Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.055
Filter
1.
Sci Rep ; 14(1): 13315, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38858439

ABSTRACT

Exposure to high, marginally lethal doses or higher of ionizing radiation, either intentional or accidental, results in injury to various organs. Currently, there is only a limited number of safe and effective radiation countermeasures approved by US Food and Drug Administration for such injuries. These approved agents are effective for only the hematopoietic component of the acute radiation syndrome and must be administered only after the exposure event: currently, there is no FDA-approved agent that can be used prophylactically. The nutraceutical, gamma-tocotrienol (GT3) has been found to be a promising radioprotector of such exposure-related injuries, especially those of a hematopoietic nature, when tested in either rodents or nonhuman primates. We investigated the nature of injuries and the possible protective effects of GT3 within select organ systems/tissues caused by both non-lethal level (4.0 Gy), as well as potentially lethal level (5.8 Gy) of ionizing radiation, delivered as total-body or partial-body exposure. Results indicated that the most severe, dose-dependent injuries occurred within those organ systems with strong self-renewing capacities (e.g., the lymphohematopoietic and gastrointestinal systems), while in other tissues (e.g., liver, kidney, lung) endowed with less self-renewal, the pathologies noted tended to be less pronounced and less dependent on the level of exposure dose or on the applied exposure regimen. The prophylactic use of the test nutraceutical, GT3, appeared to limit the extent of irradiation-associated pathology within blood forming tissues and, to some extent, within the small intestine of the gastrointestinal tract. No distinct, global pattern of bodily protection was noted with the agent's use, although a hint of a possible radioprotective benefit was suggested not only by a lessening of apparent injury within select organ systems, but also by way of noting the lack of early onset of moribundity within select GT3-treated animals.


Subject(s)
Dietary Supplements , Radiation-Protective Agents , Animals , Radiation-Protective Agents/pharmacology , Vitamin E/pharmacology , Vitamin E/analogs & derivatives , Acute Radiation Syndrome/prevention & control , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/pathology , Chromans/pharmacology , Male , Radiation Injuries, Experimental/prevention & control , Radiation Injuries, Experimental/pathology , Macaca mulatta , Liver/drug effects , Liver/radiation effects , Liver/pathology
2.
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
3.
J Microencapsul ; 41(4): 255-268, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38647544

ABSTRACT

The aim is to investigate the possible pulmonary protective effect of vanillic acid (VA) in liposome-TPGS nanoparticles, to overcome VA's poor bioavailability. VA was successfully extracted. Liposomes were prepared using thin film hydration. Central composite design was adopted for optimisation of liposomes to get the maximum entrapment efficiency (EE%) and the minimum mean diameter, where the liposomes were further modified with TPGS, and tested for PDI, zeta-potential, and in-vitro drug release. In-vivo study on mice with LPS-acute pulmonary toxicity was tested. TPGS-modified VA-liposomes showed EE% of 69.35 ± 1.23%, PS of 201.7 ± 3.23 nm, PDI of 0.19 ± 0.02, and zeta-potential of -32.2 ± 0.32 mv. A sustained drug release of the TPGS-modified VA-liposomes was observed compared to standard VA, and a pulmonary-protective effect through decreasing miR-217 expression with subsequent anti-inflammatory effect through suppression of MAPK and PI3K/NF-κB pathways was also demonstrated in the current study. TPGS-modified VA-liposomes showed an enhanced bioavailability and a sustained drug release with promising pulmonary protective effects against acute pulmonary injury diseases.


Subject(s)
Liposomes , MicroRNAs , NF-kappa B , Vanillic Acid , Vitamin E , Animals , NF-kappa B/metabolism , Vanillic Acid/pharmacology , Vanillic Acid/analogs & derivatives , Vitamin E/chemistry , Vitamin E/pharmacology , Vitamin E/analogs & derivatives , Mice , Signal Transduction/drug effects , Male , Lung/drug effects
4.
Sci Rep ; 14(1): 5757, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38459144

ABSTRACT

Despite remarkable scientific progress over the past six decades within the medical arts and in radiobiology in general, limited radiation medical countermeasures (MCMs) have been approved by the United States Food and Drug Administration for the acute radiation syndrome (ARS). Additional effort is needed to develop large animal models for improving the prediction of clinical safety and effectiveness of MCMs for acute and delayed effects of radiation in humans. Nonhuman primates (NHPs) are considered the animal models that reproduce the most appropriate representation of human disease and are considered the gold standard for drug development and regulatory approval. The clinical and histopathological effects of supralethal, total- or partial-body irradiations (12 Gy) of NHPs were assessed, along with possible protective actions of a promising radiation MCM, gamma-tocotrienol (GT3). Results show that these supralethal radiation exposures induce severe injuries that manifest both clinically as well as pathologically, as evidenced by the noted functionally crippling lesions within various major organ systems of experimental NHPs. The MCM, GT3, has limited radioprotective efficacy against such supralethal radiation doses.


Subject(s)
Acute Radiation Syndrome , Chromans , Medical Countermeasures , Radiation-Protective Agents , Vitamin E/analogs & derivatives , Animals , United States , Humans , Vitamin E/pharmacology , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/pathology , Disease Models, Animal , Radiation-Protective Agents/pharmacology , Macaca mulatta
5.
Eur J Med Chem ; 269: 116346, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38518524

ABSTRACT

Considering the increasing risk of nuclear attacks worldwide, the development of develop potent and safe radioprotective agents for nuclear emergencies is urgently needed. γ-tocotrienol (GT3) and δ-tocotrienol (DT3) have demonstrated a potent radioprotective effect by inducing the production of granulocyte-colony stimulating factor (G-CSF) in vivo. However, their application is limited because of their low bioavailability. The utilization of ester prodrugs can be an effective strategy for modifying the pharmacokinetic properties of drug molecules. In this study, we initially confirmed that DT3 exhibited the most significant potential for inducing G-CSF effects among eight natural vitamin E homologs. Consequently, we designed and synthesized a series of DT3 ester and ether derivatives, leading to improved radioprotective effects. The metabolic study conducted in vitro and in vivo has identified DT3 succinate 5b as a prodrug of DT3 with an approximately seven-fold higher bioavailability compared to DT3 alone. And DT3 ether derivative 8a were relatively stable and approximately 4 times more bioavailable than DT3 prototype. Furthermore, 5b exhibited superior ability to mitigate radiation-induced pancytopenia, enhance the recovery of bone marrow hematopoietic stem and progenitor cells, and promote splenic extramedullary hematopoiesis in sublethal irradiated mice. Similarly, 8a shown potential radiation protection, but its radiation protection is less than DT3. Based on these findings, we identified 5b as a DT3 prodrug, and providing an attractive candidate for further drug development.


Subject(s)
Hematopoietic System , Prodrugs , Radiation Protection , Vitamin E/analogs & derivatives , Animals , Mice , Granulocyte Colony-Stimulating Factor/pharmacology , Esters/pharmacology , Ethers , Prodrugs/pharmacology , Granulocytes
6.
Biochem Biophys Res Commun ; 704: 149661, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38417343

ABSTRACT

To date only four recombinant growth factors, including Filgrastim (rhG-CSF), have been approved by FDA as radiomitigators to ameliorate hematopoietic acute radiation syndrome (H-ARS). These approved agents are not stable under room-temperature, needing to be stored at 2-8 °C, and would not be feasible in a mass casualty scenario where rapid and cost-effective intervention is crucial. Delta-tocotrienol (δ-T3H), the most potent G-CSF-inducing agent among vitamin E isoforms, exhibited efficiency and selectivity on G-CSF production in comparison with TLR and STING agonists in mice. Five-dose δ-T3H was utilized as the optimal therapeutic regimen due to long-term G-CSF production and the best peripheral blood (PB) recovery of irradiated mice. Comparable with rhG-CSF, sequential administration of δ-T3H post-irradiation improved hematologic recovery and accelerated the regeneration of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) in the bone marrow (BM) and spleen of 6.5Gy irradiated mice; and consistently enhanced repopulation of BM-HSCs. In 4.0Gy irradiated nonhuman primates, δ-T3H exhibited comparable efficacy as rhG-CSF to promote PB recovery and colony-formation of BM-HPCs. Altogether, we demonstrated that sequential administration of delta-tocotrienol ameliorates radiation-induced myelosuppression in mice and non-human primates through inducing G-CSF production, indicated δ-T3H as a promising radiomitigator for the management of H-ARS, particularly in a mass casualty scenario.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Vitamin E , Animals , Mice , Bone Marrow/pathology , Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/drug effects , Granulocyte Colony-Stimulating Factor/metabolism , Primates , Recombinant Proteins/pharmacology , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use
7.
Radiat Res ; 201(5): 371-383, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38253059

ABSTRACT

A complex cascade of systemic and tissue-specific responses induced by exposure to ionizing radiation can lead to functional impairment over time in the surviving population. Current methods for management of survivors of unintentional radiation exposure episodes rely on monitoring individuals over time for the development of adverse clinical symptoms due to the lack of predictive biomarkers for tissue injury. In this study, we report on changes in metabolomic and lipidomic profiles in multiple tissues of nonhuman primates (NHPs) that received either 4.0 Gy or 5.8 Gy total-body irradiation (TBI) of 60Co gamma rays, and 4.0 or 5.8 Gy partial-body irradiation (PBI) from LINAC-derived photons and were treated with a promising radiation countermeasure, gamma-tocotrienol (GT3). These include small molecule alterations that correlate with radiation effects in the jejunum, lung, kidney, and spleen of animals that either survived or succumbed to radiation toxicities over a 30-day period. Radiation-induced metabolic changes in tissues were observed in animals exposed to both doses and types of radiation, but were partially alleviated in GT3-treated and irradiated animals, with lung and spleen being most responsive. The majority of the pathways protected by GT3 treatment in these tissues were related to glucose metabolism, inflammation, and aldarate metabolism, suggesting GT3 may exert radioprotective effects in part by sparing these pathways from radiation-induced dysregulation. Taken together, the results of our study demonstrate that the prophylactic administration of GT3 results in metabolic and lipidomic shifts that likely provide an overall advantage against radiation injury. This investigation is among the first to highlight the use of a molecular phenotyping approach in a highly translatable NHP model of partial- and total-body irradiation to determine the underlying physiological mechanisms involved in the radioprotective efficacy of GT3.


Subject(s)
Macaca mulatta , Metabolomics , Whole-Body Irradiation , Animals , Whole-Body Irradiation/adverse effects , Male , Metabolome/radiation effects , Vitamin E/metabolism , Vitamin E/analogs & derivatives , Radiation-Protective Agents/pharmacology , Gamma Rays/adverse effects , Chromans
8.
J Agric Food Chem ; 70(37): 11689-11703, 2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36094395

ABSTRACT

The influence of pumpkin seed roasting conditions (110-140 °C) and screw-pressing on the formation of previously undescribed Δ7-phytosterol oxidation products and tocopherylquinone adducts with nucleophilic phosphatidylethanolamine species was investigated. The roasting process of pumpkin seed paste at a temperature above 120 °C for 30 min considerably enhanced the formation of Δ7-oxysterols. Targeted analysis [electron impact mass spectrometry (MS), 1D-nuclear magnetic resonance] led to the identification of five novel markers of pumpkin paste roasting, among which (3ß,5α,22E,24S)-stigmasta-7,22-dien-6-one-3-ol (6-oxo-α-spinasterol), stereoisomers of (3ß,5α,22E)-7,8-epoxystigmast-22-en-3-ol (7,8-epoxy-α-spinasterol), and (3ß,5α)-22,23-epoxystigmast-7-en-3-ol (7,8-epoxy-α-spinasterol) were reported in edible oils for the first time. Simulated culinary processing provided novel stereoisomers of (3ß,5α,22E)-stigmasta-7,22-dien-3,6-diol, unusual (3ß,5α,22E)-stigmasta-7,22-dien-6,15-dione-3-ol, and (5α,22E)-stigmasta-7,22-dien-3-one accompanied by minor stereoisomers of (3ß,5α)-7,8;22,23-diepoxystigmastan-3-ol. Moreover, a clear relationship between the pumpkin seed oil stability index and synergistic effect of glycerophospholipids with present tocochromanols was found. High-resolution atmospheric pressure chemical ionization-MS experiments clearly demonstrated the formation of various γ-tocopherylquinone adducts with primary amines, namely, octylamine. The mitigation strategy of potentially detrimental oxysterols from pumpkin seed oil included optimization of processing parameters while maintaining the formation of desirable sensory-active compounds.


Subject(s)
Cucurbita , Oxysterols , Phytosterols , Amines , Bone Screws , Phosphatidylethanolamines , Plant Oils/chemistry , Temperature , Vitamin E/analogs & derivatives
9.
Mol Immunol ; 151: 126-133, 2022 11.
Article in English | MEDLINE | ID: mdl-36126500

ABSTRACT

γ-Tocotrienol (GT3), a member of the vitamin E family, is well known for its medicinal value in clinical treatments. However, the role of GT3 in T helper 17 (Th17)/regulatory T cell (Treg) differentiation and function is not fully understood. Here, we demonstrated that GT3 suppressed Th17 differentiation in vitro by inhibiting signal transducer and activator of transcription 3 (STAT3) phosphorylation in the interleukin 6 (IL-6)/Janus kinase (JAK)/STAT3 signaling pathway. GT3 also inhibited HIF1A expression in Th17 metabolism. Additionally, we showed that GT3 treatment inhibited disease aggravation in an imiquimod (IMQ)-induced psoriasis-like mouse model by reducing the percentage of Th17 cells in the spleen in vivo. The findings of this study demonstrated the effects of GT3 on Th17 cells through the STAT3 signaling pathway.


Subject(s)
Janus Kinases , STAT3 Transcription Factor , Animals , Cell Differentiation , Chromans , Imiquimod/pharmacology , Interleukin-17/metabolism , Interleukin-6/metabolism , Mice , STAT3 Transcription Factor/metabolism , Signal Transduction , Th17 Cells , Vitamin E/analogs & derivatives , Vitamin E/metabolism , Vitamin E/pharmacology
10.
Complement Ther Med ; 70: 102866, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35933083

ABSTRACT

OBJECTIVE: We aimed to compare the efficacy of δ-tocotrienol with α-tocopherol in the treatment of patients with non-alcoholic fatty liver disease (NAFLD). DESIGN AND INTERVENTIONS: This study was a double-blinded, active-controlled trial. The patients with NAFLD were randomly assigned to receive either δ-tocotrienol 300 mg or α-tocopherol 268 mg twice daily for 48 weeks. ENDPOINTS: The primary endpoints were change from baseline in fatty liver index (FLI), liver-to-spleen attenuation ratio (L/S ratio), and homeostatic model assessment for insulin resistance (HOMA-IR) at 48 weeks. Key secondary endpoints were change in markers of inflammation, oxidative stress, and hepatocyte apoptosis. Clinical assessment, biochemical analysis, and computed tomography scan of the liver were conducted at baseline, 24 and 48 weeks. RESULTS: A total of 100 patients (δ-tocotrienol = 50, α-tocopherol = 50) were randomized and included in the intention to treat analysis. Compared with baseline, there was a significant improvement (p < .001) in FLI, L/S ratio, HOMA-IR, and serum malondialdehyde in both groups at 48 weeks that was not significant between the two groups. However, there was a significantly greater decrease in body weight, serum interleukin-6, tumor necrosis factor-alpha, leptin, cytokeratin-18, and increase in adiponectin in the δ-tocotrienol group compared to the α-tocopherol group at 48 weeks (p < .05). No adverse events were reported. CONCLUSION: δ-tocotrienol and α-tocopherol exerted equally beneficial effects in terms of improvement in hepatic steatosis, oxidative stress, and insulin resistance in patients with NAFLD. However, δ-tocotrienol was more potent than α-tocopherol in reducing body weight, inflammation, and apoptosis associated with NAFLD. TRIAL REGISTRATION: Sri Lankan Clinical Trials Registry (https://slctr.lk/SLCTR/2019/038).


Subject(s)
Insulin Resistance , Non-alcoholic Fatty Liver Disease , Biomarkers , Body Weight , Double-Blind Method , Humans , Inflammation , Liver , Vitamin E/analogs & derivatives , alpha-Tocopherol
11.
Biochem Biophys Res Commun ; 617(Pt 2): 33-40, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35689840

ABSTRACT

Programmed death-ligand 1 (PD-L1), a critical immune checkpoint ligand, is commonly overexpressed on the surface of many tumor types including lung and prostate cancer. PD-L1 can exert cancer-promoting activity through either suppressing T cell-mediated immune response or activating tumor-intrinsic signaling. Here, we demonstrated that ß-tocotrienol (ß-T3), an isomer of vitamin E, effectively inhibited PD-L1 expression both in vitro and in vivo, which was mechanistically associated inactivating JAK2/STAT3 pathway. Down-regulating PD-L1 expression by ß-T3 led to enhanced immune response and inactivation of PD-L1-induced tumor-intrinsic signaling, which in turn contributed to its anticancer activity. This study uncovered a novel mechanism involved in the anticancer effect of ß-T3.


Subject(s)
B7-H1 Antigen , Neoplasms , B7-H1 Antigen/metabolism , Cell Line, Tumor , Humans , Male , Vitamin E/analogs & derivatives , Vitamin E/pharmacology
12.
Int J Mol Sci ; 23(9)2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35563033

ABSTRACT

The gastrointestinal (GI) system is highly susceptible to irradiation. Currently, there is no Food and Drug Administration (FDA)-approved medical countermeasures for GI radiation injury. The vitamin E analog gamma-tocotrienol (GT3) is a promising radioprotector in mice and nonhuman primates (NHP). We evaluated GT3-mediated GI recovery in total-body irradiated (TBI) NHPs. Sixteen rhesus macaques were divided into two groups; eight received vehicle and eight GT3 24 h prior to 12 Gy TBI. Proximal jejunum was assessed for structural injuries and crypt survival on day 4 and 7. Apoptotic cell death and crypt cell proliferation were assessed with TUNEL and Ki-67 immunostaining. Irradiation induced significant shortening of the villi and reduced mucosal surface area. GT3 induced an increase in crypt depth at day 7, suggesting that more stem cells survived and proliferated after irradiation. GT3 did not influence crypt survival after irradiation. GT3 treatment caused a significant decline in TUNEL-positive cells at both day 4 (p < 0.03) and 7 (p < 0.0003). Importantly, GT3 induced a significant increase in Ki-67-positive cells at day 7 (p < 0.05). These data suggest that GT3 has radioprotective function in intestinal epithelial and crypt cells. GT3 should be further explored as a prophylactic medical countermeasure for radiation-induced GI injury.


Subject(s)
Acute Radiation Syndrome , Chromans , Radiation-Protective Agents , Vitamin E , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/prevention & control , Animals , Chromans/therapeutic use , Disease Models, Animal , Intestines/pathology , Intestines/radiation effects , Ki-67 Antigen , Macaca mulatta , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use
13.
Int J Mol Sci ; 23(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35563663

ABSTRACT

The Warburg effect is commonly recognized as a hallmark of nearly all tumors. In prostate cancer (PCa), it has been shown to be driven by PTEN loss- and Akt hyperactivation-associated upregulation of hexokinase 2 (HK2). δ-Tocotrienol (δ-TT) is an extensively studied antitumor compound; however, its role in affecting PCa glycolysis is still unclear. Herein, we demonstrated that δ-TT inhibits glucose uptake and lactate production in PTEN-deficient LNCaP and PC3 PCa cells, by specifically decreasing HK2 expression. Notably, this was accompanied by the inhibition of the Akt pathway. Moreover, the nutraceutical could synergize with the well-known hypoglycemic agent metformin in inducing PCa cell death, highlighting the crucial role of the above metabolic phenotype in δ-TT-mediated cytotoxicity. Collectively, these results unravel novel inhibitory effects of δ-TT on glycolytic reprogramming in PCa, thus providing new perspectives into the mechanisms of its antitumor activity and into its use in combination therapy.


Subject(s)
Hexokinase , Prostatic Neoplasms , Cell Line, Tumor , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Humans , Male , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vitamin E/analogs & derivatives
14.
Sci Rep ; 12(1): 3387, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35233005

ABSTRACT

Radiological incidents or terrorist attacks would likely expose civilians and military personnel to high doses of ionizing radiation, leading to the development of acute radiation syndrome. We examined the effectiveness of prophylactic administration of a developmental radiation countermeasure, γ-tocotrienol (GT3), in a total-body irradiation (TBI) mouse model. CD2F1 mice received GT3 24 h prior to 11 Gy cobalt-60 gamma-irradiation. This dose of radiation induces severe hematopoietic acute radiation syndrome and moderate gastrointestinal injury. GT3 provided 100% protection, while the vehicle control group had 100% mortality. Two-dimensional differential in-gel electrophoresis was followed by mass spectrometry and Ingenuity Pathway Analysis (IPA). Analysis revealed a change in expression of 18 proteins in response to TBI, and these changes were reversed with prophylactic treatment of GT3. IPA revealed a network of associated proteins involved in cellular movement, immune cell trafficking, and inflammatory response. Of particular interest, significant expression changes in beta-2-glycoprotein 1, alpha-1-acid glycoprotein 1, alpha-2-macroglobulin, complement C3, mannose-binding protein C, and major urinary protein 6 were noted after TBI and reversed with GT3 treatment. This study reports the untargeted approach, the network, and specific serum proteins which could be translated as biomarkers of both radiation injury and protection by countermeasures.


Subject(s)
Acute Radiation Syndrome , Radiation-Protective Agents , Acute Radiation Syndrome/drug therapy , Animals , Chromans , Gamma Rays/adverse effects , Glycoproteins/therapeutic use , Mice , Proteomics , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Vitamin E/analogs & derivatives , Whole-Body Irradiation
15.
Curr Mol Pharmacol ; 15(7): 980-986, 2022.
Article in English | MEDLINE | ID: mdl-35100963

ABSTRACT

BACKGROUND: Breast Cancer is one of the most commonly diagnosed cancers worldwide and a major cause of death among women. Although chemotherapeutic agents remain the keystones in cancer therapy, significant side effects have failed to provide a safe and tolerable treatment for cancer patients. Dietary antioxidant vitamins were extensively investigated over the past years and their relevance in cancer chemotherapy remains to be elucidated. OBJECTIVE: In the current study, we aimed to investigate the anti-proliferative and apoptotic effects of combining γ-tocotrienol, a member of the vitamin E family, with the chemotherapeutic drug etoposide in MCF-7 and MDA-MB-231 breast cancer cell lines. METHODS: The antiproliferative effect of etoposide combined with γ-tocotrienol was measured using MTS viability reagent. The pro-apoptotic effect was elucidated through Cell Death ELISA and dual Annexin V/PI staining followed by flow cytometric analysis. RESULTS: Our results showed that etoposide significantly decreased the cell growth of both cell lines, with MDA-MB-231 cells being more sensitive to etoposide treatment than MCF-7. Moreover, simultaneous treatment of both breast cancer cell lines with low doses of γ-tocotrienol and etoposide induced a synergistic antiproliferative effect (CI<1). Furthermore, the combination therapy significantly increased the percentage of total apoptotic cells in the MDA-MB-231 cell line and the degree of DNA fragmentation as compared to treatment with either compound alone. CONCLUSION: In conclusion, our results provide evidence for the profound anti-tumorigenic effect of combined etoposide and γ-tocotrienol in the breast cancer cell lines.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Chromans , Etoposide/pharmacology , Etoposide/therapeutic use , Female , Humans , MCF-7 Cells , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Vitamin E/therapeutic use
16.
Cancer Prev Res (Phila) ; 15(4): 233-245, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35144931

ABSTRACT

Vitamin E compounds, consisting of α, ß, γ, and δ forms of tocopherols and tocotrienols, display different cancer preventive activities in experimental models. Tocotrienols may have higher potential for clinical use due to their lower effective doses in laboratory studies. However, most studies on tocotrienols have been carried out using cancer cell lines. Strong data from animal studies may encourage the use of tocotrienols for human cancer prevention research. To examine the cancer inhibitory activity of different vitamin E forms, we first investigated their inhibitory activities of different vitamin E forms in prostate cancer cell lines. We found that δ-tocotrienol (δT3) was the most effective form in inhibiting cell growth at equivalent doses. Because of this in vitro potency, δT3 was further studied using prostate-specific Pten-/- (Ptenp-/-) mice. We found that 0.05% δT3 in diet reduced prostate adenocarcinoma multiplicity by 32.7%, featuring increased apoptosis and reduced cell proliferation. The inhibitory effect of 0.05% δT3 in diet was similar to that of 0.2% δ-tocopherol (δT) in diet reported previously. Our further study on the δT3-induced transcriptome changes indicated that δT3 inhibited genes in blood vessel development in the prostate of Ptenp-/- mice, which was confirmed by IHC. Together, our results demonstrate that δT3 effectively inhibits the development of prostate adenocarcinoma in Ptenp-/- mice, which involves inhibition of proliferation and angiogenesis and promotion of apoptosis. PREVENTION RELEVANCE: We demonstrated that δ-tocotrienol is the most active vitamin E form in inhibiting the growth of several prostate cancer cell lines. In transgenic Ptenp-/- mice, δ-tocotrienol inhibited the formation of prostate cancer. This result would encourage and help design clinical studies for the application of δ-tocotrienol for prostate cancer prevention.


Subject(s)
Prostate , Prostatic Neoplasms , Animals , Cell Transformation, Neoplastic , Humans , Male , Mice , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/prevention & control , Vitamin E/analogs & derivatives , Vitamin E/pharmacology
17.
J Pak Med Assoc ; 72(1): 4-7, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35099428

ABSTRACT

OBJECTIVE: To study the effects of delta-tocotrienol on glycaemic control parameters in individuals with pre-diabetes. METHODS: The randomised control trial was conducted at the Armed Forces Institute of Pathology, Rawalpindi, Pakistan, from July 15 to November 15, 2019, and comprised individuals aged 18-60 years having fasting plasma glucose of 5.6 to 6.9 mmol/L or glycosylated haemoglobin of 5.7 to 6.4%. They were randomised into group A receiving 300mg delta-tocotrienol and group B receiving a placebo once daily for 12 weeks. Weight, height, waist circumference, fasting plasma glucose, insulin and glycosylated haemoglobin were measured at the beginning and end of the trial to assess any change. Body mass index and homeostatic model assessment-insulin resistance were also calculated. Data was analysed using SPSS 21. RESULTS: Of the 77participants, 40(52%) were in group A and 37(48%) in group B. Group A showed significantly greater reduction in terms of fasting plasma glucose, glycosylated haemoglobin, insulin and homeostatic model assessment-insulin resistance index (p≤0.001) post-intervention. CONCLUSIONS: Delta-tocotrienol supplementation was found to have a significant effect in improving glycaemic control parameters in persons with pre-diabetes. Futures larger scale clinical trials are needed to confirm these findings. CLINICAL TRIAL NUMBER: SLCTR/2019/024.


Subject(s)
Prediabetic State , Blood Glucose , Dietary Supplements , Glycemic Control , Humans , Prediabetic State/drug therapy , Vitamin E/analogs & derivatives
18.
Bioorg Med Chem ; 53: 116532, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34863066

ABSTRACT

2-Prenylated benzopyrans represent a class of natural and synthetic compounds showing a wide range of significant activities. Polycerasoidol is a natural prenylated benzopyran isolated from the stem bark of Polyalthia cerasoides (Annonaceae) that exhibits dual PPARα/γ agonism and an anti-inflammatory effect by inhibiting mononuclear leukocyte adhesion to the dysfunctional endothelium. Herein, we report the synthesis of three new series of prenylated benzopyrans containing one (series 1), two (series 2, "polycerasoidol" analogs) and three (series 3, "trans-δ-tocotrienolic acid" analogs) isoprenoid units in the hydrocarbon side chain at the 2-position of the chroman-6-ol (6-hydroxy-dihydrobenzopyran) scaffold. Isoprenoid moieties were introduced through a Grignard reaction sequence, followed by Johnson-Claisen rearrangement and subsequent Wittig olefination. hPPAR transactivation activity and the structure activity relationships (SAR) of eleven novel synthesized 2-prenylated benzopyrans were explored. PPAR transactivation activity demonstrated that the seven-carbon side chain analogs (series 1) displayed selectivity for hPPARα, while the nine-carbon side chain analogs (polycerasoidol analogs, series 2) did so for hPPARγ. The side chain elongation to 11 or 13 carbons (series 3) resulted in weak dual PPARα/γ activation. Therefore, 2-prenylated benzopyrans of seven- and nine-carbon side chain (polycerasoidol analogs) are good lead compounds for developing useful candidates to prevent cardiovascular diseases associated with metabolic disorders.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzopyrans/pharmacology , PPAR alpha/agonists , PPAR gamma/agonists , Vitamin E/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Vitamin E/chemical synthesis , Vitamin E/chemistry , Vitamin E/pharmacology
19.
Nat Prod Res ; 36(18): 4661-4671, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34852702

ABSTRACT

Chemical investigation of Allophylus africanus P. Beauv fruits led to the isolation of a new δ-tocotrienol, 3α-hydroxy-δ-tocotrienol (1) together with eight known compounds (2-9). Compound (1) was allylated (1a) and prenylated (1 b and 1c) to give three new semi-synthesized derivatives which were fully characterized as: 6-O-allyl-3α-hydroxy-δ-tocotrienol (1a), 6-O-prenyl-3α-hydroxy-δ-tocotrienol (1 b) and 6-O,5-C-diprenyl-3α-hydroxy-δ-tocotrienol (1c). The structures of compounds were established using comprehensive spectroscopic analysis including UV, MS, 1 D NMR, 2 D NMR and by comparison with the corresponding literature data. Compound (1) and its semi-synthetic derivatives (1a-c) were tested for their antioxydant activity using DPPH radical scavenging assay and also for their cytotoxicity using human cervix carcinoma KB-3-1 cell lines. The results showed that compound (1) exhibited antioxidant activity with an IC50 value of 0.25 µM compared to the reference control trolox (26 µM); and good cytotoxic activity with IC50 values of 97 µM compared to the reference (+)-griseofulvin (IC50 between17-21 µM).


Subject(s)
Antioxidants , Sapindaceae , Antioxidants/chemistry , Antioxidants/pharmacology , Female , Humans , Seeds , Vitamin E/analogs & derivatives
20.
Food Funct ; 13(1): 398-410, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34908071

ABSTRACT

γ-Tocotrienol (γ-T3), an isoprenoid phytochemical, has shown the promotion of osteoblast proliferation and differentiation in our previous study. In this study, its underlying mechanism was investigated through regulating the Wnt/ß-catenin signaling pathway in MC3T3-E1 cells. Comparative experiment results showed that γ-T3, not α-tocopherol (α-TOC) increased more significantly the viability and differentiation in MC3T3-E1 cells. After that, the cells were incubated with 10 mM LiCl, or 4 µM γ-T3 with or without 1 µM XAV-939. γ-T3 at 4 µM stimulated the Wnt/ß-catenin signaling pathway by increasing the expression and nuclear accumulation of ß-catenin, and the expressions of their downstream factors, such as cyclin-D1, c-Myc, BMP2 and BMP-4 in MC3T3-E1 cells. γ-T3 not only upregulated the viability, induced G0/G1 to the S phase, and promoted the expressions of PCNA (Proliferating Cell Nuclear Antigen) and Ki-67, but also increased ALP activity and the expressions of ON, OPN and OCN. Moreover, the effects of γ-T3 on the MC3T3-E1 cells resembled the actions of LiCl, an activator of the Wnt/ß-catenin signaling pathway. Notably, all these effects of γ-T3 on the MC3T3-E1 cells were completely blocked by the Wnt/ß-catenin signaling pathway inhibitor XAV-939. Our data demonstrated that γ-T3 can target ß-catenin to enhance the Wnt/ß-catenin signaling pathway, which led to increased expressions of the downstream cell proliferation and cell cycle-associated (cyclin D1 and c-myc), and cell differentiation-associated (BMP-2 and BMP-4) target genes, and ultimately promoted MC3T3-E1 cell proliferation and differentiation. Therefore, γ-T3 may be a potential agent to prevent and reverse osteoporosis due to its safety and powerful abilities of osteogenesis.


Subject(s)
Cell Differentiation/drug effects , Chromans/pharmacology , Vitamin E/analogs & derivatives , Wnt Signaling Pathway/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Mice , Osteoblasts/drug effects , Vitamin E/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...