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










Publication year range
1.
ACS Appl Mater Interfaces ; 15(2): 2538-2551, 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36548054

ABSTRACT

The assembly of α-synuclein (αS) oligomers is recognized as the main pathological driver of synucleinopathies. While the elimination of toxic αS oligomers shows promise for the treatment of Parkinson's disease (PD), the discovery of αS oligomer degradation drugs has been hindered by the lack of proper drug screening tools. Here, we report a drug screening platform for monitoring the efficacy of αS-oligomer-degrading drugs using amyloid-shelled gold nanocomplexes (ASGNs). We fabricate ASGNs in the presence of dopamine, mimicking the in vivo generation process of pathological αS oligomers. To test our platform, the first of its kind for PD drugs, we use αS-degrading proteases and various small molecular substances that have shown efficacy in PD treatment. We demonstrate that the ASGN-based in vitro platform has strong potential to discover effective αS-oligomer-targeting drugs, and thus it may reduce the attrition problem in drug discovery for PD treatment.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Amyloid/metabolism , Amyloidogenic Proteins
2.
Transl Neurodegener ; 10(1): 49, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876226

ABSTRACT

Alzheimer's disease (AD) is the most serious age-related neurodegenerative disease and causes destructive and irreversible cognitive decline. Failures in the development of therapeutics targeting amyloid-ß (Aß) and tau, principal proteins inducing pathology in AD, suggest a paradigm shift towards the development of new therapeutic targets. The gram-negative bacteria and lipopolysaccharides (LPS) are attractive new targets for AD treatment. Surprisingly, an altered distribution of gram-negative bacteria and their LPS has been reported in AD patients. Moreover, gram-negative bacteria and their LPS have been shown to affect a variety of AD-related pathologies, such as Aß homeostasis, tau pathology, neuroinflammation, and neurodegeneration. Moreover, therapeutic approaches targeting gram-negative bacteria or gram-negative bacterial molecules have significantly alleviated AD-related pathology and cognitive dysfunction. Despite multiple evidence showing that the gram-negative bacteria and their LPS play a crucial role in AD pathogenesis, the pathogenic mechanisms of gram-negative bacteria and their LPS have not been clarified. Here, we summarize the roles and pathomechanisms of gram-negative bacteria and LPS in AD. Furthermore, we discuss the possibility of using gram-negative bacteria and gram-negative bacterial molecules as novel therapeutic targets and new pathological characteristics for AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Gram-Negative Bacteria/metabolism , Humans , Lipopolysaccharides
3.
Heliyon ; 7(11): e08433, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34877426

ABSTRACT

Psoriasis is a chronic autoimmune disease with an unknown etiology and highly limited treatment strategies. The drugs currently used in the treatment of psoriasis are rarely recommended for long-term use owing to the serious side effects. Although different targets have been identified for controlling psoriasis, the role of epigenetic modifications as therapeutic targets is yet to be elucidated. Here, we investigated the therapeutic potential of 8-hydroxyquinoline-5-carboxylic acid (IOX1), a novel drug with a genetic target, in psoriasis. The daily topical administration of IOX1 in a mouse model of imiquimod (IMQ)-induced psoriatic inflammation reduced inflammatory reactions in the skin and lowered the PASI score. Furthermore, intraperitoneally injected IOX1 repressed the inflammatory status induced by IMQ in psoriatic mice by reducing the mRNA levels of pro-inflammatory cytokines, restoring splenocyte populations, and regulating macrophage polarization. Our findings indicate the remedial effects of IOX1 on dermatitis psoriasis and the potential of IOX1 as a therapeutic compound in psoriasis.

4.
Sci Rep ; 11(1): 2942, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536477

ABSTRACT

Sepsis is caused by organ dysfunction initiated by an unrestrained host immune response to infection. The emergence of antibiotic-resistant bacteria has rapidly increased in the last decades and has stimulated a firm research platform to combat infections caused by antibiotic-resistant bacteria that cannot be eradicated with conventional antibiotics. Strategies like epigenetic regulators such as lysine demethylase (Kdm) has received attention as a new target. Thus, we sought to investigate the epigenetic mechanisms in sepsis pathophysiology with the aim of discovering new concepts for treatment. A transcriptome analysis of dendritic cells during their inflammatory state identified Kdm as a critical molecule in sepsis regulation. Next, 8-hydroxyquinoline-5-carboxylic acid (IOX1) ability to control endotoxemia induced by Lipopolysaccharide and bacterial sepsis was demonstrated. IOX1 has been shown to regulate endotoxemia and sepsis caused by Escherichia coli and carbapenem-resistant Acinetobacter baumannii and has also contributed to the suppression of multidrug-resistant bacterial growth through the inhibition of DNA Gyrase. These findings show that IOX1 could be a component agent against bacterial sepsis by functioning as a broad-spectrum antibiotic with dual effects.


Subject(s)
Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/drug therapy , Hydroxyquinolines/pharmacology , Sepsis/drug therapy , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , DNA Gyrase/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Hydroxyquinolines/therapeutic use , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Sepsis/immunology , Sepsis/microbiology
5.
Biochem Biophys Res Commun ; 543: 1-7, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33588136

ABSTRACT

Muscle wasting caused by catabolic reactions in skeletal muscle is commonly observed in patients with sepsis. Myostatin, a negative regulator of muscle mass, has been reported to be upregulated in diseases associated with muscle atrophy. However, the behavior of myostatin during sepsis is not well understood. Herein, we sought to investigate the expression and regulation of myostatin in skeletal muscle in mice inoculated with gram-negative bacteria. Interestingly, the protein level of myostatin was found to increase in the muscle of septic mice simultaneously with an increase in the levels of follistatin, NF-κΒ, myogenin, MyoD, p- FOXO3a, and p-Smad2. Furthermore, the inhibition of myostatin by YK11 repressed the levels of pro-inflammatory cytokines and organ damage markers in the bloodstream and in the major organs of mice, which originally increased in sepsis; thus, myostatin inhibition by YK11 decreased the mortality rate due to sepsis. The results of this study suggest that YK11 may help revert muscle wasting during sepsis and subdue the inflammatory environment, thereby highlighting its potential as a preventive agent for sepsis-related muscle wasting.


Subject(s)
Escherichia coli Infections/drug therapy , Escherichia coli/drug effects , Muscle, Skeletal/drug effects , Muscular Atrophy/prevention & control , Myostatin/antagonists & inhibitors , Norpregnadienes/pharmacology , Sepsis/drug therapy , Animals , Cachexia/metabolism , Cachexia/pathology , Cachexia/prevention & control , Cytokines/metabolism , Disease Models, Animal , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Male , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , NF-kappa B/metabolism , Sepsis/metabolism , Sepsis/pathology
6.
Int Immunopharmacol ; 73: 482-490, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31173970

ABSTRACT

There is currently no specific drug for the treatment of sepsis and antibiotic administration is considered the best option, despite numerous issues. Therefore, the development of drugs to control the pathogen-induced inflammatory responses associated with sepsis is essential. To address this, our study examined the transcriptomes of lipopolysaccharide (LPS)-induced dendritic cells (DCs), identifying TANK-binding kinase1 (Tbk1) as a key factor involved in the inflammatory response. These data suggested drug repositioning of the Tbk1 inhibitor CYT387, currently used for the treatment of myelofibrosis and some cancers, as a candidate for regulating the LPS-induced inflammatory response. CYT387 also inhibited pro-inflammatory cytokine and surface molecule expression by mature DCs after LPS exposure. These effects correlated with both Akt phosphorylation and IκBα degradation. Finally, CYT387 demonstrated therapeutic effects in LPS-induced endotoxemia and Escherichia coli K1-induced mouse models of sepsis and decreased the expression of pro-inflammatory cytokines. In conclusion, our study suggests that drug repositioning of CYT387 may serve as a potential therapeutic for sepsis.


Subject(s)
Benzamides/therapeutic use , Endotoxemia/drug therapy , Escherichia coli Infections/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Sepsis/drug therapy , Animals , Benzamides/pharmacology , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Repositioning , Endotoxemia/immunology , Escherichia coli Infections/immunology , Female , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Sepsis/immunology , Transcriptome/drug effects
8.
Sci Rep ; 9(1): 3817, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30846839

ABSTRACT

The development of novel peptide antibiotics with potent activity against multidrug-resistant Gram-negative bacteria and anti-septic activity is urgently needed. In this study, we designed short, 12-meric antimicrobial peptides by substituting amino acids from the N-terminal 12 residues of the papiliocin (Pap12-1) peptide to alter cationicity and amphipathicity and improve antibacterial activity and bacterial membrane interactions. Pap12-6, with an amphipathic α-helical structure and Trp12 at the C-terminus, showed broad-spectrum antibacterial activity, especially against multidrug-resistant Gram-negative bacteria. Dye leakage, membrane depolarization, and electron microscopy data proved that Pap12-6 kills bacteria by permeabilizing the bacterial membrane. Additionally, Pap12-6 significantly reduced the secretion of NO, TNF-α, and IL-6 and secreted alkaline phosphatase reporter gene activity confirmed that Pap12-6 shows anti-inflammatory activity via a TLR4-mediated NF-κB signaling pathway. In a mouse sepsis model, Pap12-6 significantly improved survival, reduced bacterial growth in organs, and reduced LPS and inflammatory cytokine levels in the serum and organs. Pap12-6 showed minimal cytotoxicity towards mammalian cells and controlled liver and kidney damage, proving its high bacterial selectivity. Our results suggest that Pap12-6 is a promising peptide antibiotic for the therapeutic treatment of Gram-negative sepsis via dual bactericidal and immunomodulatory effects on the host.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacterial Infections/drug therapy , Sepsis/drug therapy , Animals , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Drug Development , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/metabolism , Gram-Negative Bacterial Infections/metabolism , Interleukin-6/metabolism , Mice , Microbial Sensitivity Tests , Nitric Oxide/metabolism , Sepsis/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
J Mol Med (Berl) ; 97(5): 647-658, 2019 05.
Article in English | MEDLINE | ID: mdl-30848296

ABSTRACT

Sepsis is a disease responsible for the death of almost all critical patients. Once infected by virus or bacteria, patients can die due to systemic inflammation within a short period of time. Cytokine storm plays an essential role in causing organ dysfunction and septic shock. Thus, inhibition of cytokine secretion is considered very important in sepsis therapy. In this study, we found that TFP, an antipsychotic drug mainly used to treat schizophrenia by suppressing dopamine secretion, inhibited cytokine release from activated immune cells both in vitro and in vivo. Trifluoperazine (TFP) decreased the levels of pro-inflammatory cytokines without altering their transcription level. In LPS-induced endotoxemia and cecal content injection (CCI) models, TFP intraperitoneal administration improved survival rate. Thus, TFP was considered to inhibit the secretion of proteins through a mechanism similar to that of W7, a calmodulin inhibitor. Finally, we confirmed that TFP treatment relieved organ damage by estimating the concentrations of aspartate transaminase (AST), alanine transaminase (ALT), and blood urea nitrogen (BUN) in the serum. Our findings were regarded as a new discovery of the function of TFP in treating sepsis patients. KEY MESSAGES: • TFP inhibits LPS-induced activation of DCs by suppressing pro-inflammatory cytokine. • Treatment of TFP increases survival of LPS-induced endotoxemia and CCI sepsis models. • TFP exerted a protective effect against tissue or organ damage in animal models.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Drug Repositioning , Sepsis/drug therapy , Trifluoperazine/therapeutic use , Animals , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Female , Lipopolysaccharides/immunology , Mice, Inbred C57BL , Sepsis/immunology , Sepsis/pathology
10.
Mol Cells ; 42(3): 262-269, 2019 Mar 31.
Article in English | MEDLINE | ID: mdl-30841024

ABSTRACT

The porcine myeloid antimicrobial peptide (PMAP), one of the cathelicidin family members, contains small cationic peptides with amphipathic properties. We used a putative lysozyme originated from the bacteriophage P22 (P22 lysozyme) as a fusion partner, which was connected to the N-terminus of the PMAP36 peptide, to markedly increase the expression levels of recombinant PMAP36. The PMAP36-P22 lysozyme fusion protein with high solubility was produced in Escherichia coli. The final purified yield was approximately 1.8 mg/L. The purified PMAP36-P22 lysozyme fusion protein exhibited antimicrobial activity against both Gram-negative and Gram-positive bacteria (Staphylococcus aureus, Salmonella enterica serovar Typhimurium, Pseudomonas aeruginosa, and Bacillus subtilis). Furthermore, we estimated its hemolytic activity against pig erythrocytes as 6% at the high concentration (128 µM) of the PMAP36-P22 lysozyme fusion protein. Compared with the PMAP36 peptide (12%), our fusion protein exhibited half of the hemolytic activity. Overall, our recombinant PMAP36-P22 lysozyme fusion protein sustained the antimicrobial activity with the lower hemolytic activity associated with the synthetic PMAP36 peptide. This study suggests that the PMAP36-P22 lysozyme fusion system could be a crucial addition to the plethora of novel antimicrobials.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Muramidase/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Bacteria/drug effects , Bacteria/ultrastructure , Cell Membrane Permeability/drug effects , Female , Hemolysis/drug effects , Immunologic Factors/pharmacology , Mice, Inbred C57BL , Microbial Sensitivity Tests , Muramidase/chemistry , Plasmids/metabolism , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Swine , Cathelicidins
11.
J Immunother Cancer ; 7(1): 60, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30819254

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are professional antigen presenting cells (APCs), which can activate antigen-specific CD8+ T cell immunity, resulting in tumor clearance. Immature DCs are usually stimulated by various adjuvants through their immune receptors. Among them, Toll-like receptor 4 (TLR4) has an important role in activating DCs to cause their maturation. In fact, TLR4 is well-known to induce innate and adaptive immune responses against various external microbial or internal damage associated molecular patterns (DAMP). LPS is widely regarded as a strong stimulator of TLR4 signaling. However, LPS is inappropriate for use in humans since it is an endotoxin. Unfortunately, other TLR4 ligands such as HMGB1 or heat shock proteins have weak adjuvant effects. Therefore, there is a need to identify novel, biocompatible, strong, TLR4 ligands. METHODS: 40S ribosomal protein S3 (RPS3) was screened through pull-down assay using TLR4. BMDCs from wild type (WT) and TLR4 knock-out mice were treated by RPS3 to identify the activation and maturation of DCs. T cell generation including memory T cells, tumor prevention, and treatment experiments were performed with BMDCs based vaccination. Also, human DCs originated from patients were treated by RPS3 to confirm the activation and maturation of DCs. RESULTS: In this study, we identified 40S ribosomal protein S3 (RPS3) through a pull-down assay using a variety of human cancer cell-derived proteins that could bind to TLR4. RPS3 was released from tumor cells following treatment with an anticancer drug, and it was shown that the released RPS3 binds to TLR4. Recombinant RPS3 induced maturation and activation of DCs, and following pulsing with tumor specific antigens, these DCs could be used as a vaccine to significantly increase tumor specific CD8+IFN-γ+ T cells, and provide both tumor prevention and tumor treatment effects. The effect of RPS3 on DC maturation and its utility as a vaccine were shown to be dependent on TLR4 using TLR4 knockout mice. CONCLUSIONS: This study therefore proved that human cancer cell-derived RPS3, a novel TLR4 ligand, has great potential as an adjuvant in tumor-specific antigen DC-based vaccines.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Ribosomal Proteins/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cytokines/metabolism , Female , HMGB1 Protein/metabolism , Humans , Immunologic Memory , Ligands , Mice , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Recombinant Proteins , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
12.
Front Immunol ; 10: 3063, 2019.
Article in English | MEDLINE | ID: mdl-32038618

ABSTRACT

Probiotics can be an effective treatment for atopic dermatitis (AD), while their mechanism of action is still unclear. Here, we induced AD in mice with 2,4-dinitrochlorobenzene and administrated YK4, a probiotic mixture consisting of Lactobacillus acidophilus CBT LA1, L. plantarum CBT LP3, Bifidobacterium breve CBT BR3, and B. lactis CBT BL3. Then, we have validated the underlying mechanism for the alleviation of AD by YK4 from the intestinal and systematic immunological perspectives. Administration of YK4 in AD mice alleviated the symptoms of AD by suppressing the expression of skin thymic stromal lymphopoietin and serum immunoglobulin E eliciting excessive T-helper (Th) 2 cell-mediated responses. YK4 inhibited Th2 cell population through induce the proportion of Th1 cells in spleen and Treg cells in Peyer's patches and mesenteric lymph node (mLN). CD103+ dendritic cells (DCs) in mLN and the spleen were significantly increased in AD mice administered with YK4 when compared to AD mice. Furthermore, galectin-9 was significantly increased in the gut of AD mice administered with YK4. In vitro experiments were performed using bone marrow-derived DCs (BMDC) and CD4+ T cells to confirm the immune mechanisms of YK4 and galectin-9. The expression of CD44, a receptor of galectin-9, together with programmed death-ligand 1 was significantly upregulated in BMDCs following treatment with YK4. IL-10 and IL-12 were upregulated when BMDCs were treated with YK4. Cytokines together with co-receptors from DCs play a major role in the differentiation and activation of CD4+ T cells. Proliferation of Tregs and Th1 cell activation were enhanced when CD4+T cells were co-cultured with YK4-treated BMDCs. Galectin-9 appeared to contribute at least partially to the proliferation of Tregs. The results further suggested that DCs treated with YK4 induced the differentiation of naïve T cells toward Th1 and Tregs. At the same time, YK4 alleviated AD symptoms by inhibiting Th2 response. Thus, the present study suggested a potential role of YK4 as an effective immunomodulatory agent in AD patients.


Subject(s)
Dermatitis, Atopic/etiology , Dermatitis, Atopic/metabolism , Dietary Supplements , Galectins/metabolism , Immunomodulation , Probiotics/administration & dosage , Animals , Cytokines/metabolism , Dermatitis, Atopic/pathology , Disease Models, Animal , Female , Humans , Mice , Phenotype , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
13.
Cardiovasc Toxicol ; 19(3): 244-254, 2019 06.
Article in English | MEDLINE | ID: mdl-30519910

ABSTRACT

This study investigated vildagliptin-induced vasodilation and its related mechanisms using phenylephrine induced precontracted rabbit aortic rings. Vildagliptin induced vasodilation in a concentration-dependent manner. Pretreatment with the large-conductance Ca2+-activated K+ channel blocker paxilline, ATP-sensitive K+ channel blocker glibenclamide, and inwardly rectifying K+ channel blocker Ba2+ did not affect the vasodilatory effects of vildagliptin. However, application of the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine significantly reduced the vasodilatory effects of vildagliptin. In addition, application of either of two sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitors, thapsigargin or cyclopiazonic acid, effectively inhibited the vasodilatory effects of vildagliptin. These vasodilatory effects were not affected by pretreatment with adenylyl cyclase, protein kinase A (PKA), guanylyl cyclase, or protein kinase G (PKG) inhibitors, or by removal of the endothelium. From these results, we concluded that vildagliptin induced vasodilation via activation of Kv channels and the SERCA pump. However, other K+ channels, PKA/PKG-related signaling cascades associated with vascular dilation, and the endothelium were not involved in vildagliptin-induced vasodilation.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Muscle, Smooth, Vascular/drug effects , Potassium Channels, Voltage-Gated/agonists , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Vildagliptin/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Enzyme Activation , Male , Muscle, Smooth, Vascular/enzymology , Potassium Channels, Voltage-Gated/metabolism , Rabbits , Signal Transduction
14.
Oncoimmunology ; 7(10): e1472187, 2018.
Article in English | MEDLINE | ID: mdl-30288341

ABSTRACT

Dendritic cell (DC)-based vaccines are recognized as a promising immunotherapeutic strategy against cancer. Various adjuvants are often incorporated to enhance the modest immunogenicity of DC vaccines. More specifically, many of the commonly used adjuvants are derived from bacteria. In the current study, we evaluate the use of apoptosis inhibitor 5 (API5), a damage-associated molecular pattern expressed by many human cancer cells, as a novel DC vaccine adjuvant. We showed that API5 can prompt activation and maturation of DCs and activate NFkB by stimulating the Toll-like receptor signaling pathway. We also demonstrated that vaccination with API5-treated DCs pulsed with OVA, E7, or AH1-A5 peptides led to the generation of OVA, E7, or AH1-A5-specific CD8 + T cells and memory T cells, which is associated with long term tumor protection and antitumor effects in mice, against EG.7, TC-1, and CT26 tumors. Additionally, we determined that API5-mediated DC activation and immune stimulation are dependent on TLR4. Lastly, we showed that the API5 protein sequence fragment that is proximal to its leucine zipper motif is responsible for the adjuvant effects exerted by API5. Our data provide evidence that support the use of API5 as a promising adjuvant for DC-based therapies, which can be applied in combination with other cancer therapies. Most notably, our results further support the continued investigation of human-based adjuvants.

15.
Cancer Res ; 78(21): 6247-6256, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30115698

ABSTRACT

Chemotherapy is commonly used in the treatment of ovarian cancer, yet most ovarian cancers harbor inherent resistance or develop acquired resistance. Therefore, novel therapeutic approaches to overcome chemoresistance are required. In this study, we developed a hyaluronic acid-labeled poly(d,l-lactide-co-glycolide) nanoparticle (HA-PLGA-NP) encapsulating both paclitaxel (PTX) and focal adhesion kinase (FAK) siRNA as a selective delivery system against chemoresistant ovarian cancer. The mean size and zeta potential of the HA-PLGA-NP were 220 nm and -7.3 mV, respectively. Incorporation efficiencies for PTX and FAK siRNA in the HA-PLGA-NPs were 77% and 85%, respectively. HA-PLGA-NP showed higher binding efficiency for CD44-positive tumor cells as compared with CD44-negative cells. HA-PLGA (PTX+FAK siRNA)-NP caused increased cytotoxicity and apoptosis in drug-resistant tumor cells. Treatment of human epithelial ovarian cancer tumor models HeyA8-MDR (P < 0.001) and SKOV3-TR (P < 0.001) with HA-PLGA (PTX+FAK siRNA)-NP resulted in significant inhibition of tumor growth. Moreover, in a drug-resistant, patient-derived xenograft (PDX) model, HA-PLGA (PTX+FAK siRNA)-NP significantly inhibited tumor growth compared with PTX alone (P < 0.002). Taken together, HA-PLGA-NP acts as an effective and selective delivery system for both the chemotherapeutic and the siRNA in order to overcome chemoresistance in ovarian carcinoma.Significance: These findings demonstrate the efficacy of a novel, selective, two-in-one delivery system to overcome chemoresistance in epithelial ovarian cancer. Cancer Res; 78(21); 6247-56. ©2018 AACR.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Focal Adhesion Kinase 1/metabolism , Hyaluronan Receptors/chemistry , Nanoparticles/chemistry , Paclitaxel/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , RNA, Small Interfering/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial/drug therapy , Cell Line, Tumor , Cell Survival , Drug Carriers/chemistry , Drug Resistance, Neoplasm , Female , Gene Silencing , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism
16.
Nutrients ; 10(7)2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29976865

ABSTRACT

Toll-like receptor 2 (TLR2) responses are involved in various inflammatory immune disorders. Phloretin is a naturally occurring dietary flavonoid that is abundant in fruit. Here, we investigated whether the anti-inflammatory activity of phloretin is mediated through TLR2 pathways, and whether phloretin acts as an inhibitor of TLR2/1 heterodimerization using the TLR2/1 agonist Pam3CSK4. We tested the effects of phloretin on tumor necrosis factor (TNF)-α production induced by various TLRs using known TLR-specific agonists. Phloretin significantly inhibited Pam3CSK4-induced TRL2/1 signaling in Raw264.7 cells compared to TLR signaling induced by the other agonists tested. Therefore, we further tested the effects of phloretin in human embryonic kidney (HEK) 293-hTLR2 cells induced by Pam3CSK4, and confirmed that phloretin has comparable inhibition of TLR2/1 heterodimerization to that induced by the known TLR2 inhibitor CU-CPT22. Moreover, phloretin reduced the secretion of the inflammatory cytokines TNF-α and interleukin (IL)-8 in Pam3CSK4-induced HEK293-hTLR2 cells, whereas it did not significantly reduce these cytokines under Pam2CSK4-induced activation. Western blot results showed that phloretin significantly suppressed Pam3CSK4-induced TLR2 and NF-κB p65 expression. The molecular interactions between phloretin and TLR2 were investigated using bio-layer interferometry and in silico docking. Phloretin bound to TLR2 with micromolar binding affinity, and we proposed a binding model of phloretin at the TLR2⁻TLR1 interface. Overall, we confirmed that phloretin inhibits the heterodimerization of TLR2/1, highlighting TLR2 signaling as a therapeutic target for treating TLR2-mediated inflammatory immune diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/prevention & control , Macrophages/drug effects , Phloretin/pharmacology , Toll-Like Receptor 1/antagonists & inhibitors , Toll-Like Receptor 2/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inflammation/metabolism , Interleukin-8/metabolism , Lipopeptides/pharmacology , Macrophages/metabolism , Mice , Molecular Docking Simulation , Phloretin/chemistry , Phloretin/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , RAW 264.7 Cells , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 1/chemistry , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Drug Deliv ; 25(1): 1394-1402, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29890852

ABSTRACT

Angiogenesis plays an essential role in the growth and metastasis of tumor cells, and the modulation of angiogenesis can be an effective approach for cancer therapy. We focused on silencing the angiogenic gene PLXDC1 as an important factor for anti-angiogenesis tumor therapy. Herein, we developed PLXDC1 small interfering siRNA (siRNA)-incorporated chitosan nanoparticle (CH-NP/siRNA) coated with hyaluronic acid (HA) to target the CD44 receptor on tumor endothelial cells. This study aimed to improve targeted delivery and enhance therapeutic efficacy for tumor anti-angiogenesis. The HA-CH-NP/siRNA was 200 ± 10 nm in size with a zeta potential of 26.4 mV. The loading efficiency of siRNA to the HA-CH-NP/siRNA was up to 60%. The selective binding of HA-CH-NP/siRNA to CD44-positive tumor endothelial cells increased by 2.1-fold compared with that of the CD44 nontargeted CH-NP/siRNA. PLXDC1 silencing by the HA-CH-NP/siRNA significantly inhibited tumor growth in A2780 tumor-bearing mice compared with that in the control group (p < .01), and mRNA expression of PLXDC1 was significantly reduced in the HA-CH-NP/siRNA-treated group. Furthermore, treatment with HA-CH-NP/siRNA resulted in significant inhibition of cell proliferation (p < .001), reduced microvessel density (p < .001), and increased cell apoptosis (p < .001). This study demonstrates that HA-CH-NP/siRNA is a highly selective delivery platform for siRNA, and has broad potential to be used in anti-angiogenesis tumor therapy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Chitosan/chemistry , Endothelial Cells/drug effects , Hyaluronan Receptors/genetics , Nanoparticles/chemistry , Neoplasm Proteins/genetics , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Receptors, Cell Surface/genetics , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Female , Gene Silencing/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronic Acid/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/administration & dosage , Particle Size , RNA, Messenger/genetics , Xenograft Model Antitumor Assays/methods
18.
J Nat Prod ; 81(6): 1435-1443, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29851490

ABSTRACT

Sepsis is a systemic inflammatory response to pathogenic infection that currently has no specific pharmaceutical interventions. Instead, antibiotics administration is considered the best available option, despite increasing drug resistance. Alternative strategies are therefore urgently required to prevent sepsis and strengthen the host immune system. One such option is tamarixetin (4'- O-methylquercetin), a naturally occurring flavonoid derivative of quercetin that protects against inflammation. The purpose of this study was to determine whether the anti-inflammatory effects of tamarixetin protect against the specific inflammatory conditions induced in lipopolysaccharide (LPS) or Escherichia coli K1 models of sepsis. Our study showed that tamarixetin reduced the secretion of various inflammatory cytokines by dendritic cells after activation with LPS. It also promoted the secretion of the anti-inflammatory cytokine interleukin (IL)-10 and specifically increased the population of IL-10-secreting immune cells in LPS-activated splenocytes. Tamarixetin showed general anti-inflammatory effects in mouse models of bacterial sepsis and decreased bacteria abundance and endotoxin levels. We therefore conclude that tamarixetin has superior anti-inflammatory properties than quercetin during bacterial sepsis. This effect is associated with an increased population of IL-10-secreting immune cells and suggests that tamarixetin could serve as a specific pharmaceutical option to prevent bacterial sepsis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Disaccharides/pharmacology , Interleukin-10/metabolism , Quercetin/analogs & derivatives , Sepsis/drug therapy , Animals , Cytokines/metabolism , Dendritic Cells/drug effects , Escherichia coli/pathogenicity , Female , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Quercetin/pharmacology , Sepsis/metabolism
19.
Article in English | MEDLINE | ID: mdl-29637049

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is an extremely successful pathogen with multifactorial ability to control the host immune response. Insights into the Mtb factors modulating host response are required for the discovery of novel vaccine antigen targets as well as a better understanding of dynamic interactions between the bacterial factors and host cells. Here, we exploited the functional role of Mtb GrpE, a cofactor of heat-shock protein 70 (HSP70), in promoting naïve CD4+/CD8+T cell differentiation toward Th1-type T-cell immunity through interaction with dendritic cells (DCs). GrpE functionally induced DC maturation by up-regulating the expression of cell surface molecules (CD80, CD86, and MHC class I and II) and production of several pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6, and IL-12p70) in DCs. These effects of GrpE in DC activation were initiated upon binding to Toll-like receptor 4 (TLR4) followed by activation of downstream MyD88-, TRIF-, MAPK-, and NF-κB-dependent signaling pathways. GrpE-activated DCs displayed an excellent capacity to effectively polarize naïve CD4+ and CD8+ T cells toward Th1-type T-cell immunity with the dose-dependent secretion of IFN-γ and IL-2 together with increased levels of CXCR3 expression. Notably, GrpE-stimulated DCs induced the proliferation of GrpE-specific Th1-type effector/memory CD4+/CD8+CD44highCD62Llow T cells from the spleen of Mtb-infected mice in a TLR4-dependent manner. Collectively, these results demonstrate that GrpE is a novel immune activator that interacts with DCs, in particular, via TLR4, to generate Th1-biased memory T cells in an antigen-specific manner. GrpE may contribute to the enhanced understanding of host-pathogen interactions as well as providing a rational basis for the discovery of new potential targets to develop an effective tuberculosis vaccine.


Subject(s)
Bacterial Proteins/immunology , Dendritic Cells/immunology , Heat-Shock Proteins/immunology , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Toll-Like Receptor 4/immunology , Tuberculosis/immunology , Animals , Bacterial Proteins/genetics , Cell Differentiation , Female , Heat-Shock Proteins/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , NF-kappa B/genetics , NF-kappa B/immunology , Toll-Like Receptor 4/genetics , Tuberculosis/genetics , Tuberculosis/microbiology
20.
Clin Exp Pharmacol Physiol ; 45(2): 205-212, 2018 02.
Article in English | MEDLINE | ID: mdl-28945283

ABSTRACT

Amitriptyline, a tricyclic antidepressant (TCA) drug, is widely used in treatment of psychiatric disorders. However, the side effects of amitriptyline on vascular K+ channels remain to be determined. Therefore, we investigated the effect of the tricyclic antidepressant and serotonin reuptake inhibitor amitriptyline on voltage-dependent K+ (Kv) channels in freshly isolated rabbit coronary arterial smooth muscle cells, using the whole-cell patch clamp technique. The Kv current amplitudes were inhibited by amitriptyline in a concentration-dependent manner, with an apparent IC50 value of 2.2 ± 0.14 µmol/L and a Hill coefficient of 0.87 ± 0.03. Amitriptyline shifted the activation curve to a more positive potential, but had no significant effect on the inactivation curve, suggesting that amitriptyline altered the voltage sensitivity of Kv channels. Pretreatment with Kv1.5 and Kv1.2 channel inhibitors did not alter the inhibitory effect of amitriptyline on Kv channels. Additionally, application of train pulses (1 and 2 Hz) did not affect amitriptyline-induced inhibition of Kv currents, which suggested that the action of amitriptyline on Kv channels was not use (state)-dependent. From these results, we concluded that amitriptyline inhibited the channels in a concentration-dependent, but state-independent manner.


Subject(s)
Amitriptyline/pharmacology , Coronary Vessels , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Potassium Channel Blockers , Animals , Antidepressive Agents, Tricyclic/pharmacology , Potassium Channels/metabolism , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...