Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(19): e2306684, 2024 May.
Article in English | MEDLINE | ID: mdl-38482992

ABSTRACT

Cryotherapy leverages controlled freezing temperature interventions to engender a cascade of tumor-suppressing effects. However, its bottleneck lies in the standalone ineffectiveness. A promising strategy is using nanoparticle therapeutics to augment the efficacy of cryotherapy. Here, a cold-responsive nanoplatform composed of upconversion nanoparticles coated with silica - chlorin e6 - hyaluronic acid (UCNPs@SiO2-Ce6-HA) is designed. This nanoplatform is employed to integrate cryotherapy with photodynamic therapy (PDT) in order to improve skin cancer treatment efficacy in a synergistic manner. The cryotherapy appeared to enhance the upconversion brightness by suppressing the thermal quenching. The low-temperature treatment afforded a 2.45-fold enhancement in the luminescence of UCNPs and a 3.15-fold increase in the photodynamic efficacy of UCNPs@SiO2-Ce6-HA nanoplatforms. Ex vivo tests with porcine skins and the subsequent validation in mouse tumor tissues revealed the effective HA-mediated transdermal delivery of designed nanoplatforms to deep tumor tissues. After transdermal delivery, in vivo photodynamic therapy using the UCNPs@SiO2-Ce6-HA nanoplatforms resulted in the optimized efficacy of 79% in combination with cryotherapy. These findings underscore the Cryo-PDT as a truly promising integrated treatment paradigm and warrant further exploring the synergistic interplay between cryotherapy and PDT with bright upconversion to unlock their full potential in cancer therapy.


Subject(s)
Hyaluronic Acid , Nanoparticles , Photochemotherapy , Animals , Photochemotherapy/methods , Mice , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Cryotherapy/methods , Chlorophyllides , Porphyrins/chemistry , Porphyrins/administration & dosage , Disease Models, Animal , Photosensitizing Agents/administration & dosage , Administration, Cutaneous , Silicon Dioxide/chemistry , Swine
2.
Biomater Res ; 27(1): 45, 2023 May 13.
Article in English | MEDLINE | ID: mdl-37173721

ABSTRACT

Cancer is a disease caused by abnormal cell growth that spreads through other parts of the body and threatens life by destroying healthy tissues. Therefore, numerous techniques have been employed not only to diagnose and monitor the progress of cancer in a precise manner but also to develop appropriate therapeutic agents with enhanced efficacy and safety profiles. In this regard, molecularly imprinted polymers (MIPs), synthetic receptors that recognize targeted molecules with high affinity and selectivity, have been intensively investigated as one of the most attractive biomaterials for theragnostic approaches. This review describes diverse synthesis strategies to provide the rationale behind these synthetic antibodies and provides a selective overview of the recent progress in the in vitro and in vivo targeting of cancer biomarkers for diagnosis and therapeutic applications. Taken together, the topics discussed in this review provide concise guidelines for the development of novel MIP-based systems to diagnose cancer more precisely and promote successful treatment. Molecularly imprinted polymers (MIPs), synthetic receptors that recognize targeted molecules with high affinity and selectivity, have been intensively investigated as one of the most attractive biomaterials for cancer theragnostic approaches. This review describes diverse synthesis strategies to provide the rationale behind these synthetic antibodies and provides a selective overview of the recent progress in the in vitro and in vivo targeting of cancer biomarkers for diagnosis and therapeutic applications. The topics discussed in this review aim to provide concise guidelines for the development of novel MIP-based systems to diagnose cancer more precisely and promote successful treatment.

3.
Biosensors (Basel) ; 13(3)2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36979518

ABSTRACT

Over the past two decades, lanthanide-based upconversion nanoparticles (UCNPs) have been fascinating scientists due to their ability to offer unprecedented prospects to upconvert tissue-penetrating near-infrared light into color-tailorable optical illumination inside biological matter. In particular, luminescent behavior UCNPs have been widely utilized for background-free biorecognition and biosensing. Currently, a paramount challenge exists on how to maximize NIR light harvesting and upconversion efficiencies for achieving faster response and better sensitivity without damaging the biological tissue upon laser assisted photoactivation. In this review, we offer the reader an overview of the recent updates about exciting achievements and challenges in the development of plasmon-modulated upconversion nanoformulations for biosensing application.


Subject(s)
Biosensing Techniques , Lanthanoid Series Elements , Nanoparticles , Luminescence
4.
Sci Rep ; 13(1): 101, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596838

ABSTRACT

Tranilast, an anti-allergic drug used in the treatment of bronchial asthma, was identified as an inhibitor of the transcription factor Forkhead box O-1 (FoxO-1) by high throughput chemical library screening in the present study. Based on FoxO-1's role in apoptotic cell death and differentiation, we examined the effect of tranilast on palmitic acid (PA)-induced cell damage in INS-1 cells. Tranilast substantially inhibited lipoapoptosis and restored glucose-stimulated insulin secretion under high PA exposure. Moreover, PA-mediated downregulation of PDX-1, MafA, and insulin expression was attenuated by tranilast. PA-induced oxidative and ER stress were also reduced in the presence of tranilast. These protective effects were accompanied by increased phosphorylation and decreased nuclear translocation of FoxO-1. Conversely, the effects of tranilast were diminished when treated in transfected cells with FoxO-1 phosphorylation mutant (S256A), suggesting that the tranilast-mediated effects are associated with inactivation of FoxO-1. Examination of the in vivo effects of tranilast using wild type and diabetic db/db mice showed improved glucose tolerance along with FoxO-1 inactivation in the pancreas of the tranilast-treated groups. Thus, we report here that tranilast has protective effects against PA-induced lipotoxic stress in INS-1 cells, at least partly, via FoxO-1 inactivation, which results in improved glucose tolerance in vivo.


Subject(s)
Insulin-Secreting Cells , Palmitic Acid , Mice , Animals , Palmitic Acid/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Apoptosis , Glucose/metabolism
5.
Adv Drug Deliv Rev ; 188: 114419, 2022 09.
Article in English | MEDLINE | ID: mdl-35810884

ABSTRACT

In the past decade, upconversion (UC) nanomaterials have been extensively investigated for the applications to photomedicines with their unique features including biocompatibility, near-infrared (NIR) to visible conversion, photostability, controllable emission bands, and facile multi-functionality. These characteristics of UC nanomaterials enable versatile light delivery for deep tissue biophotonic applications. Among various stimuli-responsive delivery systems, the light-responsive delivery process has been greatly advantageous to develop spatiotemporally controllable on-demand "smart" photonic medicines. UC nanomaterials are classified largely to two groups depending on the photon UC pathway and compositions: inorganic lanthanide-doped UC nanoparticles and organic triplet-triplet annihilation UC (TTA-UC) nanomaterials. Here, we review the current-state-of-art inorganic and organic UC nanomaterials for photo-medicinal applications including photothermal therapy (PTT), photodynamic therapy (PDT), photo-triggered chemo and gene therapy, multimodal immunotherapy, NIR mediated neuromodulations, and photochemical tissue bonding (PTB). We also discuss the future research direction of this field and the challenges for further clinical development.


Subject(s)
Nanoparticles , Nanostructures , Photochemotherapy , Delivery of Health Care , Humans
6.
Eur J Pharmacol ; 928: 175085, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35679889

ABSTRACT

Browning, a white to brown-like (beige) adipocyte conversion, offers a promising therapeutic strategy for the treatment of human obesity. In the present study, the effects of sodium salicylate, a nonsteroidal anti-inflammatory drug, on adipocyte browning were investigated. We found sodium salicylate altered the macrophage phenotype to M2 in RAW264.7 cells, mediated by up-regulation of heme oxygenase-1 (HO-1), and sodium salicylate-treated conditioned medium from macrophages (Sal-M2 CM) induced browning of fully differentiated 3T3-L1 adipocytes. Conversely, the conditioned medium obtained from macrophages when treated with sodium salicylate in the presence of either ZnPP (a HO-1 inhibitor) or HO-1 siRNA did not induce browning. In association with macrophage HO-1 induction by sodium salicylate, iron production also increased, and deferoxamine (an iron chelator) blunted the browning effects of Sal-M2 CM, suggesting that iron may play a role in the Sal-M2 CM-induced browning. The in vivo browning effects of sodium salicylate were confirmed in ob/ob mice, whereas in vivo macrophage depletion by clodronate as well as HO-1 blockade by either ZnPP or adeno-associated virus carrying HO-1 shRNA (AAV-HO-1 shRNA) attenuated the browning effects of sodium salicylate. These results reveal sodium salicylate induces browning in vitro and in vivo by up-regulating HO-1 thus promoting M2 polarization.


Subject(s)
Adipocytes, Brown , Adipocytes, White , Heme Oxygenase-1 , Macrophages , Sodium Salicylate , 3T3-L1 Cells , Animals , Culture Media, Conditioned , Heme Oxygenase-1/metabolism , Iron , Membrane Proteins , Mice , RNA, Small Interfering/pharmacology , Sodium Salicylate/pharmacology , Up-Regulation
7.
Adv Exp Med Biol ; 1351: 109-124, 2022.
Article in English | MEDLINE | ID: mdl-35175614

ABSTRACT

Graphene and graphene-based materials have been attracted in the past few years for biomedical applications due to their physicochemical and biological properties such as large surface area, chemical and mechanical stability, excellent conductivity, and good biocompatibility. Graphene-based materials not only surface modified graphene-based materials like graphene oxide (GO) or reduced graphene oxide (rGO) but also other structural forms like fullerene, carbon nanotubes, and graphite have been applied to advanced drug delivery systems. In this chapter, we review on the application of graphene-based materials in the drug delivery system with their physicochemical properties, methods for the preparation of graphene-based carriers, followed by analysis about their biodistribution and biosafety whether they are suitable as drug delivery carriers.


Subject(s)
Graphite , Nanostructures , Nanotubes, Carbon , Drug Carriers/chemistry , Drug Delivery Systems , Graphite/chemistry , Nanostructures/chemistry , Tissue Distribution
8.
Biomater Res ; 25(1): 24, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34321111

ABSTRACT

Various non-invasive administrations have recently emerged as an alternative to conventional needle injections. A transdermal drug delivery system (TDDS) represents the most attractive method among these because of its low rejection rate, excellent ease of administration, and superb convenience and persistence among patients. TDDS could be applicable in not only pharmaceuticals but also in the skin care industry, including cosmetics. Because this method mainly involves local administration, it can prevent local buildup in drug concentration and nonspecific delivery to tissues not targeted by the drug. However, the physicochemical properties of the skin translate to multiple obstacles and restrictions in transdermal delivery, with numerous investigations conducted to overcome these bottlenecks. In this review, we describe the different types of available TDDS methods, along with a critical discussion of the specific advantages and disadvantages, characterization methods, and potential of each method. Progress in research on these alternative methods has established the high efficiency inherent to TDDS, which is expected to find applications in a wide range of fields.

9.
Front Chem ; 9: 699284, 2021.
Article in English | MEDLINE | ID: mdl-34169061

ABSTRACT

The localized surface plasmon resonance of metallic nanoparticles has attracted much attention owing to its unique characteristics, including the enhancement of signals in sensors and photothermal effects. In particular, hollow gold nanostructures are highly promising for practical applications, with significant advantages being found in their material properties and structures: 1) the interaction between the outer surface plasmon mode and inner cavity mode leads to a greater resonance, allowing it to absorb near-infrared light, which can readily penetrate tissue; 2) it has anti-corrosiveness and good biocompatibility, which makes it suitable for biomedical applications; 3) it shows a reduced net density and large surface area, allowing the possibility of nanocarriers for drug delivery. In this review, we present information on the classification, characteristics, and synthetic methods of hollow gold nanostructures; discuss the recent advances in hollow gold nanostructures in biomedical applications, including biosensing, bioimaging, photothermal therapy, and drug delivery; and report on the existing challenges and prospects for hollow gold nanostructures.

10.
Materials (Basel) ; 14(6)2021 Mar 18.
Article in English | MEDLINE | ID: mdl-33803897

ABSTRACT

Hyaluronate (HA) has been widely investigated for noninvasive topical drug delivery of chemical drugs and biopharmaceuticals. However, previous noninvasive delivery systems have been facilitated mostly by chemical conjugation of drugs with HA, which can cause reduced therapeutic efficacy and safety issues in chemically modified drugs. Here, HA nanogels were synthesized by crosslinking via "click" chemistry for noninvasive topical delivery of a model drug without chemical modification. The model-drug-encapsulating HA nanogels could be uptaken to the skin melanoma cells in vitro by HA-mediated endocytosis. In addition, histological analysis showed that HA nanogels could be topically delivered to the deep skin and tongue tissues through the noninvasive delivery routes. Taken together, HA nanogels could be effectively used for the noninvasive topical delivery of various therapeutic drugs.

11.
Eur J Pharmacol ; 899: 174011, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33705803

ABSTRACT

Forkhead transcription factor forkhead box O1 (FoxO1) plays an important role in glucose and lipid metabolism, contributing to the pathogenesis of metabolic disorders. This study aimed to discover a novel FoxO1 inhibitor as a potential new anti-diabetic drug candidate, and describes the biological effects of JY-2, 5-(2,4-dichlorophenyl)-3-(pyridin-2-yl)-1,2,4-oxadiazole in vitro and in vivo. JY-2 inhibited FoxO1 transcriptional activity in a concentration-dependent manner, with an IC50 value of 22 µM. The inhibitory effects of JY-2 on FoxO3a and FoxO4 appeared to be weaker than that on FoxO1. Consistent with its inhibitory effect on FoxO1, JY-2 reduced the palmitic acid (PA)-stimulated mRNA expression of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), two key enzymes involved in gluconeogenesis in HepG2 cells. In association with the reduced expression of lipid metabolism genes, triglyceride accumulation was also reduced by JY-2, as determined by Oil Red O staining. In addition, JY-2 restored PA-impaired glucose-stimulated insulin secretion (GSIS), in conjunction with an increased mRNA expression of PDX1, MafA, and insulin in INS-1 cells. The in vivo efficacy of JY-2 was examined using C57BL/6J, db/db, and high fat-diet induced obese and diabetic (DIO) mice models, and showed that JY-2 improved glucose tolerance, in parallel with a reduced mRNA expression of gluconeogenic genes. Pharmacokinetic analysis revealed that JY-2 exhibited excellent oral bioavailability (98%), with little adverse effects. These results demonstrated that the novel FoxO1 inhibitor, JY-2, may exert beneficial anti-diabetic effects and that it warrants further investigation as a novel anti-diabetic drug candidate.


Subject(s)
Diabetes Mellitus/drug therapy , Gluconeogenesis/drug effects , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Oxadiazoles/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus/enzymology , Diabetes Mellitus/pathology , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Hep G2 Cells , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Hypoglycemic Agents/pharmacokinetics , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Obesity/complications , Oxadiazoles/pharmacokinetics , Palmitic Acid/toxicity , Rats , Signal Transduction
12.
Sci Rep ; 10(1): 2165, 2020 Feb 04.
Article in English | MEDLINE | ID: mdl-32015443

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Sci Rep ; 9(1): 1236, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718686

ABSTRACT

Telmisartan is a well-known anti-hypertensive drug acting as an angiotensin 2 receptor blocker (ARB), but it also possesses partial PPARγ agonistic activity and induces insulin sensitivity. In the present study, we investigated the effects of telmisartan on macrophage polarization in association with its browning capacity, because PPARγ plays a key role in M2 polarization and in the browning of white adipocytes. Telmisartan induced M2 marker expression in murine macrophages concentration dependently, which was confirmed by flow cytometry. Both PPARγ and PPARδ activations appear to be responsible for telmisartan-induced M2 polarization. Telmisartan-treated conditioned medium (Tel-CM) of RAW264.7 cells and of bone marrow derived macrophages (BMDM) induced the expressions of browning markers in fully differentiated white adipocytes with reduced lipid droplets, and increased oxygen consumption rate and mitochondrial biogenesis. Levels of catecholamines (CA) released into the conditioned medium as well as intracellular tyrosine hydroxylase (TH) mRNAs were found to be increased by telmisartan, and browning effects of Tel-CM were lessened by ß3 receptor antagonist (L-748,337), suggesting CA secreted into CM play a role in Tel-CM-induced adipocyte browning. Acute administration of telmisartan (2 weeks, p.o.) to C57BL/6J mice increased the expressions of browning markers and M2 markers in white adipose tissues, whereas macrophage depletion by clodronate liposome pretreatment attenuated the telmisartan-induced expressions of browning markers. Together, telmisartan was observed to induce the browning of fully differentiated white adipocytes, at least in part, via PPAR activation-mediated M2 polarization.


Subject(s)
Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Macrophage Activation/drug effects , Macrophages/immunology , Telmisartan/pharmacology , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Catecholamines/metabolism , Cell Differentiation , Clodronic Acid/pharmacology , Culture Media, Conditioned/metabolism , Humans , Insulin Resistance , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Obesity/drug therapy , PPAR gamma/agonists , PPAR gamma/metabolism , RAW 264.7 Cells , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Telmisartan/therapeutic use
14.
Eur J Pharmacol ; 832: 120-128, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-29782861

ABSTRACT

Foenumoside B (FSB), a bioactive component isolated from the Lysimachia foenum-graecum extract (LFE), has been shown to possess anti-inflammatory effects, but the underlying molecular mechanisms involved have not been elucidated. Accordingly, the authors investigated the mechanisms responsible for the anti-inflammatory effects of FSB in murine macrophages activated by LPS. FSB suppressed the LPS-induced expressions of iNOS and COX-2 at protein and mRNA levels and consequently decreased NO and PGE2 production in RAW264.7 and primary macrophages. FSB also reduced the LPS-induced inductions of TNF-α, IL-6 and IL-1ß at protein and mRNA levels. Studies of the molecular mechanisms involved in the anti-inflammatory effects of FSB showed that it inhibited the transcriptional activities of NF-κB and AP-1, and the nuclear translocation of NF-κB via inhibition of the phosphorylations of AKT, p38 and STAT3. In a sepsis model, pretreatment with FSB inhibited the LPS-stimulated mRNA and protein levels of proinflammatory mediators, such as, iNOS, COX-2, TNF-α, IL-6 and IL-1ß in plasma and liver. Importantly, FSB increased the survival rate of mice in the LPS-induced sepsis model. Taken together, these results show that the anti-inflammatory effects of FSB against LPS-induced inflammatory conditions are associated with inhibitions of the phosphorylations of AKT, p38 and STAT3 followed by the transcriptional suppressions of NF-κB and AP-1, and thus, reduced expressions of pro-inflammatory genes.


Subject(s)
Lipopolysaccharides/toxicity , Macrophages/drug effects , NF-kappa B/metabolism , Primulaceae/chemistry , Saponins/pharmacology , Shock, Septic/drug therapy , Transcription Factor AP-1/metabolism , Animals , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Disease Models, Animal , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Saponins/isolation & purification , Saponins/therapeutic use , Shock, Septic/immunology , Shock, Septic/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Sci Rep ; 7(1): 5025, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694473

ABSTRACT

Leukotriene B4 (LTB4) production via the 5-lipoxygenase (5-LO) pathway contributes to the development of insulin resistance in adipose and hepatic tissues, but the role of LTB4 in skeletal muscle is relatively unknown. Here, the authors investigated the role of LTB4 in C2C12 myotubes in palmitic acid (PA)-induced ER stress, inflammation and insulin resistance. PA (750 µM) evoked lipotoxicity (ER stress, oxidative stress, inflammation and insulin resistance) in association with LTB4 production. 5-LO inhibition reduced all the lipotoxic effects induced by PA. On the other hand, PA did not induce cysteinyl leukotrienes (CysLTs), which themselves had no effect on ER stress and inflammation. The beneficial effects of 5-LO suppression from PA-induced lipotoxicity were related with AMPK activation. In ob/ob mice, once daily oral administration of zileuton (50, 100 mg/kg) for 5 weeks improved insulin resistance, increased AMPK phosphorylation, and reduced LTB4 and ER stress marker expression in skeletal muscle. These results show that 5-LO inhibition by either zileuton or 5-LO siRNA protects C2C12 myotubes from PA-induced lipotoxicity, at least partly via AMPK activation, and suggest that the in vivo insulin-sensitizing effects of zileuton are in part attributable to its direct action on skeletal muscle via LTB4 downregulation followed by AMPK activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hydroxyurea/analogs & derivatives , Lipoxygenase Inhibitors/administration & dosage , Muscle Fibers, Skeletal/metabolism , Palmitic Acid/adverse effects , Animals , Arachidonate 5-Lipoxygenase/metabolism , Cell Line , Endoplasmic Reticulum Stress/drug effects , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacology , Insulin Resistance , Leukotriene B4/metabolism , Lipoxygenase Inhibitors/pharmacology , Mice , Muscle Fibers, Skeletal/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects
16.
Chem Biol Interact ; 260: 196-207, 2016 Dec 25.
Article in English | MEDLINE | ID: mdl-27720946

ABSTRACT

Previously, we reported that 6-(3,4-dihydro-1H-isoquinolin-2-yl)-N-(6-methylpyridin-2-yl) nicotinamide (DIMN) analogues inhibited the growth of prostate cancer cells as an anti-androgenic compound. In the present study, we evaluated cytotoxic effects of these DIMN derivatives and found that DIMN-26 most potently inhibited the proliferation of the LNCap-LN3 androgen-dependent and DU145 androgen-independent prostate cancer cells through induction of G2/M phase cell cycle arrest and subsequent apoptosis. The G2/M phase arrest was found due to increases in the activation of cdc2 (also known as cyclin-dependent kinase 1, CDK1)/cyclin B1 complex. DIMN-26 also induced apoptosis in LNCap-LN3 and DU145 prostate cancer cells through activation of caspase-3, -8, and -9, and cleavage of poly(ADP-ribose) polymerase-1 (PARP-1). In addition, DIMN-26 caused the dephosphorylation and mitochondrial accumulation of Bad protein and induced the loss of mitochondria membrane potential, consequently releasing cytochrome c into the cytosol of the cell. Furthermore, overexpression of AKT protein significantly reduced DIMN-26-induced PARP-1 cleavage and p-Bad decrease and cdc2 activation. In addition, DIMN-26 inhibited the 5α-dihydrotestosterone (DHT)-induced cell growth and proliferation and nuclear translocation and transcriptional activities of androgen receptor (AR) in LNCap-LN3 prostate cancer cells. Consistent with these findings, DIMN-26 significantly inhibited the DHT-induced expression of AR-response genes (ARGs), such as prostate-specific antigen (PSA), AR, ß2-microglobulin (B2M), selenoprotein P (SEPP1), and ste20-related proline-alanine-rich kinase (SPAK) in LNCap-LN3 prostate cancer cells. Taken together, these results suggest that DIMN-26 plays a therapeutic role not only in induction of G2/M arrest and apoptosis but also in suppression of androgen receptor signaling in androgen-dependent and androgen-independent prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Down-Regulation/drug effects , Isoquinolines/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Androgens/pharmacology , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoquinolines/chemistry , Isoquinolines/therapeutic use , Male , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
17.
J Pharmacol Exp Ther ; 358(1): 3-13, 2016 07.
Article in English | MEDLINE | ID: mdl-27189969

ABSTRACT

Berberine, a major isoquinoline alkaloid found in medicinal herbs, has been reported to possess anti-inflammatory effects; however, the underlying mechanisms responsible for its actions are poorly understood. In the present study, we investigated the inhibitory effects of berberine and the molecular mechanisms involved in lipopolysaccharide (LPS)-treated RAW 264.7 and THP-1 macrophages and its effects in LPS-induced septic shock in mice. In both macrophage cell types, berberine inhibited the LPS-induced nitric oxide (NO) production and inducible NO synthase (iNOS) protein expression, but it had no effect on iNOS mRNA transcription. Suppression of LPS-induced iNOS protein expression by berberine occurred via a human antigen R (HuR)-mediated reduction of iNOS mRNA stability. Molecular data revealed that the suppression on the LPS-induced HuR binding to iNOS mRNA by berberine was accompanied by a reduction in nucleocytoplasmic HuR shuttling. Pretreatment with berberine reduced LPS-induced iNOS protein expression and the cytoplasmic translocation of HuR in liver tissues and increased the survival rate of mice with LPS-induced endotoxemia. These results show that the suppression of iNOS protein expression by berberine under LPS-induced inflammatory conditions is associated with a reduction in iNOS mRNA stability resulting from inhibition of the cytoplasmic translocation of HuR.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Berberine/pharmacology , ELAV-Like Protein 1/metabolism , Macrophages/drug effects , Nitric Oxide Synthase Type II/biosynthesis , RNA Stability/drug effects , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Berberine/administration & dosage , Berberine/therapeutic use , Cell Line , ELAV-Like Protein 1/genetics , Gene Knockdown Techniques , Humans , Immunohistochemistry , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Nitric Oxide Synthase Type II/genetics , Protein Binding , Real-Time Polymerase Chain Reaction , Shock, Septic/enzymology , Shock, Septic/prevention & control
18.
PLoS One ; 11(5): e0155432, 2016.
Article in English | MEDLINE | ID: mdl-27176632

ABSTRACT

Lysimachia foenum-graecum extract (LFE) and its active component foenumoside B (FSB) have been shown to inhibit adipocyte differentiation, but their mechanisms were poorly defined. Here, we investigated the molecular mechanisms responsible for their anti-adipogenic effects. Both LFE and FSB inhibited the differentiation of 3T3-L1 preadipocytes induced by peroxisome proliferator-activated receptor-γ (PPARγ) agonists, accompanied by reductions in the expressions of the lipogenic genes aP2, CD36, and FAS. Moreover, LFE and FSB inhibited PPARγ transactivation activity with IC50s of 22.5 µg/ml and 7.63 µg/ml, respectively, and showed selectivity against PPARα and PPARδ. Rosiglitazone-induced interaction between PPARγ ligand binding domain (LBD) and coactivator SRC-1 was blocked by LFE or FSB, whereas reduced NCoR-1 binding to PPARγ by rosiglitazone was reversed in the presence of LFE or FSB. In vivo administration of LFE into either ob/ob mice or KKAy mice reduced body weights, and levels of PPARγ and C/EBPα in fat tissues. Furthermore, insulin resistance was ameliorated by LFE treatment, with reduced adipose tissue inflammation and hepatic steatosis. Thus, LFE and FSB were found to act as PPARγ antagonists that improve insulin sensitivity and metabolic profiles. We propose that LFE and its active component FSB offer a new therapeutic strategy for metabolic disorders including obesity and insulin resistance.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Cell Differentiation/drug effects , PPAR gamma/antagonists & inhibitors , Primulaceae/chemistry , Saponins/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Carrier Proteins , Cell Line , Humans , Male , Mice , Models, Molecular , Molecular Conformation , PPAR gamma/chemistry , PPAR gamma/metabolism , Protein Binding , Saponins/chemistry
19.
Cell Signal ; 28(8): 788-97, 2016 08.
Article in English | MEDLINE | ID: mdl-27049873

ABSTRACT

Bortezomib is an anti-cancer agent that induces ER stress by inhibiting proteasomal degradation. However, the effects of bortezomib appear to be dependent on its concentration and cellular context. Since ER stress is closely related to type 2 diabetes, the authors examined the effects of bortezomib on palmitic acid (PA)-induced ER stress in C2C12 murine myotubes. At low concentrations (<20nM), bortezomib protected myotubes from PA (750µM)-induced ER stress and inflammation. Either tunicamycin or thapsigargin-induced ER stress was also reduced by bortezomib. In addition, reduced glucose uptake and Akt phosphorylation induced by PA were prevented by co-treating bortezomib (10nM) both in the presence or absence of insulin. These protective effects of bortezomib were found to be associated with reduced JNK phosphorylation. Furthermore, bortezomib-induced AMPK phosphorylation, and the protective effects of bortezomib were diminished by AMPK knockdown, suggesting that AMPK activation underlies the effects of bortezomib. The in vivo administration of bortezomib at nontoxic levels (at 50 or 200µg/kg, i.p.) twice weekly for 5weeks to ob/ob mice improved insulin resistance, increased AMPK phosphorylation, reduced ER stress marker levels, and JNK inhibition in skeletal muscle. The study shows that bortezomib reduces ER stress, inflammation, and insulin resistance in vitro and in vivo, and suggests that bortezomib has novel applications for the treatment of metabolic disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bortezomib/pharmacology , Endoplasmic Reticulum Stress/drug effects , Inflammation/pathology , Insulin Resistance , Muscle Fibers, Skeletal/enzymology , Palmitic Acid/toxicity , Animals , Cell Line , Cytoprotection/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Male , Mice, Obese , Models, Biological , Muscle Fibers, Skeletal/drug effects , Thapsigargin/pharmacology , Tunicamycin/pharmacology
20.
PLoS One ; 10(9): e0139093, 2015.
Article in English | MEDLINE | ID: mdl-26418009

ABSTRACT

Rosiglitazone is a well-known anti-diabetic drug that increases insulin sensitivity via peroxisome proliferator-activated receptor γ (PPARγ) activation, but unfortunately it causes bone loss in animals and humans. A previous study showed that prolyl hydroxylase domain protein (PHD) plays a role in rosiglitazone-induced adipocyte differentiation. Based on the inverse relationship between adipocyte and osteoblast differentiation, we investigated whether PHD is involved in the effects of rosiglitazone on osteoblast differentiation. Rosiglitazone inhibited osteoblast differentiation in a concentration-dependent manner, and in parallel induced three PHD isoforms (PHD1, 2, and 3). PHD inhibitors and knockdown of each isoform prevented the inhibitory effects of rosiglitazone on osteoblast differentiation and increased the expression of Runx2, a transcription factor essential for osteoblastogenesis. MG-132, a proteasomal inhibitor also prevented the rosiglitazone-induced degradation of Runx2. Furthermore, both increased PHD isoform expressions and reduced osteoblast differentiation by rosiglitazone were prevented by PPARγ antagonists, indicating these effects were mediated via PPARγ activation. In vivo oral administration of rosiglitazone to female ICR mice for 8 weeks reduced bone mineral densities and plasma alkaline phosphatase (ALP) activity, and increased PHD expression in femoral primary bone marrow cells and the ubiquitination of Runx2. Together, this suggests that the rosiglitazone-induced suppression of osteoblast differentiation is at least partly induced via PPARγ-mediated PHD induction and subsequent promotion of the ubiquitination and degradation of Runx2.


Subject(s)
Cell Differentiation/drug effects , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Osteoblasts/cytology , Osteogenesis/drug effects , Procollagen-Proline Dioxygenase/metabolism , Thiazolidinediones/pharmacology , Animals , Blotting, Western , Cells, Cultured , Female , Hypoglycemic Agents/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Immunoenzyme Techniques , Mice , Mice, Inbred ICR , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteogenesis/physiology , PPAR gamma/agonists , Procollagen-Proline Dioxygenase/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone
SELECTION OF CITATIONS
SEARCH DETAIL
...