifested versus the alkyl and alkylthio substituents. According to these findings, the WF3F:[6,6]-phenyl-C71-butyric acid methyl ester (PC71BM)-based organic photovoltaic device is a rare example that features a high power conversion efficiency (PCE) of 17.34% under 500 lx indoor light-emitting diode light source with a high open-circuit voltage (VOC) of 0.69 V, due to the suppression of the voltage losses, and a PCE of 9.44% at 1 sun (100 mW/cm2) conditions, simultaneously.

16.
Biomaterials ; 217: 119298, 2019 10.
Article in English | MEDLINE | ID: mdl-31280073

ABSTRACT

Genetically engineered cells via CRISPR/Cas9 system can serve as powerful sources for cancer immunotherapeutic applications. Furthermore, multiple genetic alterations are necessary to overcome tumor-induced immune-suppressive mechanisms. However, one of the major obstacles is the technical difficulty with efficient multiple gene manipulation of suspension cells due to the low transfection efficacy. Herein, we established a carrier-free multiplexed gene editing platform in a simplified method, which can enhance the function of cytotoxic CD8+ T cells by modulating suspension cancer cells. Our multiple Cas9 ribonucleoproteins (RNPs) enable simultaneous disruption of two programmed cell death 1 (PD-1) ligands, functioning as negative regulators in the immune system, by accessing engineered Cas9 proteins with abilities of complexation and cellular penetration. In addition, combination with electroporation enhanced multiple gene editing efficacy, compared with that by treatment of multiple Cas9 RNPs alone. This procedure resulted in high gene editing at multiple loci of suspension cells. The treatment of multiple Cas9 RNPs targeting both ligands strongly improved Th1-type cytokine production of cytotoxic CD8+ T cells, resulting in synergistic cytotoxic effects against cancer. Simultaneous suppression of PD-L1 and PD-L2 on cancer cells via our developed editing system allows effective anti-tumor immunity. Furthermore, the treatment of multiple Cas9 RNPs targeting PD-L1, PD-L2, and TIM-3 had approximately 70-90% deletion efficacy. Thus, our multiplexed gene editing strategy endows potential clinical utilities in cancer immunotherapy.


Subject(s)
Gene Editing , Suspensions/chemistry , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Proliferation , Clathrin/metabolism , Cytotoxicity, Immunologic , Endocytosis , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunity , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Ribonucleoproteins/metabolism
17.
Chemistry ; 25(24): 6154-6161, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-30801818

ABSTRACT

Monomeric perylene diimide (PDI) small molecules display a high absorption coefficient and crystallinity in solid-state thin films due to strong π-π interactions between the molecules. To take advantage of these exciting properties of PDIs, N,N'-bis(1-ethylpropyl)perylene-3,4,9,10-tetracarboxylic diimide (EP-PDI) was mixed with a binary blend of PTB7 and PC71 BM to fabricate an efficient ternary blend, which were in turn used to produce organic photovoltaic (OPV) devices well suited to indoor applications (PTB7=poly({4,8-bis[(2-ethylhexyl)oxy]benzo[1,2-b:4,5-b']dithiophene-2,6-diyl}{3-fluoro-2-[(2-ethylhexyl)carbonyl]thieno[3,4-b]thiophenediyl}), PC71 BM=[6,6]-phenyl-C71 -butyric acid methyl ester). We varied the PC71 BM/EP-PDI weight ratio to investigate the influence of EP-PDI on the optical, electrical, and morphological properties of the PTB7:PC71 BM:EP-PDI ternary blend. Compared with the reference PTB7:PC71 BM binary blend, the ternary blends showed strong optical absorption in the wavelength range in which the spectra of indoor LED lamps show their strongest peaks. The addition of EP-PDI to the binary blend was found to play an important role in altering the morphology of the blend in such a way as to facilitate charge transport in the resulting ternary blend. Apparently, as a result, the optimal PTB7:PC71 BM:EP-PDI-based inverted OPV device exhibited a power conversion efficiency (PCE) of 15.68 %, a fill factor (FF) of 68.5 %, and short-circuit current density (JSC ) of 56.7 µA cm-2 under 500 lx (ca. 0.17 mW cm-2 ) indoor LED light conditions.

18.
Cell Rep ; 26(5): 1357-1367.e5, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30699360

ABSTRACT

Katanin was the first microtubule (MT)-severing enzyme discovered, but how katanin executes MT severing remains poorly understood. Here, we report X-ray crystal structures of the apo and ATPγS-bound states of the catalytic AAA domain of human katanin p60 at 3.0 and 2.9 Å resolution, respectively. Comparison of the two structures reveals conformational changes induced by ATP binding and how such changes ensure hexamer stability. Moreover, we uncover structural details of pore loops (PLs) and show that Arg283, a residue unique to katanin among MT-severing enzymes, protrudes from PL1 and lines the entry of the catalytic pore. Functional studies suggest that PL1 and Arg283 play essential roles in the recognition and remodeling of the glutamylated, C-terminal tubulin tail and regulation of axon growth. In addition, domain-swapping experiments in katanin and spastin suggest that the non-homologous N-terminal region, which contains the MT-interacting and trafficking domain and a linker, confers specificity to the severing process.


Subject(s)
Glutamates/metabolism , Katanin/chemistry , Katanin/metabolism , Microtubules/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , Axons/metabolism , HeLa Cells , Humans , Mice, Inbred ICR , Models, Molecular , Mutation/genetics , Protein Domains , Protein Multimerization , Sensory Receptor Cells/metabolism , Spastin/metabolism
19.
Sci Rep ; 8(1): 10909, 2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30026594

ABSTRACT

Nm23-H1/NDPK-A is a tumor metastasis suppressor having NDP kinase (NDPK) activity. Nm23-H1 is positively associated with prolonged disease-free survival and good prognosis of cancer patients. Approaches to increasing the cellular levels of Nm23-H1 therefore have significance in the therapy of metastatic cancers. We found a small molecule, (±)-trans-3-(3,4-dimethoxyphenyl)-4-[(E)-3,4-dimethoxystyryl]cyclohex-1-ene, that activates Nm23, hereafter called NMac1. NMac1 directly binds to Nm23-H1 and increases its NDPK activity. Employing various NMac1 derivatives and hydrogen/deuterium mass spectrometry (HDX-MS), we identified the pharmacophore and mode of action of NMac1. We found that NMac1 binds to the C-terminal of Nm23-H1 and induces the NDPK activation through its allosteric conformational changes. NMac1-treated MDA-MB-231 breast cancer cells showed dramatic changes in morphology and actin-cytoskeletal organization following inhibition of Rac1 activation. NMac1 also suppressed invasion and migration in vitro, and metastasis in vivo, in a breast cancer mouse model. NMac1 as an activator of NDPK has potential as an anti-metastatic agent.


Subject(s)
Breast Neoplasms/drug therapy , NM23 Nucleoside Diphosphate Kinases/metabolism , Neoplasm Metastasis/drug therapy , Small Molecule Libraries/administration & dosage , Up-Regulation , Allosteric Regulation/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Treatment Outcome , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/metabolism
20.
ACS Nano ; 12(8): 7750-7760, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30028587

ABSTRACT

Cas9 ribonucleoprotein (RNP)-mediated delivery has emerged as an ideal approach for in vivo applications. However, the delivery of Cas9 RNPs requires electroporation or lipid- or cationic-reagent-mediated transfection. Here, we developed a carrier-free Cas9 RNP delivery system for robust gene editing in vivo. For simultaneous delivery of Cas9 and a guide RNA into target cells without the aid of any transfection reagents, we established a multifunctional Cas9 fusion protein (Cas9-LMWP) that forms a ternary complex with synthetic crRNA:tracrRNA hybrids in a simple procedure. Cas9-LMWP carrying both a nuclear localization sequence and a low-molecular-weight protamine (LMWP) enables the direct self-assembly of a Cas9:crRNA:tracrRNA ternary complex (a ternary Cas9 RNP) and allows for the delivery of the ternary Cas9 RNPs into the recipient cells, owing to its intrinsic cellular and nuclear translocation ability with low immunogenicity. To demonstrate the potential of this system, we showed extensive synergistic anti-KRAS therapy (CI value: 0.34) via in vitro and in vivo editing of the KRAS gene by the direct delivery of multifunctional Cas9 RNPs in lung cancer. Thus, our carrier-free Cas9 RNP delivery system could be an innovative platform that might serve as an alternative to conventional transfection reagents for simple gene editing and high-throughput genetic screening.


Subject(s)
CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/metabolism , Gene Editing , Genetic Therapy , Lung Neoplasms/therapy , RNA/genetics , A549 Cells , Animals , Apoptosis , CRISPR-Associated Protein 9/genetics , Cell Movement , Cell Survival , Drug Delivery Systems , Female , Gene Transfer Techniques , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...