Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
ACS Nano ; 17(17): 17554-17567, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37643221

ABSTRACT

Localized expression of immunomodulatory molecules can stimulate immune responses against tumors in the tumor microenvironment while avoiding toxicities associated with systemic administration. In this study, we developed a polyethylenimine-modified porous silica nanoparticle (PPSN)-based delivery platform carrying cytokine mRNA for local immunotherapy in vivo. Our delivery platform was significantly more efficient than FDA-approved lipid nanoparticles for localized mRNA translation. We observed no off-target translation of mRNA in any organs and no evidence of systemic toxicity. Intratumoral injection of cytokine mRNA-loaded PPSNs led to high-level expression of protein within the tumor and stimulated immunogenic cancer cell death. Additionally, combining cytokine mRNA with an immune checkpoint inhibitor enhanced anticancer responses in several murine cancer models and enabled the inhibition of distant metastatic tumors. Our results demonstrate the potential of PPSNs-mediated mRNA delivery as a specific, effective, and safe platform for mRNA-based therapeutics in cancer immunotherapy.


Subject(s)
Interleukin-2 , Nanoparticles , Animals , Mice , Interleukin-2/genetics , Porosity , Cytokines , RNA, Messenger/genetics , Silicon Dioxide
2.
Article in English | MEDLINE | ID: mdl-36913611

ABSTRACT

Cancer vaccine is one of the immunotherapeutic strategies aiming to effectively deliver cancer antigens to professional antigen-presenting cells such as dendritic cells (DCs), macrophages, and B cells to elicit a cancer-specific immune response. Despite the advantages of the cancer vaccine that can be applied to various cancer types, the clinical approach is limited due to the non-specific or adverse immune responses, stability, and safety issues. In this study, we report an injectable nanovaccine platform based on large-sized (∼350 nm) porous silica nanoparticles (PSNs). We found that large-sized PSNs, called PS3, facilitated the formation of an antigen supply depot at the site of injection so that a single injection of PSN-based nanovaccine elicited sufficient tumor-specific cell-mediated and humoral immune response. As a result, antigen-loaded PS3 induced successful tumor regression in prophylactic and therapeutic vaccination.

3.
Nat Commun ; 13(1): 4568, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35931667

ABSTRACT

Artificial, synthetic chaperones have attracted much attention in biomedical research due to their ability to control the folding of proteins and peptides. Here, we report bio-inspired multifunctional porous nanoparticles to modulate proper folding and intracellular delivery of therapeutic α-helical peptide. The Synthetic Nano-Chaperone for Peptide (SNCP) based on porous nanoparticles provides an internal hydrophobic environment which contributes in stabilizing secondary structure of encapsulated α-helical peptides due to the hydrophobic internal environments. In addition, SNCP with optimized inner surface modification not only improves thermal stability for α-helical peptide but also supports the peptide stapling methods in situ, serving as a nanoreactor. Then, SNCP subsequently delivers the stabilized therapeutic α-helical peptides into cancer cells, resulting in high therapeutic efficacy. SNCP improves cellular uptake and bioavailability of the anti-cancer peptide, so the cancer growth is effectively inhibited in vivo. These data indicate that the bio-inspired SNCP system combining nanoreactor and delivery carrier could provide a strategy to expedite the development of peptide therapeutics by overcoming existing drawbacks of α-helical peptides as drug candidates.


Subject(s)
Molecular Chaperones , Peptides , Amino Acid Sequence , Molecular Chaperones/metabolism , Peptides/chemistry , Protein Conformation, alpha-Helical , Protein Folding , Protein Structure, Secondary
4.
Biomaterials ; 280: 121257, 2022 01.
Article in English | MEDLINE | ID: mdl-34839122

ABSTRACT

Recent strategies in cancer immunotherapy based on interleukin-2 (IL-2) are generally focused on reducing regulatory T cell (Treg) development by modifying IL-2 receptor alpha (IL-2Rα) domain. However, the clinical utility of high-dose IL-2 treatment is mainly limited by severe systemic toxicity. We find that peritumorally injectable 'BALLkine-2', recombinant human IL-2 (rIL-2) loaded porous nanoparticle, dramatically reduces systemic side effects of rIL-2 by minimizing systemic IL-2 exposure. Notably, in cynomolgus monkeys, subcutaneous (SC)-injection of BALLkine-2 not only dramatically reduces systemic circulation of rIL-2 in the blood, but also increases half-life of IL-2 compared to IV- or SC-injection of free rIL-2. Peritumorally-injected BALLkine-2 enhances intratumoral lymphocyte infiltration without inducing Treg development and more effectively synergizes with PD-1 blockade than high-dose rIL-2 administration in B16F10 melanoma model. BALLkine-2 could be a highly potent therapeutic option due to higher anti-tumor efficacy with lower and fewer doses and reduced systemic toxicity compared to systemic rIL-2.


Subject(s)
Melanoma , Nanoparticles , Humans , Immunotherapy , Interleukin-2/therapeutic use , Melanoma/drug therapy , Recombinant Proteins/therapeutic use , T-Lymphocytes, Regulatory
5.
Nanomedicine ; 32: 102316, 2021 02.
Article in English | MEDLINE | ID: mdl-33068744

ABSTRACT

Among various strategies to treat neurodegenerative disorders, cell replacement therapies have drawn much attention recently. Such a trend led to the increase in demand for the rare and specialized cells, followed by the outburst development of various cell reprogramming strategies. However, several limitations on these conventional methods remain to be solved, including the genetic instability of the viral vectors and the high cytotoxicity or poor performance of the non-viral carriers. Therefore, non-viral methods need to be developed to ensure safe and efficient cell reprogramming. Here, we introduce a polymer-modified nano-reagent (Polymer-functionalized Nanodot, PolyN) for the safe and efficient, non-viral direct cell reprogramming. PolyN facilitated the highly efficient contemporary overexpression of the transgene compared to the conventional reagent. With our nano-reagent, we demonstrated the SOX2-mediated cell reprogramming and successfully generated the neuron-like cell from the human fibroblast.


Subject(s)
Cellular Reprogramming , Fibroblasts/cytology , Nanoparticles/chemistry , Neurons/cytology , Polymers/chemistry , DNA/genetics , Gene Transfer Techniques , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Neural Stem Cells/cytology , Plasmids/genetics , Transfection
6.
Sci Adv ; 6(22): eaaz8201, 2020 05.
Article in English | MEDLINE | ID: mdl-32523995

ABSTRACT

Direct-acting agents against viral components are considered as the most promising candidates for the successful antiviral therapeutics. To date, no direct-acting drugs exist for the treatment against dengue virus (DV) infection, which can develop into life-threatening diseases. RNA-dependent RNA polymerase (RdRp), an RNA virus-specific enzyme highly conserved among various viral families, has been known as the broad-range antiviral drug target. Here, we developed an RNA-based graphene biosensor system [RNA nano-graphene oxide system (RANGO)] to enable the fluorescence-based quantitative analysis of the RdRp enzyme activity. We used the RANGO system to a high-throughput chemical screening to identify novel direct-acting antiviral drug candidates targeting DV RdRp from the FDA-approved small-molecule library. RANGO accelerated the massive selection of drug candidates. We found that one of the selected hit compounds, montelukast, showed antiviral activity in vitro and in vivo by directly inhibiting replication of DV and thus relieved related symptoms.

7.
Nanoscale ; 12(11): 6385-6393, 2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32134425

ABSTRACT

Skin fibrosis occurs in a variety of human diseases but the current anti-fibrosis treatments are not sufficient. One major cause of fibrotic diseases shared across diverse organ fibrosis is uncontrolled overexpression of the connective tissue growth factor (CTGF, also known as CCN2). Here, we examine the anti-fibrotic activity of RNAi therapy utilizing siRNA against CTGF with a new drug delivery system (DDS), 'DegradaBALL', which is based on porous nanoparticles, for durable CTGF gene silencing. DegradaBALL is a modular DDS having many favorable properties for RNA delivery such as effective intracellular uptake, convenient drug loading, biocompatibility, sustained release profile and biodegradability. DegradaBALL loaded with siCTGF, named 'LEM-S401', showed highly durable and effective CTGF gene-silencing in TGF-ß induced lung fibrosis and skin fibrosis model cells, A549 and HaCaT, respectively. In addition, LEM-S401 induced knockdown of collagen types I and III, which are excess extracellular matrix components in fibrotic skin in addition to CTGF in the mouse wound healing model. Most importantly, we showed that LEM-S401 effectively inhibited the formation of hypertrophic scars in wound-associated dermal fibrosis mouse models, during both the epidermis recovery and tissue remodeling process. Our findings suggest that LEM-S401 could be a highly potent therapeutic option for skin fibrotic diseases.


Subject(s)
Connective Tissue Growth Factor , RNA Interference , RNA, Small Interfering , Skin Diseases , Skin/metabolism , A549 Cells , Animals , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type III/biosynthesis , Collagen Type III/genetics , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Fibrosis , Humans , Male , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Skin/pathology , Skin Diseases/genetics , Skin Diseases/metabolism , Skin Diseases/pathology , Skin Diseases/therapy
8.
Sci Rep ; 8(1): 11413, 2018 07 30.
Article in English | MEDLINE | ID: mdl-30061704

ABSTRACT

MicroRNAs (miRNAs) are important regulatory RNAs that control gene expression in various biological processes. Therefore, control over the disease-related miRNA expression is important both for basic research and for a new class of therapeutic modality to treat serious diseases such as cancer. Here, we present a high-throughput screening strategy to identify small molecules that modulate miRNA expression in living cells. The screen enables simultaneous monitoring of the phenotypic cellular changes associated with the miRNA expression by measuring quantitative fluorescent signals corresponding to target miRNA level in living cells based on a novel biosensor composed of peptide nucleic acid and nano-sized graphene oxide. In this study, the biosensor based cellular screening of 967 compounds (including FDA-approved drugs, enzyme inhibitors, agonists, and antagonists) in cells identified four different classes of small molecules consisting of (i) 70 compounds that suppress both miRNA-21 (miR-21) expression and cell proliferation, (ii) 65 compounds that enhance miR-21 expression and reduce cell proliferation, (iii) 2 compounds that suppress miR-21 expression and increase cell proliferation, and (iv) 21 compounds that enhance both miR-21 expression and cell proliferation. We further investigated the hit compounds to correlate cell morphology changes and cell migration ability with decreased expression of miR-21.


Subject(s)
Biosensing Techniques/methods , Graphite/chemistry , High-Throughput Screening Assays/methods , MicroRNAs/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Reproducibility of Results , Small Molecule Libraries/analysis
9.
ACS Appl Mater Interfaces ; 9(40): 34634-34640, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-28921950

ABSTRACT

An improved cell conversion strategy for neural differentiation of mouse embryonic stem (mES) cells is developed by incorporating functionalized mesoporous silica nanoparticle (MSN) as an efficient delivery carrier of retinoic acid (RA), which is a pleiotropic factor required for initiation of neural differentiation. Traditional RA-mediated neural differentiation methods required either preactivation of the cells to the differentiating state by embryoid body (EB) formation or repetitive treatment of the differentiation factor. Our modified cell conversion system involves only singular treatment of the RA/MSN complex, which simplified the whole process and accelerated neural induction to be finished within 6 days with high quality. With our new method, neural cells were successfully derived from mES cells with stable expression of neurite marker gene.


Subject(s)
Mouse Embryonic Stem Cells , Animals , Cell Differentiation , Embryoid Bodies , Mice , Nanoparticles , Tretinoin
10.
Hepatology ; 62(4): 1160-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26154152

ABSTRACT

UNLABELLED: Enhanced expression of the cancer stem cell (CSC) marker, CD133, is closely associated with a higher rate of tumor formation and poor prognosis in hepatocellular carcinoma (HCC) patients. Despite its clinical significance, the molecular mechanism underlying the deregulation of CD133 during tumor progression remains to be clarified. Here, we report on a novel mechanism by which interleukin-6/signal transducer and activator of transcription 3 (IL-6/STAT3) signaling up-regulates expression of CD133 and promotes HCC progression. STAT3 activated by IL-6 rapidly bound to CD133 promoter and increased protein levels of CD133 in HCC cells. Reversely, in hypoxic conditions, RNA interference silencing of STAT3 resulted in decrease of CD133 levels, even in the presence of IL-6, with a concomitant decrease of hypoxia-inducible factor 1 alpha (HIF-1α) expression. Active STAT3 interacted with nuclear factor kappa B (NF-κB) p65 subunit to positively regulate the transcription of HIF-1α providing a mechanistic explanation on how those three oncogenes work together to increase the activity of CD133 in a hypoxic liver microenvironment. Activation of STAT3 and its consequent induction of HIF-1α and CD133 expression were not observed in Toll-like receptor 4/IL-6 double-knockout mice. Long-term silencing of CD133 by a lentiviral-based approach inhibited cancer cell-cycle progression and suppressed in vivo tumorigenicity by down-regulating expression of cytokinesis-related genes, such as TACC1, ACF7, and CKAP5. We also found that sorafenib and STAT3 inhibitor nifuroxazide inhibit HCC xenograft formation by blocking activation of STAT3 and expression of CD133 and HIF-1α proteins. CONCLUSION: IL-6/STAT3 signaling induces expression of CD133 through functional cooperation with NF-κB and HIF-1α during liver carcinogenesis. Targeting STAT3-mediated CD133 up-regulation may represent a novel, effective treatment by eradicating the liver tumor microenvironment.


Subject(s)
Antigens, CD/physiology , Carcinoma, Hepatocellular/etiology , Glycoproteins/physiology , Interleukin-6/physiology , Liver Neoplasms/etiology , Peptides/physiology , STAT3 Transcription Factor/physiology , Up-Regulation , AC133 Antigen , Animals , Cell Hypoxia , Humans , Mice , Mice, Inbred C57BL
11.
Nat Commun ; 5: 5633, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25476420

ABSTRACT

The 26S proteasome is the primary machinery that degrades ubiquitin (Ub)-conjugated proteins, including many proteotoxic proteins implicated in neurodegeneraton. It has been suggested that the elevation of proteasomal activity is tolerable to cells and may be beneficial to prevent the accumulation of protein aggregates. Here we show that purified proteasomes can be directly transported into cells through mesoporous silica nanoparticle-mediated endocytosis. Proteasomes that are loaded onto nanoparticles through non-covalent interactions between polyhistidine tags and nickel ions fully retain their proteolytic activity. Cells treated with exogenous proteasomes are more efficient in degrading overexpressed human tau than endogenous proteasomal substrates, resulting in decreased levels of tau aggregates. Moreover, exogenous proteasome delivery significantly promotes cell survival against proteotoxic stress caused by tau and reactive oxygen species. These data demonstrate that increasing cellular proteasome activity through the direct delivery of purified proteasomes may be an effective strategy for reducing cellular levels of proteotoxic proteins.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , tau Proteins/metabolism , Cell Line , Cell Survival , Humans , Oxidative Stress , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Aggregates , tau Proteins/genetics
12.
Biochim Biophys Acta ; 1843(9): 2037-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24861866

ABSTRACT

Although an in vitro 3D environment cannot completely mimic the in vivo tumor site, embedding tumor cells in a 3D extracellular matrix (ECM) allows for the study of cancer cell behaviors and the screening of anti-metastatic reagents with a more in vivo-like context. Here we explored the behaviors of MDA-MB-231 breast cancer cells embedded in 3D collagen I. Diverse tumor environmental conditions (including cell density, extracellular acidity, or hypoxia as mimics for a continuous tumor growth) reduced JNKs, enhanced TGFß1/Smad signaling activity, induced Snail1, and reduced cortactin expression. The reduced JNKs activity blocked efficient formation of invadopodia labeled with actin, cortactin, or MT1-MMP. JNKs inactivation activated Smad2 and Smad4, which were required for Snail1 expression. Snail1 then repressed cortactin expression, causing reduced invadopodia formation and prominent localization of MT1-MMP at perinuclear regions. MDA-MB-231 cells thus exhibited less efficient collagen I degradation and invasion in 3D collagen I upon JNKs inhibition. These observations support a signaling network among JNKs, Smads, Snail1, and cortactin to regulate the invasion of MDA-MB-231 cells embedded in 3D collagen I, which may be targeted during screening of anti-invasion reagents.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Collagen Type I/pharmacology , Cortactin/metabolism , Pseudopodia/metabolism , Transcription Factors/metabolism , Tumor Microenvironment/drug effects , Actins/metabolism , Animals , Breast Neoplasms/enzymology , Cattle , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Cell Nucleus/metabolism , Cortactin/genetics , Female , Gels , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 14/metabolism , Neoplasm Invasiveness , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins c-jun/metabolism , Pseudopodia/drug effects , Signal Transduction , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription, Genetic , Transforming Growth Factor beta1/metabolism
13.
Biochem Pharmacol ; 86(7): 950-9, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23962443

ABSTRACT

Aberrantly activated signal transducer and activator of transcription (STAT) proteins are implicated with human cancers and represent essential roles for cancer cell survival and proliferation. Therefore, the development of small-molecule inhibitors of STAT signaling bearing pharmacological activity has therapeutic potential for the treatment of human cancers. In this study, we identified sophoraflavanone G as a novel small-molecule inhibitor of STAT signaling in human cancer cells. Sophoraflavanone G inhibited tyrosine phosphorylation of STAT proteins in Hodgkin's lymphoma and tyrosine phosphorylation of STAT3 in solid cancer cells by inhibiting phosphorylation of the Janus kinase (JAK) proteins, Src family tyrosine kinases, such as Lyn and Src, Akt, and ERK1/2. In addition, sophoraflavanone G inhibited STAT5 phosphorylation in murine-bone-marrow-derived pro-B cells transfected with translocated Ets Leukemia (TEL)-JAKs and cytokine-induced rat pre-T lymphoma cells, as well as STAT5b reporter activity in TEL-JAKs and STAT5b reporter systems. Sophoraflavanone G also inhibited nuclear factor-κB (NF-κB) signaling in multiple myeloma cells. Furthermore, sophoraflavanone G inhibited cancer cell proliferation and induced apoptosis by regulating the expression of apoptotic and anti-apoptotic proteins. Our data suggest that sophoraflavanone G is a novel small-molecule inhibitor of STAT signaling by targeting upstream signals of STATs that may have therapeutic potential for cancers caused by persistently activated STAT proteins.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Flavanones/pharmacology , STAT Transcription Factors/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/metabolism , Drug Screening Assays, Antitumor/methods , Humans , NF-kappa B/metabolism , Phosphorylation/drug effects , STAT Transcription Factors/genetics , Signal Transduction/drug effects , Tyrosine/metabolism
14.
Immunology ; 140(3): 288-300, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23746113

ABSTRACT

The naive T-cell pool in peripheral lymphoid tissues is fairly stable in terms of number, diversity and functional capabilities in spite of the absence of prominent stimuli. This stability is attributed to continuous tuning of the composition of the T-cell pool by various homeostatic signals. Despite extensive research into the link between signal transducer and activator of transcription 3 (Stat3) and T-cell survival, little is known about how Stat3 regulates homeostasis by maintaining the required naive T-cell population in peripheral lymphoid organs. We assessed whether the elimination of Stat3 in T cells limits T-cell survival. We demonstrated that the proportion and number of single-positive thymocytes as well as T cells in the spleen and lymph nodes were significantly decreased in the Stat3-deficient group as a result of the enhanced susceptibility of Stat3-deleted T lymphocytes to apoptosis. Importantly, expression of the anti-apoptotic Bcl-2 and Bcl-xL was markedly decreased in Stat3-deleted single-positive thymocytes and T lymphocytes, suggesting that Stat3 helps to maintain the T-cell pool in the resting condition by promoting the expression of Bcl-2 family genes. These findings suggest the importance of Stat3 in the integration of homeostatic cues for the maintenance and functional tuning of the T-cell pool.


Subject(s)
Gene Expression Regulation , Precursor Cells, T-Lymphoid/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Survival/genetics , Cells, Cultured , Homeostasis , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism
15.
Mol Cells ; 35(5): 410-20, 2013 May.
Article in English | MEDLINE | ID: mdl-23649461

ABSTRACT

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) produced by the oxidative burst in activated macrophages and neutrophils cause oxidative stressimplicated diseases. Quercetin is flavonoid that occurs naturally in plants and is widely used as a nutritional supplement due to its antioxidant and anti-inflammatory properties. In this study, we investigated antioxidant activities and mechanisms of action in zymosan-induced macrophages of quercetin and quercetin-related flavonoids such as quercitrin, isoquercitrin, quercetin 3-O-ß-(2″-galloyl)-rhamnopyranoside (QGR) and quercetin 3-O-ß-(2″-galloyl)-glucopyranoside (QGG) as well as gallic acid, a building moiety of QGR and QGG. QGR and QGG exhibited stronger antioxidant activities compared with quercetin, whereas quercitrin, isoquercitrin and gallic acid exhibited weak-tono antioxidant activities, assessed by 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging, superoxide production, superoxide scavenging, nitric oxide (NO) production, peroxynitrite (ONOO(-)) scavenging and myeloperoxidase (MPO) activity. Regarding mechanisms, the quercetincontaining flavonoids QGR and QGG differentially targeted compared with quercetin in the NF-κB signaling pathway that inhibited the DNA binding activity of the NF-κB complex without affecting the degradation and phosphorylation of IκBα and NF-κB phosphorylation. In addition, QGR and QGG inhibited CRE and activator protein (AP-1) transcriptional activity and JNK phosphorylation by inhibiting the cAMP/protein kinase A (PKA) and protein kinase C (PKC) signaling in a different manner than quercetin. Our results showed that although QGR and QGG exhibited stronger antioxidant activities than querce-tin in macrophages, their mechanisms of action in terms of the NF-κB, PKA and PKC signaling pathways were different.


Subject(s)
Antioxidants/metabolism , Integrases/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Quercetin/analogs & derivatives , Signal Transduction , Transcription Factor AP-1/metabolism , Animals , Biphenyl Compounds/metabolism , Mice , NF-kappa B/antagonists & inhibitors , Peroxynitrous Acid/metabolism , Picrates/metabolism , Quercetin/pharmacology , Transcription Factor AP-1/antagonists & inhibitors
16.
Biochem Biophys Res Commun ; 435(4): 685-90, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23702480

ABSTRACT

Bone marrow stromal cell antigen 2 (BST-2) is a type II transmembrane protein that is known to be a therapeutic target in several types of cancer. However, despite its clinical importance, the roles of BST-2 expression have remained elusive. Here, we found that BST-2 expression is up-regulated in tamoxifen-resistant MCF-7 human breast cancer (TRM-7) cells, resulting in enhanced invasiveness and migration. Matrigel and wound healing assays also showed that overexpression of BST-2 increased invasion and migration in MCF-7 cells, whereas invasion and migration were decreased by the silencing of BST-2 in TRM-7 cells. In addition, B16F10 cells expressing BST-2 showed increased metastatic melanoma nodule growth in a lung metastasis mouse model. Furthermore, BST-2 expression and promoter activity were regulated by activated signal transducer and activator of transcription 3 (STAT3). Taken together, our results indicate that BST-2 is an important factor in the invasiveness and motility of tamoxifen-resistant breast cancer cells, and that its expression and activity are regulated by activated STAT3. Therefore, regulation of BST-2 is a potential therapeutic target for tamoxifen-resistant breast cancer.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Melanoma/pathology , Melanoma/secondary , Membrane Glycoproteins/metabolism , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Drug Interactions , Drug Resistance, Neoplasm/drug effects , Female , Humans , Lung Neoplasms/secondary , Melanoma/drug therapy , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Treatment Outcome
17.
Eur Respir J ; 41(5): 1147-56, 2013 May.
Article in English | MEDLINE | ID: mdl-22835616

ABSTRACT

Recently, statins have been shown to have anti-inflammatory effects on lung inflammatory diseases. However, the mechanisms of action of simvastatin in viral pneumonia have yet to be elucidated, although viral infection remains a considerable health threat. In this study, we hypothesised that simvastatin inhibits polyinosinic-polycytidylic acid (poly I:C)-induced airway inflammation, such as RANTES (regulated on activation, normal T-cell expressed and secreted) expression and inflammatory cell recruitment. In bronchial cells, the effect of simvastatin on poly I:C-induced RANTES expression and signal transducer and activator of transcription (STAT)3-mediated signal transduction was determined using an ELISA and short hairpin (sh)RNA system. In a poly I:C-induced pneumonia mouse model, immunological changes in the lungs after simvastatin inhalation, such as inflammatory cell recruitment and cytokine/chemokine release, were examined. In poly I:C-stimulated bronchial cells, RANTES secretion was increased by STAT3 activation, and simvastatin suppressed poly I:C-induced STAT3 activation, resulting in inhibition of RANTES expression. In BALB/c mice stimulated with inhaled poly I:C, RANTES expression and neutrophil infiltration into the airway were elevated. However, simvastatin treatment attenuated STAT3 activation, RANTES release and subsequent neutrophilia in the lungs. These findings suggest that simvastatin inhibits airway inflammation, but there are other mechanisms that need to be fully elucidated.


Subject(s)
Chemokine CCL5/metabolism , Neutrophils/drug effects , Pneumonia/chemically induced , Poly I-C/adverse effects , Simvastatin/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Bronchi/cytology , Cell Line, Tumor , Disease Models, Animal , Doxycycline/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/cytology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation , Lung/drug effects , Mice , Mice, Inbred BALB C , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism
18.
J Immunol ; 189(1): 365-72, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22623332

ABSTRACT

α-Enolase (ENO1) is a multifunctional glycolytic enzyme expressed abundantly in the cytosol. It has been implicated in autoimmune and inflammatory diseases. Serum Abs against ENO1 were reported in rheumatoid arthritis (RA). Cell-surface expression of ENO1 has been found to be increased rapidly in response to inflammatory stimuli, but its expression and function has not been reported in RA. In this study, we show that cell-surface expression of ENO1 is increased on monocytes and macrophages isolated from RA patients but not on those from osteoarthritis patients, and Ab against ENO1 can stimulate these cells to produce higher amounts of proinflammatory mediators, such as TNF-α, IL-1 α/ß, IFN-γ, and PGE(2) via p38 MAPK and NF-κB pathway. The frequency of ENO1-positive cells in synovial fluid mononuclear cells was higher than PBMCs. ENO1-positive cells were also found in the inflamed synovium from RA patients and arthritic ankle tissues of mice with collagen-induced arthritis. Taken together, these findings suggest that Abs against ENO1 present in RA sera may stimulate monocytes and macrophages expressing cell-surface ENO1 and contribute to production of proinflammatory mediators during the effector phase of synovial inflammation.


Subject(s)
Arthritis, Rheumatoid/enzymology , Biomarkers, Tumor/biosynthesis , DNA-Binding Proteins/biosynthesis , Macrophages/enzymology , Monocytes/enzymology , Phosphopyruvate Hydratase/biosynthesis , Synovial Membrane/enzymology , Tumor Suppressor Proteins/biosynthesis , Amino Acid Sequence , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Biomarkers, Tumor/physiology , Cells, Cultured , Collagen/administration & dosage , DNA-Binding Proteins/physiology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred DBA , Molecular Sequence Data , Monocytes/immunology , Monocytes/pathology , Phosphopyruvate Hydratase/physiology , Synovial Membrane/immunology , Synovial Membrane/pathology , Tumor Suppressor Proteins/physiology
19.
Anticancer Res ; 30(2): 481-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20332458

ABSTRACT

The initiation and growth of hepatocellular carcinoma (HCC) are closely linked to chronic inflammation. Not only is cyclin D1 overexpressed, but it is also related to aggressive progression in HCC. However, the mechanism of expression cyclin D1, a cell-cycle regulator of paramount importance, in the tumor microenvironment remains unknown. Here, we investigated the mechanism of cyclin D1 expression induced by interleukin-6 (IL-6) and whether 3-[3,4-dihydroxy-phenyl]-acrylic acid 2-[3,4-dihydroxy-phenyl]-ethyl ester (CADPE), a derivate of caffeic acid, suppresses cyclin D1 expression. CADPE significantly inhibited IL-6-induced signal transducer and activator of transcription 3 (STAT3) activity in the Huh7 HCC cell line and attenuated IL-6-induced cyclin D1 transcription. Moreover, overexpression of constitutively active STAT3 increased cyclin D1 transcriptional activity and protein expression, whereas overexpression of a dominant-negative STAT3 deletion mutant (STAT3 (1-588)) reduced cyclin D1 transcriptional activity. In addition, CADPE effectively deacetylated histone 4 and prevented STAT3 recruitment to the cyclin D1 promoter, consistent with a role for the CADPE target, STAT3, in the regulation of cyclin D1 transcription. Collectively, these results indicate that CADPE suppresses cyclin D1 expression in HCC cells by blocking both IL-6-mediated STAT3 activation and recruitment of STAT3 to the cyclin D1 promoter.


Subject(s)
Caffeic Acids/pharmacology , Carcinoma, Hepatocellular/metabolism , Cyclin D1/metabolism , Interleukin-6/pharmacology , Liver Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Blotting, Western , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Luciferases/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
20.
Biol Pharm Bull ; 32(11): 1870-4, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19881300

ABSTRACT

Fraxinellone and sauchinone, isolated from natural substance, are known to have an anti-inflammatory effect in inflammatory conditions. However, the anti-inflammatory actions of these compounds have been insufficiently demonstrated in viral-induced neuroinflammation. A viral component (double-stranded (ds)RNA) triggers a toll-like receptor 3-dependent inflammatory response that stimulates pro-inflammatory mediators in the brain. In present study, we initially examined the biological effects of fraxinellone and sauchinone on anti-inflammatory actions in dsRNA-stimulated microglia. Both compounds inhibited dsRNA-induced inducible nitric oxide synthase (iNOS) expression, a major pro-inflammatory enzyme. To demonstrate the mechanism of inhibitory effect on iNOS expression, we further examined the signaling pathway induced by dsRNA in microglia. Our data show that dsRNA promotes the expression of signal transducers and activators of transcription (STAT)1/3 identified as major inflammatory transcription factors as well as activates c-Jun N-terminal kinase (JNK) in an early time. Moreover, both compounds suppressed activation of JNK-STAT1/3 signaling pathway. These results suggest that an anti-inflammatory effect by fraxinellone and sauchinone is mediated via blockade of the JNK-STAT1/3-iNOS signaling pathway in viral-infected microglia.


Subject(s)
Benzofurans/pharmacology , Benzopyrans/pharmacology , Dioxoles/pharmacology , Microglia/drug effects , Nitric Oxide Synthase Type II/metabolism , RNA, Double-Stranded/physiology , Animals , Base Sequence , Blotting, Western , DNA Primers , Mice , Microglia/enzymology , Toll-Like Receptor 3/genetics , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...