Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Int Immunopharmacol ; 99: 107624, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34343939

ABSTRACT

Hepatocellular carcinoma (HCC) is an inflammation-induced and chemotherapy-resistant common liver cancer, and a major cause of death. Some natural products have been found to be used as photosensitizers in photodynamic therapy of HCC. Due to its specific molecular structure diversities and biological activities, current status of HCC treatment with nature production remains unsatisfactory, owing largely to the toxicity, side effect and inefficiency to drug targeting. Herein, we show a nanoparticle-based broad-spectrum anti-inflammatory strategy that naïve neutrophil membrane-coated PLGA nanoparticles (NM-HB NPs) were constructed for synchronous nearinfrared fluorescence (NIR FL) imaging and photodynamic therapy (PDT) for HCC. Moreover, NM-HB NPs inhibited the expression of JUNB and promoted the ROS production. JUNB depletion enhanced the anti-HCC effect of NM-HB NPs. Importantly, it was shown that NM-HB NPs are well targeted to the tumor site and overcomes the blood circulation and immune elimination in vivo and vitro. In a mouse model of HCC, the neutrophil membrane-coated nanoparticles (NM-HB NPs) show significant therapeutic efficacy by PDT and suppressing tumor tissue increase. All results demonstrated that NM coated HB NPs representing a viable and effective treatment option for HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cell Membrane , Liver Neoplasms/drug therapy , Nanoparticles/administration & dosage , Neutrophils , Perylene/analogs & derivatives , Quinones/administration & dosage , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cell Line , Humans , Interleukin-6/blood , Interleukin-6/genetics , Liver Neoplasms/blood , Liver Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Nuclear Export Signals , Perylene/administration & dosage , Photochemotherapy , Rats , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
2.
Pharm Biol ; 58(1): 686-694, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32658590

ABSTRACT

CONTEXT: Hydroxysafflor yellow A (HSYA) has been shown to have neuroprotective effects in cerebral infarction. However, its underlying roles in apoptosis and inflammation in Parkinson's disease (PD) are unknown. OBJECTIVE: The present study investigates the effects and underlying mechanisms of HSYA on dopaminergic (DA) neurodegeneration, inflammation, and apoptosis. MATERIALS AND METHODS: The PD model was established by 2 µL of 6-hyroxydopamine (6-OHDA) (3 µg/µL) striatal injection in C57BL/6J mice with different doses of HSYA (2, 4, or 8 mg/kg). In vitro, after being treated with HSYA for 1 h, SH-SY5Y cells were exposed to 6-OHDA for 24 h before analysis. Expression of tyrosine hydroxylase (TH) in substantia nigra (SN) and corpus striatum (STR) was evaluated by immunohistochemistry (IHC) and western blot. In addition, apoptosis-related and inflammatory proteins were examined by western blot. RESULTS: Administration of HSYA significantly reduced the Apomorphine (APO)-induced rotation, decreased from 122.5 ± 15.1 (6-OHDA group) to 47.2 ± 14.3 (8 mg/kg HSYA group). HSYA partially restored a deficit in the SN and STR of PD mice brains in TH. Furthermore, western blot analysis revealed that HSYA reduced inflammatory proteins, including iNOS, COX-2 and NF-κB and attenuated the elevation of DA neuronal apoptosis observed in PD. In vitro assays showed that HSYA reduced the levels of p-p38 and p-JNK and increased that of p-ERK in 6-OHDA-leisoned SH-SY5Y cells. CONCLUSIONS: These findings indicate that HSYA protects against 6-OHDA induced DA neurodegeneration partly by regulating the MAPK inflammatory signalling pathway and apoptosis which highlight its therapeutic potential in the treatment of PD.


Subject(s)
Chalcone/analogs & derivatives , Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Quinones/pharmacology , Animals , Apoptosis/drug effects , Chalcone/administration & dosage , Chalcone/pharmacology , Corpus Striatum/drug effects , Dopamine/metabolism , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Oxidopamine/toxicity , Parkinsonian Disorders/physiopathology , Quinones/administration & dosage , Signal Transduction/drug effects
3.
ACS Appl Mater Interfaces ; 11(48): 44989-44998, 2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31755268

ABSTRACT

Tumor hypoxia severely limits the therapeutic efficacy of solid tumors in photodynamic therapy. One strategy is to develop photosensitizers with simultaneously high efficiency in photodynamic (PDT) and photothermal therapies (PTT) in a single natural-origin phototheranostic agent to overcome this problem. However, less attention has been paid to the natural-origin phototheranostic agent with high PDT and PTT efficiencies even though they have negligible side effects and are environmentally sustainable in comparison with many reported phototheranostic agents. In addition, almost all clinical applied photosensitizers are of natural origin so far. Herein, we synthesized a natural product-based hypocrellin derivative (AETHB), with a high singlet oxygen quantum yield of 0.64 as an efficient photosensitizer different from commercially available porphyrin-based photosensitizers. AETHB is further assembled with human serum albumin to construct nanoparticles (HSA-AETHB NPs) with a high photothermal conversion efficiency (more than 50%). As-prepared HSA-AETHB NPs have shown good water solubility and biocompatibility, pH and light stability, wide absorption (400-750 nm), and NIR emission centered at 710 nm. More importantly, HSA-AETHB NPs can be applied for fluorescent/photoacoustic dual-mode imaging and simultaneously highly efficient PDT/PTT in hypoxic solid tumors. Therefore, this natural-origin multifunctional phototheranostic agent is showing very promising for effective, precise, and safe cancer therapy in clinical applications.


Subject(s)
Hyperthermia, Induced , Neoplasms/therapy , Perylene/analogs & derivatives , Photochemotherapy , Quinones/chemistry , Serum Albumin/chemistry , Animals , Cell Line, Tumor , Female , Humans , Infrared Rays , Mice , Mice, Nude , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Perylene/administration & dosage , Perylene/chemistry , Phenol , Quinones/administration & dosage , Theranostic Nanomedicine
4.
Molecules ; 24(15)2019 Jul 25.
Article in English | MEDLINE | ID: mdl-31349553

ABSTRACT

In a recent study, we described the neuroprotective properties of VCE-003.2-an aminoquinone derivative of the non-psychotropic phytocannabinoid cannabigerol (CBG)-administered intraperitoneally (i.p.) in an inflammatory model of Parkinson's disease (PD). We also demonstrated that these properties derive from its activity on the peroxisome proliferator-activated receptor-γ, in particular at a regulatory site within this receptor type. In the present study, we wanted to further confirm this neuroprotective potential using an oral lipid formulation of VCE-003.2, developed to facilitate the clinical development of this phytocannabinoid derivative. To this end, we evaluated VCE-003.2, administered orally at two doses (10 and 20 mg/kg), to mice subjected to unilateral intrastriatal injections of lipopolysaccharide (LPS), a classic model of inflammation-driven neuronal deterioration that recapitulates characteristics of PD. The administration of VCE-003.2 to these mice showed, as expected, poor activity in the different motor tests (rotarod, computer-aided actimeter) used in experimental parkinsonism, in general due to the lack of evident changes in these behaviors by LPS lesion. However, VCE-003.2, at 20 mg/kg, was highly active in improving the changes detected in LPS-lesioned mice in the cylinder rearing test. In addition, the histopathological analysis of the basal ganglia revealed a trend towards recovery at 20 mg/kg VCE-003.2 in the loss of tyrosine hydroxylase-containing nigrostriatal neurons, as well as a complete reduction in the elevated LAMP-1 immunolabeling (reflecting autophagy impairment) caused by LPS lesion. These effects were not seen at 10 mg/kg. This was associated with a partial reduction in the intense glial reactivity provoked by LPS in the substantia nigra, in particular the astroglial reactivity labeled with glial fibrillary acidic protein. The analysis using qPCR in the striatum of proinflammatory mediators, such as tumor necrosis factor-α, interleukin-1ß, inducible nitric oxide synthase, and cyclooxygenase-2, showed that the marked elevations provoked by the LPS lesion tended to be, in general, attenuated by VCE-003.2 treatment, with the greatest effects normally found with the highest dose of 20 mg/kg. In summary, our data confirm the neuroprotective potential of an oral formulation of VCE-003.2 against neuronal injury in an in vivo model of PD based on neuroinflammation, and this study opens the possibility to further the development of oral VCE-003.2 in the clinic.


Subject(s)
Cannabinoids/pharmacology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , PPAR gamma/agonists , Quinones/pharmacology , Administration, Oral , Animals , Biomarkers , Cannabinoids/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Female , Immunohistochemistry , Inflammation Mediators , Mice , Neurons/pathology , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders , Quinones/administration & dosage , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
5.
Mar Drugs ; 17(3)2019 Mar 17.
Article in English | MEDLINE | ID: mdl-30884884

ABSTRACT

An ilimquinone (IQ) mixture isolated from Hippiospongia metachromia, consisting of IQ and epi-ilimaquinone (epi-IQ), exerts anti-HIV, anti-microbial, anti-inflammatory, and anti-cancer effects. An HPLC-MS/MS method was developed for simultaneous determination of the two epimers in rat plasma, separating them using a biphenyl column. Ascorbic acid is added during the sample preparation to ensure the stability of both isomers. The plasma concentrations of the isomers were monitored following intravenous and oral administration of the IQ mixture in rats as well as the individual epimers that were separately orally administered. Compare to IQ, epi-IQ was much more stable in rat plasma, likely due to its configurations of decalin. Both substances decayed in more than bi-exponential pattern, with an elimination rate constant of 1.2 h-1 for IQ and 1.7 h-1 for epi-IQ. The epi-IQ was distributed more widely than IQ by about two-fold. Consequently, the clearance of epi-IQ was greater than that of IQ by about three-fold. The oral absolute bioavailability for IQ was 38%, and, that for epi-IQ, was 13%. Although the systemic exposure of IQ was greater than that of epi-IQ by ~8.7-fold, the clearance of each isomer was similar when administered either orally or intravenously, when normalized for bioavailability. The stereo-specific behavior of the isomers appears to originate from differences in both their tissue distribution and gastrointestinal permeability.


Subject(s)
Porifera/chemistry , Quinones/chemistry , Quinones/pharmacokinetics , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacokinetics , Animals , Biological Availability , Chromatography, High Pressure Liquid/methods , Isomerism , Male , Quinones/administration & dosage , Quinones/blood , Rats , Rats, Sprague-Dawley , Sesquiterpenes/administration & dosage , Sesquiterpenes/blood , Tandem Mass Spectrometry/methods
6.
J Pharm Biomed Anal ; 166: 291-294, 2019 Mar 20.
Article in English | MEDLINE | ID: mdl-30684930

ABSTRACT

Ilimaquinone, a metabolite isolated from the marine sponge Hippiospongia metachromia, has antimicrobial, cytotoxic, anti-HIV, anti-inflammatory, and anti-cancer activities. A new quantitative analytical method for determination of ilimaquinone in rat plasma using HPLC-MS/MS was developed and validated. Ascorbic acid was added to ensure the stability of ilimaquinone in plasma. After protein precipitation using acetonitrile plus diclofenac as an internal standard, the analytes were chromatographed on a biphenyl column with a mobile phase of methanol and water (8:2, v/v, including 0.1% formic acid). This method was successfully applied in a pharmacokinetic study of ilimaquinone after oral administration in rats.


Subject(s)
Quinones/pharmacokinetics , Sesquiterpenes/pharmacokinetics , Administration, Oral , Animals , Chromatography, High Pressure Liquid , Drug Stability , Porifera/chemistry , Quinones/administration & dosage , Quinones/blood , Quinones/isolation & purification , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Sesquiterpenes/administration & dosage , Sesquiterpenes/blood , Sesquiterpenes/isolation & purification , Tandem Mass Spectrometry
7.
Drug Deliv ; 25(1): 1779-1789, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30338719

ABSTRACT

Nonhealing chronic wounds on foot induced by diabetes is a complicated pathologic process. They are mainly caused by impaired neovascularization, neuropathy, and excessive inflammation. A strategy, which can accelerate the vessel network formation as well as inhibit inflammatory response at the same time, makes it possible for effective diabetic ulcers treatment. Co-delivery of multiple drugs with complementary bioactivity offers a strategy to properly treat diabetic wound. We previously demonstrated that hydroxysafflor yellow A (HSYA) could accelerate diabetic wound healing through promoting angiogenesis and reducing inflammatory response. In order to further enhance blood vessel formation, a pro-angiogenic molecular called deferoxamine (DFO) was topically co-administrated with HSYA. The in vitro results showed that the combination of DFO and HSYA exerted synergistic effect on enhancing angiogenesis by upregulation of hypoxia inducible factor-1 alpha (HIF-1α) expression. The interpenetrating polymer networks hydrogels, characterized by good breathability and water absorption, were designed for co-loading of DFO and HSYA aiming to recruit angiogenesis relative cells and upgrade wound healing in vivo. Both DFO and HSYA in hydrogel have achieved sustained release. The in vivo studies indicated that HSYA/DFO hydrogel could accelerate diabetic wound healing. With a high expression of Hif-1α which is similar to that of normal tissue. The noninvasive US/PA imaging revealed that the wound could be recovered completely with abundant blood perfusion in dermis after given HSYA/DFO hydrogel for 28 days. In conclusion, combination of pro-angiogenic small molecule DFO and HSYA in hydrogel provides a promising strategy to productively promote diabetic wound healing as well as better the repair quality.


Subject(s)
Chalcone/analogs & derivatives , Deferoxamine/administration & dosage , Drug Delivery Systems , Neovascularization, Physiologic/drug effects , Quinones/administration & dosage , Siderophores/administration & dosage , Wound Healing/drug effects , Administration, Topical , Animals , Chalcone/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Drug Therapy, Combination , Humans , Male , Rats, Sprague-Dawley
8.
Food Funct ; 9(8): 4443-4456, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-30070297

ABSTRACT

The inflammatory environment has been demonstrated to be strongly associated with the progression of osteoarthritis (OA). HSYA, the main active component in the medical and edible dual purpose plant safflower, has previously showed significant anti-inflammatory effects in several diseases. In the current study, the protective effects of HSYA in the inhibition of OA development and its underlying mechanism were examined by both in vitro and in vivo experiments. Our data indicated that interleukin-1 beta (IL-1ß) induced over-production of pro-inflammatory cytokines, such as nitric oxide (NO) and prostaglandin E2 (PGE2); also, the expression of cyclooxygenase-2 (COX-2), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS) were all inhibited by pretreatment with HSYA in a dose-dependent manner (2.5 to 40 µM). Furthermore, HSYA attenuated IL-1ß-induced degradation of the extracellular matrix (ECM) by decreasing the expression of metalloproteinases (MMPs) and thrombospondin motifs 5 (ADAMTS5). Mechanistically, HSYA suppressed IL-1ß-induced activation of the nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) cascades. Meanwhile, molecular docking studies revealed that HSYA has excellent binding abilities to p65, extracellular signal-regulated kinase (ERK), p38 and c-Jun N-terminal kinase (JNK). In addition, the protective effects of HSYA were observed in a surgically induced mouse OA model. In summary, this study provides evidence that HSYA can be applied as a potential therapeutic agent in the treatment of OA.


Subject(s)
Chalcone/analogs & derivatives , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Osteoarthritis/prevention & control , Quinones/administration & dosage , ADAMTS5 Protein/genetics , ADAMTS5 Protein/metabolism , Animals , Chalcone/administration & dosage , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Biochem Pharmacol ; 157: 304-313, 2018 11.
Article in English | MEDLINE | ID: mdl-30076848

ABSTRACT

Systemic sclerosis (SSc) or scleroderma is a chronic multi-organ autoimmune disease characterized by vascular, immunological, and fibrotic abnormalities. The etiology of SSc is unknown, but there is growing evidence that dysfunction of the endocannabinoid system (ECS) plays a critical role in its development. Since the semi-synthetic cannabinoquinoid VCE-004.8 could alleviate bleomycin (BLM)-induced skin fibrosis, we have investigated an oral lipid formulation (EHP-101) of this dual PPARγ/CB2 receptors activator for the prevention of skin- and lung fibrosis and of collagen accumulation in BLM challenged mice. Immunohistochemistry analysis of the skin showed that EHP-101 could prevent macrophage infiltration as well as the expression of Tenascin C (TNC), vascular cell adhesion molecule 1 (VCAM1), and the α-smooth muscle actin (SMA). EHP-101 could also prevent the reduced expression of vascular CD31 typical of skin fibrosis. RNAseq analysis of skin biopsies showed a clear effect of EHP-101 in the inflammatory and epithelial-mesenchymal transition transcriptomic signatures. TGF-ß-regulated genes [matrix metalloproteinase-3 (Mmp3), cytochrome b-245 heavy chain (Cybb), lymphocyte antigen 6E (Ly6e), vascular cell adhesion molecule-1 (Vcam1) and Integrin alpha-5 (Itga5)] were induced in BLM mice and repressed by EHP-101 treatment. By intersecting differentially expressed genes in EHP-101-treated mice with a dataset of human scleroderma intrinsic genes, 53 overlapped genes were discovered, including biomarkers of SSc like the C-C motif chemokine 2 (Ccl2) and the interleukin 13 receptor subunit alpha 1 (IL-13Ra1) genes. Taken together, these data provide a rationale for further developing VCE-004.8 as an orally active agent to alleviate scleroderma and, possibly, other fibrotic diseases as well.


Subject(s)
Cannabidiol/analogs & derivatives , Lung/pathology , Skin/pathology , Administration, Oral , Animals , Bleomycin , Blood Vessels/drug effects , Collagen/analysis , Female , Fibrosis , Lung/drug effects , Mice, Inbred BALB C , Phenotype , Quinones/administration & dosage , Quinones/therapeutic use , Skin/blood supply , Skin/drug effects , Skin/metabolism , Transcriptome/drug effects
10.
Pharmacol Res ; 134: 40-50, 2018 08.
Article in English | MEDLINE | ID: mdl-29787870

ABSTRACT

Given the high and increasing prevalence of obesity, the safe and effective treatment of obesity would be beneficial. Here, we examined whether oral hydroxysafflor yellow A (HSYA), an active compound from the dried florets of Carthamus tinctorius L., can reduce high-fat (HF) diet-induced obesity in C57BL/6 J mice. Our results showed that the average body weight of HF group treated by HSYA was significantly lower than that of the HF group (P < 0.01). HSYA also reduced fat accumulation, ameliorated insulin resistance, restored glucose homeostasis, reduced inflammation, enhanced intestinal integrity, and increased short-chain fatty acids (SCFAs) production in HF diet-fed mice. Sequencing of 16S rRNA genes in fecal samples demonstrated that HSYA reversed HF diet induced gut microbiota dysbiosis. Particularly, HSYA increased the relative abundances of genera Akkermansia and Romboutsia, as well as SCFAs-producing bacteria, including genera Butyricimonas and Alloprevotella, whereas it decreased the phyla Firmicutes/Bacteroidetes ratio of HF diet-fed mice. Additionally, serum metabolomics analysis revealed that HSYA increased lysophosphatidylcholines (lysoPCs), L-carnitine and sphingomyelin, and decreased phosphatidylcholines in mice fed a HF diet, as compared to HF group. These changed metabolites were mainly linked with the pathways of glycerophospholipid metabolism and sphingolipid metabolism. Spearman's correlation analysis further revealed that Firmicutes was positively while Bacteroidetes and Akkermansia were negatively correlated with body weight, fasting serum glucose and insulin. Moreover, Akkermansia and Butyricimonas had positive correlations with lysoPCs, suggestive of the role of gut microbiota in serum metabolites. Our findings suggest HSYA may be a potential therapeutic drug for obesity and the gut microbiota may be potential territory for targeting of HSYA.


Subject(s)
Anti-Obesity Agents/administration & dosage , Bacteria/drug effects , Chalcone/analogs & derivatives , Energy Metabolism/drug effects , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Obesity/prevention & control , Quinones/administration & dosage , Weight Loss/drug effects , Adiposity/drug effects , Administration, Oral , Animals , Bacteria/growth & development , Bacteria/metabolism , Biomarkers/blood , Blood Glucose/metabolism , Chalcone/administration & dosage , Diet, High-Fat , Disease Models, Animal , Fatty Acids, Volatile/blood , Insulin/blood , Insulin Resistance , Intestines/microbiology , Male , Metabolomics/methods , Mice, Inbred C57BL , Obesity/blood , Obesity/microbiology , Obesity/physiopathology
11.
Cancer Sci ; 109(6): 1958-1969, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29617063

ABSTRACT

Photodynamic therapy (PDT) is considered an innovative and attractive modality to treat ovarian cancer. In the present study, a biodegradable polymer poly (ethylene glycol) (PEG)-poly (lactic acid)(PLA)-folate (FA-PEG-PLA) was prepared in order to synthesize an active-targeting, water-soluble and pharmacomodulated photosensitizer nanocarrier. Drug-loading content, encapsulation efficiency, in vitro and in vivo release were characterized, in which hypocrellin B (HB)/FA-PEG-PLA micelles had a high encapsulation efficiency and much slower control release for drugs compared to free drugs (P < .05). To evaluate the targeting ability of the HB/FA-PEG-PLA micelles, a cellular uptake study in vitro was carried out, which showed significantly enhanced uptake of HB/FA-PEG-PLA micelles in SKOV3 (FR+) compared to A2780 cancer cells (FR-). The enhanced uptake of HB/FA-PEG-PLA micelles to cancer cells resulted in a more effective post-PDT killing of SKOV3 cells compared to plain micelles and free drugs. Binding and uptake of HB/FA-PEG-PLA micelles by SKOV3 cells were also observed in vivo after ip injection of folate-targeted micelles in tumor-bearing ascitic ovarian cancer animals. Drug levels in ascitic tumor tissues were increased 20-fold (P < .001), which underscored the effect of a regional therapy approach with folate targeting. Furthermore, the HB-loaded micelles were mainly distributed in kidney and liver (the main clearance organs) in biodistribution. These results showed that our newly developed PDT photosensitizer HB/FA-PEG-PLA micelles have a high drug-loading capacity, good biocompatibility, controlled drug release, and enhanced targeting and antitumor effect, which is a potential approach to future targeting ovarian cancer therapy.


Subject(s)
Micelles , Ovarian Neoplasms/therapy , Perylene/analogs & derivatives , Polymers/chemistry , Quinones/administration & dosage , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Drug Carriers/chemistry , Drug Liberation , Female , Folic Acid/analogs & derivatives , Folic Acid/chemistry , Humans , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Perylene/administration & dosage , Perylene/chemistry , Perylene/pharmacokinetics , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Polyesters/chemistry , Polyethylene Glycols/chemistry , Quinones/chemistry , Quinones/pharmacokinetics , Rats, Sprague-Dawley , Tissue Distribution
12.
Nanomedicine ; 14(5): 1643-1654, 2018 07.
Article in English | MEDLINE | ID: mdl-29689372

ABSTRACT

In this work, a theranostic nanocage system was developed for the targeted delivery of the anti-cancer agents camptothecin (CPT) and luotonin A (LuA). The core of the nanocage system (Fe3O4@OA-AD-SP NCs) was formed by biogenically synthesized Fe3O4 nanoparticles (NPs) decorated with a model anti-cancer drug (AD) and biosurfactant saponin (SP). The Fe3O4@OA-AD-SP NCs showed a high lipophilic AD loading efficiency (>80%) and a controlled pH-responsive drug release in stimulated cancerous cells in pH 6.4 media buffer. In addition, Fe3O4@OA-AD-SP NCs exhibited better serum protein binding efficacy at physiological pH values (7.4), furthering the important role of SP surface decoration. Particularly, these NCs showed better chemotherapeutic efficacy when examined in MCF-7 and HeLa cancer cell lines with a specific targeting capacity. Therefore, this study provides a new nano platform based on magnetic targeting and pH responsive lipophilic anticancer drug delivery to the cancer site.


Subject(s)
Camptothecin/pharmacology , Ferric Compounds/chemistry , Magnetic Fields , Nanocomposites/administration & dosage , Neoplasms/drug therapy , Pyrroles/pharmacology , Quinones/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/administration & dosage , Camptothecin/chemistry , Cell Proliferation/drug effects , Drug Delivery Systems , HeLa Cells , Humans , MCF-7 Cells , Nanocomposites/chemistry , Pyrroles/administration & dosage , Pyrroles/chemistry , Quinones/administration & dosage , Quinones/chemistry , Theranostic Nanomedicine
13.
Eur J Pharmacol ; 823: 72-78, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29408092

ABSTRACT

To investigate the effects of Hydroxysafflor Yellow A (HSYA), which is derived from safflower, on the proliferation, migration and angiogenesis of cells in vitro and its potential efficacy in vivo when topically applied to a diabetic wound. Human umbilical vein endothelial cells (HUVECs) and mouse macrophage cells (RAW264.7) were used to evaluate angiogenesis and anti-inflammatory activities, respectively. The influence of HSYA on the wound scratch assay was investigated in keratinocytes. A splinted excisional wound model in rats with TIDM induced by streptozotocin was used to assess the effects of wound healing. Collagen disposition and secretion of vascular growth factors (VEGF) as well as transforming growth factor-ß1 (TGF-ß1) were evaluated by an ELISA assay and histological staining. The in vitro results showed that HSYA could significantly enhance both the neovascularization of HUVECs and the migration of keratinocytes. It showed the significant inhibitory effect on nitric oxide production, indicating the anti-inflammatory activity of HSYA. In vivo, the topical application of HSYA significantly enhanced the wound closure rate, and the time to complete wound closure was 17 days, whereas 30 days were needed with PBS treatment. Further, treatment with HSYA exhibited significant granulation tissue formation with higher collagen content, re-epithelialization and angiogenesis according to Masson's trichrome staining evaluation, VEGE and TGF-ß1 ELISA measurement. In conclusion, HSYA application could be considered a promising therapeutic strategy for treating chronic non-healing diabetic foot ulcers.


Subject(s)
Chalcone/analogs & derivatives , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Quinones/administration & dosage , Quinones/pharmacology , Wound Healing/drug effects , Administration, Topical , Animals , Chalcone/administration & dosage , Chalcone/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kinetics , Male , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Curr Drug Deliv ; 15(7): 1009-1019, 2018.
Article in English | MEDLINE | ID: mdl-29268684

ABSTRACT

OBJECTIVE: The aim of this study was to develop a novel W/O microemulsion for a natural extract of Wen-Luo-Tong (WLT) containing mainly icariin, hydroxysafflor yellow A (HSYA) and gallic acid to be applied to skin as a potential treatment for peripheral neuropathy. METHODS: The oil phase was selected on the basis of affinity with the surfactant and co-surfactant. Pseudo-ternary diagrams were constructed to optimize microemulsions and finally stability studies were performed on the selected formulations. Droplet sizes were analyzed by using a zetasizer and were found to be within the desired range. Selected microemulsions with acceptable viscosities, containing 5%, 8% and 10% of water extract solution, were used for in vitro skin penetration studies using Franz diffusion cells and excised rat skin. New LC-MS/MS and UPLC-Q-TOF/MS methods were employed for quantitative and qualitative analysis. RESULTS: The optimized formulation (ME-4) consisting of 10% (w/w) water extract solution, 60% isopropyl myristate, 30%(w/w) Smix: Propylene glycol (5:2) significantly increased the cumulative permeated amounts of HSYA, icariin and gallic acid compared with the water extract solution controls. CONCLUSION: This novel formulation also increased the number of components penetrating rat skin. Ten components were detected in the Franz cell receptor solution using a UPLC-Q-TOF/MS system after the application of formulation ME-4 for 24h on the skin in vitro. However, only one component was detected after applying the control. Therefore, the microemulsion ME-4 was selected for future in vivo pharmacodynamic studies.


Subject(s)
Emulsions , Skin Absorption , Administration, Cutaneous , Animals , Chalcone/administration & dosage , Chalcone/analogs & derivatives , Chalcone/chemistry , Chalcone/pharmacokinetics , Chromatography, High Pressure Liquid , Emulsions/administration & dosage , Emulsions/chemistry , Emulsions/pharmacokinetics , Flavonoids/administration & dosage , Flavonoids/chemistry , Flavonoids/pharmacokinetics , Gallic Acid/administration & dosage , Gallic Acid/chemistry , Gallic Acid/pharmacokinetics , Male , Molecular Weight , Myristates/administration & dosage , Myristates/chemistry , Myristates/pharmacokinetics , Permeability , Propylene Glycol/administration & dosage , Propylene Glycol/chemistry , Propylene Glycol/pharmacokinetics , Quinones/administration & dosage , Quinones/chemistry , Quinones/pharmacokinetics , Rats, Sprague-Dawley , Skin/metabolism , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacokinetics , Tandem Mass Spectrometry
15.
Int J Pharm ; 535(1-2): 164-171, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29107614

ABSTRACT

Hydroxysafflor yellow A (HSYA) is the main bioactive flavonoid extracted from the flower of Carthamus tinctorius L., which is widely used in traditional Chinese medicine for the treatment of myocardial ischemia and cerebral ischemia. HSYA has high water solubility but poor intestinal membrane permeability, resulting in low oral bioavailability. Currently, only HSYA sodium chloride injection has been approved for clinical use and oral formulations are urgently needed. In this study, HSYA solid lipid nanoparticles (SLNs) with the structure of w/o/w were prepared by a warm microemulsion process using approved drug excipients for oral delivery to increase the oral absorption of HSYA. The optimized HSYA SLNs are spherical with an average size of 214nm and the encapsulation efficiency is 55%. HSYA SLNs exhibited little cytotoxicity in Caco-2 and Hela cells, but increased the oral absorption of HSYA about 3.97-fold in rats, compared to HSYA water solution. In addition, cycloheximide pretreatment significantly decreased the oral absorption of HSYA delivered by SLNs. Importantly, the pharmacodynamics evaluation demonstrated that SLNs further decreased the infarct areas in rats. In conclude, SLNs could be a promising delivery system to enhance the oral absorption and pharmacological activities of HSYA.


Subject(s)
Chalcone/analogs & derivatives , Drug Carriers/chemistry , Flavonoids/administration & dosage , Lipids/chemistry , Nanoparticles/chemistry , Quinones/administration & dosage , Administration, Oral , Animals , Brain Ischemia/drug therapy , Caco-2 Cells , Cell Survival/drug effects , Chalcone/administration & dosage , Chalcone/pharmacology , Chalcone/therapeutic use , Drug Liberation , Flavonoids/pharmacology , Flavonoids/therapeutic use , HeLa Cells , Humans , Male , Medicine, Chinese Traditional , Oral Mucosal Absorption , Quinones/pharmacology , Quinones/therapeutic use , Rats, Sprague-Dawley
16.
Anticancer Res ; 37(8): 4103-4109, 2017 08.
Article in English | MEDLINE | ID: mdl-28739694

ABSTRACT

BACKGROUND/AIM: Kinamycin F is a bacterial metabolite which contains an unusual and potentially reactive diazo group that is known for its ability to inhibit cell growth. In this study, the potential anti-tumor activity of kinamycin F was investigated in three human osteosarcoma cell lines, MG-63, U-2 OS and HOS as an antitumor agent with a potentially novel target. MATERIALS AND METHODS: Proliferation and cell viability were measured in three human osteosarcoma cell lines by commercially available kits. We also evaluated the effects of the drug on cell cycle progression using the Muse™ Cell Analyzer. Caspase-3 activity was determined by a fluorometric EnzChek assay kit. Finally, following treatment with kinamycin F the protein levels of cyclin D3, cyclin A and cdK-2 were examined. RESULTS: Kinamycin F induced a concentration-dependent cell death in all the three cell lines. Flow cytometry revealed that kinamycin F treatment at 1 µM concentration significantly increased the cell population in the G2/M-phase (60-65%). Kinamycin F activated caspase 3 in all the three cell lines, clearly demonstrating that the growth inhibitory effect of kinamycin F can be attributed to apoptosis induction. Finally, kinamycin F suppressed osteosarcoma cell proliferation affecting cyclin A and D3 expression. CONCLUSION: Understanding the mechanism by which kinamycin F exerts its ability to inhibit cell growth may be a step forward in the development of new therapeutic strategies for the treatment of OS.


Subject(s)
Cell Proliferation/drug effects , Osteosarcoma/drug therapy , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , G2 Phase/drug effects , Humans , Osteosarcoma/pathology , Quinones/administration & dosage
17.
J Exp Clin Cancer Res ; 35(1): 149, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27658586

ABSTRACT

BACKGROUND: Hsp90 proteins are important therapeutic targets for many anti-cancer drugs in clinical trials. Geldanamycin (GA) was identified as the first natural inhibitor of Hsp90, increasing evidence suggests that GA was not a good choice for clinical trials. In this study, we investigated two new non-benzoquinone geldanamycin analogs of Hsp90 inhibitors, DHQ3 and 17-demethoxy-reblastatin (17-DR), to explore the molecular mechanisms of their anti-cancer activity in vivo and vitro. METHODS: MTT and colony formation assays were used to measure cell viability. Flow cytometry, DAPI staining, ATP assay, electron microscopy, western blots, siRNAs transfection and immunofluorescence were used to determine the molecular mechanism of DHQ3- or 17-DR-induced different forms of death in human breast cancer MDA-MB-231 cells. Malachite green reagent was used to measure ATPase activity of the analogs. RESULTS: DHQ3 and 17-DR presented efficiently inhibitory effect in MDA-MB-231 cell lines, and DHQ3 induced necroptosis by activation of the RIP1-RIP3-MLKL necroptosis cascade. And DHQ3-induced cell death was inhibited by a necroptosis inhibitor, necrostatin-1 (Nec-1), but not by a caspase inhibitor z-VAD-fmk. On the other hand, 17-DR induced apoptosis in MDA-MB-231 cells, indicating a caspase-dependent killing mechanism. We further demonstrated that down-regulation of RIP1 and RIP3 by siRNA protected against DHQ3 but not 17-DR induced cell death. These results were confirmed by electron microscopy. DHQ3 and 17-DR induced the degradation of Hsp90 client proteins, and they showed strong antitumor effects in MDA-MB-231 cell-xenografted nude mice. CONCLUSIONS: These findings supported that DHQ3 and 17-DR induce different forms of death in some cancer cell line via activation of different pathways. All of the results provided evidence for its anti-tumorigentic action with low hepatotoxicity in vivo, making them promising anti-breast cancer agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzoquinones/administration & dosage , Breast Neoplasms/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/administration & dosage , Quinones/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lactams, Macrocyclic/pharmacology , MCF-7 Cells , Mice , Nuclear Pore Complex Proteins/metabolism , Quinones/pharmacology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
18.
Int J Mol Med ; 38(1): 83-94, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27176815

ABSTRACT

In clinical practice, the traditional Chinese medicinal herbs, Radix Salvia Miltiorrhiza and Carthamus tinctorius L., are usually prescribed in combination due to their significant cardioprotective effects. However, the mechanisms responsible for these combined effects remain unknown. Thus, in this study, we investigated the mechanisms responsible for the combined effects of Danshensu (DSS) and hydroxysafflor yellow A (HSYA) by establishing a rat model of myocardial ischemia/reperfusion (MI/R), as well as a model of hypoxia/reoxygenation (H/R) using H9c2 cells. The combination index (CI) was calculated using the median-effect method. DSS and HSYA in combination led to a CI value of <1 as regards infarct size in vivo and cell viability in vitro. The rats with MI/R injury that were treated with DSS and/or HSYA were found to have significantly lower levels of creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI) and malondialdehyde (MDA), and a lower expressoin of 8-hydroxydeoxyguanosine (8-OHdG), and markedly enhanced superoxide dismutase (SOD) activity. Our in vitro experiments revealed that the cells treated with DSS and/or HSYA had a reduced lactate dehydrogenase (LDH) activity and a decreased percentage of cell apoptosis (increased Bcl-2/Bax ratio, decreased expression of cleaved caspase-3). DSS and HSYA increased the expression of heme oxygenase-1 (HO-1), the phosphorylation of Akt and the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). Furthermore, the Akt inhibitor, LY294002, partially hampered the expression of Nrf2 and HO-1. The HO-1 inhibitor, zinc protoporphyrin IX (ZnPP­IX), did not decrease the expression of p-Akt and Nrf2, although it abolished the anti-apoptotic and antioxidant effects of DSS and HSYA. The findings of our study thus demonstrate that DSS and HSYA confer synergistic cardioprotective effects through the Akt/Nrf2/HO-1 signaling pathway, to certain extent, by enhancing the antioxidant defense system and exerting anti-apoptotic effects.


Subject(s)
Cardiotonic Agents/therapeutic use , Chalcone/analogs & derivatives , Heme Oxygenase-1/metabolism , Lactates/therapeutic use , Myocardial Reperfusion Injury/drug therapy , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinones/therapeutic use , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Caspase 3/metabolism , Cell Line , Chalcone/administration & dosage , Chalcone/pharmacology , Chalcone/therapeutic use , Creatine Kinase, MB Form/metabolism , Drug Synergism , In Situ Nick-End Labeling , Lactates/administration & dosage , Lactates/pharmacology , Malondialdehyde/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protoporphyrins/pharmacology , Quinones/administration & dosage , Quinones/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Troponin I/metabolism , bcl-2-Associated X Protein/metabolism
19.
Anticancer Res ; 36(5): 2105-11, 2016 May.
Article in English | MEDLINE | ID: mdl-27127110

ABSTRACT

AIM: We screened nine 1,2-dihydro-quinolines using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assay in HepG2 and SMMC cells and identified ethyl-2-cyano-2-(2- (methoxycarbonyl)allyl)quinoline-1(2H)-carboxylate (compound 5) as a potential anticancer agent. In order to further develop its anticancer therapeutic potential, we incorporated this agent into cationic liposomes for delivery to tumor cells. The characteristics of liposomes, their cytotoxicity and cellular uptake by tumor cells were investigated. We demonstrated that cationic 1,2-dioleoyl-3- trimethyl-ammonium-propane containing liposomes (cLips) loaded with compound 5 has superior antitumor activity compared to neutral liposomes. These data suggest cLip-compound 5 to be a promising agent that warrants further evaluation.


Subject(s)
Antineoplastic Agents/administration & dosage , Liposomes , Quinones/administration & dosage , Cations , Cell Line, Tumor , Humans , Microscopy, Confocal
20.
J Photochem Photobiol B ; 158: 113-21, 2016 May.
Article in English | MEDLINE | ID: mdl-26967521

ABSTRACT

To increase the therapeutic efficacy of photodynamic therapy (PDT) in treating lung cancer, we developed both photosensitizer and anticancer drug encapsulated hyaluronic acid-ceramide nanoparticles. Based on our previous study, a co-delivery system of photosensitizers and anticancer agents greatly improves the therapeutic effect of PDT. Furthermore, hyaluronic acid-ceramide-based nanoparticles are ideal targeting carriers for lung cancer. In vitro phototoxicity in A549 (human lung adenocarcinoma) cells and in vivo antitumor efficacy in A549 tumor-bearing mice treated with hypocrellin B (HB)-loaded nanoparticles (HB-NPs) or hypocrellin B and paclitaxel loaded nanoparticles (HB-P-NPs) were evaluated. Cell viability assay, microscopic analysis and FACS analysis were performed for the in vitro studies and HB-P-NPs showed enhanced phototoxicity compared with HB-NPs. In the animal study, the tumor volume change and the histological analysis was studied and the anticancer efficacy improved in the order of free HB

Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Ceramides/chemistry , Hyaluronic Acid/chemistry , Lung Neoplasms/drug therapy , Paclitaxel/administration & dosage , Perylene/analogs & derivatives , Photochemotherapy , Quinones/administration & dosage , Animals , Heterografts , Mice , Perylene/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...