Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 722
Filter
Add more filters

Publication year range
1.
Molecules ; 27(4)2022 Feb 20.
Article in English | MEDLINE | ID: mdl-35209214

ABSTRACT

Anti-rheumatoid arthritis (RA) effects of α-tocopherol (α-T) have been shown in human patients in a double-blind trial. However, the effects of α-T and its derivatives on fibroblast-like synoviocytes (FLS) during the pathogenesis of RA remain unclear. In the present study, we compared the expression levels of genes related to RA progression in FLS treated with α-T, succinic ester of α-T (TS), and phosphate ester of α-T (TP), as determined via RT-PCR. The mRNA levels of interleukin (IL)-6, tumor necrosis factor-α (TNF-α), matrix metalloproteinase (MMP)-3, and MMP-13 were reduced by treatment with TP without cytotoxicity, while α-T and TS did not show such effects. Furthermore, intraperitoneal injection of TP ameliorated the edema of the foot and joint and improved the arthritis score in laminarin-induced RA model mice. Therefore, TP exerted anti-RA effects through by inhibiting RA-related gene expression.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Gene Expression Regulation/drug effects , alpha-Tocopherol/analogs & derivatives , Animals , Arthritis, Rheumatoid/chemically induced , Cytokines/biosynthesis , Glucans/toxicity , Humans , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Mice , alpha-Tocopherol/pharmacology
2.
Nano Lett ; 20(4): 2219-2229, 2020 04 08.
Article in English | MEDLINE | ID: mdl-31823615

ABSTRACT

Distal metastases of tumors result from the interaction between "seeds" (circulating tumor cells, CTCs) and "soil" (premetastatic niche, PMN). Various strategies focus on CTC inhibition, but only a few strategies inhibit PMN formation. The main predisposition of PMN formation in melanoma lies in the pulmonary recruitment of granulocytic myeloid-derived suppressor cells (G-MDSCs, CD11b+Ly6G+ cells) induced by tumors, which increase vascular permeability by secreting matrix metalloproteinase-9 (MMP-9) and result in immunosuppression by secreting interleukin-10 (IL-10) in premetastatic lungs. Here, a micellar hypotoxic low molecular weight heparin-tocopherol succinate nanoparticle (LMWH-TOS nanoparticle, LT NP) was established and investigated for its influence on PMN formation in this study. We first demonstrated that the hydrophilic segment LMWH in LT NPs can inhibit early pulmonary recruitment of G-MDSCs through interrupting their extravasation by inhibiting P-selectin/PSGL-1-mediated adhesion between vascular endothelial cells and G-MDSCs. In addition, the hydrophobic segment (TOS) in LT NPs significantly inhibited the expression of MMP-9 in G-MDSCs. As a result, the drug-free nanoparticles could maintain the normal microenvironment of lungs, thus effectively inhibiting implantation and colonization of CTCs. Further, phenylboronic acid (PBA)-modified and doxorubicin/immunopotentiator α-galactosylceramide (αGC)-coloaded nanoparticles (PLT/DOX/αGC NPs) were exploited. PBA modification achieved targeted chemotherapy by binding to overexpressed sialic acid residues on the tumor cell surface. This nanosystem effectively inhibited the postoperative metastasis and tumor recurrence simultaneously. Our work provides a proof of concept that the prevention of PMN formation through interfering G-MDSCs with self-delivery nanosystems is a safe and effective antimetastasis strategy.


Subject(s)
Endothelial Cells/drug effects , Lung Neoplasms/prevention & control , Melanoma, Experimental/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Nanoparticles/therapeutic use , Neoplastic Cells, Circulating/drug effects , Animals , Endothelial Cells/pathology , Heparin, Low-Molecular-Weight/therapeutic use , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Micelles , Myeloid-Derived Suppressor Cells/pathology , Nanomedicine , Neoplastic Cells, Circulating/pathology , Tumor Microenvironment/drug effects , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/therapeutic use
3.
Pharm Dev Technol ; 25(4): 492-509, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31903817

ABSTRACT

In this study, pH-triggered polymeric micelle comprising α-tocopherol (TOC) and heparin (HEP) was developed and loaded with docetaxel (DTX). The amphiphilic copolymer was synthesized by grafting TOC onto HEP backbone by a pH-cleavable bond. DTX-loaded micelles were characterized in terms of critical micelle concentration (CMC), particle size, zeta potential, entrapment efficiency (EE), pH-responsive behavior, and drug release. In vitro cytotoxicity of the micelles against breast cancer cells was investigated by MTT assay. The cellular uptake of coumarin-loaded micelles was also evaluated. Furthermore, the pharmacokinetics of DTX-loaded micelles was evaluated and compared with that of Taxotere®.HEP-CA-TOC copolymers showed low CMC values and high EE. At pH 7.4, the micelles remained stable in size and shape, whereas considerable changes in particle size and morphology were observed at pH 5.5. DTX-loaded micelles showed pH-dependent drug release profiles. Coumarin-loaded micelles showed higher cellular uptake than free coumarin. Therefore, the DTX-loaded micelles showed more toxicity against breast cancer cells than free DTX. A significant increase in T1/2 ß, AUC0-∞ and MRT was observed in DTX-loaded micelle treated group as compared to the group treated with Taxotere®.The results suggest that the pH-sensitive HEP-modified micelles could be promising for enhanced intracellular drug delivery of DTX for cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Delayed-Action Preparations/chemistry , Docetaxel/administration & dosage , Heparin/analogs & derivatives , alpha-Tocopherol/analogs & derivatives , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Docetaxel/pharmacokinetics , Docetaxel/pharmacology , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Mice , Micelles
4.
Nanotechnology ; 30(8): 085101, 2019 Feb 22.
Article in English | MEDLINE | ID: mdl-30523865

ABSTRACT

To increase the efficacy of small molecule chemotherapeutic drug (SMCD) and reduce its toxic and side effects, we selected two model drugs doxorubicin (DOX) and chloroquine (CQ). DOX is a SMCD and CQis a chemosensitizer with autophagy inhibition. Poly(lactic-co-glycolic acid) (PLGA) and alpha-tocopherol polyethylene glycol 1000 succinate were chosen as delivery carriers to design and prepare a novel type of drug co-delivery single-nanoparticles by emulsification-solvent volatilisation, named NPDOX+CQ. The physicochemical properties of NPDOX+CQ were characterised. Then A549 cells and A549/Taxol cells were used for the in vitro anti-cancer effect study. At the same time, cellular uptake, intracellular migration and anti-cancer mechanism of nanoparticles were studied. The NPs showed a uniform spherical shape with good dispersibility, and both drugs had good encapsulation efficiency and loading capacity. In all formulations, NPDOX+CQ showed the highest in vitro cytotoxicity. The results showed that NPs could protect drugs from being recognised and excluded by P-glycoprotein (P-gp). Moreover, the results of the mechanistic study demonstrated that NPs were transported by autophagy process after being taken up by the cells. Therefore, during the migration of NPDOX+CQ, CQ could exert its efficacy and block autophagy so that DOX would not be hit by autophagy. Western Blot results showed that NPDOX+CQ had the best inhibition effect of autophagy. It can be concluded that the system can prevent the drug from being recognised and excluded by P-gp, and CQ blocks the process of autophagy so that the DOX is protected and more distributed to the nucleus of multidrug resistance (MDR) cell. The NPDOX+CQ constructed in this study provides a feasible strategy for reversing MDR in tumour cells.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Chloroquine/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Neoplasms/drug therapy , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Chloroquine/pharmacokinetics , Chloroquine/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Delivery Systems , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Nanoparticles/chemistry , Neoplasms/metabolism , Polyethylene Glycols/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , alpha-Tocopherol/analogs & derivatives
5.
Nanomedicine ; 21: 102010, 2019 10.
Article in English | MEDLINE | ID: mdl-31195135

ABSTRACT

The phosphorylation of (+) alpha tocopherol produces adhesive nanostructures that interact with oral biofilms to restrict their growth. The aim of this work was to understand if these adhesive (+) alpha tocopheryl phosphate (α-TP) nanostructures could also control macrophage responses to the presence of oral bacteria. The (+) α-TP planar bilayer fragments (175 nm ±â€¯21 nm) formed in a Trizma®/ethanol vehicle swelled when exposed to the cell lines (maximum stabilized size = 29 µm). The swelled (+) α-TP aggregates showed selective toxicity towards THP-1 macrophages (LD50 = 304 µM) compared to human gingival fibroblasts (HGF-1 cells; LD50 > 5 mM), and they inhibited heat killed bacteria stimulated MCP-1 production in both macrophages (control 57.3 ±â€¯18.1 pg/mL vs (+) α-TP 6.5 ±â€¯3.2 pg/mL) and HGF-1 cells (control 673.5 ±â€¯133 pg/mL vs (+) α-TP - 463.9 ±â€¯68.9 pg/mL).


Subject(s)
Macrophages/drug effects , Mouth/drug effects , Nanostructures/administration & dosage , alpha-Tocopherol/analogs & derivatives , Biofilms/drug effects , Cell Adhesion/drug effects , Cell Line , Chemokine CCL2/genetics , Gingiva/drug effects , Gingiva/growth & development , Gingiva/microbiology , Gingiva/pathology , Hepatocyte Growth Factor/genetics , Humans , Macrophages/metabolism , Macrophages/microbiology , Monocytes/drug effects , Monocytes/microbiology , Mouth/growth & development , Mouth/microbiology , Mouth/pathology , Nanostructures/chemistry , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/genetics , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
6.
Bioconjug Chem ; 29(2): 255-266, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29268009

ABSTRACT

Two novel α-tocopheryl-lipoic acid conjugates (TL1 and TL2) were synthesized for the anticancer drug, doxorubicin (DOX), delivery. Both conjugates were able to form stable nanovesicles. The critical aggregation concentration (CAC) was determined using 4-(N,N-dimethylamino)cinnamaldehyde (DMACA) as a fluorescence probe. Formation of highly packed nanovesicles was characterized by 1,6-diphenyl-1,3,5-hexatriene (DPH) fluorescence anisotropy and microviscosity measurements. The morphologies of nanovesicles were visualized by transmission electron microscopy (TEM) and atomic force microscopy (AFM). The response of nanovesicles to reducing environment of cells was probed by the addition of dithiothreitol (DTT), which was followed by the increase in the hydrodynamic diameter under dynamic light scattering (DLS) measurements. The encapsulation efficiency of a commonly used anticancer drug, doxorubicin (DOX), in nanovesicles was found to be ∼60% and ∼55% for TL1 and TL2, respectively (TL1-DOX and TL2-DOX). Also, the cumulative drug (DOX) release from DOX-encapsulated nanovesicles in response to biological reducing agent glutathione (GSH) was ∼50% and ∼40% for TL1-DOX and TL2-DOX, respectively, over a period of 10 h. Both TL1-DOX and TL2-DOX delivered the anticancer drug, doxorubicin (DOX), across the DOX-sensitive and DOX-resistant HeLa (HeLa-DOXR) cells in an efficient manner and significantly more efficaciously than the drug alone treatments, especially in HeLa-DOXR cells. The nanovesicle mediated DOX treatment also showed significantly higher cell death when compared to DOX alone treatment in HeLa-DOXR cells. Blood compatibility of the nanovesicles was supported from clotting time, hemolysis, and red blood cell (RBC) aggregation experiments for their potential in vivo applications. Concisely, we present biocompatible and responsive nanovesicles for efficacious drug delivery to drug-sensitive and drug-resistant cancer cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Delayed-Action Preparations/chemistry , Doxorubicin/administration & dosage , Nanocapsules/chemistry , Thioctic Acid/chemistry , alpha-Tocopherol/analogs & derivatives , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , HeLa Cells , Humans , Neoplasms/drug therapy , Oxidation-Reduction
7.
J Vasc Surg ; 67(4): 1263-1273.e1, 2018 04.
Article in English | MEDLINE | ID: mdl-28571880

ABSTRACT

OBJECTIVE: Considering the poor efficacy of local intramuscular injections with endothelial progenitor cells (EPCs) for critical limb ischemia in patients with diabetes, the study aimed to investigate the effect of α-tocopherol (α-T) and α-tocopherol phosphate (α-TP) on apoptosis and angiogenesis in a rat model under oxidative stress conditions. METHODS: Primary EPCs from Sprague-Dawley rats were harvested and treated with α-T and α-TP for 24 hours. Gene transcription and protein expression were evaluated by real-time polymerase chain reaction and Western blot, respectively. Cell apoptosis, migration, and tube formation ability were detected by flow cytometry, Transwell assay (Chemicon International, Temecula, Calif), and Matrigel-based angiogenesis assay (Corning Inc, Corning, NY). The in vivo experiments were carried out using 30 single hind limb ischemic models of diabetic rats that were treated with allogeneic EPCs. Capillary density was evaluated by immunohistochemistry. RESULTS: α-T and α-TP attenuated high glucose/hypoxia-induced cell apoptosis by promoting Bcl-2 and Akt and inhibiting nuclear factor κB p65, JNK, Notch-1, and p38MAPK genes. Furthermore, α-T and α-TP promoted the transcription and expression of vascular endothelial growth factor receptor 2 and decreased the transcription and expression of Tie-2 and Notch-1 in EPCs under high-glucose/hypoxic conditions. Moreover, α-T and especially α-TP enhanced the migratory activity of EPCs under high-glucose/hypoxic conditions. Capillary density of ischemic hind limbs was increased on day 14 after administration of EPCs pretreated with α-T and α-TP. CONCLUSIONS: α-T, especially α-TP, possesses therapeutic potential in the inhibition of apoptosis and increases the migratory capacity of EPCs under high-glucose/hypoxic conditions. It promotes angiogenesis by upregulating Bcl-2, Akt, and vascular endothelial growth factor receptor 2 and decreasing nuclear factor κB p65, p38MAPK, Notch-1, JNK, and Tie-2.


Subject(s)
Apoptosis/drug effects , Bone Marrow Cells/drug effects , Endothelial Progenitor Cells/drug effects , Glucose/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , alpha-Tocopherol/analogs & derivatives , Angiogenic Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cell Hypoxia , Cell Movement/drug effects , Cells, Cultured , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/surgery , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Endothelial Progenitor Cells/transplantation , Hindlimb , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Ischemia/surgery , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stem Cell Niche , Time Factors , alpha-Tocopherol/pharmacology
8.
Nanomedicine ; 14(7): 2307-2316, 2018 10.
Article in English | MEDLINE | ID: mdl-29410321

ABSTRACT

'Soft' nanomaterials have the potential to produce substantive antibiofilm effects. The aim of this study was to understand the oral antimicrobial activity of soft nanomaterials generated from alpha-tocopherol (α-T) and alpha-tocopherol phosphate (α-TP). (+) α-TP formed planar bilayer islands (175 ± 21 nm, -14.9 ± 3.5 mV) in a Trizma® buffer, whereas (+) α-T formed spherical liposomes (563 ± 1 nm, -10.5 ± 0.2 mV). The (+) α-TP bilayers displayed superior Streptococcus oralis biofilm growth retardation, a more substantive action, generated a superior adsorption to hydroxyapatite and showed an enhanced inhibition of multi-species bacterial saliva biofilm growth (38 ± 7µm vs 58 ± 18 µm, P ˂ 0.05) compared to (+) α-T. Atomic force microscopy data indicated that the ability of the 'soft' α-TP nanomaterials to transition into planar bilayer structures upon contact with interfaces facilitated their adhesive properties and substantive antimicrobial effects.


Subject(s)
Anti-Infective Agents/administration & dosage , Biofilms/drug effects , Lipid Bilayers/chemistry , Saliva/microbiology , Streptococcus mutans/drug effects , Streptococcus oralis/drug effects , alpha-Tocopherol/analogs & derivatives , Adhesives , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Biofilms/growth & development , Humans , Liposomes/administration & dosage , Liposomes/chemistry , Microscopy, Atomic Force , Mouth/microbiology , Streptococcus mutans/growth & development , Streptococcus oralis/growth & development , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
9.
Parasitol Res ; 117(10): 3177-3182, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30030625

ABSTRACT

α-Tocopheryl succinate (α-TOS), a derivative of vitamin E, is synthesized by esterification of α-tocopherol. It has been reported that α-TOS inhibits the mitochondrial complex II resulting in generation of reactive oxygen species, which triggers selective apoptosis in a large number of cancer cells, while it appears largely non-toxic towards normal cells. Plasmodium parasites are well known to have high sensitivity to oxidative stress. Thus, α-TOS is suspected to impact Plasmodium parasites by oxidative stress. In this study, to ascertain whether α-TOS is an appropriate candidate for an anti-malarial drug, C57BL/6J mice were infected with P. yoelii 17XL and P. berghei ANKA, a lethal strain of rodent malaria and experimental cerebral malaria (ECM), and treated with several concentrations of α-TOS by intraperitoneal administration on 1, 3, 5, and 7 days post infection (dpi). In addition, the permeability of the blood brain barrier (BBB) was examined by Evans blue staining in ECM on 7 dpi. As a result of α-TOS treatment, parasitemia was decreased and survival rate was significantly increased in mice infected with both parasites. Furthermore, the intensity of Evans blue staining on brains taken from α-TOS-treated mice was weaker than that of untreated mice. This means that α-TOS might inhibit the breakdown of BBB and progress of cerebral malaria. These findings indicate that vitamin E derivatives like α-TOS might be a potential candidate for treatment drugs against malaria.


Subject(s)
Antimalarials/administration & dosage , Malaria, Cerebral/drug therapy , alpha-Tocopherol/analogs & derivatives , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/parasitology , Humans , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Parasitemia/drug therapy , Plasmodium yoelii/drug effects , Plasmodium yoelii/physiology , Reactive Oxygen Species/metabolism , alpha-Tocopherol/administration & dosage
10.
Int J Mol Sci ; 19(6)2018 05 23.
Article in English | MEDLINE | ID: mdl-29882845

ABSTRACT

Amphiphilic polymer of α-tocopherol succinate modified glycol chitosan (TS-GC) was successfully constructed by conjugating α-tocopherol succinate to the skeleton of glycol chitosan and characterized by Fourier-transform infrared (FT-IR) and proton nuclear magnetic resonance (¹H-NMR). In aqueous milieu, the conjugates self-assembled to micelles with the critical aggregation concentration of 7.2 × 10−3 mg/mL. Transmission electron microscope (TEM) observation and dynamic light scattering (DLS) measurements were carried out to determine the physicochemical properties of the micelles. The results revealed that paclitaxel (PTX)-loaded TS-GC micelles were spherical in shape. Moreover, the PTX-loaded micelles showed increased particle sizes (35 nm vs. 142 nm) and a little reduced zeta potential (+19 mV vs. +16 mV) compared with blank micelles. The X-ray diffraction (XRD) spectra demonstrated that PTX existed inside the micelles in amorphous or molecular state. In vitro and in vivo tests showed that the PTX-loaded TS-GC micelles had advantages over the Cremophor EL-based formulation in terms of low toxicity level and increased dose, which suggested the potential of the polymer as carriers for PTX to improve their delivery properties.


Subject(s)
Chitosan/chemistry , Drug Delivery Systems , Micelles , Paclitaxel/administration & dosage , Polymers/chemistry , Animals , Antineoplastic Agents/pharmacology , Chitosan/chemical synthesis , Female , Glycerol/analogs & derivatives , Glycerol/pharmacology , Humans , Injections, Intravenous , MCF-7 Cells , Male , Mice , Particle Size , Polymers/chemical synthesis , Proton Magnetic Resonance Spectroscopy , Rabbits , Spectroscopy, Fourier Transform Infrared , Static Electricity , X-Ray Diffraction , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/chemical synthesis , alpha-Tocopherol/chemistry
11.
J Cell Biochem ; 118(7): 1855-1867, 2017 07.
Article in English | MEDLINE | ID: mdl-28059487

ABSTRACT

The CD36 scavenger receptor binds several ligands and mediates ligand uptake and ligand-dependent signal transduction and gene expression, events that may involve CD36 internalization. Here we show that CD36 internalization in THP-1 monocytes is triggered by α-tocopherol (αT) and more strongly by α-tocopheryl phosphate (αTP) and EPC-K1, a phosphate diester of αTP and L-ascorbic acid. αTP-triggered CD36 internalization is prevented by the specific covalent inhibitor of selective lipid transport by CD36, sulfo-N-succinimidyl oleate (SSO). Moreover, SSO inhibited the CD36-mediated uptake of 14C-labelled αTP suggesting that αTP binding and internalization of CD36 is involved in cellular αTP uptake, whereas the uptake of αT was less affected. Similar to that, inhibition of selective lipid transport of the SR-BI scavenger receptor resulted mainly in reduction of αTP and not αT uptake. In contrast, uptake of αT was mainly inhibited by Dynasore, an inhibitor of clathrin-mediated endocytosis, suggesting that the differential regulatory effects of αTP and αT on signaling may be influenced by their different routes of uptake. Interestingly, αTP and EPC-K1 also reduced the neutral lipid content of THP-1 cells and the phagocytosis of fluorescent Staphylococcus aureus bioparticles. Moreover, induction of the vascular endothelial growth factor (VEGF) promoter activity by αTP occurred via CD36/PI3Kγ/Akt, as it could be inhibited by specific inhibitors of this pathway (SSO, Wortmannin, AS-605240). These results suggest that αTP activates PI3Kγ/Akt signaling leading to VEGF expression in monocytes after binding to and/or transport by CD36, a receptor known to modulate angiogenesis in response to amyloid beta, oxLDL, and thrombospondin. J. Cell. Biochem. 118: 1855-1867, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
CD36 Antigens/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Vascular Endothelial Growth Factor A/metabolism , alpha-Tocopherol/analogs & derivatives , Ascorbic Acid/analogs & derivatives , Ascorbic Acid/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Monocytes/drug effects , Monocytes/metabolism , Monocytes/microbiology , Phagocytosis/drug effects , Signal Transduction/drug effects , Staphylococcus aureus/physiology , THP-1 Cells/drug effects , THP-1 Cells/metabolism , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , alpha-Tocopherol/pharmacology
12.
J Am Chem Soc ; 139(44): 15801-15811, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29043787

ABSTRACT

Reactive oxygen species (ROS) and their associated byproducts have been traditionally associated with a range of pathologies. It is now believed, however, that at basal levels these molecules also have a beneficial cellular function in the form of cell signaling and redox regulation. Critical to elucidating their physiological role is the opportunity to visualize and quantify the production of ROS with spatiotemporal accuracy. Armed with a newly developed, extremely sensitive fluorogenic α-tocopherol analogue (H4BPMHC), we report herein the observation of steady concentrations of lipid peroxyl radicals produced in live cell imaging conditions. Imaging studies with H4BPMHC indicate that the rate of production of lipid peroxyl radicals in HeLa cells under basal conditions is 33 nM/h within the cell. Our work further provides indisputable evidence on the antioxidant role of Vitamin E, as lipid peroxidation was suppressed in HeLa cells both under basal conditions and in the presence of Haber-Weiss chemistry, generated by the presence of cumyl hydroperoxide and Cu2+ in solution, when supplemented with the α-tocopherol surrogate, PMHC (2,2,5,7,8-pentamethyl-6-hydroxy-chromanol, an α-tocopherol analogue lacking the phytyl tail). H4BPMHC has the sensitivity needed to detect trace changes in oxidative status within the lipid membrane, underscoring the opportunity to illuminate the physiological relevance of lipid peroxyl radical production during cell homeostasis and disease.


Subject(s)
Fluorescent Dyes/chemistry , Optical Imaging/methods , Peroxides/metabolism , Reactive Oxygen Species/metabolism , alpha-Tocopherol/analogs & derivatives , Fluorescent Dyes/metabolism , HeLa Cells , Homeostasis , Humans , Lipid Peroxidation , Oxidation-Reduction , Peroxides/analysis , Reactive Oxygen Species/analysis , alpha-Tocopherol/metabolism
13.
Clin Exp Pharmacol Physiol ; 44 Suppl 1: 107-116, 2017 12.
Article in English | MEDLINE | ID: mdl-28744946

ABSTRACT

α-Tocopheryl phosphate (TP) is a naturally occurring form of vitamin E found in the body. In the present study we compared the ability of an α-TP mixture (TPM) against a standard vitamin E supplement, α-tocopherol acetate (TA) on the development of atherosclerotic lesions in ApoE-deficient mice. Mice were maintained on either a normal chow diet for 24 weeks (Normal Diet), vs a group in which the final 8 weeks of the 24-week period mice were placed on a high fat (21%), high cholesterol (0.15%) challenge diet (HFHC), to exacerbate atherosclerotic lesion development.. The difference in these two control groups established the extent of the diet-induced atherosclerotic lesion development. Mice in the various treatment groups received either TA (300 mg/kg chow) or TPM (6.7-200 mg/kg chow) for 24 weeks, with TPM treatment resulting in dose-dependent significant reductions in atherosclerotic lesion formation and plasma levels of pro-inflammatory cytokines. TA-treated mice, with the tocopherol equivalent TPM dose (200 mg/kg chow), showed no significant reduction in plasma lipid levels or evidence for aortic lesion regression. At this TPM equivalent TA dose, a 44% reduction in aortic lesion formation was observed. In addition, these TPM treated mice, also showed a marked reduction in aortic superoxide formation and decreased circulating plasma levels of known pro-inflammatory markers IL-6, MCP-1, IL-1ß, IFN-γ and TNF-α. These findings indicate that TPM treatment slows progression of atherosclerotic lesions in ApoE-deficient mice with this effect potentially involving reduced oxidative stress and decreased inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Plaque, Atherosclerotic , alpha-Tocopherol/analogs & derivatives , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Cytokines/blood , Diet, High-Fat , Disease Models, Animal , Inflammation Mediators/blood , Male , Mice, Knockout, ApoE , Oxidative Stress/drug effects , Superoxides/metabolism , alpha-Tocopherol/pharmacology
14.
Cell Mol Neurobiol ; 36(6): 1015-1022, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26749581

ABSTRACT

α-Tocopheryl phosphate (αTP) is a phosphorylated form of α-tocopherol. Since it is phosphorylated in the hydroxyl group that is essential for the antioxidant property of α-tocopherol, we hypothesized that αTP would modulate the antioxidant system, rather than being an antioxidant agent per se. α-TP demonstrated antioxidant activity in vitro against iron-induced oxidative stress in a mitochondria-enriched fraction preparation treated with 30 or 100 µM α-TP. However, this effect was not observed ex vivo with mitochondrial-enriched fraction from mice treated with an intracerebroventricular injection of 0.1 or 1 nmol/site of αTP. Two days after treatment (1 nmol/site αTP), peroxiredoxin 2 (Prx2) and glutathione reductase (GR) expression and GR activity were decreased in cerebral cortex and hippocampus. Glutathione content, glutathione peroxidase, and thioredoxin reductase activities were not affected by αTP. In conclusion, the persistent decrease in GR and Prx2 protein content is the first report of an in vivo effect of αTP on protein expression in the mouse brain, potentially associated to a novel and biologically relevant function of this naturally occurring compound.


Subject(s)
Antioxidants/pharmacology , Brain/drug effects , Glutathione Reductase/metabolism , Oxidative Stress/drug effects , Peroxiredoxins/metabolism , alpha-Tocopherol/analogs & derivatives , Animals , Antioxidants/metabolism , Brain/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , alpha-Tocopherol/pharmacology
15.
Bioorg Med Chem ; 24(12): 2754-61, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27161877

ABSTRACT

Previously prepared fluorescent derivatives of α-tocopherol have shown tremendous utility in both in vitro exploration of the mechanism of ligand transfer by the α-tocopherol transfer protein (α-TTP) and the intracellular transport of α-tocopherol in cells and tissues. We report here the synthesis of a 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) containing α-tocopherol analog having extended conjugation with an alkenyl thiophene group that extends the absorption and emission maxima to longer wavelengths (λex=571nm and λem=583nm). The final fluorophore thienyl-ene-BODIPY-α-tocopherol, 2, binds to recombinant human α-TTP with a Kd=8.7±1.1nM and is a suitable probe for monitoring the secretion of α-tocopherol from cultured Mcf7#189 cells.


Subject(s)
Boron Compounds/chemistry , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/analysis , Animals , Boron Compounds/chemical synthesis , Boron Compounds/metabolism , Carrier Proteins/metabolism , Cell Line , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Humans , Protein Binding , Rats , alpha-Tocopherol/metabolism
16.
J Biol Chem ; 289(47): 32488-98, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25278024

ABSTRACT

Cytochrome c is a multifunctional hemoprotein in the mitochondrial intermembrane space whereby its participation in electron shuttling between respiratory complexes III and IV is alternative to its role in apoptosis as a peroxidase activated by interaction with cardiolipin (CL), and resulting in selective CL peroxidation. The switch from electron transfer to peroxidase function requires partial unfolding of the protein upon binding of CL, whose specific features combine negative charges of the two phosphate groups with four hydrophobic fatty acid residues. Assuming that other endogenous small molecule ligands with a hydrophobic chain and a negatively charged functionality may activate cytochrome c into a peroxidase, we investigated two hydrophobic anionic analogues of vitamin E, α-tocopherol succinate (α-TOS) and α-tocopherol phosphate (α-TOP), as potential inducers of peroxidase activity of cytochrome c. NMR studies and computational modeling indicate that they interact with cytochrome c at similar sites previously proposed for CL. Absorption spectroscopy showed that both analogues effectively disrupt the Fe-S(Met(80)) bond associated with unfolding of cytochrome c. We found that α-TOS and α-TOP stimulate peroxidase activity of cytochrome c. Enhanced peroxidase activity was also observed in isolated rat liver mitochondria incubated with α-TOS and tBOOH. A mitochondria-targeted derivative of TOS, triphenylphosphonium-TOS (mito-VES), was more efficient in inducing H2O2-dependent apoptosis in mouse embryonic cytochrome c(+/+) cells than in cytochrome c(-/-) cells. Essential for execution of the apoptotic program peroxidase activation of cytochrome c by α-TOS may contribute to its known anti-cancer pharmacological activity.


Subject(s)
Cytochromes c/chemistry , Peroxidase/chemistry , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/chemistry , Animals , Apoptosis/drug effects , Apoptosis/genetics , Binding Sites/genetics , Cell Line , Cytochromes c/genetics , Cytochromes c/metabolism , Enzyme Activation/drug effects , Horses , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Male , Mice, Knockout , Models, Molecular , Molecular Structure , Peroxidase/metabolism , Protein Binding , Protein Structure, Tertiary , Spectrophotometry , Vitamins/chemistry , Vitamins/metabolism , Vitamins/pharmacology , alpha-Tocopherol/pharmacology
17.
J Cell Biochem ; 116(3): 398-407, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25290554

ABSTRACT

In several studies, vitamin E has been observed to influence angiogenesis and vasculogenesis. We recently showed that the phosphorylated form of α-tocopherol (αT), α-tocopheryl phosphate (αTP), increases the expression of the vascular endothelial growth factor (VEGF). Thus, αTP may act as an active lipid mediator increasing VEGF expression, angiogenesis, and vasculogenesis. Here, we investigated the molecular signaling mechanisms by which αTP induces VEGF expression using cultured HEK293 cells as model system. αT and more so αTP increased VEGF-promoter activity in a phosphatidylinositol-3-kinase gamma (PI3Kγ)-dependent manner. In contrast, after overexpression of PI3Kγ and/or protein kinase B (PKB), VEGF promoter activity was inhibited by αT and more so by αTP. Inhibition by αT and αTP was dependent on the lipid kinase activity of PI3Kγ, whereas an induction was seen with the protein kinase activity, consistent with a model in which PKB inhibition by αT or αTP occurs only when activated at the plasma membrane and possibly involves a phosphatase such as PHLPP1. PI3Kγ-induced VEGF expression was reduced when the human tocopherol-associated protein 1 (hTAP1/SEC14L2) was overexpressed suggesting formation of an inactive PI3Kγ/hTAP1 heterodimer, that could be reactivated by αT and more so by αTP. We suggest a novel signaling mechanism by which αTP stimulates PI3Kγ activity by stimulating hTAP-mediated phosphatidylinositol exchange and presentation to the enzyme and/or dissociation of an inactive heterodimer. At cellular level, hTAP may act as sensor for intracellular lipid information (location, type, and amount of lipid) and translate it into responses of PI3K-mediated signaling and gene expression.


Subject(s)
Carrier Proteins/metabolism , Lipids/chemistry , Lipoproteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/metabolism , Vascular Endothelial Growth Factor A/genetics , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
J Nutr ; 145(10): 2347-54, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26290004

ABSTRACT

BACKGROUND: Studies examining vitamin E intake and the percentage of the population meeting dietary guidelines do not distinguish between natural (RRR-α-tocopherol) and synthetic (all-rac-α-tocopherol) intake, even though these different isomeric forms differ in bioactivity. OBJECTIVE: This study aimed to determine the effect of RRR-α-tocopherol vs. all-rac-α-tocopherol intake on the percentage of the population meeting the vitamin E recommendation and on plasma α-tocopherol stereoisomer distribution. METHODS: With the use of data from the Irish National Adult Nutrition Survey (NANS), this study examined the percentage of the Irish population meeting the European Union (EU) RDA for vitamin E of 12 mg/d, correcting for a bioactivity difference in all-rac- vs. RRR-α-tocopherol, where 1 mg of all-rac-α-tocopherol is considered to be equivalent to 1:1.36 (0.74) mg in the EU RDA. In a subcohort of supplement users and nonusers, plasma α- and γ-tocopherol concentrations and α-tocopherol stereoisomer distribution were measured. Receiver operating characteristic (ROC) curve analysis was conducted to determine ability to discriminate supplement user types. RESULTS: Analysis of the NANS showed that 100% of participants still met the recommended intake of 12 mg/d, after all-rac-α-tocopherol intake was corrected for α-tocopherol equivalent bioactivity. In the subcohort analysis, the percentage of plasma RRR-α-tocopherol was significantly lower in high all-rac-α-tocopherol supplement (>11 mg/d) users (82%) compared with nonusers and with high RRR-α-tocopherol supplement (>35 mg/d) users (91% and 93% respectively, P < 0.01). High RRR-α-tocopherol supplement users had a significantly higher plasma α-tocopherol than low all-rac-α-tocopherol supplement (<2.5 mg/d) users (34 vs. 25 µmol/L, P = 0.01). ROC analysis demonstrated an ability to distinguish between RRR- and all-rac-α-tocopherol consumers, which may be useful in investigating the potential effect of RRR- and all-rac-α-tocopherol intake on health. CONCLUSIONS: This study demonstrated that the percentage of the population meeting the vitamin E recommendation was unaffected when all-rac-α-tocopherol intake was corrected for α-tocopherol equivalent bioactivity. all-rac-α-Tocopherol intake led to a decrease in the percentage of plasma RRR-α-tocopherol relative to RRR-α-tocopherol intake.


Subject(s)
Diet , Dietary Supplements , Patient Compliance , Recommended Dietary Allowances , Vitamin E/administration & dosage , alpha-Tocopherol/blood , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , Cross-Sectional Studies , Diet/adverse effects , European Union , Female , Humans , Ireland , Male , Middle Aged , Nutrition Surveys , Nutritive Value , ROC Curve , Stereoisomerism , Young Adult , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/chemistry , gamma-Tocopherol/analogs & derivatives , gamma-Tocopherol/blood , gamma-Tocopherol/chemistry
19.
Biomacromolecules ; 16(5): 1566-81, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25848887

ABSTRACT

α-Tocopheryl succinate (α-TOS) is a well-known mitochondrially targeted anticancer compound, however, it is highly hydrophobic and toxic. In order to improve its activity and reduce its toxicity, new surfactant-free biologically active nanoparticles (NP) were synthesized. A methacrylic derivative of α-TOS (MTOS) was prepared and incorporated in amphiphilic pseudoblock copolymers when copolymerized with N-vinylpyrrolidone (VP) by free radical polymerization (poly(VP-co-MTOS)). The selected poly(VP-co-MTOS) copolymers formed surfactant-free NP by nanoprecipitation with sizes between 96 and 220 nm and narrow size distribution, and the in vitro biological activity was tested. In order to understand the structure-activity relationship three other methacrylic monomers were synthesized and characterized: MVE did not have the succinate group, SPHY did not have the chromanol ring, and MPHY did not have both the succinate group and the chromanol ring. The corresponding families of copolymers (poly(VP-co-MVE), poly(VP-co-SPHY), and poly(VP-co-MPHY)) were synthesized and characterized, and their biological activity was compared to poly(VP-co-MTOS). Both poly(VP-co-MTOS) and poly(VP-co-MVE) presented triple action: reduced cell viability of cancer cells with little or no harm to normal cells (anticancer), reduced viability of proliferating endothelial cells with little or no harm to quiescent endothelial cells (antiangiogenic), and efficiently encapsulated hydrophobic molecules (nanocarrier). The anticancer and antiangiogenic activity of the synthesized copolymers is demonstrated as the active compound (vitamin E or α-tocopheryl succinate) do not need to be cleaved to trigger the biological action targeting ubiquinone binding sites of complex II. Poly(VP-co-SPHY) and poly(VP-co-MPHY) also formed surfactant-free NP that were also endocyted by the assayed cells; however, these NP did not selectively reduce cell viability of cancer cells. Therefore, the chromanol ring of the vitamin E analogues has an important role in the biological activity of the copolymers. Moreover, when succinate moiety is substituted and vitamin E is directly linked to the macromolecular chain through an ester bond, the biological activity is maintained.


Subject(s)
Nanoparticles/chemistry , Structure-Activity Relationship , Vitamin E/chemical synthesis , alpha-Tocopherol/chemical synthesis , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Methacrylates/chemical synthesis , Methacrylates/chemistry , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Polymers/chemical synthesis , Polymers/chemistry , Surface-Active Agents/chemistry , Vitamin E/analogs & derivatives , Vitamin E/chemistry , Vitamin E/pharmacology , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
20.
Mol Nutr Food Res ; 68(10): e2300657, 2024 May.
Article in English | MEDLINE | ID: mdl-38698718

ABSTRACT

SCOPE: Tocomonoenols (T1) are little-known vitamin E derivatives naturally occurring in foods. Limited knowledge exists regarding the cellular uptake and metabolism of α-tocomonoenol (αT1) and none about that of γ-tocomonoenol (γT1). METHODS AND RESULTS: The study investigates the cytotoxicity, uptake, and metabolism of αT1 and γT1 in HepG2 cells compared to the α- and γ-tocopherols (T) and -tocotrienols (T3). None of the studied tocochromanols are cytotoxic up to 100 µmol L-1. The uptake of the γ-congeners is significantly higher than that of the corresponding α-forms, whereas no significant differences are observed based on the degree of saturation of the sidechain. Carboxymethylbutyl-hydroxychromans (CMBHC) are the predominant short-chain metabolites of all tocochromanols and conversion is higher for γT1 than αT1 as well as for the γ-congeners of T and T3. The rate of metabolism increases with the number of double bonds in the sidechain. The rate of metabolic conversion of the T1 is more similar to tocopherols than to that of the tocotrienols. CONCLUSION: This is the first evidence that both αT1 and γT1 follow the same sidechain degradation pathway and exert similar rates of metabolism than tocopherols. Therefore, investigation into the biological activities of tocomonoenols is warranted.


Subject(s)
Chromans , Vitamin E , Humans , Hep G2 Cells , Chromans/pharmacology , Vitamin E/pharmacology , Vitamin E/analogs & derivatives , Vitamin E/metabolism , Vitamin E/pharmacokinetics , gamma-Tocopherol/metabolism , gamma-Tocopherol/pharmacology , Tocotrienols/pharmacology , Tocotrienols/metabolism , Tocotrienols/pharmacokinetics , Cell Survival/drug effects , alpha-Tocopherol/pharmacology , alpha-Tocopherol/metabolism , alpha-Tocopherol/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL