GET THE APP

Comparative Evaluation of NS-5 Mixture and its Components on Supe
Clinical & Experimental Cardiology

Clinical & Experimental Cardiology
Open Access

ISSN: 2155-9880

+44 1300 500008

Research Article - (2015) Volume 6, Issue 7

Comparative Evaluation of NS-5 Mixture and its Components on Superoxide Production in HUVEC, and Inflammatory Biomarkers in Humans

Asaf A Qureshi1*, Dilshad A Khan2, Wajiha Mahjabeen2, Neerupma Silswal1 and Nilofer Qureshi1,3
1Department of Basic Medical Sciences, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA
2Department of Chemical Pathology and Endocrinology, Armed Forces Institute of Pathology, and National University of Medical Science, Rawalpindi, Pakistan, 64000, Pakistan
3Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
*Corresponding Author: Asaf A Qureshi, Ph.D., Department of Basic Medical Sciences, 2411 Holmes Street, School of Medicine, University of Missouri, Kansas City, MO 64108, USA

Abstract

Background: Inhibitory effects of NS-5 mixture of resveratrol, quercetin, δ-tocotrienol, nicotinic acid on several inflammatory and cardiovascular risk factors have been reported in normal cholesterolemic and hypercholesterolemic humans. The hypothesis was that combination of cholesterol-lowering and inflammatory-reducing properties of NS-5 mixture would be more effective than its individual components in reducing the serum levels of several biomarkers of cardiovascular disease in humans. However, effects of NS-5 mixture and its components did not report effects on cytokines, gene expression, and microRNAs were not reported in previous publication. As this area is gaining importance in the understanding of various transcriptional factors and signal pathways, which regulate several biomarkers in various diseases.

Aims: Modulation of NS-5 mixture, and its components were evaluated on superoxide production in HUVEC in vitro, and on serum levels of total cholesterol, NO, CRP, TAS, plasma cytokines, gene expression, miRNAs in vivo in normal cholesterolemic and hypercholesterolemic humans.

Study design: Study was carried out as double-blind randomized, trial of NS-5 mixture, resveratrol, quercetin, and δ-tocotrienol in free-living healthy and hypercholesterolemic humans.

Results: The NS-5 mixture, resveratrol, quercetin, δ-tocotrienol, or nicotinic acid treatments caused reduction in superoxide production (11% to 24%; P<0.01) in HUVEC. These reductions were more pronounced with LPSstimulated HUVEC (26% to 40%; P<0.01) compared to predose values. These findings were further supported by decreases (P<0.01) in serum total cholesterol levels of NS-5 treated group (24%) versus resveratrol (18%), quercetin (20%), and δ-tocotrienol (22%) in hypercholesterolemic humans, followed by reduction of NO, CRP and increases in TAS in normal cholesterolemic and hypercholesterolemic humans. There was significant (P<0.001) down-regulation in pro-inflammatory cytokines and gene expression of resistin, IL-2α, IL-6, IL-12, IL-18, TNF-α, and others, that are normally involved in pathogenesis of atherosclerosis, diabetes, and aging processes. The plasma inflammatory miRNAs (miR-101a, miR-125a, miR-155, miR-223) were down-regulated as compared to predose values. The elevated levels of miRNA-146a during senescence were down-regulated after treatment with these compounds.

Conclusions: This is the first report that describes the effects of NS-5 mixture, its components on proteomics, gene expression and levels of miRNAs in normal cholesterolemic and hypercholesterolemic humans. Results suggest that NS-5 mixture and its components are potent agents in the reduction of superoxide production, cardiovascular risk factors and inflammatory biomarkers, which are modulated by NF-κB. Maximum inhibition in superoxide production and other risk factors was observed with NS-5 mixture as compared with its individual components, thus supporting our hypothesis.

Keywords: NS-5 mixture; Resveratrol; Quercetin, δ-tocotrienol; Inflammatory biomarkers; Serum total Cholesterol; NO; hsCRP; Total antioxidant status; Plasma cytokines; Circulatory miRNAs

Abbreviations

AHA Step-1 diet: American Heart Association Step-1 diet; NO: Nitric Oxide; CRP: C-Reactive Protein; TAS: Total Antioxidant Status; miRNAs: microRNAs; mRNAs: messenger Ribonucleic Acids

Introduction

We have recently reported that serum NO levels increase with age, and are significantly higher in hypercholesterolemic humans as compared with normal cholesterolemic humans [1]. The serum NO levels decrease by dietary supplementation with a mixture of transresveratrol, trans-pterostilbene, quercetin, δ-tocotrienol, and nicotinic acid (NS-5) plus AHA Step-1 diet. Levels of other biomarkers of cardiovascular risk factors, C-reactive protein (CRP), and γ-glutamyltransferase (γ-GT) activity were also reduced via this dietary supplementation in these two groups. The NS-5 mixture treatment had no significant effect on serum total cholesterol, LDL-cholesterol, or triglyceride levels in normal cholesterolemic humans. However, NS-5 mixture plus AHA Step-1 diet, when fed to hypercholesterolemic humans, reduced serum total cholesterol, LDL-cholesterol, triglyceride levels, several inflammatory biomarker levels, and increased the total antioxidant status [2]. These results were based on our hypothesis that combining the cholesterol-lowering and inflammation-reducing properties of naturally-occurring compounds including, resveratrol, pterostilbene, quercetin, δ-tocotrienol, and nicotinic acid mixture (NS-5) would greatly increase their inhibitory effect on the serum production of NO and the consequential beneficial effect in reducing cardiovascular risk factors in humans.

This previous study lacked information regarding the effects of NS-5 mixture on various cytokines/proteins, their gene expression, and circulatory microRNAs [2], an area is gaining importance in the understanding of various transcriptional factors and signaling pathways, known to regulate molecules involved in cardiovascular and other diseases [3,4]. In order to verify our hypothesis, first comparative effects of NS-5 mixture versus each of its component in vitro were carried out, followed by in vivo studies in humans. Human umbilical vein endothelial cells (HUVEC) were selected for the in vitro study to examine the relative inhibitory effects of NS-5 mixture, and its main components (resveratrol, quercetin, δ-tocotrienol, and nicotinic acid; (Figure 1) on superoxide production with and without LPS-stimulation. The LPS HUVEC (resulting in increased levels of nitric oxide) were included to imitate the impact of these compounds as observed in hypercholesterolemic and normal cholesterolemic humans. Pterostilbene was not included, because it is a dimethoxy ester of phenolic (hydroxyl) groups of resveratrol, and has similar positive, and physiological effects as resveratrol [5].

clinical-experimental-cardiology-Chemical-structures

Figure 1: Chemical structures of dietary nutritional supplements used in the study.

In our earlier publications, the important role played by endothelial vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in atherogenesis has been described [2]. Several histochemical studies have indicated that adhesion molecules are expressed in human atherosclerotic plaques, and have also suggested a role for adhesion molecules in atherosclerosis and risk of future myocardial infarction [6,7]. These studies have provided the basis to consider anti-adhesion therapy as a new method of reducing the risk of developing cardiovascular disease. Hence, attempts to lower the production of adhesion molecules have received wide attention. As a result, several in vitro studies have indicated that polyphenols and several other naturally-occurring antioxidants, such as flavonoids, vitamins, and tocotrienols represent potential inhibitors of adhesion molecule expression [8,9]. Expression of these adhesion molecules has been shown to depend much on the transcriptional activation of NF- κB, an antioxidant-sensitive transcription factor [10]. The naturally-occurring compounds that inhibit endothelial adhesion molecule expression have been of great interest in view of the role that adhesion molecules play in atherogenesis.

Over the last several years, our research has focused on roles of several naturally occurring compounds in inflammatory response [11]. We have reported several compounds that have the ability to inhibit inflammatory biomarkers, largely through their capacity to inhibit NF-κB activation [5,11]. As mentioned above, NO level decreased by dietary supplementation of NS-5 mixture of resveratrol, pterostilbene, quercetin, δ-tocotrienol and nicotinic acid (vitamin B3) [1,2]. In addition, the consumption of NS-5 mixture caused significant reductions in the levels of serum CRP, and γ-GT activity (risk factors), as well as increased the serum levels of TAS in normal cholesterolemic and hypercholesterolemic humans [2].

Hypercholesterolemia is a disorder of elevated serum total cholesterol, resulting in progressive loss of arterial flexibility and formation of atheromatous plaque, leading to the narrowing of blood vessels, particularly arteries of the heart [12]. The major risk factors for atherosclerosis include elevated levels of serum total cholesterol, LDL-cholesterol and inflammation of coronary arteries caused in part, by increased levels of C-reactive protein (CRP), which is now considered the best indicator for the risk of coronary heart disease [13-15]. However, physiological and genetic factors also contribute to the progression of heart disease [16].

Circulating miRNAs are novel biomarkers for diverse cardiovascular diseases, including acute myocardial infarction, heart failure, coronary artery disease, diabetes, stroke, and hypertension [17-19]. Recently, a comprehensive review has reported the relationship between various cytokines and miRNAs, describing in detail how miRNAs mediate some of the known functions of these cytokines [19].

The present study evaluated the effects of NS-5 mixture, and its individual components on the production of superoxide in vitro in HUVECs, in the presence or absence of LPS. In vivo effects on serum levels of total cholesterol, NO, CRP, TAS, plasma cytokines, gene expression, and plasma microRNAs associated with aging process, cardiovascular, and other diseases were evaluated in normal cholesterolemic and hypercholesterolemic humans.

Materials and Methods

The study was carried out in the Department of Chemical Pathology & Endocrinology, Armed Forces Institute of Pathology (AFIP), Rawalpindi, Pakistan in collaboration with the Department of Basic Medical Sciences, University of Missouri-Kansas City, MO, USA The study protocol was registered and approved by the Institutional Review Board of AFIP, Rawalpindi, Pakistan. The study was carried out under FDA approved IND number 36906.

Materials

Primary cultures of HUVEC and endothelial cell culture media (EGM) were purchased from American Type Culture Collection (Rockville, MD) and Clonetics, BioWhittaker (Walkersville, MD). Reflection™ autoradiography films were purchased from NEN Life Science Research Products (Boston, MA). Capsules (250 mg/capsule) of trans-Resveratrol were a gift from “Mega Resveratrol”, 60 Newtown Road # 32 (Danbury CT, USA). Quercetin was obtained from Alfa Aesar (Johnson Matthey Co. Lancastor, UK), and nicotinic acid (niacin, vitamin B3) was purchased from VOIGT Global Distribution Inc. P.O.Box. 1130 (Lawrence, Kansas, USA). Delta-Gold 125 mg soft gels from annatto seeds (consisting of 90% δ-tocotrienol + 10% g-tocotrienol) were supplied by American River Nutrition, Inc. (Hadley, MA. USA). Serum total cholesterol levels were estimated by using reagent kits from Sigma Chemical Co. (St. Louis, USA). Pure total RNA was obtained from the EDTA treated fresh whole blood by using “total RNA purification kit # 17200 (NORGEN Bioteck Corporation, Thorold, ON, Canada). The various plasma cytokines, cDNA, and miRNA were estimated by using Signosis, Inc. (1700 Wyatt Drive Suite 10-12. Santa Clara, CA, 95054). Human Cytokine Elisa Plate Array I (chemiluminescence), Catalog number EA-4001, and for gene expression analyses and customized Human cDNA Plate Array for messenger ribonucleic acid (mRNA), Catalog Number APUM000416, were used (Signosis, Inc.). The estimation of circulating microRNAs (miRNAs) was carried out using customized miRNA Direct Hybridization Plate Array chemiluminescence; Catalog Number Inv-00465 according to the manufacturer’s instructions (Signosis, Inc.).

Measurement of intracellular superoxide production by various compounds in HUVEC

Cell Culture: The stock solutions of each compound were prepared (1 mg/mL) in 95% ethanol and stored at -20°C. The working solutions (10 μM) of NS-5 mixture and its components were prepared by diluting the appropriate volume of each stock solution in culture medium to give the final ethanol concentration of 0.1% (v/v). Monolayers of HUVEC were maintained in culture dishes pre-coated with collagen type IV at 37°C, 5% CO2 and 95% air in EGM supplemented with 2% fetal bovine serum (FBS), 10 ng/mL human epidermal growth factor, 1 μg/mL hydrocortisone, and 12 μg/mL bovine brain extract. Cells between the second and sixth passage were used for experimentation. Upon reaching confluency, cells were pretreated with NS-5 mixture and its components for 60 min and then treated second set of 1-6 with LPS (100 ng/mL) for 30 min followed by incubation with dihydroethidium dye (5 μM) specific for superoxide detection, and images were captured by epifluorescence microscope under constant exposure time and gain. The control cells received only 0.1% ethanol (v/v) 95% in culture medium.

Composition of nutritional supplement NS-5 mixture

Each 250 mg capsule of NS-5 mixture contained resveratrol+quercetin+δ-tocotrienol+nicotinic acid (50 mg of each)+corn starch (50 mg). The resveratrol, quercetin, δ-tocotrienol, nicotinic acid were purchased, as described in the Materials. The 250 mg/capsule of corn starch was used as placebo group. The capsulation of starch (placebo) and packing (30 capsules / bottle) was carried out at “Kabco Inc.” (New Jersey, USA).

The present study was carried out as double blind randomized, placebo control trial of dietary nutritional supplements (NS-5 mixture), and its main components (resveratrol, quercetin, δ-tocotrienol) in freeliving healthy adults and hypercholesterolemic humans of Wah Cantt, Pakistan. Nicotinic acid (vitamin B3) treatment was not included in current investigation, as its’ pharmacological effects as an effective hypolipidemic agent in human studies have been well documented [20]. All participants signed an informed-consent form, which was approved by Institutional Review Board of the Medical College. The study protocol was approved by institutional review committee of Army Medical College (Rawalpindi, Pakistan). The study was carried out under a FDA approved IND number 36906.

Experimental design

Study group # (1): The participants (n=25; 15 males/10 females) were recruited from free-living healthy population (age>50 years and serum cholesterol levels 4.61 ± 0.07 mmol/L) from the “Senior Citizen’s Community Centre” at Wah Cantt, Pakistan. Prospective participants were grouped according to cholesterol level (>median<) and subgrouped by sex, and randomly divided into five groups of placebo, NS-5 mixture, resveratrol, quercetin, and δ-tocotrienol for treatments.

Study group # (2): The hypercholesterolemic participants (age >50 years and serum cholesterol levels 6.57 ± 0.14 mmol/L) also consisted of (n=25 subjects; 15 males/10 females. They were also randomly divided into five groups of the same treatments plus AHA Step-1 diet as for Study group # 1.

Inclusion criteria

Physical examination was carried out for each participant, which included the participant’s height, weight, serum lipid parameters, glucose, alanine aminotransferase (ALT), systolic and diastolic blood pressure at rest, history of significant diseases, medications (any statin drugs, nitrates, calcium antagonist, angiotension-converting enzyme [ACE] inhibitors, and diuretics) and smoking. Height and body mass index (BMI, kg/m2) was used as a measure of relative body weight. Venous blood samples were drawn at screening after overnight fast (12 h; 8 pm-8 am). At screening the participants were counseled to follow their normal dietary intake of study group # 1 or AHA step-1 diet (intake of <30% fat and <300 mg/d cholesterol) of study group # 2. Initial screening was accomplished during baseline phase I, ranging from three to four weeks. Venous blood samples were drawn at the termination of the baseline phase, and at week 6 after the treatments phase. Processed samples were coded and held at -72°C until analyses were carried out following the completion of treatment phases. All relevant investigations were carried out in the department of pathology, Army Medical College, NUST, Rawalpindi, Pakistan, 64000.

Exclusion criteria

Body weight should be >125% of Metropolitan Life relative weight, and participants having elevated levels of serum glutamate-pyruvate or glutamate-oxaloacetate transaminase activity, blood urea nitrogen or fasting glucose, or hypertensive disease were excluded. Liver function tests, thyroid stimulating hormone (TSH), serum urea and fasting plasma glucose were analyzed to exclude a liver, renal, and thyroid disorders and diabetes mellitus.

Each participant in study group # 1 was individually counselled to take his or her normal daily diet and for participants in study group # 2, AHA Step-1 diet was recommended. Participants of study group # 2 were also advised to stop using cholesterol-lowering drugs or antioxidants and were counselled individually to modify food intake to meet the goals of the AHA Step-1 diet. Human subjects were asked to stop the intake of whole milk, butter, cheese, eggs, animal fat and ice cream. In order to ascertain full compliance of dietary recommendation and intake of nutritional supplements, participants were contacted by telephone during each phase.

Study group # 1 (Free-living normal cholesterolemic humans)

The study consisted of two phases (phase I and II). All the human subjects were screened at baseline during the first four weeks (phase I). Three-day diet records were taken prior to start, and every week during each phase of the study to monitor dietary intake of each subject. The human subjects were then randomized into five groups 1-4. Human subjects of group 1 were given one capsule (250 mg) of placebo (starch), group 1 (NS-5 mixture), 2 (resveratrol), 3 (quercetin), and two capsules of (δ-tocotrienol; 125 mg/capsule) daily for four weeks.

Study group # 2 (hypercholesterolemic humans)

This experiment also consisted of two phases (I and II); the first phase, free choice of diet. Phase (baseline) was followed by a second phase, during which all participants were counselled to follow the American Heart Association Step-1 diet (AHA Step-1 diet), and all human subjects were divided into five groups. During phase II, subjects of group 2 were administered 1 capsule of placebo (starch), 5 (NS-5 mixture), 6 (resveratrol), 7 (quercetin), 8 (δ-tocotrienol; two 125 mg/ capsule) plus AHA Step-1 diet for four weeks.

Blood samples collection

From the subjects of both studies, two sets of venous blood (10 mL of each) were drawn: one set in ethylene diamine tetra acetate (EDTA) glazed tube to get plasma for the purification of messenger RNAs, and a second serum samples set after overnight fast at the end of feeding period. Processed samples were coded and held at -72°C until analyses were carried for all the treatments.

Biochemical analysis

Estimation of serum levels of total cholesterol and C-reactive protein (CRP): All laboratory analyses were performed at the department of chemical pathology, Army medical college, according to validated standard procedures of the laboratory. Serum levels of total cholesterol were measured by cholesterol oxidase method (CHOD. POD). Serum hS-CRP was analyzed by two-site sequential chemiluminescent immunometric assay kit (Seimen, LA, California, USA) on Immulite 1000 (Immulite, Diagnostic Product Corporation, USA) according to the manufacturer’s directions. The analytical sensitivity was 0.1 mg/L. Elevated CRP was defined for values greater than 4.0 mg/L.

Estimation of serum levels of nitric oxide (NO): Serum nitrate was carried out by colorimetric assay based on Griess reagent, using a standard kit procedure (Cayman kit, Ann Arbor, MI.) at 540 nm on ELISA reader (Diamate 710, UK). Serum nitrate was measured as nitrite after enzymatic reduction by incubating with nitrate reductase and NADPH. After incubation, the reaction mixture was deproteinized and Griess reagent was added. After 10 min of color development (deep purple) at room temperature, the absorbance was measured with a micro-plate reader at 540 nm. Values obtained by this procedure represented the sum of nitrate and nitrite. CV of the method was 4.1%.

Estimation of serum levels of total antioxidant status (TAS): Serum total antioxidant status (TAS) was estimated by a kinetic colorimetric assay kit (Randox, Crumlin, UK), on the automated clinical chemistry analyzer, Selectra E (Vita Lab, Netherland), following the previously reported method [21]. Serum TAS present in the sample causes a decrease in 2,2’-Azino-di-[3-ethylbenthiazoline sulphonate] (ABTS) formation (radical cation) which develop a relatively stable blue green color when incubated with peroxidase (metmyoglobin) and H2O2. The decrease in color formation of ABTS is proportional to the concentration of TAS, which is measured at 600 nm.

Analyses of total RNA from EDTA treated whole blood after feeding NS-5 mixture and its components of normal cholesterolemic and hypercholesterolemic humans: The pure total RNA was extracted from EDTA treated fresh whole blood of human subjects fed NS-5 or it components (250 mg/d) for four weeks, by using total RNA purification kit # 17200 (NORGEN Bioteck Corporation, Thorold, ON, Canada). The purity of total RNA was carried out by measuring the absorption at several wavelengths using a Thermo Scientific NanoDrop 1000 Spectrophotometer. The purity of total RNA was determined by the ratio of 260/280 (2.02-2.04). The plasma miRNAs were also purified from plasma of participants of both study groups by using Plasma/Serum Circulating miRNA Purification Mini Kit (Slurry Format) Product # 51000 (NORGEN Bioteck Corporation, Thorold, ON, Canada).

Estimation of human plasma cytokines, cDNA, and miRNA: The various plasma cytokines/proteins were estimated by using Human Cytokine Elisa Plate Array I (chemiluminescence), Catalog number EA- 4001 (Signosis, Inc., Santa Clara, CA, 95054). The gene expression was carried out by extracting mRNA from each sample, then converted to cDNA and plated on a cytokine cDNA array plate Customized Human cDNA Plate Array, Catalog Number AP-UM000416 (Signosis, Inc.). Assays for estimating the plasma cytokines/protein and gene expression of messenger RNAs were carried out according to the protocols provided by Signosis, Inc. The incubation of each assay mixture at various temperatures was carried out by using Enviro-Genie Shaker/ incubator (Enviro-Genie Industries, Bohemia, NY). The intensity of chemiluminescence was detected using a Microplate Luminometer (GloMax Promega, Madison, WI) at 500 nm, and luminescence was monitored over 20 min period. Estimation of circulating miRNAs was carried out using “Customized miRNA Direct Hybridization Plate Array”, chemiluminescence; Catalog Number Inv-00465 (Signosis, Inc., 1700 Wyatt Drive Suite 10-12. Santa Clara, CA, 95054).

Statistical analysis

Statistical analysis was performed using SPSS 16 (SPSS Inc, Chicago). Continuous, normally distributed variables were summarized as means ± SD, and percent differences were calculated from baseline values of each inflammatory marker or lipid parameter analysis of one-way variance was used to test whether changes in serum total cholesterol, NO, CRP, and TAS occur during the course of supplementation, and whether there were between- and within-subject differences. As all observations were required, available degrees of freedom were reduced by this statistical approach. Paired Student’s t-test was applied for normally distributed variables for percentage values of cytokines/ proteins, and their gene expression and miRNAs. A two tailed P value <0.05 was considered significant. Data are reported as mean ± SD (Standard Deviation).

Results

The data presented was an average of three to four estimations of each sample. The values of cytokines, gene expression and miRNAs were based on percentages for comparative purposes. The physical characteristics of all the participants of pre-dose values of normal cholesterolemic and hypercholesterolemic humans are reported in Table 1. There were no changes in the body weight, height, body mass index, serum creatinine, serum glucose, serum alanine aminotransferase, systolic and diastolic blood pressure at the end of post-dose treatment of normal cholesterolemic and hypercholesterolemic humans. The results were based on pre-dose versus post-dose of respective groups rather than values compared with placebo group. There was no change in the values of the placebo group before and after treatment in normal cholesterolemic humans. However, there was a 4% decrease after treatment+AHA Step-1 diet of placebo group in hypercholesterolemic humans (data not shown).

# Parameters Normal cholesterolemic (Means ± SD) Hypercholesterolemic
(Means ± SD)
1 Age 61.51 ± 7.46 60.82 ± 6.06
2 Males/Females (n) 3/2 3/2
3 Height(meter) 1.71 ± 0.09 1.72 ± 0.08
4 Weight(kg) 72.97 ± 9.49 77.88 ± 8.95
5 BMI(kg/m2) 24.79 ± 2.05 26.43 ± 3.10
6 Systolic BP (mmHg) 136.43 ± 6.81 136.76 ± 7.67
7 DiastolicBP (mmHg) 84.71 ± 5.41 87 ± 6.96
8 Serum Creatinine(mmol/L) 90.77 ± 10.30 96.73 ± 12.47
9 Serum ALT(U/L) 32.14 ± 9.72 35.09 ± 5.73
10 SerumTotalCholesterol (mmol/L) 4.61 ±0.07 6.57±0.14
12 Serum Triglyceride (mmol/L) 1.47 ± 0.67 1.51 ± 0.48
13 Serum Glucose (mmol/L) 4.10 ± 0.22 4.91 ± 0.21
14 Serum NO (mmol/L) 6.58 ± 0.16 12.95 ± 0.24
15 Serum CRP(mg/L) 2.22 ± 0 .09 4.96 ± 0.17
16 Serum TAS (mmol/L) 1.39 ± 0.06 1.44 ± 0.06

Table 1: Baseline characteristics of normal cholesterolemic and hypercholesterolemic humans.

Dietary supplement of NS-5 mixture and its components inhibit superoxide production with and without LPS-stimulated HUVEC

The superoxide productions were significantly inhibited by treatments with NS-5 mixture components in human umbilical vein endothelial cells (HUVEC). The maximum reduction was observed with NS-5 mixture (24%; P<0.001) as compared to resveratrol (14%), quercetin (11%), δ-tocotrienol (13%) and nicotinic acid (16%) at P<0.01, respectively (Figure 2). In order to confirm these findings, the same treatments were repeated with and without LPS-stimulated HUVEC for the production of superoxide. As expected, the LPS-stimulation resulted in a significant increase in superoxide production (954 vs 1250), and there were maximum decreases of 40% (P<0.01) with NS-5 mixture, resveratrol (26%; P<0.01), quercetin (33%; P<0.01), δ-tocotrienol (38%; P<0.01), and nicotinic acid (27%; P<0.01) treatments compared to vehicle+LPS (Figure 3). The maximum inhibition in the superoxide production again was observed with NS-5 mixture treatment compared to its individual components. It is also interesting to note that inhibition of superoxide productions was much more pronounced in LPSstimulated HUVEC (Figures 2 and 3), which also imitated the results as observed with normal cholesterolemic versus hypercholesterolemic humans [1,2]. The images captured by epifluorescence microscopy of all these treatments confirm these results as shown in Figure 4.

clinical-experimental-cardiology-superoxide

Figure 2: Dietary supplement of NS-5 mixture and its main components inhibit intracellular superoxide production in human umbilical vein endothelial cells (HUVEC): The working solutions (10 μM) of NS-5 mixture and its components were prepared by diluting the appropriate volume of each stock solution in culture medium to give the final ethanol concentration of 0.1% (v/v). Monolayers of HUVEC were maintained in culture dishes pre- coated with collagen type IV at 37ºC, 5% CO2 and 95% air in EGM supplemented with 2% fetal bovine serum (FBS), 10 ng/ml human epidermal growth factor, 1 μg/ ml hydrocortisone, and 12 μg/ml bovine brain extract. Cells between second and six passages were used for experimentation. Cells were pretreated with NS-5 mixture or its components (set of 1-6) for 60 min, followed by incubation with dihydroethidium dye (5 μM) specific for superoxide detection, and images were captured by epifluorescence microscope under constant exposure time and gain. The control cells received only 0.1% ethanol (v/v) 95% in culture medium. Data are mean fluorescence intensity of the cells (n=10-15) ± SD (standard deviation). Percentages of each treatment compared to control group (vehicle) of each treatment is above the column. Values in a column not sharing a common symbol are significantly different of various groups at ¶P< 0.001; §, ‡ P < 0.01..

clinical-experimental-cardiology-stock-solution

Figure 3: Dietary supplement of NS-5 mixture and its main components inhibit intracellular superoxide production in LPS-stimulated HUVEC: The working solutions (10 μM) of NS-5 mixture and its components were prepared by diluting the appropriate volume of each stock solution in culture medium to give the final ethanol concentration of 0.1% (v/v). Monolayers of HUVEC were maintained in culture dishes pre-coated with collagen type IV at 37°C, 5% CO2 and 95% air in EGM supplemented with 2% fetal bovine serum (FBS), 10 ng/ml human epidermal growth factor, 1 μg/ml hydrocortisone, and 12 μg/ml bovine brain extract. Cells of third passage were used for experimentation. Cells were pretreated with NS-5 mixture or its components (set of 1-6 and 7-12) for 60 min and then set 7-12 were treated with LPS (100 ng/ml) for 30 min, followed by incubation of both sets with dihydroethidium dye (5 μM) specific for superoxide detection, and images were captured by epifluorescence microscope under constant exposure time and gain. The control cells received only 0.1% ethanol (v/v) 95% in culture medium. Data are mean fluorescence intensity of the cells (n=12-16) ± SD (standard deviation). Percentages of each treatment compared to control group (vehicle) of each treatment is above the column. Values in a column not sharing a common symbol are significantly different of various groups at set 1-6, ¶, § P < 0.02; and set 7-12, ¶, §, ‡ P < 0.01.

clinical-experimental-cardiology-Dietary-supplement

Figure 4: Dietary supplement of NS-5 mixture and its main components on dihdroethidium staining for superoxide production with and without LPSstimulated HUVEC.

Effects of NS-5 mixture and its components on cardiovascular risk factors in normal cholesterolemic and hypercholesterolemic humans

Although, recent publications [1,2] described the effects of dietary supplementation of NS-5 mixture on several cardiovascular risk factors, including lipid parameters, now the comparative effects of NS-5 mixture versus resveratrol, quercetin, and δ-tocotrienol were compared only on selected four risk factors including serum levels of total cholesterol, nitric oxide, CRP, and TAS in normal cholesterolemic and hypercholesteromic humans. Serum total cholesterol levels were not changed with the consumption of NS-5 mixture or any of its components in normal cholesterolemic humans, however, there were significant (P<0.01) reduction in hypercholesterolemic humans with NS-5 (24%), resveratrol (18%), quercetin (20%), and δ-tocotrienol (22%), compared to pre-dose values (Figure 5).

clinical-experimental-cardiology-Serum-total

Figure 5: Serum total cholesterol levels were decreased in hypercholesterolemic humans who were administered NS-5 mixture or its individual component plus AHA Step-1 diet, but not in freeliving normal cholesterolemic humans receiving NS-5 mixture or its components: Columns 1-4 represent subjects of group # 1 of normal cholesterolemic humans were administered one capsule of 250 mg/d of NS-5 mixture, or resveratrol, or quercetin, or δ-tocotrienol to normal cholesterolemic participants for four weeks. The columns 5-8 represent group # 2 of hypercholesterolemic participants were transferred to AHA Step-1 diet plus one capsule of 250 mg/d of NS-5 mixture or resveratrol, or quercetin or δ-tocotrienol for four weeks. Data are means ± SD (standard deviation). Percentages were compared to pre-dose vs post-dose of each treatment is above the column. Values in a column not sharing a common symbol are significantly different of various groups at ¶, § P < 0.01.

There were also significant reductions in the serum levels of NO after consumption of NS-5 mixture (36, 42%; P<0.01) and its components, resveratrol (24, 28%; P<0.01), quercetin (31, 37%; P<0.01), δ-tocotrienol (32, 39%; P<0.01), respectively in normal cholesterolemic and hypercholesterolemic humans, compared to pre-dose values (Figure 6). Levels of serum CRP were also followed a similar pattern as observed with NO levels in these two groups. The NS-5 mixture consumption resulted in significant (P<0.01) reduction in CRP with NS-5 mixture (39, 50%), resveratrol (24, 40%), quercetin (35, 47%), and δ-tocotrienol (36, 38%), respectively compared to pre-dose values (Figure 7). The serum levels of total antioxidant status were increased after consumption of NS-5 mixture (29%; 33%, P<0.01), resveratrol (16, 22% P<0.05, P<0.01), quercetin (19 P<0.05, 27%, P<0.01), and δ-tocotrienol (20% P<0.05, 30%, P<0.01), respectively compared to predose values in both groups (Figure 8). All these results clearly indicated that the consumption of NS-5 mixture caused maximum reduction compared to its individual components predicted in our hypothesis.

clinical-experimental-cardiology-nitric-oxide

Figure 6: Serum nitric oxide (NO) levels were decreased in free-living normal cholesterolemic (NS-5 mixture or its individual component) and hypercholesterolemic subjects who were administered NS-5 mixture or its individual component plus AHA Step-1 diet: Columns 1-4 represent subjects of group # 1 of normal cholesterolemic humans were administered one capsule of 250 mg/d of NS-5 mixture, or resveratrol, or quercetin, or δ- tocotrienol to normal cholesterolemic participants for four weeks. The columns 5-8 represent group # 2 of hypercholesterolemic participants were transferred to AHA Step-1 diet plus one capsule of 250 mg/d of NS-5 mixture or resveratrol, or quercetin or δ-tocotrienol for four weeks. Data are means ± SD (standard deviation). Percentages were compared to pre-dose vs postdose of each treatment is above the column. Values in a column not sharing a common symbol are significantly different of various groups at ¶, §, ‡ P < 0.01.

clinical-experimental-cardiology-hypercholesterolemic

Figure 7: Serum C-reactive protein (CRP) levels were decreased in freeliving normal cholesterolemic (NS-5 mixture or its individual component) and hypercholesterolemic humans who were administered NS-5 mixture or its individual component plus AHA Step-1 diet: Columns 1-4 represent subjects of group # 1 of normal cholesterolemic humans were administered one capsule of 250 mg/d of NS-5 mixture, or resveratrol, or quercetin, or δ-tocotrienol to normal cholesterolemic participants for four weeks. The columns 5-8 represent group # 2 of hypercholesterolemic participants were transferred to AHA Step-1 diet plus one capsule of 250 mg/d of NS-5 mixture or resveratrol, or quercetin or δ-tocotrienol for four weeks. Data are means ± SD (standard deviation). Percentages were compared to pre-dose vs postdose of each treatment is above the column. Values in a column not sharing a common symbol are significantly different of various groups at ¶, § P < 0.01..

clinical-experimental-cardiology-cholesterolemic

Figure 8: Serum total antioxidant status (TAS) levels were increased in free-living normal cholesterolemic (NS-5 mixture or its individual component) and hypercholesterolemic humans who were administered NS-5 mixture or its individual component plus AHA Step-1 diet: Columns 1-4 represent subjects of group # 1 of normal cholesterolemic subjects were administered one capsule of 250 mg/d of NS-5 mixture, or resveratrol, or quercetin, or δ-tocotrienol to normal cholesterolemic participants for four weeks. The columns 5-8 represent group # 2 of hypercholesterolemic participants were transferred to AHA Step-1 diet plus one capsule of 250 mg/d of NS-5 mixture or resveratrol, or quercetin or δ-tocotrienol for four weeks. Data are means ± SD (standard deviation). Percentages were compared to pre-dose vs post-dose of each treatment is above the column. Values in a column not sharing a common symbol are significantly different from various groups at ¶, § P < 0.01; ‡P<0.05.

Modulation of plasma cytokines/proteins, their gene expression, microRNAs by NS-5 mixture, and its components in normal cholesterolemic and hypercholesterolemic humans

As pointed out in the introduction, the present study was carried out to determine the effects of NS-5 mixture and its components on proteomic, genomic and microRNAs in humans, which was lacking in our earlier publications [1,2]. The effects of δ-tocotrienol on the functions of resistin, IL-1α, IL-2, IL-6, IL-8, IL-10, IL-12, TNF-α, IFN-γ, and miRNA-10b, R-16-1, R-18a, R-20a, R-29a, R-125a, R-133, R-223, R-372, R-214 have been reported in our recent publications [3,4]. The values of δ-tocotrienol of these cytokines and miRNAs were included in the present tables for comparison purposes, and their descriptions or functions were not included in this paper. Moreover, the effects of NS-5 mixture and its components on protein expression of cytokines, genes, and miRNAs were divided under different categories (cardiovascular, inflammation, cancer, diabetes and aging), and main functions were also included in for normal cholesterolemic (Tables 2,4,6), and for hypercholesterolemic humans (Tables 3,5,7). The values were based on percentages of pre-dose compared to post-dose values for each marker. The post-dose value was calculated based on their respective pre-dose value for each of estimation (regarded as 100%).

      NS-5 Resveratrol Quercetin d-Tocotrienol  
# Cytokines‡ Pre-dose Post-dose Post-dose Post-dose Post-dose¶ Functions
      % % % %  
I Cardiovascular disease:
1 Resistin§ 100 71 ± 1.4** 84 ± 1.5** 88 ± 1.8** 81 ± 1.6** Regulatory role in insulin resistance, diabetes, atherosclerosis.
2 IL-2 100 67 ± 2.7** 51 ± 1.9** 65 ± 1.6** 75 ± 0.7** For growth, proliferation, & differentiation of T cells to (Effector T) cells.
3 IL-6 100 75 ± 1.2** 77 ± 1.4** 83 ± 2.4** 83 ± 1.2** Regulates the immune response, hematopoiesis and inflammation.
4 IL-8 100 68 ± 2.9** 69 ± 3.3** 77 ± 2.2* 75 ± 0.3** Produced by macrophages & epithelial cells, potent angiogenic factor.
5 IL-12 100 74 ± 2.9** 82 ± 2.8* 84 ± 2.7* 75 ± 2.9** It plays a key role in the activities of natural killer cells and T lymphocytes.
6 1L-17a 100 44 ± 3.3** 60 ± 2.4** 74 ± 3.1** 49 ± 3.4** This gene is a pro-inflammatory cytokine produced by activated T cells.
7 IL-18 100 72 ± 1.4** 85 ± 1.7** 50 ± 3.9** 85 ± 1.8* It acts as an angiogenic factor in many diseases.
8 COX-2 100 68 ± 3.2** 76 ± 1.5** 75 ± 3.6** 74 ± 2.9** It is an enzyme, which is responsible for the production of prostanoids.
9 GAPDH 100 70 ± 1.1** 73 ± 2.3** 77 ± 2.8** 80 ± 2.5** Key role in glycolysis & new host defense mechanism against HIV.
10 IP-10 100 58 ± 1.7** 83 ± 2.9* 82 ± 2.6* 57 ± 2.1** It acts as potent inhibitor of angiogenesis in vivo.
II Inflammation:
11 TNF-a 100 63 ± 1.2** 71 ± 1.3* 68 ± 2.1* 42 ± 2.7* Cytokines produced by macrophages/monocytes during inflammation.
12 INF-g 100 67 ± 2.4** 75 ± 3.3** 73 ± 2.1** 67 ± 2.9** Potent mediators of host defense and homoeostasis.
13 MIP-1a 100 62 ± 1.8** 70 ± 2.3** 73 ± 3.5** 68 ± 1.9** A subfamily of chemokines that exhibit pro-inflammatory activities.
14 NOS-2 100 65 ± 1.8** 75 ± 1.5** 63 ± 3.9** 65 ± 2.4** It is involved significantly in many vascular functions.
15 VCAM-1 100 61 ± 1.7** 68 ± 2.7** 63 ± 4.1** 50 ± 3.8** It is involved in inflammatory-linkage.
16 MCP-1 100 78 ± 1.5** 87 ± 1.7** 80 ± 2.9* 75 ± 1.4** It is a chemokine.
III Cancer            
17 IGF-1 100 78 ± 1.1** 84 ± 2.2** 86 ± 2.7* 75 ± 3.1* It is a potent mitogen.
18 P-53 100 197 ± 4.2** 135 ± 3.2** 156 ± 3.9* 141 ± 4.1* It regulates the cell-cycle and acts as a tumor suppressor.
19 FAS-1 100 231 ± 6.7* 123 ± 3.4* 127 ± 2.1* 135 ± 3.2* It forms death-inducing signaling complex (DISC) upon ligand binding
20 VEGF 100 66 ± 2.3* 71 ± 2.4* 79 ± 3.1* 73 ± 3.6* Role in stabilizing the endothelial cell adhesion & signal transduction.
21 CCND-1 100 62 ± 1.8** 76 ± 2.3* 68 ± 3.5* 65 ± 1.5** Controls progression of cells by activating cyclin-dependent kinase.
IV Diabetes
22 PAI-1 100 22 ± 2.2** 46 ± 2.3** 70 ± 1.6** 63 ± 2.8** Associated with atherosclerosis and have increased levels in diabetics.
V Aging            
23 IL-1a 100 57 ± 2.1** 75 ± 2.6** 76 ± 2.9** 77 ± 1.7** Agonist mediating inflammatory and immuno-modulatory effects.
24 IL-10 100 72 ± 2.6** 73 ± 4.1** 66 ± 1.5** 62 ± 1.6** Cytokine with pleiotropic effects in immuno-regulation & inflammation.
* - **Values in a row sharing a common asterisk are significantly different at *P<0.01; **P<0.001.
¶The effect of δ-tocotrienol on resistin, IL-2, IL-6, IL-8, IL-10, IL-12, TNF-α and IFN-γ has been reported recently (AAQ et al. BJMMR. 6(4): 351-366, JCEC. 2015; 6:4. 1000367.
‡The cytokines IL-12, IL-17α, IL-18 (inflammation); IL-2 (cancer); and resistin, IL-6, IL-17α (diabetes) also involved in these diseases.
§Resistin = Obesity-mediated insulin resistance and Type 2 diabetes Mellitus; IL-2: Interleukin-2; IL-6: Interleukin-6, 8, 12, 17α, 18; COX-2: Cyclooxygenase-2; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; TNF-α: Tumor Necrosis Factor-α; INF-γ: Interferon-γ; MIP-1α: Macrophage Inflammatory Protein-1α; NOS-2: Nitric Oxide Synthase-2; VCAM-1: Vascular Cell Adhesion Molecule-1; MCP-1: Monocyte Chemotactic Protein-1; IGF-1: Insulin-like Growth Factor-1; P-53: P-53 tumor suppressor protein; FAS-1: Fatty Acid Synthetase-1; VGEF: Vascular Endothelial Growth Factor; CCND-1: Cyclin D-1; PAI-1: Plasminogen Activator Inhibitor-1; IL-1α: Interleukin-1α; IL-10: Interleukin-10.

Table 2: Effects of NS- 5, resveratrol, quercetin, and δ-tocotrienol (Pre-dose vs Post-dose) on plasma cytokines/proteins of normal cholesterolemic humans.

      NS-5 Resveratrol Quercetin d-Tocotrienol  
# Cytokines‡ Pre-dose Post-dose Post-dose Post-dose Post-dose¶ Functions
      % % % %  
I Cardiovascular disease:
1 Resistin 100 86 ± 1.6** 91 ± 1.7* 88 ± 2.1* 93 ± 2.7* Regulatory role in insulin resistance, diabetes, atherosclerosis.
2 IL-2 100 89 ± 1.8* 94 ± 2.2* 89 ± 4.2* 92 ± 2.9* For growth, proliferation, & differentiation of T cells.
3 IL-6 100 63 ± 1.8** 69 ± 2.4** 85 ± 1.3** 78 ± 1.3** Cytokine regulates immune response, hematopoiesis & inflammation.
4 IL-8 100 75 ± 2.4** 80 ± 1.4** 79 ± 2.4** 75 ± 3.0** Chemokine produced by macrophages, epithelial cells, &angiogenic factor.
5 IL-12 100 73 ± 2.4** 80 ± 1.2** 92 ± 1.3* 76 ± 0.9** Plays a role in the activities of natural killer cells & T lymphocytes.
6 1L-17a 100 69 ± 2.1** 72 ± 2.1** 62 ± 0.8** 72 ± 1.5** Pro-inflammatory cytokine produced by activated T cells.
7 IL-18 100 70 ± 3.4** 83 ± 1.1** 82 ± 2.3** 76±1.3** It acts as an angiogenic factor in many diseases.
8 COX-2 100 73 ± 2.5** 82 ± 2.8** 81 ± 5.6** 70 ± 1.5** It is an enzyme, which is responsible for the production of prostanoids.
9 GAPDH 100 85 ± 7.8* 65 ± 3.3** 71 ± 4.3** 74 ± 1.7** Plays a role in glycolysis & new host defense mechanism against HIV.
10 IP-10 100 80 ± 1.1** 93 ± 0.9** 84 ± 0.7** 84 ± 2.7** It acts as potent inhibitors of angiogenesis in vivo.
II Inflammation
11 TNF-a 100 75 ± 1.7** 85 ± 2.4** 81 ± 1.8** 86 ± 2.6** Cytokine produced by macrophages/monocytes during inflammation.
12 INF-g 100 75 ± 1.7** 93 ± 1.9* 80 ± 3.1** 82 ± 2.5** Potent mediators of host defense and homoeostasis.
13 MIP-1a 100 82 ± 2.1** 93 ± 0.7* 85 ± 2.2** 90 ± 2.3* Chemokines that exhibit a variety of pro-inflammatory activities.
14 NOS-2 100 72 ± 2.8** 75 ± 1.7** 71 ± 1.2** 75 ± 1.9** It is involved significantly in many vascular functions.
15 VCAM-1 100 51 ± 2.7** 64 ± 4.1** 60 ± 2.4** 60 ± 3.1** It is involved in inflammatory-linkage.
16 MCP-1 100 71 ± 3.8** 82 ± 3.3* 73 ± 2.8** 79 ± 3.3* It is a chemokine
III Cancer            
17 IGF-1 100 79 ± 2.7** 90 ± 1.5* 79 ± 1.2** 79 ± 1.1** It is a potent mitogen.
18 P-53 100 135 ± 2.2** 126 ± 4.1** 120 ± 5.1** 150 ± 2.9** It regulates the cell-cycle & acts as a tumor suppressor.
19 FAS-1 100 145 ± 4.0** 140 ± 4.5** 141 ± 1.9** 164 ± 4.9** It forms death-inducing signaling complex (DISC) upon ligand binding
20 VEGF 100 79 ± 2.4** 79 ±4.0** 82 ± 4.1** 78 ± 2.4** Role in stabilizing the endothelial cell adhesion & signal transduction.
21 CCND-1 100 65 ± 2.7** 69 ± 1.6** 68±3.8** 68±1.3** Controls progression of cells by activating cyclin-dependent kinase.
IV Diabetes
22 PAI-1 100 82±1.3** 86±4.6** 59 ±3.1** 84±1.2* Involved in atherosclerosis & diabetes.
V Aging            
23 IL-1a 100 60 ±1.3** 68±3.5** 82±1.5** 75±3.2** It is an agonist mediating inflammatory and immuno-modulatory effects.
24 IL-10 100 34±1.7** 69±3.0** 52±3.7** 65±1.2** Pleiotropic effects in immuno-regulation and inflammation.
* - **Values in a row sharing a common asterisk are significantly different at *P<0.01; **P<0.001.
¶The effect of δ-tocotrienol on resistin, IL-2, IL-6, IL-8, IL-10, IL-12, TNF-αand IFN-γ has been reported recently (AAQ et al. BJMMR. 6(4): 351-366, JCEC. 2015.
‡The cytokines IL-12, IL-17α, IL-18 (inflammation); IL-2 (cancer); and resistin, IL-6, IL-17α(diabetes) also involved in these diseases.
§Resistin: Obesity-mediated insulin resistance and Type 2 diabetes Mellitus; IL-2: Interleukin-2; IL-6: Interleukin-6, 8, 12, 17α, 18; COX-2: Cyclooxygenase-2;
GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; TNF-α: Tumor Necrosis Factor-α; INF-γ: Interferon-γ; MIP-1α: Macrophage Inflammatory Protein-1α; NOS-2: Nitric Oxide Synthase-2; VCAM-1: Vascular Cell Adhesion Molecule-1; MCP-1: Monocyte Chemotactic Protein-1; IGF-1: Insulin-like Growth Factor-1; P-53: P-53 tumor suppressor protein; FAS-1: Fatty Acid Synthetase-1; VGEF: Vascular Endothelial Growth Factor, CCND-1: Cyclin D-1; PAI-1: Plasminogen Activator Inhibitor-1; IL-1α: Interleukin-1α; IL-10: Interleukin-10.

Table 3: Effects of NS-5, resveratrol, quercetin, and  δ-tocotrienol (Pre-dose vs Post-dose) on plasma cytokines/proteins in hypercholesterolemic humans.

  NS-5 Resveratrol Quercetin d-Tocotrienol
# Genes¶¶ Pre-dose Post-dose Post-dose Post-dose Post-dose‡ Descriptions
      % % % %  
I Cardiovascular disease
1 Resistin 100 48 ± 3.22** 76 ± 1.12** 57 ± 1.35** 73 ± 2.81** Pathogenesis of obesity-mediatedinsulin resistanceand type2diabetes mellitus
2 IL-2 100 53 ± 2.07** 88 ± 2.48* 64 ± 2.59** 72 ± 2.88** Interleukin-2
3 IL-6 100 56 ± 3.06** 71 ± 0.68** 85 ± 0.91** 68 ± 2.87* Interleukin-6
4 IL-8 100 68 ± 1.22** 94 ± 2.90* 95 ± 0.76* 95 ± 0.21* Interleukin-8
5 IL-12 100 85 ± 1.93** 78 ± 1.51** 70 ± 2.12** 81 ± 0.89** Interleukin-12
6 1L-17a 100 66 ± 3.44** 83 ± 1.56** 83 ± 0.46** 85 ± 1.62** Insulin-enhances nitricoxide,&NF-κB.
7 IL-18 100 45 ± 3.08** 59 ± 1.30** 85 ± 1.34** 79 ± 3.00** Interleukin-18
8 9 COX-2 100 41 ± 2.78** 73 ± 1.44** 55 ± 1.36** 65 ± 1.62** Cyclooxygenase-2
9 GAPDH 100 29 ± 2.59** 81 ± 1.36** 73 ± 0.88** 78 ± 2.96** Glyceraldehyde-3-phosphate dehydrogenase
10 IP-10 100 21 ± 2.15** 88 ± 1.97* 74 ± 1.74** 72 ± 0.94** Interferon-Inducible Protein-10
II Inflammation
11 TNF-a 100 48 ± 2.30** 63 ± 1.31** 65 ± 2.44** 83 ± 2.08** Tumor Necrosis Factor-a
12 INF-g 100 59 ± 2.36** 67 ± 1.10** 71 ± 1.31** 75 ± 1.60** Interferon-g
13 MIP-1a 100 57 ± 3.94** 87 ± 1.13* 81 ± 3.20** 74 ± 2.24** MacrophageInflammatoryProtein-1a
14 NOS-2 100 56 ± 0.41** 57 ± 1.94** 67 ± 1.72** 67 ± 2.11** Nitric oxidesynthase-2
15 VCAM-1 100 70 ± 2.01** 90 ± 3.76* 72 ± 2.93** 84 ± 0.70** Vascularcelladhesionmolecule
16 MCP-1 100 69 ± 4.99** 83 ± 2.83** 77 ± 2.17** 76 ± 2.02** MonocyteChemotacticProtein-1
III Cancer
17 IGF-1 100 68 ± 3.56** 74 ± 2.33** 70 ± 2.99** 71 ± 2.93** Insulin-likeGrowthFactor-1
18 P-53 100 217 ± 4.87** 113 ± 1.19* 120 ± 1.96** 239 ± 8.66** P-53 tumor suppressor protein
19 FAS-1 100 157 ± 3.72** 119 ± 2.78** 151 ± 3.07** 129 ± 1.94** Fattyacid synthetase-1
20 VEGF 100 71 ± 2.51** 73 ± 1.78** 83 ± 0.85** 90 ± 0.47* VascularEndothelialGrowthFactor
21 CCND-1 100 40 ± 5.33** 71 ± 6.04** 54 ± 1.91** 67 ± 3.71** CyclinD-1
IV Diabetes
22 PAI-1 100 26 ± 1.21** 81 ± 0.48** 71 ± 4.79** 71 ± 2.37** PlasmenogenActivatorInhibitor-1
V Aging
23 IL-1a 100 70 ± 1.67** 84 ± 1.52* 84 ± 2.09* 80 ± 0.76** Interleukin-1a
24 IL-10 100 76 ± 1.44** 96 ± 1.58* 88 ± 4.12* 69 ± 1.42** Interleukin-10
* - **Values in a row sharing a common asterisk are significantly different at *P<0.01; **P<0.001.
‡The effect of δ-tocotrienol on resistin, IL-2, IL-6, IL-8, IL-10, IL-12, TNF-α and IFN-γ has been reported recently (AAQ et al. BJMMR. 6(4): 351-366, and JCEC. 2015; 6:4. 1000367).
¶The mRNAs IL-12, IL-17α, IL-18 (inflammation); IL-2 (cancer); and resistin, IL-6, IL-17α (diabetes) also involved in these diseases.<

Table 4: Effects of NS-5, resveratrol, quercetin, and δ-tocotrienol (Pre-dose vs Post-dose) on gene expression of messenger RNAs (mRNAs) of normal cholesterolemic humans.

  NS-5 Resveratrol Quercetin d-Tocotrienol
# Genes¶ Pre-dose Post-dose Post-dose Post-dose Post-dose‡ Descriptions
      % % % %  
I Cardiovascular disease
1 Resistin 100 39 ± 3.02** 43 ± 1.21** 52 ± 1.96** 72 ± 1.04** Pathogenesis of obesity-mediatedinsulin
resistanceand type2diabetes mellitus
2 IL-2 100 28 ± 1.99** 73 ± 1.52* 79 ± 2.64** 71 ± 1.71** Interleukin-2
3 IL-6 100 56 ± 2.20** 76 ± 2.02** 61 ± 0.89** 62 ± 1.36** Interleukin-6
4 IL-8 100 65 ± 1.22** 69 ± 1.22** 77 ± 2.62** 67 ± 2.36** Interleukin-8
5 IL-12 100 18 ± 0.62** 78 ± 1.03** 70 ± 2.06** 71 ± 3.40** Interleukin-12
6 1L-17a 100 63 ± 2.17** 82 ± 1.86** 69 ± 0.89** 84 ± 0.54** Insulin-enhances nitric oxide,&NF-κB.
7 IL-18 100 75 ± 1.24** 59 ± 1.30** 85 ± 1.34** 79 ± 3.00** Interleukin-18
8 COX-2 100 69 ± 1.35** 69 ± 0.83** 61 ± 2.68** 70 ± 1.52** Cyclooxygenase-2
9 GAPDH 100 82 ± 1.76** 88 ± 2.03* 88 ± 1.00* 71 ± 2.66** Glyceraldehyde-3-phosphate dehydrogenase
10 IP-10 100 83 ± 1.61** 37 ± 1.85** 30 ± 8.50** 19 ± 2.64** Interferon-Inducible Protein-10
II Inflammation
11 TNF-a 100 52 ± 1.80** 57 ± 2.11** 27 ± 0.87** 87 ± 0.94* Tumor Necrosis Factor-a
12 INF-g 100 61 ± 2.57** 73 ± 2.48** 45 ± 3.0** 64 ± 1.90** Interferon-g
13 MIP-1a 100 62 ± 1.95** 73 ± 1.55** 67 ± 0.46** 64 ± 1.53** MacrophageInflammatoryProtein-1a
14 NOS-2 100 61 ± 2.02** 62 ± 1.62** 76 ± 0.81** 64 ± 4.24** Nitric oxidesynthase-2
15 VCAM-1 100 83 ± 2.36** 83 ± 2.59** 72 ± 0.72** 84 ± 0.91** Vascularcelladhesionmolecule
16 MCP-1 100 35 ± 2.44** 60 ± 3.22** 36 ± 2.83** 39 ± 0.66** MonocyteChemotacticProtein-1
III Cancer
17 IGF-1 100 64 ± 4.03** 74 ± 1.99** 65 ± 1.49** 61 ± 3.69** Insulin-likeGrowthFactor-1
18 P-53 100 165 ± 2.43** 3.27** 126 ± 0.57** 197 ± 2.96** P-53 tumor suppressor protein
19 FAS-1 100 155 ± 3.65** 148 ± 1.44** 129 ± 1.41** 132 ± 0.89** Fattyacid synthetase-1
20 VEGF 100 65 ± 2.09** 73 ± 3.84** 71 ± 0.93** 78 ± 1.74* VascularEndothelialGrowthFactor
21 CCND-1 100 27 ± 4.89** 48 ± 1.37** 71 ± 0.59** 63 ± 3.31** CyclinD-1
IV Diabetes
22 PAI-1 100 33 ± 1.61** 76 ± 1.15** 57 ± 2.93** 72 ± 1.02** PlasmenogenActivatorInhibitor-1
V Aging
23 IL-1a 100 45 ± 3.15** 72 ± 1.84** 53 ± 1.99** 66 ± 2.76** Interleukin-1a
24 IL-10 100 17 ± 1.91** 45 ± 1.79** 57 ± 2.66** 45 ± 0.66** Interleukin-10
 - V alues in a row sharing a common asterisk are significantly different at P<0.01; P<0.001. ‡
The effect of δ-tocotrienol on resistin, IL-2, IL-6, IL-8, IL-10, IL-12, TNF-α and IFNγ has been reported recently (AAQ et al. BJMMR. 6(4): 351-366, and JCEC. 2015. 6:4. 1000367).
The mRNAs IL-12, IL-17α, IL-18 (inflammation); IL-2 (cancer); and resistin, IL-6, IL-17α (diabetes) also involved in these diseases.

Table 5: Effects of NS-5, resveratrol, quercetin, and δ-tocotrienol (Pre-dose vs Post-dose) on gene expression of messengerRNAs (mRNAs) of hypercholesterolemic humans.

  NS-5 Resveratrol Quercetin d-Tocotrienol
‡MicroRNAs Pre-dose Post-dose Post-dose Post-dose Post-dose
    % % % % %
I Cardiovascular disease:
a Down-regulation
1 miRNA-7a 100 43 ± 1.02** 74 ± 1.83** 78 ± 1.76** 61 ± 0.66**
2 miRNA-10b 100 34 ± 2.40** 87 ± 0.64* 91 ±1.13* 42 ± 4.17**
3 miRNA-15a 100 68 ± 2.07** 92 ± 1.89* 67 ± 1.49** 87 ± 0.24*
4 miRNA-16 100 61 ± 1.66** 87 ± 0.44* 74 ± 1.16** 71 ± 1.24**
5 miRNA-20a 100 61 ± 0.97** 83 ± 0.75* 93 ± 0.98* 71 ± 1.07**
6 miRNA-21 100 43 ± 2.14** 79 ± 0.62** 50 ± 1.50** 40 ± 2.06**
7 miRNA-29a 100 83 ± 0.97* 87 ± 2.32* 74 ± 1.09** 84 ± 0.41**
8 miRNA-92a 100 36 ± 2.25** 79 ± 0.88** 41 ± 1.68** 55 ± 1.91**
9 miRNA-126a 100 71 ± 0.79** 91 ± 1.45* 67 ± 6.08** 82 ± 0.52*
10 miRNA-133a 100 35 ± 1.52** 84 ± 1.52* 44 ± 2.44** 72 ± 0.76**
11 miRNA-200 100 64 ± 1.09** 70 ± 2.22** 79 ± 0.77** 84 ± 0.69*
12 miRNA-206 100 48 ± 0.98** 58 ± 1.98** 70 ± 1.41** 78 ± 0.91**
b Up-regulation
13 miRNA-499 100 127 ± 1.20** 122 ± 0.57** 144 ± 4.39** 132 ± 2.00**
II Inflammation
c Up-regulation
14 miRNA-373 100 176 ± 1.96** 164 ± 4.20** 136 ± 1.79** 132 ± 1.30**
15 miRNA-93 100 341 ± 3.38** 331 ± 4.14** 129 ± 2.50** 310 ± 5.12**
16 miRNA-192 100 362 ± 1.96** 229 ± 7.91** 357 ± 5.26** 282 ± 1.98**
17 miRNA-216a 100 367 ± 5.89** 220 ± 5.44** 319 ± 6.81** 177 ± 5.06**
18 miRNA-503 100 259 ± 4.77** 240 ± 4.93** 121 ± 1.29** 130 ± 3.11**
  Down-regulation
d
19 miRNA-101a 100 55 ± 1.20** 63 ± 2.18** 84 ± 1.06* 66 ± 1.37**
20 miRNA-125a 100 64 ± 1.85** 68 ± 1.22** 87 ± 0.79* 86 ± 1.60*
21 miRNA-155 100 23 ± 2.53** 66 ± 4.81** 57 ± 0.63** 26 ± 2.05**
22 miRNA-223 100 62 ± 2.36** 40 ± 5.06** 46 ± 1.44** 82 ± 1.81*
III Aging
e Down-regulation
23 miRNA-146a 100 21 ± 1.98** 44 ± 4.48** 33 ± 3.31** 26 ± 1.93**
* - **Values in a row sharing a common asterisk are significantly different at *P<0.01; **P<0.001.
The Effects of δ-tocotrienol on miRNAs-7a, miR-15a, miR-20a, miR-21, miR-29a, miR-92a, miR-200, miR-206 (in BJMMR. 2015; 6(4): 351-366), and miR-16-1, miR-125, miR-133a, miR-155, miR-223, miR-372, miR-10b, miR-18a, miR-214 (in JCEC. 2015; in press) have been reported. These miRNAs of δ-tocotrienol were included for comparison purpose only.

Table 6: The effects of various compounds on plasma circulating microRNAs (miRNA), the novel biomarkers for cardiovascular disease, cancer, diabetes, and aging process in normal cholesterolemic humans.

  NS-5 Resveratrol Quercetin d-Tocotrienol
  ‡MicroRNA Pre-dose Post-dose Post-dose Post-dose Post-dose
% % % % %
I Cardiovascular disease
a Up-regulation
1 miRNA-7a 100 128 ± 1.41** 124 ± 2.55** 116 ± 1.24* 113 ± 1.74*
2 miRNA-10b 100 203 ± 1.03** 123 ± 2.00** 180 ±1.52** 153 ± 5.25**
3 miRNA-15a 100 147 ± 3.14** 131 ± 1.16** 127 ± 0.66** 120 ± 0.53**
4 miRNA-16 100 231 ± 2.83** 209 ± 1.90** 127 ± 1.04** 131 ± 1.26**
5 miRNA-20a 100 252 ± 2.4**2 209 ± 7.73** 146 ± 6.08** 176 ± 5.15**
6 miRNA-21 100 242 ± 0.94** 192 ± 5.20** 123 ± 1.02** 129 ± 3.33**
7 miRNA-29a 100 217 ± 1.49** 123 ± 1.63** 115 ± 5.33* 206 ± 2.48**
8 miRNA-92a 100 245 ± 1.09** 163 ± 2.64** 119 ± 0.88* 239 ± 4.07**
9 miRNA-126a 100 251 ± 1.78** 174 ± 4.65** 167 ± 2.84** 158 ± 2.10**
10 miRNA-133a 100 237 ± 2.52** 130 ± 5.23** 125 ± 3.69** 195 ± 3.40**
11 miRNA-200 100 168 ± 3.60** 125 ± 1.89** 136 ± 0.91** 147 ± 5.56**
12 miRNA-206 100 148 ± 2.78** 133 ± 1.25** 119 ± 1.44* 122 ± 2.77*
b Down-regulation
13 miRNA-499 100 42 ± 1.88** 58 ± 1.97** 71 ± 1.65** 54 ± 3.96**
II Inflammation
c Up-regulation
14 miRNA-373 100 182 ± 4.61** 123 ± 2.05** 174 ± 2.41** 170 ± 4.39**
15 miRNA-93 100 391 ± 1.79** 150 ± 3.35** 122 ± 2.84** 332 ± 2.78**
16 miRNA-192 100 245 ± 4.76** 181 ± 1.32** 204 ± 3.61** 219 ± 3.45**
17 miRNA-216a 100 289 ± 6.85** 250 ± 3.98** 250 ± 4.95** 147 ± 2.47**
18 miRNA-503 100 345 ± 1.70** 271 ± 1.60** 163 ± 2.10** 127 ± 1.61**
d Down-regulation
19 miRNA-101a 100 71 ± 0.96** 82 ± 1.06* 42 ± 1.57** 84 ± 1.96*
20 miRNA-125a 100 36 ± 2.47** 64 ± 5.20** 53 ± 1.39** 67 ± 0.93**
21 miRNA-155 100 39 ± 2.92** 72 ± 2.53** 47 ± 3.37** 46 ± 2.16**
22 miRNA-223 100 22 ± 1.34** 74 ± 0.84** 67 ± 1.05** 29 ± 2.01**
III Aging
e Down-regulation
23 miRNA-146a 100 33 ± 2.46** 71 ± 3.02** 53 ± 3.33** 36 ± 1.10**
* - **Values in a row sharing a common asterisk are significantly different at *P<0.01; **P<0.001.
The Effects of δ-tocotrienol on miRNAs-7a, miR-15a, miR-20a, miR-21, miR-29a, miR-92a, miR-200, miR-206 (in BJMMR. 2015; 6(4): 351-366), and miR-16-1, miR-125, miR-133a, miR-155, miR-223, miR-372, miR-10b, miR-18a, miR-214 (in JCEC. 2015; 4:6. 1000367). have been reported. These miRNAs of δ-tocotrienol were included for comparison purpose only.

Table 7: The effects of variouscom pounds on plasma circulating microRNAs (miRNA), the novel biomarkers for cardiovascular disease, cancer, diabetes, and aging process in hypercholesterolemic humans.

All cytokines (not described previously- IL-17α, IL-18, COX- 2, GAPDH, IP-10, MIP-1α, NOS-2, VCAM-1, P53, FAS-1, VEGF, CCND-1, PAI-1) were down-regulated maximally with NS-5 mixture treatment group, compared to resveratrol, quercetin and δ-tocotrienol treatment groups, except IL-2 (resveratrol), IL-18, IL-10 (quercetin), TNF-α, VCAM-1, and IL-10 with δ-tocotrienol treatment group in normal cholesterolemic humans (Table 2). The Protein 53 and FAS-1 were up-regulated with all four treatments, which are associated with cancer (Table 2). In hypercholesterolemic humans, NS-5 mixture treatment group followed the same trend, as was observed with normal cholesterolemic humans, except GAPDH showed better downregulation with resveratrol (35%), quercetin (29%), δ-tocotrienol (24%), IL-17α (quercetin), COX-2, P-53, FAS-1 (d-tocotrienol) compared to NS-5 treatment group (Table 3). Similarly, NS-5 mixture showed much more pronounced down-regulation or up-regulation in the gene expression of these cytokines compared to remaining other treatment groups (resveratrol, quercetin, δ-tocotrienol) both in normal cholesterolemic and hypercholesterolemic humans as shown in Tables 4 and 5, respectively.

The dysregulation of miRNAs play a crucial role in the development of cardiovascular disease cancer, diabetes and inflammation. Several studies have provided evidence showing that miRNAs participate in regulating cell cycle progression, proliferation, stem cell gene expression, and stress-induced responses. Cluster 1 - 23 miRNAs were also divided into cardiovascular, inflammation and aging groups in normal cholesterolemic humans (Table 6). miRNA 1-12 cluster was associated with cardiovascular disease, and significantly downregulated with NS-5 mixture treatment (17% - 66%; mostly P<0.001) compared to resveratrol (8 - 42%; P<0.01), quercetin (7 - 59%; P<0.01 and 0.001) and δ-tocotrienol (13-60%; P<0.001), respectively based on pre-dose to post-dose treatments (Table 7). MicroRNAs associated with inflammation (miR-373, miR-93, miR-192, miR-216a and miR- 503 (P<0.001) were up-regulated with the above mentioned treatments, and the best impact was again with NS-5 mixture group compared to resveratrol, quercetin and δ-tocotrienol treatments (Table 6). The maximum significant (P<0.001) down-regulation was observed in miRNA-146a associated with aging (79%), (56%), (67%), and (74%) with NS-5 mixture, resveratrol, quercetin and δ-tocotrienol treatments, respectively compared to their respective pre-dose treatments (Table 6).

On the other hand, a cluster of miRNAs 1-12 of cardiovascular disease (miR-7a, miR-10b, miR-15a, miR-16, miR-20a, miR-21, miR-29a, miR-92a, miR-26a, miR-133a, miR-200, miR-206) were significantly (P<0.001) up-regulated instead of being down-regulated as observed in normal cholesterolemic humans with NS-5 mixture, resveratrol, quercetin, and d-tocotrienol compared to their respective pre-dose percentage values in hypercholesterolemic humans (Table 7). The down-regulation of miRNAs associated with cardiovascular disease, inflammation and aging were similar to those found in normal cholesterolemic humans (Table 7). All these results clearly demonstrate that inhibition of superoxide production in HUVEC, biomarkers, or down-regulation of various cytokines, their gene expression, and miRNAs in normal cholesterolemic and hypercholesterolemic humans was maximal with NS-5 mixture, as compared to its individual components, resveratrol, quercetin and δ-tocotrienol, thus validating our original hypothesis.

Discussion

The dietary supplementation of NS-5 mixture and its components (resveratrol, quercetin, δ-tocotrienol, nicotinic acid) treatments caused a significant reduction in superoxide production (11% to 24%) in HUVEC. These reductions were much more pronounced when these compounds were tested in LPS-stimulated HUVEC (12% to 40%) compared to pre-dose values. The maximal inhibition in superoxide production was with NS-5 treatment group compared to its individual components, which proved our hypothesis that a combined mixture (NS-5) of various compounds will be more effective than its individual components. These findings were further supported by serum total cholesterol levels of NS-5 mixture treated group (24%) versus resveratrol (18%), quercetin (20%), and δ-tocotrienol (22%) treated groups in hypercholesterolemic humans (Figure 4), and a similar trend in the reduction of serum levels of NO, CRP. An increase in TAS was observed in normal cholesterolemic and hypercholesterolemic humans (Figures 5-7).

These results suggest that NS-5 mixture and its components are potent and effective agents in the reduction of cellular superoxide production in HUVEC, and cardiovascular risk factors and inflammatory biomarkers in humans, which are modulated by redox sensitive transcription factor NF-κB [11,22]. NF-κB regulates several molecules involved in atherosclerosis, including ICAM-1 and VCAM- 1 [23], and it was reported that these compounds reduced NF-κB activation, suggesting that these compounds may also act on adhesion molecule expression by blocking activation of NF-κB [11].

Atherosclerosis is associated with an impairment of endothelium dependent relaxations, which represent the bioavailability of nitric oxide (NO) produced from endothelial NO synthase (eNOS). Among various mechanisms implicated in the impaired endothelium-dependent relaxations in atherosclerosis, superoxide generated from dysfunctional eNOS has attracted much attention. Experimental studies in vitro have revealed that NO from eNOS constitutes an antiatherogenic molecule. A deficiency in eNOS was demonstrated to accelerate atherosclerotic lesion formation in eNOS knockout mice [24]. In contrast, eNOS overexpression with hypercholesterolemia may promote atherogenesis via increased superoxide generation from dysfunctional eNOS as reported by Kawashima, and also observed in the present results [25].

Moreover, aging is a devastating physiological phenomenon that leads to deterioration in normal functioning of the body, resulting in multiple detrimental changes, ultimately decreasing the quality of life [26]. The anti-aging properties of resveratrol have conflicting reports. It was reported that a mixture of resveratrol combined with 5% quercetin and 5% rice bran phytate (longevinex; the ingredients were micronized to increase the bioavailability) fed rats produced desirable positive effects, and revealed superior cardiac performance, reduced infarct size, and induction of survival signals evidenced by increased Bcl2/ Bax ratio and enhanced Akt phosphorylation. In contrast, LC3-II and Beclin were increased significantly after longevinex treatment [27]. This confirms the hypothesis that resveratrol in combination with other compounds is more effective rather than being administered alone.

Our earlier studies have revealed that δ-tocotrienol affects several different signaling pathways and other factors involved in inflammation (TNF-α, IL-1α, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, resistin, INF-γ, IGF-1, VEGF) in hypercholesterolemic subjects, which are important inflammatory biomarkers [3,4]. In our earlier studies, in regard to cytokines/proteins, the main focus was, their association with cardiovascular disease [3,4]. The present study demonstrates that NS-5 mixture and its components caused down-regulation or upregulation of these and other several cytokines/proteins, including their gene expression in normal cholesterolemic and hypercholesterolemic humans.

It is well known that IL-17α plays important roles in inflammation and the immune response. It is involved in the pathogenesis of various diseases (allergies, autoimmune diseases, malignancy, and protective roles against infectious diseases) and promotes induction of cytotoxic T-lymphocytes responses against cancer [28]. It induces stromal cells to produce pro- inflammatory and hematopoietic cytokines, and also enhances the surface expression of ICAM-1/intracellular adhesion molecule-1. The increasing level of IL-17α is associated with cardiovascular disease via possible regulation of aortic VCAM- 1 expression and T cell content [28], which was down-regulated significantly with treatment of NS-5 and its components. The treatments of NS-5, and its components have down-regulated interleukin-18 (IL- 18), which is synthesized by various cell types as an inactive precursor molecule that becomes functional following cleavage by caspase-1, and may be a potential therapeutic target for angiogenesis- directed diseases.

The down-regulation by NS-5 mixture and its components on several cytokines/proteins were observed in COX-2, GAPDH, IP-10, MIP-1α, NOS-2, VCAM-1, MCP-1, VEGF, CCND-1, and PAI-1 and up-regulated in protein-53 and FAS-1 (Tables 2-7). Cyclooxygenase-2 is an enzyme that is responsible for the formation of prostanoids, and is a cardio-protective protein that can remove ischemia. GAPDH catalyzes the energy-yielding step in carbohydrate metabolism, and the production of ATP and pyruvate. IP-10 functions as an antimicrobial peptide in innate immunity, and MIP-1α, chemokines are the major factors produced by macrophages after stimulation with bacterial endotoxins. They stimulate human granulocytes, which can lead to acute neutrophilic inflammation, and also induce the synthesis and release of IL-1, IL-6 and TNF-α, from fibroblasts and macrophages.

Nitric oxide synthase-2 (NOS-2) synthesized the metastable free radical nitric oxide, which plays critical role as a mediator of vasodilation in blood vessels. The vasodilator action of nitric oxide plays a key role in renal control of extracellular fluid homeostasis and is essential for the regulation of blood flow and blood pressure [29]. VCAM-1 is expressed in individuals with minimum coronary disease and angina. The primary function of VCAM-1 is the mediation of leukocyte-endothelial cell adhesion and signal transduction. Monocyte Chemotactic Protein-1 (MCP-1) is one of the key chemokines that play a major role in selectively recruiting monocytes, neutrophils, and lymphocytes. It also regulates migration and infiltration of monocytes/ macrophages. Migration of monocytes from the blood stream across the vascular endothelium is required for routine immunological surveillance of tissues, and in response to inflammation.

Serum VEGF level is high in an advanced disease of cancer patients. Angiogenesis is the growth of new blood vessels and is an important natural process occurring in the body. IL-6 and IL-8 have been shown to promote angiogenesis [30]. These results clearly demonstrate that NS-5 mixture, and its components possess anti-angiogenic activity by reducing expression of angiogenic promoters. Therefore, they may act as potential agents for the prevention of cancer progression through angiogenesis [31]. Cyclin-1 (CCND-1) is a family of proteins that control the progression of cells by activating cyclin-dependent kinase (CDK) enzyme. The oscillation of the cyclins, namely fluctuations in cyclins gene expression and destruction by the ubiquitin-mediated proteasome pathway, induce oscillation in CDK activity to drive the cell cycle. Plasminogen Activator Inhibitor-1 (PAI-1) is mainly produced by endothelial cells, and also synthesized by adipose tissue, and an increased level of PAI-1 has been found with a number of atherosclerotic factors, and patients with insulin resistance syndrome and diabetes mellitus.

Protein53 (P53) is a tumor suppressor protein that works by regulating the cell cycle, and its activity stops the formation of tumors. FAS-1 protein forms the death-inducing signaling complex (DISC) upon ligand binding. It may act as a cell adhesion molecule. Vascular Endothelial Growth Factor (VEGF) plays an important role in regulating major angiogenic processes such as prolification, migration, differentiation and apoptosis [32].

The circulating miRNAs are novel biomarkers for diverse cardiovascular diseases, including acute myocardial infarction, heart failure, coronary artery disease, diabetes, stroke, hypertension, and acute pulmonary embolism [17]. The impacts of NS-5 mixture and its components on a cluster of 1-23 miRNAs has been shown in normal cholesterolemic humans and hypercholesterolemic humans (Table 7). The plasma high levels of Let-7a expression may be an indicator of impaired cell cycle function in old people with normal cholesterol levels, and this miRNA was significantly down-regulated with the treatment of NS-5, and its components. Most of the highly expressed miRNA that were lower in the blood of patients with coronary artery disease are known to be expressed in endothelial cells (miR-126 and members of the miR-17-92 cluster, namely miR-17, miR-19b, miR-20a, and miR-106a) and down-regulated in several cell types [33]. Plasma levels of miR-126, miR-29a, miR-92a were regulated with NS-5 mixture, and each of its components in hypercholesterolemic humans. In normal cholesterolemic humans, these miRNAs were down-regulated compared to their pre-dose values. MicroRNAs (miR-125a, miR-155, miR-216a) associated with inflammation that were up-regulated in normal cholesterolemic as well as patients with coronary artery disease were down-regulated with the treatments of NS-5 mixture, and its components.

It is reported that over 75 miRNAs have different type of expression. MicroRNA-20b and miRNA-21 were implicated in cardiac remodeling (anti-angiogenic), but now they are also linked with pancreatic cancer and osteogenic differentiation [34,35]. Moreover, miRNA-21 is also known as a good biomarker for inflammation. Plasma miRNA analyses revealed reduction in the levels of R-15a, R-20b, R-21, and R-223 by the treatments of NS-5 and its components in normal cholesteromic humans only, as these miRNAs were associated with diabetes mellitus type 2 and also with cardiovascular disease [36]. It was reported that high glucose concentration reduced the level of miR-126a in endothelial cells. The reduction of miR-126a was confined to circulating vesicles in the plasma of type 2 diabetes patients [37]. These finding might explain the impaired peripheral angiogenic signaling in patients with diabetes mellitus. MicroR-126a also promotes blood vessel growth, one of the most predictive markers of diabetes.

As pointed out earlier miRNAs are novel post-transcriptional modulator of gene expression with potential roles as regulators of skeletal muscle mass and function possibly by contributing to reduced muscle cell renewal and regeneration in the aging human muscle. A characteristic of aging is loss of skeletal muscle known as sarcopenia [38]. Aging is the predominant risk factor for developing cardiovascular disease. Many miRNAs appear to be dysregulated during cellular senescence, aging and disease [38]. However, only few miRNAs have been linked to age-related changes in cellular and organ functions. More recent evidence has showed that miRNAs can contribute in the regulation of longevity, and miRNA-71 remarkably involved in longevity in a cell-non-autonomous manner. A member of the Let-7 family is down-regulated with advanced aging, which consists of the known function in the suppression of the steroid hormone receptor daf-12 [39].

Recently, a review has reported the relationship between various cytokines and miRNAs, where the regulation of miRNAs by proand anti-inflammatory cytokines and the regulatory cytokines were discussed, describing in detail how miRNAs mediate some of the known functions of these cytokines [19]. The regulatory correlation between plasma levels of several pro- and anti-inflammatory cytokines (TNF-α, IL-1α, IL-6, IL-10, IL-12, IFN-γ) and miRNAs (miR-29a, miR-92a, miR- 125a, miR-155) modulated by treatments of NS-5 and its components was described in our earlier reports [3,4]. The first example of miRNA induction by IL-1 was demonstrated in a human monocytic leukemia cell line THO1, when miR-146a and miR-146b were induced in an NF-κB-dependent manner by repressing IL-1 signaling, IRAK1 and TRAF6, thereby creating a negative feed-back loop by switching off or fine-tuning the pro-inflammatory response [40]. This was corroborated by later studies, which showed that over-expression of miR-146 could reduce the expression of IL-1-induced inflammatory cytokines in both epithelial and primary fibroblasts cells [41]. Induction of miR-155 has also been shown to modulate IL-1 signaling [42]. The enforced expression of miR-155 in rheumatoid arthritis synovial fibroblasts could block the production of IL-1-induced matrix metalloproteinase 3 [43]. From the studies, it appears that the IL-1-induction of miRNAs plays an important role in its negative regulation and highlights how manipulation of miRNAs could present new avenues for the therapy of IL-1α- associated diseases.

Similarly, TNF-α is a pro-inflammatory cytokine and is one of the principle mediators of the inflammatory disorders similar to those associated with IL-1α, such as rheumatoid arthritis, inflammatory bowel disease and psoriasis. Elevated levels of TNF-α are associated with many inflammatory disorders [3]. TNF-α can induce a subset of miRNAs and current evidence suggests that miRNAs play a role in mediating TNF-α messenger RNA stabilization. TNF-α can positively influence its own expression by inducing miR-155 and down-regulation of miR-125b treatments of NS-5, and its components as shown in the present study.

Recent evidence suggests a role for miRNAs in IL-6-mediated cell survival and IL-6 signaling modulation. Up-regulation of miR-21 and let-7 miRNA members was found in malignant myeloma, hepatocellular and cholineangiocyte cells due to over-expression of IL-6, which was down-regulated by the treatments of NS-5 and its components. IL-6 may also modulate its own signaling pathway via induction of the miR-17 - miR-92 cluster [4]. In several studies, it was demonstrated how the manipulation miRNA expression through over-expression or repression is sufficient to elicit a response independent of the original emphasizing an important function in miRNA biology [44,45].

Interleukin-10 is an anti-inflammatory cytokine that is crucial for dampening the inflammatory response after pathogen invasion as described earlier [3]. One particular mechanism of action is to down-regulate pro-inflammatory genes such as those encoding IL-1, TNF-α, and IL-6. The IL-10-mediated inhibition of miR-155 led to an increase in SHIP1 expression, supporting the role of IL-10 as an anti-inflammatory mediator, as well as identifying a novel gene target for IL-10 [3,46]. Interferons (IFN) are cytokines that plays a key role in host defense against viral invasion and may have anti-viral and therapeutic potential against HIV infection [47]. All these highlight the hidden complexity of miRNAs induction and regulation of targets by cytokines, and suggest that similar mechanisms may exist for other cytokine signaling components [19].

Conclusions

The NS-5 mixture, and its components (resveratrol, quercetin, δ-tocotrienol, nicotinic acid) treatments caused significant reduction in superoxide production (12% to 19%) in HUVEC. These reductions were much more pronounced in LPS-stimulated HUVEC (26% to 40%), compared to pre-dose values. The maximum inhibition in superoxide production was with NS-5 treatment group compared to its individual components. These findings were further supported by serum total cholesterol levels of NS-5 treated group (24%) versus resveratrol, quercetin, and d-tocotrienol (18%-22%) treated groups only in hypercholesterolemic humans. A similar trend was observed in the reduction of serum levels of NO, and CRP while an increase in TAS was observed in normal cholesterolemic and hypercholesterolemic humans. There were significant reductions in the levels of proinflammatory cytokines and gene expressions of resistin, IL-2α, IL- 6, IL-12, IL-18, TNF-α, and others, involved in the pathogenesis of atherosclerosis, diabetes, and ageing processes in both groups of subjects. The plasma circulating miRNAs associated with inflammation and cardiovascular disease (miR-92a, miR-126-a, miR-133a, miR- 146-a, miR-155, miR-223, miR-101a, miR-499) of these treatments in normal cholesterolemic subjects were down-regulated compared to their respective pre-dose values. However, a cluster of 1-12 miRNAs (miR-7a, miR-10b, miR-15a, miR-16, miR-20a, miR-21, miR-29a, miR- 126a, miR-133a, miR-200, miR-206) associated with cardiovascular disease were down-regulated in hypercholesterolemic humans, and up-regulated with NS-5 mixture and its components treatments. The miRNA-146a significantly increased during senescence, whereas the study compounds treatment significantly decreased these elevated levels of miRNA-146a. Several miRNAs have been implicated in the epigenetic regulation of key metabolic, inflammatory, cancer, and key anti-angiogenic pathways in type 2 diabetes, and those have been influenced by this study treatments. As far as safety and tolerability of NS-5 or other treatments are concerned, none of the participants reported any adverse events or reactions either during or after the course of the study using these naturally-occurring compounds. The present results indicated that NS-5 mixture is the most effective inhibitor/modulator of superoxide production and other several risk with cardiovascular and factors associated other diseases compared to its individual components (resveratrol, quercetin, and δ-tocotrienol), thus confirming our hypothesis, which is also supported by other investigators [27,48,49].

Acknowledgements

We thank Ms. AnneM. Trias for editing the manuscript, and the president of Mega Resveratrol (60 Newton Toad # 32, Danbury CT, USA) for supplying 250 mg capsules of trans for the present and other studies in our lab. We also thank American River Nutrition, Inc. for donating the DeltaGold softgels (125 mg per capsule, American River Nutrition, Inc. Hadley, MA USA). The study was supported by Advanced Medical Research (AMR), Madison, Wisconsin, USA. The study was carried out under an FDA-approved IND # 36906. This study was supported in part from grant numbers GM102631 and GM102631S1 (N.Q.).

Conflict of Interest

A.A. Qureshi, D.A. Khan, W. Mahjabeen, N. Silswal, and N. Qureshi have no conflict of interest to declare.

References

  1. Qureshi AA , Khan DA, Mahjabeen W, Papasian CJ, Qureshi N (2012) Suppression of Nitric Oxide Production and Cardiovascular Risk Factors in Healthy Seniors and Hypercholesterolemic Subjects by a Combination of Polyphenols and Vitamins. J ClinExpCardiolog S5: 8.
  2. Qureshi AA, Khan DA, Mahjabeen W, Papasian CJ, Qureshi N (2013) Nutritional supplement-5 with a combination of proteasome inhibitors (resveratrol, quercetin, d-tocotrienol) modulate age-related biomarkers and cardiovascular lipid parameters in human subjects. J ClinExpCardiolog 4: 238.
  3. Qureshi AA, Khan DA, Mahjabeen W, Qureshi, N (2015) Dose-dependent modulation of lipid parameters, cytokines and RNA by d-tocotrienol in hypercholesterolemic subjects restricted to AHA Step-1 diet. British Journal of Medicine and Medical Research 6: 351-366.
  4. Qureshi AA, Khan DA, Mahjabeen W, Trias AM, Silswal N, et al. (2015) Impact of d-tocotrienol on inflammatory biomarkers and oxidative stress in hypercholesterolemic subjects. J ClinExpCardiolog 6: 367.
  5. Qureshi AA, Guan XQ, Reis JC, Papasian CJ, Jabre S, et al. (2012) Inhibition of nitric oxide and inflammatory cytokines in LPS- stimulated murine macrophages by resveratrol, a potent proteasome inhibitor. LipidsHealth Dis 11: 76.
  6. Poston RN ,Haskard DO, Coucher JR, Gall NP, Johnson-Tidey RR (1992) Expression of intercellular adhesion molecule-1 in atherosclerotic plaques. Am J Pathol 140: 665-673.
  7. Ridker PM, Heinekens CH, Roitman-Johnson B, Stampfer MJ, Allen J (1998) Plasma concentration of soluble intercellular adhesion molecule-1 and risk of future myocardial infarction in apparently healthy mem. Lancet 351: 88-92.
  8. Cominacini L ,Garbin U, Pasini AF, Davoli A, Campagnola M, et al. (1997) Antioxidants inhibit the expression of intercellular cell adhesion molecule-1 and vascular cell adhesion molecule-1 induced by oxidized LDL on human umbilical vein endothelial cells. Free RadicBiol Med 22: 117-127.
  9. Theriault A , Chao JT, Gapor A (2002) Tocotrienol is the most effective vitamin E for reducing endothelial expression of adhesion molecules and adhesion to monocytes. Atherosclerosis 160: 21-30.
  10. Weber C ,Erl W, Pietsch A, Ströbel M, Ziegler-Heitbrock HW, et al. (1994) Antioxidants inhibit monocyte adhesion by suppressing nuclear factor-kappa B mobilization and induction of vascular cell adhesion molecule-1 in endothelial cells stimulated to generate radicals. ArteriosclerThromb 14: 1665-1673.
  11. Qureshi AA, Tan X, Reis JC, Badr MZ, Papassion CJ, et al. (2011) Suppression of nitric oxide induction and pro-inflammatory cytokines by novel preteasome inhibitors in various experimental models. Lipids Health Dise 1: 177.
  12. Kerte'sz A, Bombicz M, Priksz D, Balla J, Balla G, et al. (2013) Adverse impact of diet-induced hypercholesterolemia on cardiovascular tissue homestasis in a rabbit model: Time-dependent changes in cardiac parameters. Int J MolSci 14: 19086-19108.
  13. Pepys MB , Hirschfield GM (2003) C-reactive protein: a critical update. J Clin Invest 111: 1805-1812.
  14. Ferri C , Croce G, Cofini V, De Berardinis G, Grassi D, et al. (2007) C-reactive protein: interaction with the vascular endothelium and possible role in human atherosclerosis. Curr Pharm Des 13: 1631-1645.
  15. Kaur G ,Rao LV, Agrawal A, Pendurthi UR (2007) Effect of wine phenolics on cytokine-induced C-reactive protein expression. J ThrombHaemost 5: 1309-1317.
  16. Berliner JA ,Navab M, Fogelman AM, Frank JS, Demer LL, et al. (1995) Atherosclerosis: basic mechanisms. Oxidation, inflammation, and genetics. Circulation 91: 2488-2496.
  17. Xu J , Zhao J, Evan G, Xiao C, Cheng Y, et al. (2012) Circulating microRNAs: novel biomarkers for cardiovascular diseases. J Mol Med (Berl) 90: 865-875.
  18. Small EM , Frost RJ, Olson EN (2010) MicroRNAs add a new dimension to cardiovascular disease. Circulation 121: 1022-1032.
  19. McCoy CE (2011) The role of miRNAs in cytokine signaling. Front Biosci (Landmark Ed) 16: 2161-2171.
  20. Gille A ,Bodor ET, Ahmed K, Offermanns S (2008) Nicotinic acid: pharmacological effects and mechanisms of action. Annu Rev PharmacolToxicol 48: 79-106.
  21. Miller NJ , Rice-Evans C, Davies MJ, Gopinathan V, Milner A (1993) A novel method for measuring antioxidant capacity and its application to monitoring the antioxidant status in premature neonates. ClinSci (Lond) 84: 407-412.
  22. Makarov SS1 (2000) NF-kappaB as a therapeutic target in chronic inflammation: recent advances. Mol Med Today 6: 441-448.
  23. Collins T , Read MA, Neish AS, Whitley MZ, Thanos D, et al. (1995) Transcriptional regulation of endothelial cell adhesion molecules: NF-kappa B and cytokine-inducible enhancers. FASEB J 9: 899-909.
  24. Knowles JW , Reddick RL, Jennette JC, Shesely EG, Smithies O, et al. (2000) Enhanced atherosclerosis and kidney dysfunction in eNOS(-/-)Apoe(-/-) mice are ameliorated by enalapril treatment. J Clin Invest 105: 451-458.
  25. Kawashima S , Yokoyama M (2004) Dysfunction of endothelial nitric oxide synthase and atherosclerosis. ArteriosclerThrombVascBiol 24: 998-1005.
  26. Mukherjee S , Ray D, Lekli I, Bak I, Tosaki A, et al. (2010) Effects of Longevinex (modified resveratrol) on cardioprotection and its mechanisms of action. Can J PhysiolPharmacol 88: 1017-1025.
  27. Butcher M ,Galkina E (2011) Current views on the functions of interleukin-17A-producing cells in atherosclerosis. ThrombHaemost 106: 787-795.
  28. Yoon Y , Song J, Hong SH, Kim JQ (2000) Plasma nitric oxide concentrations and nitric oxide synthase gene polymorphisms in coronary artery disease. ClinChem 46: 1626-1630.
  29. Weng-Yew W ,Selvaduray KR, Ming CH, Nesaretnam K (2009) Suppression of tumor growth by palm tocotrienols via the attenuation of angiogenesis. Nutr Cancer 61: 367-373.
  30. Caine GJ , Lip GY, Stonelake PS, Ryan P, Blann AD (2004) Platelet activation, coagulation and angiogenesis in breast and prostate carcinoma. ThrombHaemost 92: 185-190.
  31. Selvaduray KR, Radhakrishnan AK, Kutty MK, Nesaretnam K (2012) Palm tocotrienols decrease levels of pro-angiogenic markers in human umbilical vein endothelial cells (HUVEC) and murine mammary cancer cells. Genes Nutr 7: 53-71.
  32. Fichtlscherer S , De Rosa S, Fox H, Schwietz T, Fischer A, et al. (2010) Circulating microRNAs in patients with coronary artery disease. Circ Res 107: 677-684.
  33. Springett G, Neuger AM, Centeno BA, Hutchinson T, Jump H, et al. (2012) Delta-Tocotrienol in subjects with resectable pancreatic exocrine neoplasia. 2nd International Tocotrienol Symposium, Long Beach, CA.
  34. Muhammad N , Luke DA, Shuid AN, Mohamed N, Soelaiman IN (2013) Tocotrienol supplementation in postmenopausal osteoporosis: evidence from a laboratory study. Clinics (Sao Paulo) 68: 1338-1343.
  35. Mukhopadhyay P , Das S, Ahsan MK, Otani H, Das DK (2012) Modulation of microRNA 20b with resveratrol and longevinex is linked with their potent anti-angiogenic action in the ischaemic myocardium and synergestic effects of resveratrol and γ-tocotrienol. J Cell Mol Med 16: 2504-2517.
  36. Zampetaki A ,Kiechl S, Drozdov I, Willeit P, Mayr U, et al. (2010) Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res 107: 810-817.
  37. North BJ, Sinclear DA (2012) The interaction between aging and cardiovascular disease. Circulation Research 110: 1097-1108.
  38. Dimmeler S,Nicotera P (2013) MicroRNAs in age-related diseases. EMBO Mol Med 5: 180-190.
  39. TaganovKD ,Boldin MP, Chang KJ, Baltimore D (2006) NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. ProcNatlAcadSci U S A 103: 12481-12486.
  40. Perry MM, Moschos SA, Williams AE, Shepherd NJ, Larner-Svensson HM, et al. (2008) Rapid changes in microRNA-146a expression negatively regulates the IL-1beta- induced inflammatory response in human lung alveolar epithelial cells. J Immunol 180: 5689-8698.
  41. Ceppi M , Pereira PM, Dunand-Sauthier I, Barras E, Reith W, et al. (2009) MicroRNA-155 modulates the interleukin-1 signaling pathway in activated human monocyte-derived dendritic cells. ProcNatlAcadSci U S A 106: 2735-2740.
  42. Stanczyk J ,Pedrioli DM, Brentano F, Sanchez-Pernaute O, Kolling C, et al. (2008) Altered expression of MicroRNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis. Arthritis Rheum 58: 1001-1009.
  43. BraconiC , Huang N, Patel T (2010) MicroRNA-dependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology 51: 881-890.
  44. Brock M ,Trenkmann M, Gay RE, Michel BA, Gay S, et al. (2009) Interleukin-6 modulates the expression of the bone morphogenic protein receptor type II through a novel STAT3-microRNA cluster 17/92 pathway. Circ Res 104: 1184-1191.
  45. McCoy CE ,Sheedy FJ, Qualls JE, Doyle SL, Quinn SR, et al. (2010) IL-10 inhibits miR-155 induction by toll-like receptors. J BiolChem 285: 20492-20498.
  46. Sadler AJ , Williams BR (2008) Interferon-inducible antiviral effectors. Nat Rev Immunol 8: 559-568.
  47. Juhasz B , Das DK, Kertesz A, Juhasz A, Gesztelyi R, et al. (2011) Reduction of blood cholesterol and ischemic injury in the hypercholesteromic rabbits with modified resveratrol, longevinex. [corrected] Mol Cell Biochem 348: 199-203.
  48. Juhasz B , Kertész A, Balla J, Balla G, Szabo Z, et al. (2013) Cardioprotective effects of sour cherry seed extract (SCSE) on the hypercholesterolemic rabbit heart. Curr Pharm Des 19: 6896-6905.
Citation: Qureshi AA, Khan DA, Mahjabeen W, Silswal N, Qureshi N (2015) Comparative Evaluation of NS-5 Mixture and its Components on Superoxide Production in HUVEC, and Inflammatory Biomarkers in Humans. J Clin Exp Cardiolog 6:388.

Copyright: © 2015 Qureshi AA, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top